1
|
Zhu E, Li X, Liu C, Pal NR. Boosting Drug-Disease Association Prediction for Drug Repositioning via Dual-Feature Extraction and Cross-Dual-Domain Decoding. J Chem Inf Model 2025. [PMID: 40278791 DOI: 10.1021/acs.jcim.5c00070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
The extraction of biomedical data has significant academic and practical value in contemporary biomedical sciences. In recent years, drug repositioning, a cost-effective strategy for drug development by discovering new indications for approved drugs, has gained increasing attention. However, many existing drug repositioning methods focus on mining information from adjacent nodes in biomedical networks without considering the potential inter-relationships between the feature spaces of drugs and diseases. This can lead to inaccurate encoding, resulting in biased mined drug-disease association information. To address this limitation, we propose a new model called Dual-Feature Drug Repurposing Neural Network (DFDRNN). DFDRNN allows the mining of two features (similarity and association) from the drug-disease biomedical networks to encode drugs and diseases. A self-attention mechanism is utilized to extract neighbor feature information. It incorporates two dual-feature extraction modules: the single-domain dual-feature extraction (SDDFE) module for extracting features within a single domain (drugs or diseases) and the cross-domain dual-feature extraction (CDDFE) module for extracting features across domains. By utilizing these modules, we ensure more appropriate encoding of drugs and diseases. A cross-dual-domain decoder is also designed to predict drug-disease associations in both domains. Our proposed DFDRNN model outperforms six state-of-the-art methods on four benchmark data sets, achieving an average AUROC of 0.946 and an average AUPR of 0.597. Case studies on three diseases show that the proposed DFDRNN model can be applied in real-world scenarios, demonstrating its significant potential in drug repositioning.
Collapse
Affiliation(s)
- Enqiang Zhu
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou 510006, China
| | - Xiang Li
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou 510006, China
| | - Chanjuan Liu
- School of Computer Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Nikhil R Pal
- Electronics and Communication Sciences Unit, Indian Statistical Institute, Calcutta 700108, India
| |
Collapse
|
2
|
de Rabelo MBO, Freitas KM, de Oliveira LC, Frade ACM, Filho JDDS, Heiden G, Fernandes GW, Silva IT, Teixeira MM, Braga FC, Pádua RM. Activity of Sesquiterpene Lactones and Umbelliferone From Campovassouria cruciata on SARS-CoV-2 Replication and on the Release of Pro-Inflammatory Cytokines in Lung Cells. Chem Biodivers 2025:e202402824. [PMID: 40220351 DOI: 10.1002/cbdv.202402824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/16/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025]
Abstract
The investigation of natural products is a valid strategy to identify compounds active against coronaviruses. We herein report the screening of Baccharis and Campovassouria plant species against the murine betacoronavirus murine hepatitis virus-3 (MHV-3), assays that can be carried out in BSL-2 facilities. These genera occur exclusively in the Americas and are a source of secondary metabolites with antiviral and anti-inflammatory activity. Campovassouria cruciata emerged as the most active species from the screening, and its phytochemical investigation afforded the discovery of a new isomer of leptocarpin, 3-epi-leptocapin (1), in addition to the sesquiterpenes lactones leptocarpin (2) and arturin (3), along with umbelliferone (4). The structures were elucidated by extensive mono- and two-dimensional 1H and 13C NMR data analysis. The antiviral activity of the isolated compounds was also assayed in Calu-3 cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). All compounds inhibited SARS-CoV-2 replication, with a reduction in viral load of approximately 1.7-2.0 log when tested at 7.5 µM. To evaluate potential anti-inflammatory activity, compounds were tested in A549 cells stimulated with IFN-γ and TNF-α. Both 3-epi-leptocarpin (1) and umbelliferone (4) inhibited interleukin-6 (IL-6) and IL-8 release significantly. Compound 1 reduced the production of reactive oxygen species (ROS) in A549 cells, as assessed by flow cytometry using DCFH-DA, whereas 4 exhibited ROS values seven-fold higher than the basal level. Our results highlight C. cruciata as a source of promising bioactive compounds, which deserve future investigations to explore their potential for the development of new therapeutic agents against inflammatory and viral diseases.
Collapse
Affiliation(s)
- Maria Beatriz O de Rabelo
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kátia M Freitas
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo C de Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alessandra C M Frade
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - José D de S Filho
- CiPharma-Programa de Pós-graduação em Ciências Farmacêuticas, School of Pharmacy, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Gustavo Heiden
- Herbário ECT-Embrapa Clima Temperado, Pelotas, Rio Grande do Sul, Brazil
| | - Geraldo W Fernandes
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabella Thaís Silva
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernão C Braga
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo M Pádua
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
3
|
Mustafa S, Alsayeqh AF. Role of plant phytochemicals/extracts for the control of Dermanyssus gallinae in poultry and its zoonotic importance. Poult Sci 2025; 104:104899. [PMID: 40014968 PMCID: PMC11910102 DOI: 10.1016/j.psj.2025.104899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 03/01/2025] Open
Abstract
Dermanyssus gallinae (poultry red mite) has negative impact on poultry production, causing major economic losses by damaging the health of chickens and other bird species, globally. D. gallinae also acts as vector and reservoir of different viral and bacterial pathogens such as, E. coli, S. enteriditis, P. multocida, S. gallinarum, Newcastle disease, Avian influenza, Erysipelothrix rhusiopathiae, Eastern, Western and Venezuelan equine encephalomyelitis viruses. Some of them are important from zoonotic point of view like, S. enteritidis, Borrelia burgdorferi and avian influenza A virus. It also causes gamasoidosis in humans. For its control various acaricides have been used, but because of D. gallinae resistance against these chemicals, and environmental issues related to these acaricides have encouraged research into different alternatives. One of them are plant extracts and phytochemicals that play a major role by offering a promising option for permanent pest control because of their bioactive abilities. Important phytochemicals that are most effective against these mites are phenol, flavonoid, terpenoid, tannin, and phenylpropanoids. That give their effects by creating neurotoxicity, digestive and metabolic disruption, cuticle disruption, oxidative stress, cell membrane damage, respiratory inhibition, and hormonal disruption in these mites. This review explains the significance of various plant extracts, their phytochemicals and their methods of application against D. gallinae infestations in poultry. Different types of plant's active metabolites have acaricidal, repellent and growth inhibitory effects such as tannins, phenols, terpenoids, and flavonoids. The mode of action, effectiveness and toxicity of these extracts are also discussed. This review also discusses zoonotic importance of D. gallinae.
Collapse
Affiliation(s)
- Sahar Mustafa
- Department of Clinical Medicine and Surgery, University of Agriculture, Faisalabad, Pakistan
| | - Abdullah F Alsayeqh
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia.
| |
Collapse
|
4
|
Su C, Saha T, Sinha S, Hird CP, Smith SXY, Quiñones-Mateu ME, Das SC. Inhalable spray-dried dry powders combining ivermectin and niclosamide to inhibit SARS-CoV-2 infection in vitro. Int J Pharm 2025; 671:125302. [PMID: 39892677 DOI: 10.1016/j.ijpharm.2025.125302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
SARS-CoV-2, the virus responsible for the COVID-19 pandemic, predominantly affects the respiratory tract, underscoring the need to develop antiviral agents in an inhalable formulation that can be delivered as prophylactic and/or therapeutic drugs directly to the infection site. Since the beginning of the pandemic, our group has been exploring the possibility of developing combinations of antiviral drugs that can be delivered as inhalable therapy, including combinations of remdesivir and ebselen or remdesivir and disulfiram prepared using a spray-drying technique. In this study, we used a similar spray-drying technique to develop inhalable dry powders combining the controversial drugs ivermectin and niclosamide, which have been reported to exhibit synergistic activity against SARS-CoV-2 in vitro. The combined dry powders were within the size range of 1-5 μm, amorphous in nature and displayed characteristic morphology after spray drying. The emitted dose (ED) of the spray-dried powders ranged from 68 to 83 %, whereas the fine particle fraction (FPF) ranged between 50 and 74 %. All the prepared dry powders remained stable under different humidity conditions (<15 % RH and 53 % RH). Interestingly, the optimized combinational dry powder of ivermectin and niclosamide showed an improved cytotoxic profile (CC50 value of 45.99 µM) and enhanced anti-SARS-CoV-2 activity in vitro (EC50 of 2.67 µM) compared to the single dry powders of ivermectin (CC50 = 20.25 µM and EC50 = 8.61 µM) and niclosamide (CC50 = 21.36 µM and EC50 = 5.28 µM). In summary, we developed a stable and inhalable combinational dry powder containing ivermectin and niclosamide, capable of inhibiting SARS-CoV-2 replication in vitro, demonstrating the potential to prepare dry powders that could be developed and delivered as inhalable antiviral drugs to prevent and/or treat SARS-CoV-2 or similar respiratory viruses.
Collapse
Affiliation(s)
- Catherine Su
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Tushar Saha
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Shubhra Sinha
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Cody P Hird
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Sophie X Y Smith
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Miguel E Quiñones-Mateu
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
5
|
Liu Y, Liu Y, Yang H, Zhang L, Che K, Xing L. NTMFF-DTA: Prediction of Drug-Target Affinity Based on Network Topology and Multi-feature Fusion. Interdiscip Sci 2025:10.1007/s12539-025-00692-9. [PMID: 39998589 DOI: 10.1007/s12539-025-00692-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025]
Abstract
Predicting drug-target binding affinity (DTA) is an important step in the complex process of drug discovery or drug repositioning. A large number of computational methods proposed for the task of DTA prediction utilize single features of proteins to measure drug-protein or protein-protein interactions, ignoring multi-feature fusion between protein-related features (e.g., solvent accessibility, protein pockets, secondary structures, and distance maps, etc.). To address the aforementioned constraints, we propose a new network topology and multi-feature fusion based approach for DTA prediction (NTMFF-DTA), which deeply mines protein multiple types of data and propagates drug information across domains. Data in drug-target interactions are often sparse, and multi-feature fusion can enrich data information by integrating multiple features, thus overcoming the data sparsity problem to some extent. The proposed approach offers two main contributions: (1) constructing a relationship-aware GAT that selectively focuses on the connections between nodes and edges in the molecular graph to capture the more central roles of nodes and edges in DTA prediction and (2) constructing an information propagation channel between different feature domains of drug proteins to achieve the sharing of the importance weight of drug atoms and edges, and combining with a multi-head self-attention mechanism to capture residue-enhancing features. The NTMFF-DTA model was comparatively tested against several leading baseline technologies on commonly used datasets. Experimental show that NTMFF-DTA can effectively and accurately predict DTA and outperform existing comparative models.
Collapse
Affiliation(s)
- Yuandong Liu
- Computer Science and Technology, Shandong University of Technology, Mashang, Zibo, 255000, China
| | - Youzhi Liu
- Computer Science and Technology, Shandong University of Technology, Mashang, Zibo, 255000, China
| | - Haoqin Yang
- Department of Mechanical Engineering, Shandong University of Technology, Mashang, Zibo, 255000, China
| | - Longbo Zhang
- Computer Science and Technology, Shandong University of Technology, Mashang, Zibo, 255000, China
| | - Kai Che
- Xi'an Aeronautics Computing Technique Research Institute, AVIC, Xi'an, 710065, China
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Linlin Xing
- Computer Science and Technology, Shandong University of Technology, Mashang, Zibo, 255000, China.
| |
Collapse
|
6
|
Saha T, Masum ZU, Biswas A, Mou MA, Ahmed S, Saha T. Inhaled Dry Powder of Antiviral Agents: A Promising Approach to Treating Respiratory Viral Pathogens. Viruses 2025; 17:252. [PMID: 40007007 PMCID: PMC11860668 DOI: 10.3390/v17020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Inhaled dry powder formulations of antiviral agents represent a novel and potentially transformative approach to managing respiratory viral infections. Traditional antiviral therapies in the form of tablets or capsules often face limitations in terms of therapeutic activity, systemic side effects, and delayed onset of action. Dry powder inhalers (DPIs) provide a targeted delivery system, ensuring the direct administration of antivirals to the infection site, the respiratory tract, which potentially enhance therapeutic efficacy and minimize systemic exposure. This review explores the current state of inhaled dry powder antiviral agents, their advantages over traditional routes, and specific formulations under development. We discuss the benefits of targeted delivery, such as improved drug deposition in the lungs and reduced side effects, alongside considerations related to the formulation preparation. In addition, we summarize the developed (published and marketed) inhaled dry powders of antiviral agents.
Collapse
Affiliation(s)
- Tushar Saha
- Mastaplex Ltd., Centre for Innovation, University of Otago, Dunedin 9016, New Zealand
| | - Zia Uddin Masum
- College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA;
| | - Anik Biswas
- Materials and Nanotechnology, North Dakota State University, Fargo, ND 58105, USA;
| | - Moushumi Afroza Mou
- Department of Biological Science, St. John’s University, Queens, New York, NY 11439, USA;
| | - Sohag Ahmed
- Department of Chemistry, West Virginia University, Morgantown, WV 26506, USA;
| | - Tamal Saha
- International Centre for Diarrheal Disease Research, Bangladesh, Dhaka 1212, Bangladesh;
| |
Collapse
|
7
|
Salo JLM, Marcelo LJN, Sanchez ACA, Marcelino CP, Hazel Anne LC, Miranda KJA, Carandang RRZ. Effectiveness of Tocilizumab in COVID-19 Patients with Pneumonia: A Systematic Review. ACTA MEDICA PHILIPPINA 2025; 59:72-80. [PMID: 39967711 PMCID: PMC11831086 DOI: 10.47895/amp.vi0.8188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Background and Objective COVID-19 contributes significantly to global morbidity and mortality. Age-related comorbidities elevate the risk of severe cases. Studies have recently demonstrated that widely available medications, including tocilizumab (TCZ), can manage severe symptoms. However, its effectiveness is unclear, particularly among the older population. Therefore, this review aimed to evaluate TCZ's efficacy in managing severe pneumonia in individuals aged 50 and older. Methods We systematically search several databases and gray literature including Web of Science, CINAHL, Academic Search Complete, PsycINFO, PsycArticles, SocINDEX, CENTRAL/Cochrane Library, PubMed/MEDLINE for original research articles in English across several study designs published in the year 2020-2022. A narrative synthesis was conducted to summarize the evidence. We employed the NIH quality assessment tool for observational cohort studies to evaluate risk of bias. Additionally, we utilized GRADE to appraise the certainty of evidence. Results Among 539 screened articles, only five studies met the selection criteria. Tocilizumab's impact on severe COVID-19 pneumonia revealed a diverse effect on mortality rate, with 29% in the TCZ group, and 40% in the controls died within 30 days of intubation (OR 0.61; 95% CI, 0.27-1.36). It is also reported that TCZ was not associated with mortality, despite faster decline in pulmonary function and prolonged fever. Hospital mortality in the TCZ group was significantly lower than in the controls, and age over 60 was the only significant risk factor. Moreover, administering TCZ reduced mechanical ventilation needs, with 82% extubated compared to 53% in controls. However, 45% in TCZ group was associated with a higher ventilator-associated pneumonia rate than in the untreated group which was 20% (P < 0.001). Despite this, TCZ-treated patients had shorter hospital stays. Conclusions The effects of tocilizumab on reducing mortality risk and improving the survival rate of COVID-19 patients with pneumonia remained inconclusive. Yet, the majority of results suggested that giving tocilizumab leads to shorter hospital stays, lowers the requirement for mechanical ventilation, and decreases the likelihood of ICU transfer. Tocilizumab is linked to the incidence of secondary infections; hence, this medication should be closely monitored for side effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rogie Royce Z Carandang
- College of Pharmacy, Adamson University, Manila, Philippines
- Department of Public Health Sciences, University of Connecticut School of Medicine, Connecticut, USA
| |
Collapse
|
8
|
Jumde RP, Jézéquel G, Saramago M, Frank N, Adam S, Cunha MV, Bader CD, Gunesch AP, Köhler NM, Johannsen S, Bousis S, Pietschmann T, Matos RG, Müller R, Arraiano CM, Hirsch AKH. Dynamic Combinatorial Chemistry Unveils Nsp10 Inhibitors with Antiviral Potential Against SARS-CoV-2. Chemistry 2025; 31:e202403390. [PMID: 39676060 PMCID: PMC11739841 DOI: 10.1002/chem.202403390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
The development of antiviral drugs against the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) responsible for the recent Covid-19 pandemic is crucial, as treatment options remain limited and vaccination does not prevent (re)infection. Two relatively underexplored targets of this virus are the 3'-5' exoribonuclease (ExoN) and the 2'-O-methyltransferase (2'-O-MTase), both essential for viral viability. The non-structural proteins Nsp14 and Nsp16 exhibit enzymatic activities for ExoN and 2'-O-MTase, respectively, especially when in complex with their co-factor protein Nsp10. The study focuses on the use of target-directed dynamic combinatorial chemistry (tdDCC) to identify binders of Nsp10, aiming to disturb the protein-protein interactions (PPI) involving Nsp10-Nsp14, as well as Nsp10-Nsp16. We synthesised the hits and evaluated them to assess Nsp10 affinity, ExoN and 2'-O-MTase activities inhibition, and antiviral activity in hCoV-229E and SARS-CoV-2-infected whole-cell settings. This study reports a novel class of ExoN and/or 2'-O-MTase inhibitors exhibiting antiviral activity against coronaviruses.
Collapse
Affiliation(s)
- Ravindra P. Jumde
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Current addressGlobal Antibiotic Research & Development Partnership (GARDP)Chemin Camille-Vidart 151202GenevaSwitzerland
| | - Gwenaëlle Jézéquel
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
| | - Margarida Saramago
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Nicolas Frank
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
| | - Sebastian Adam
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
| | - Marta V. Cunha
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Chantal D. Bader
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
| | - Antonia P. Gunesch
- Institute for Experimental VirologyTwincore – Centre for Experimental and Clinical Infection ResearchFeodor-Lynen-Str. 730625HannoverGermany
| | - Natalie M. Köhler
- Institute for Experimental VirologyTwincore – Centre for Experimental and Clinical Infection ResearchFeodor-Lynen-Str. 730625HannoverGermany
| | - Sandra Johannsen
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
| | - Spyridon Bousis
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
| | - Thomas Pietschmann
- Institute for Experimental VirologyTwincore – Centre for Experimental and Clinical Infection ResearchFeodor-Lynen-Str. 730625HannoverGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical School30625HannoverGermany
- Helmholtz International Lab for Anti-infectivesCampus E 8.166123SaarbrückenGermany
| | - Rute G. Matos
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical School30625HannoverGermany
- Helmholtz International Lab for Anti-infectivesCampus E 8.166123SaarbrückenGermany
| | - Cecília M. Arraiano
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical School30625HannoverGermany
- Helmholtz International Lab for Anti-infectivesCampus E 8.166123SaarbrückenGermany
| |
Collapse
|
9
|
Latarissa IR, Khairinisa MA, Iftinan GN, Meiliana A, Sormin IP, Barliana MI, Lestari K. Efficacy and Safety of Antimalarial as Repurposing Drug for COVID-19 Following Retraction of Chloroquine and Hydroxychloroquine. Clin Pharmacol 2025; 17:1-11. [PMID: 39845335 PMCID: PMC11748038 DOI: 10.2147/cpaa.s493750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Various repurposing drugs have been tested for their efficacy on coronavirus disease 2019 (COVID-19), including antimalarial drugs. During the pandemic, Chloroquine (CQ) and Hydroxychloroquine (HCQ) demonstrated good potential against COVID-19, but further studies showed both drugs had side effects that were more dangerous than the efficacy. This made World Health Organization (WHO) ban the usage for COVID-19 patients. In this context, there is a need to explore other antimalarial drugs as potential therapies for COVID-19. This study provides a descriptive synthesis of clinical trials evaluating antimalarial drugs for COVID-19 treatment conducted after the withdrawal of CQ and HCQ. The method was a literature study using the keywords "antimalarial", "COVID-19", "SARS-CoV-2", "clinical trial", and "randomized controlled trial" on the MEDLINE, Scopus, and Cochrane databases. Inclusion criteria were published clinical trials with randomized controlled trials (RCTs) on the efficacy and safety of single antimalarial drugs for COVID-19, published in English and excluding combination therapies. The results showed 3 antimalarial drugs, namely Quinine Sulfate (QS), Atovaquone (AQ), and Artemisinin-Piperaquine (AP), had gone through clinical trial to assess efficacy and safety against COVID-19 patients. Out of the 3 drugs, only AP showed significant results in the primary outcome, which was the time required to reach undetectable levels of SARS-CoV-2. Furthermore, the intervention group took 10.6 days, and the control group took 19.3 days (p=0.001). Based on this review, AP showed significant potential as a therapy in the fight against COVID-19.
Collapse
Affiliation(s)
- Irma Rahayu Latarissa
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Miski Aghnia Khairinisa
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Ghina Nadhifah Iftinan
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Anna Meiliana
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Prodia Clinical Laboratory, Central Jakarta, Indonesia
| | - Ida Paulina Sormin
- Faculty of Pharmacy, University of 17 August 1945 Jakarta, Jakarta, Indonesia
- Prodia Diacro Laboratory, Jakarta, Indonesia
| | - Melisa Intan Barliana
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - Keri Lestari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
10
|
Paróczai D, Bikov A, Blidaru A, Bobu E, Lascu A, Mot CI, Mihaicuta S, Frent S. Comparative efficacy of repurposed drugs lopinavir-ritonavir and darunavir-ritonavir in hospitalised COVID-19 patients: insights from a tertiary centre cohort. Front Cell Infect Microbiol 2025; 14:1496176. [PMID: 39885967 PMCID: PMC11779713 DOI: 10.3389/fcimb.2024.1496176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Background Drug repurposing has become a widely adopted strategy to minimise research time, costs, and associated risks. Combinations of protease inhibitors such as lopinavir and darunavir with ritonavir have been repurposed as treatments for COVID-19. Although lopinavir-ritonavir (LPV/r) and darunavir-ritonavir (DRV/r) have shown in vitro efficacy against COVID-19, the results in human studies have been inconsistent. Therefore, our objective was to compare the efficacy of LPV/r and DRV/r in COVID-19 patients admitted to a tertiary centre in Romania. Research design and methods A clinical dataset from 417 hospitalised patients was analysed. Patients were assigned to the LPV/r, DRV/r, or control (standard-of-care) group based on clinical decisions made by the attending infectious disease specialists, aligned with national treatment protocols. Kaplan-Meier and Cox proportional hazards regression analyses were conducted to compare in-hospital mortality and to identify factors associated with clinical improvement or fatal outcomes. Results By day 10, more patients showed improvement with LPV/r and DRV/r (p=0.03 and 0.01, respectively), but only LPV/r was associated with improved survival compared to the control group (p=0.05). Factors associated with mortality included male gender (HR: 3.63, p=0.02), diabetes (HR: 2.49, p=0.03), oxygen saturation below 90% at admission (HR: 5.23, p<0.01), high blood glucose levels (HR: 3.68, p=0.01), age (HR: 1.04, p=0.02), and more than 25% lesion extension on chest CT scan (HR: 2.28, p=0.03). Conclusions LPV/r, but not DRV/r, showed a survival benefit in patients hospitalised with COVID-19, but these findings deserve further investigation in a randomised clinical trial.
Collapse
Affiliation(s)
- Dóra Paróczai
- Department of Medical Microbiology, University of Szeged, Szeged, Hungary
- Albert Szent-Györgyi Health Center, Pulmonology Clinic, University of Szeged, Deszk, Hungary
| | - András Bikov
- North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Andreea Blidaru
- Department of Infectious Diseases, Infectious Diseases and Pulmonology Clinical Hospital, Timisoara, Romania
| | - Emanuel Bobu
- Department of Pulmonology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Ana Lascu
- Department of Functional Sciences, Discipline of Pathophysiology, Centre for Translational Research and Systems Medicine, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Institute for Cardiovascular Diseases of Timisoara, Clinic for Cardiovascular Surgery, Timisoara, Romania
| | - Cristian Ion Mot
- ENT Department, Municipal Emergency Hospital Timisoara, Timisoara, Romania
- Department of Surgery, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Stefan Mihaicuta
- Department of Infectious Diseases, Infectious Diseases and Pulmonology Clinical Hospital, Timisoara, Romania
- Centre for Research and Innovation in Precision Medicine of Respiratory Diseases, Department of Pulmonology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Stefan Frent
- Department of Infectious Diseases, Infectious Diseases and Pulmonology Clinical Hospital, Timisoara, Romania
- Centre for Research and Innovation in Precision Medicine of Respiratory Diseases, Department of Pulmonology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| |
Collapse
|
11
|
Ahmed AB, Abdelrahman MM, Edrees FH. Eco-sustainable chromatographic method for the determination of favipiravir and nitazoxanide for COVID-19: application to human plasma. BMC Chem 2025; 19:11. [PMID: 39789629 PMCID: PMC11714856 DOI: 10.1186/s13065-024-01364-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19), an extremely contagious illness, has posed enormous challenges to healthcare systems around the world. Although the evidence on COVID-19 management is growing, antiviral medication is still the first line of treatment. Therefore, it is critical that effective, safe, and tolerable antivirals be available to treat early COVID-19 and stop its progression. Recently, favipiravir (FAV) has received FDA approval as safe and effective antiviral medication for COVID-19 management. Nitazoxanide (NTZ) also possesses antiviral and immunomodulating activities. Moreover, FAV and NTZ in combination are clinically used in COVID-19 treatment with reported safety, synergistic antiviral and immunomodulating effects. Despite the availability of various clinical studies on both FAV and NTZ, no existing analytical application for the simultaneous estimation of FAV and NTZ exists. As a result, the current work goal is to establish a green HPLC method for their analysis and implementation to human plasma. The developed method utilizes isocratic elution with 0.1% aqueous formic acid: ethanol (55:45, v/v) and dantrolene as internal standard. The bioanalytical validation parameters passed the FDA acceptance criteria. NEMI, eco scale, AGREE and ComplexGAPI approaches were used for qualitative and quantitative evaluation of the method's greenness.
Collapse
Affiliation(s)
- Amal B Ahmed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University (NUB), Sharq El-Nile, Beni-Suef, 62511, Egypt.
| | - Maha M Abdelrahman
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Alshaheed Shehata Ahmad Hegazy St, Beni-Suef, 62514, Egypt
| | - Fadwa H Edrees
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nile Valley University (NVU), El Fayoum, 63518, Egypt
| |
Collapse
|
12
|
Maddukuri RK, Desimalla ML, Banavathu R, Arepalli SPK. Ivermectin as a Treatment Modality in COVID-19 Patients: A Systematic Review and Meta-Analysis of Up-To-Date RCTs. Indian J Community Med 2025; 50:9-19. [PMID: 40124821 PMCID: PMC11927832 DOI: 10.4103/ijcm.ijcm_117_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/27/2024] [Indexed: 03/25/2025] Open
Abstract
The objective of this systematic review and meta-analysis is to assess the efficacy and safety of Ivermectin compared to placebo or standard of care for the treatment and prevention of COVID-19 disease. Literature search was carried out in PubMed, Scopus, and google scholar. Clinical trial registries (clinical trail.gov) and preprint servers (Medrxiv) were also searched for registered and preprint data respectively. Mortality, Clinical recovery, Clinical worsening, duration of hospital stay, and adverse events were considered as primary outcomes while viral clearance is considered as a secondary outcome. Revised Cochrane risk of bias assessment tool was used to assess the risk of bias. Random effect meta-analysis was carried out for all the outcomes. GRADE was used to rate the certainty of evidence. A total of 19 RCTs with 1111 patients were included in the analysis. Ivermectin use did was not associated with a reduction in the incidence of mortality, duration of hospitalization, clinical worsening, and incidence of adverse events nor it was associated with an increase in clinical improvement compared to either placebo or standard of care. Ivermectin also did not show any reduction in viral clearance compared to both placebo or standard of care and this was associated with substantial inconsistency. The overall certainty of the evidence was very low to low. Based on the very low to low certainty of the evidence, we consider Ivermectin ineffective in the management of COVID-19 disease, both as treatment and prophylaxis.
Collapse
Affiliation(s)
- Raghava Kalyan Maddukuri
- Department of Pharmacy Practice, Chebrolu Hanumaiah Institute of Pharmaceutical Sciences, Beside RVR and JC College of Engineering and Technology, Chandramoulipuram, Chowdavaram, Guntur, Andhra Pradesh, India
| | - Madhavi Latha Desimalla
- Department of Pharmacy Practice, Chebrolu Hanumaiah Institute of Pharmaceutical Sciences, Beside RVR and JC College of Engineering and Technology, Chandramoulipuram, Chowdavaram, Guntur, Andhra Pradesh, India
| | - Reshma Banavathu
- Department of Pharmacy Practice, Chebrolu Hanumaiah Institute of Pharmaceutical Sciences, Beside RVR and JC College of Engineering and Technology, Chandramoulipuram, Chowdavaram, Guntur, Andhra Pradesh, India
| | - Sai Pavan Kumar Arepalli
- Department of Pharmacy Practice, Chebrolu Hanumaiah Institute of Pharmaceutical Sciences, Beside RVR and JC College of Engineering and Technology, Chandramoulipuram, Chowdavaram, Guntur, Andhra Pradesh, India
| |
Collapse
|
13
|
Gawande V, Kushwaha R, Mandal AA, Banerjee S. Targeting SARS-CoV-2 Proteins: In Silico Investigation with Polypyridyl-Based Zn(II)-Curcumin Complexes. Chembiochem 2024; 25:e202400612. [PMID: 39264259 DOI: 10.1002/cbic.202400612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/13/2024]
Abstract
Herein, we have selected eight Zn(II)-based complexes viz., [Zn(bpy)(acac)Cl] (1), [Zn(phen)(acac)Cl] (2), [Zn(dppz)(acac)Cl] (3), [Zn(dppn)(acac)Cl] (4), [Zn(bpy)(cur)Cl] (5), [Zn(phen)(cur)Cl] (6), [Zn(dppz)(cur)Cl] (7), [Zn(dppn)(cur)Cl] (8), where bpy=2,2'-bipyridine, phen=1,10-phenanthroline, dppz=benzo[i]dipyrido[3,2-a:2',3'-c]phenazine, dppn=naphtho[2,3-i]dipyrido[3,2-a:2',3'-c]phenazine, acac=acetylacetonate, cur=curcumin and performed in silico molecular docking studies with the viral proteins, i. e., spike protein (S), Angiotensin-converting enzyme II Receptor protein (ACE2), nucleocapsid protein (N), main protease protein (Mpro), and RNA-dependent RNA polymerase protein (RdRp) of SARS-CoV-2. The binding energy calculations, visualization of the docking orientation, and analysis of the interactions revealed that these complexes could be potential inhibitors of the viral proteins. Among complexes 1-8, complex 6 showed the strongest binding affinity with S and ACE2 proteins. 4 exerted better binding affinity in the case of the N protein, whereas 8 presented the highest binding affinities with Mpro and RdRp among all the complexes. Overall, the study indicated that Zn(II) complexes have the potential as alternative and viable therapeutic solutions for COVID-19.
Collapse
Affiliation(s)
- Vedant Gawande
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
14
|
Metwaly AM, Elkaeed EB, Alsfouk AA, Ibrahim IM, Elkady H, Eissa IH. Repurposing FDA-approved drugs for COVID-19: targeting the main protease through multi-phase in silico approach. Antivir Ther 2024; 29:13596535241305536. [PMID: 39639531 DOI: 10.1177/13596535241305536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND The COVID-19 pandemic has created an urgent need for effective therapeutic agents. The SARS-CoV-2 Main Protease (Mpro) plays a crucial role in viral replication and immune evasion, making it a key target for drug development. While several studies have explored Mpro inhibition, identifying FDA-approved drugs with potential efficacy remains a critical research focus. PURPOSE This study aims to identify FDA-approved drugs that could inhibit SARS-CoV-2 Mpro. Using computational screening, we seek compounds that share structural similarities with a known co-crystallized ligand (PRD_002214) and exhibit strong binding affinity to the enzyme, providing viable candidates for COVID-19 treatment. RESEARCH DESIGN A systematic in silico approach was used, screening 3009 FDA-approved drugs. The initial screening focused on structural similarity to PRD_002214 (PDB ID: 6LU7), followed by molecular docking studies to predict binding affinity. Promising compounds were further analyzed through molecular dynamics (MD) simulations to evaluate their stability and interactions with Mpro over 100 ns. STUDY SAMPLE Of the 3009 FDA-approved drugs screened, 74 were selected for initial evaluation. After refinement, 28 compounds underwent docking analysis, with eight showing strong binding potential to Mpro. ANALYSIS Molecular docking assessed the binding affinity and interaction of the selected compounds with Mpro. MD simulations were conducted on the top compound, Atazanavir, to study its dynamic interactions. MM-GBSA, PLIP, and PCAT analyses were used to validate binding affinity and interactions. RESULTS Eight compounds, including Carfilzomib, Atazanavir, Darunavir, and others, exhibited promising binding affinities. Among them, Atazanavir showed the highest binding strength and was selected for further MD simulation studies. These simulations revealed that Atazanavir forms stable interactions with Mpro, demonstrating favorable binding and dynamic stability. The binding affinity was further confirmed through MM-GBSA, PLIP, and PCAT analyses, supporting Atazanavir's potential as an effective Mpro inhibitor. CONCLUSIONS In silico results suggest that Atazanavir is a promising candidate for targeting SARS-CoV-2 Mpro, with strong binding affinity and dynamic stability. These findings support its potential as a lead compound for further preclinical and clinical testing, though in vitro and in vivo validation are needed to confirm its therapeutic efficacy against COVID-19.
Collapse
Affiliation(s)
- Ahmed M Metwaly
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Aisha A Alsfouk
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ibrahim M Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
15
|
Mtambo TR, Machaba KE, Chellan N, Ramharack P, Muller CJF, Mhlongo NN, Hlengwa N. The Effect of Metformin and Hydrochlorothiazide on Cytochrome P450 3A4 Metabolism of Ivermectin: Insights from In Silico Experimentation. Int J Mol Sci 2024; 25:12089. [PMID: 39596155 PMCID: PMC11594981 DOI: 10.3390/ijms252212089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The spread of SARS-CoV-2 has led to an interest in using ivermectin (a potent antiparasitic agent) as an antiviral agent despite the lack of convincing in vivo clinical data for its use against COVID-19. The off-target prophylactic use of ivermectin adds a substantial risk of drug-drug interactions with pharmaceutical medications used to treat chronic conditions like diabetes and hypertension (metformin and hydrochlorothiazide, respectively). Therefore, this study aims to evaluate the potential drug-drug interactions between ivermectin with either metformin or hydrochlorothiazide. In silico experiments and high-throughput screening assays for CYP3A4 were conducted to understand how metformin and hydrochlorothiazide might affect CYP3A4's role in metabolizing ivermectin. The study findings indicated that hydrochlorothiazide is more stable than both ivermectin and metformin. This conclusion was further supported by root mean square fluctuation analysis, which showed that hydrochlorothiazide is more flexible. The variation in the principal component analysis scatter plot across the first three normal modes suggests hydrochlorothiazide has a more mobile conformation than ivermectin and metformin. Additionally, a strong inhibition of CYP3A4 by hydrochlorothiazide was observed, suggesting that hydrochlorothiazide's regulatory effects could significantly impede CYP3A4 activity, potentially leading to a reduced metabolism and clearance of ivermectin in the body. Concurrent administration of these drugs may result in drug-drug interactions and hinder the hepatic metabolism of ivermectin.
Collapse
Affiliation(s)
- Thuli R. Mtambo
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa 3886, South Africa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Kgothatso E. Machaba
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa 3886, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Nireshni Chellan
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Pritika Ramharack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Ndumiso N. Mhlongo
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Nokulunga Hlengwa
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa 3886, South Africa
| |
Collapse
|
16
|
Gamberi C, Leverette CL, Davis AC, Ismail M, Piccialli I, Borbone N, Oliviero G, Vicidomini C, Palumbo R, Roviello GN. Oceanic Breakthroughs: Marine-Derived Innovations in Vaccination, Therapy, and Immune Health. Vaccines (Basel) 2024; 12:1263. [PMID: 39591167 PMCID: PMC11598900 DOI: 10.3390/vaccines12111263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The vast, untapped potential of the world's oceans is revealing groundbreaking advancements in human health and vaccination. Microalgae such as Nannochloropsis spp. and Dunaliella salina are emerging as resources for recombinant vaccine development with specific and heterologous genetic tools used to boost production of functional recombinant antigens in Dunaliella salina and Nannochloropsis spp. to induce immunoprotection. In humans, several antigens produced in microalgae have shown potential in combating diseases caused by the human papillomavirus, human immunodeficiency virus, hepatitis B virus, influenza virus, Zika virus, Zaire Ebola virus, Plasmodium falciparum, and Staphylococcus aureus. For animals, microalgae-derived vaccine prototypes have been developed to fight against the foot-and-mouth disease virus, classical swine fever virus, vibriosis, white spot syndrome virus, and Histophilus somni. Marine organisms offer unique advantages, including the ability to express complex antigens and sustainable production. Additionally, the oceans provide an array of bioactive compounds that serve as therapeutics, potent adjuvants, delivery systems, and immunomodulatory agents. These innovations from the sea not only enhance vaccine efficacy but also contribute to broader immunological and general health. This review explores the transformative role of marine-derived substances in modern medicine, emphasizing their importance in the ongoing battle against infectious diseases.
Collapse
Affiliation(s)
- Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Chad L. Leverette
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Alexis C. Davis
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Moayad Ismail
- Faculty of Medicine, European University, 76 Guramishvili Ave., 0141 Tbilisi, Georgia;
| | - Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Nicola Borbone
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Giorgia Oliviero
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Caterina Vicidomini
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| | - Rosanna Palumbo
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| |
Collapse
|
17
|
Yang D, Ning J, Zhang Y, Xu X, Zhang D, Fan H, Wang J, Lu G. In vitro assessment of the anti-adenoviral activity of artemisinin and its derivatives. Virus Res 2024; 349:199448. [PMID: 39127240 PMCID: PMC11403056 DOI: 10.1016/j.virusres.2024.199448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Adenoviral infections, particularly in children, remain a significant public health issue with no approved targeted treatments. Artemisinin and its derivatives, well-known for their use in malaria treatment, have shown antiviral activities in recent studies. However, their efficacy against human adenovirus (HAdV) remains unexplored. This study aimed to assess the activity of artemisinin and its derivatives against HAdV infection in vitro using cell lines and primary cells. Our data revealed that artemisinin exhibited dose-dependent anti-HAdV activity with no apparent cytotoxicity over a wide concentration range. Mechanistically, artemisinin did not affect viral attachment or entry into target cells, nor the viral genome entry into cell nucleus. Instead, it inhibited HAdV through suppression of viral DNA replication. Comparative analysis with its derivatives, artesunate and artemisone, showed distinct cytotoxicity and anti-adenoviral profiles, with artemisone showing superior efficacy and lower toxicity. Further validation using a primary airway epithelial cell model confirmed the anti-adenoviral activity of both artemisinin and artemisone against different virus strains. Together, our findings suggest that artemisinin and its derivatives may be promising candidates for anti-HAdV treatment.
Collapse
Affiliation(s)
- Diyuan Yang
- Department of Respiratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, China; Department of Pediatric Respiratory, Guangzhou women and children's medical center liuzhou hospital, Guangxi, Liuzhou, 545006, China
| | - Jing Ning
- Department of Children's Health Care, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, 511442, China
| | - Yuyu Zhang
- Department of Respiratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Xuehua Xu
- Department of Respiratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Dongwei Zhang
- Department of Respiratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Huifeng Fan
- Department of Respiratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, China
| | - Jing Wang
- Department of Children's Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Gen Lu
- Department of Respiratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Guangzhou, 510623, China.
| |
Collapse
|
18
|
Monteonofrio L, Virdia I, Pozzi S, Quadri R, Amendolare A, Marzano F, Braile M, Sulfaro V, Paroni M, Tullo A, Soddu S, Guerrini L. Molecular mechanisms of thalidomide effectiveness on COVID-19 patients explained: ACE2 is a new ΔNp63α target gene. J Mol Med (Berl) 2024; 102:1371-1380. [PMID: 39294414 PMCID: PMC11525293 DOI: 10.1007/s00109-024-02485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024]
Abstract
COVID-19 pandemic is caused by the SARS-CoV-2 virus, whose internalization and infection are mediated by the angiotensin-converting enzyme 2 (ACE2). The identification of novel approaches to tackle this step is instrumental for the development of therapies for the management of COVID-19 and other diseases with a similar mechanism of infection. Thalidomide, a drug sadly known for its teratogenic effects, has potent immunomodulatory and anti-inflammatory properties. Treatment with this drug has been shown to improve the immune functions of COVID-19 patients and proposed for the management of COVID-19 in clinical practice through drug repositioning. Here, we investigated the molecular details linking thalidomide to ACE2 and COVID-19, showing that in conditions mimicking SARS-CoV-2-associated cytokine storm, the transcription factor ΔNp63α and ACE2 are stabilized, and IL-8 production is increased. In such conditions, we found p63 to bind to and regulate the expression of the ACE2 gene. We previously showed that ΔNp63α is degraded upon thalidomide treatment and now found that treatment with this drug-or with its analogue lenalidomide-downregulates ACE2 in a p63-dependent manner. Finally, we found that thalidomide treatment reduces in vitro infection by pseudo-SARS-CoV-2, a baculovirus pseudotyped with the SARS-CoV-2 spike protein. Overall, we propose the dual effect of thalidomide in reducing SARS-CoV-2 viral re-entry and inflammation through p63 degradation to weaken SARS-CoV-2 entry into host cells and mitigate lung inflammation, making it a valuable option in clinical management of COVID-19. KEY MESSAGES: Thalidomide treatment results in p63-dependent ACE2 downregulation. ACE2 is a p63 transcriptional target. Thalidomide reduces the "cytokine storm" associated to COVID-19. Thalidomide prevents viral re-entry of SARS-CoV-2 by p63-dependent ACE2 downregulation. Thalidomide is a modulator of SARS-CoV-2 or other ACE2-dependent infections. ACE2 is modulated by a pharmacological substance.
Collapse
Affiliation(s)
- Laura Monteonofrio
- Dipartimento Di Ricerca E Tecnologie Avanzate, Istituto Nazionale Tumori Regina Elena IRCCS, 00144, Rome, Italy
| | - Ilaria Virdia
- Dipartimento Di Ricerca E Tecnologie Avanzate, Istituto Nazionale Tumori Regina Elena IRCCS, 00144, Rome, Italy
| | - Sara Pozzi
- Dipartimento Di Bioscienze, Università Degli Studi Di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Roberto Quadri
- Dipartimento Di Bioscienze, Università Degli Studi Di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Alessandra Amendolare
- Istituto Di Biomembrane, Bioenergetica E Biotecnologie Molecolari (IBIOM), Consiglio Nazionale Delle Ricerche, 70025, Bari, Italy
| | - Flaviana Marzano
- Istituto Di Biomembrane, Bioenergetica E Biotecnologie Molecolari (IBIOM), Consiglio Nazionale Delle Ricerche, 70025, Bari, Italy
| | - Micaela Braile
- Dipartimento Di Bioscienze, Università Degli Studi Di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Virginia Sulfaro
- Dipartimento Di Bioscienze, Università Degli Studi Di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Moira Paroni
- Dipartimento Di Bioscienze, Università Degli Studi Di Milano, Via Celoria 26, 20133, Milan, Italy
| | - Apollonia Tullo
- Istituto Di Biomembrane, Bioenergetica E Biotecnologie Molecolari (IBIOM), Consiglio Nazionale Delle Ricerche, 70025, Bari, Italy
| | - Silvia Soddu
- Dipartimento Di Ricerca E Tecnologie Avanzate, Istituto Nazionale Tumori Regina Elena IRCCS, 00144, Rome, Italy
| | - Luisa Guerrini
- Dipartimento Di Bioscienze, Università Degli Studi Di Milano, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
19
|
Manchanda RK, Miglani A, Kalsi A, Brahmachari S, Rama KN, Goel P, Kaushik P, Jethani A, Nagrath C, Pangtey NY, Kannoth JT, Sharma K, Arora S, Amitav B, Roy PK, Kudiyarasu RK, Rutten L. Homeopathic Medicines in Third (Omicron) Wave of COVID-19: Prognostic Factor Research. HOMEOPATHY 2024; 113:223-230. [PMID: 38158196 DOI: 10.1055/s-0043-1776758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
BACKGROUND With the emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, such as the Omicron variant, during the third wave of the coronavirus disease 2019 (COVID-19) pandemic, there was a need to identify useful homeopathic medicines. This study aimed to identify such medicines and their indications using prognostic factor research (PFR). METHODS This was an open-label, multi-centred observational study conducted in January 2022, on confirmed COVID-19 cases. The data were collected from integrated COVID Care Centres in Delhi, India, where homeopathic medicines were prescribed along with conventional treatment. Only those cases that met a set of selection criteria were considered for analysis. The likelihood ratio (LR) was calculated for the frequently occurring symptoms of the frequently prescribed medicines. An LR of 1.3 or greater was considered meaningful. RESULTS Out of the 362 COVID-19 cases, 263 cases were selected for analysis after applying selection criteria. Common symptoms included fatigue, cough, sore throat, myalgia and headache. Twenty-one medicines were prescribed, of which nine medicines - Gelsemium sempervirens, Bryonia alba, Hepar sulphuris, Rhus toxicodendron, Pulsatilla nigricans, Arsenicum album, Belladonna, Nux vomica and Phosphorus - were frequently used. By calculating LRs, the study identified meaningful indications for these medicines. CONCLUSION Homeopathic medicines have shown promising results in the third wave of COVID-19 as an adjunct therapy. The medicines that were used in the first and second waves were found useful in the third wave also, and their indications were analogous to those found in the earlier waves. Certain new indications of some medicines were elicited in this wave, which warrant further research. However, it is important not to restrict to these medicines only and to continue data collection on COVID-19 in future waves for the improvement of the COVID-19 mini-repertory.
Collapse
Affiliation(s)
- Raj Kumar Manchanda
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, Secretary for Information and Communication, Liga Medicorum Homeopathica Internationalis, New Delhi, India
| | - Anjali Miglani
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Amrit Kalsi
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Smita Brahmachari
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Kamsali Nadigadda Rama
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Pawan Goel
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Pallavi Kaushik
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Anju Jethani
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Cheshta Nagrath
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Nirmal Yadav Pangtey
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Jithesh Thavarayil Kannoth
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Kavita Sharma
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Shelly Arora
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - B Amitav
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Pradip Kumar Roy
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Ram Kumar Kudiyarasu
- Health and Family Welfare Department, Directorate of AYUSH, Government of National Capital Territory of Delhi, New Delhi, India
| | - Lex Rutten
- Independent Practitioner, Breda, The Netherlands
| |
Collapse
|
20
|
Bustos-Hamdan A, Bracho-Gallardo JI, Hamdan-Partida A, Bustos-Martínez J. Repositioning of Antibiotics in the Treatment of Viral Infections. Curr Microbiol 2024; 81:427. [PMID: 39460768 PMCID: PMC11512906 DOI: 10.1007/s00284-024-03948-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
Drug repurposing, also known as drug repositioning, is a currently tested approach by which new uses are being assigned for already tested drugs. In this case there are antibiotics that are used to combat bacterial infections. However, antibiotics are among the drugs that have been studied for possible antiviral activities. Therefore, the aim of this work is to carry out a review of the studies of antibiotics that could be repositioned for the treatment of viral infections. Among the main antibiotics that have demonstrated antiviral activity are macrolides and glycopeptides. In addition, several antibiotics from the group of tetracyclines, fluoroquinolones, cephalosporins and aminoglycosides have also been studied for their antiviral activity. These antibiotics have demonstrated antiviral activity against both RNA and DNA viruses, including the recent pandemic virus SARS-CoV-2. Some of these antibiotics were selected in addition to its antiviral activity for their immunomodulatory and anti-inflammatory properties. Of the antibiotics that present antiviral activity, in many cases the mechanisms of action are not exactly known. The use of these antibiotics to combat viral infections remains controversial and is not generally accepted, since clinical trials are required to prove its effectiveness. Therefore, there is currently no antibiotic approved as antiviral therapy. Hence is necessary to present the studies carried out on antibiotics that can be repositioned in the future as antiviral drugs.
Collapse
Affiliation(s)
- Anaíd Bustos-Hamdan
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Jair Isidoro Bracho-Gallardo
- Maestria en Biología de la Reproducción Animal, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Aída Hamdan-Partida
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Jaime Bustos-Martínez
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico.
| |
Collapse
|
21
|
Manoharan S, Santhakumar A, Perumal E. Targeting STAT3, FOXO3a, and Pim-1 kinase by FDA-approved tyrosine kinase inhibitor-Radotinib: An in silico and in vitro approach. Arch Pharm (Weinheim) 2024:e2400429. [PMID: 39428846 DOI: 10.1002/ardp.202400429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/22/2024] [Accepted: 09/14/2024] [Indexed: 10/22/2024]
Abstract
Cancer, a multifactorial pathological condition, is primarily caused due to mutations in multiple genes. Hepatocellular carcinoma (HCC) is a form of primary liver cancer that is often diagnosed at the advanced stage. Current treatment strategies for advanced HCC involve systemic therapies which are often hindered due to the emergence of resistance and toxicity. Therefore, a multitarget approach might prove more effective in HCC treatment. The present study focuses on targeting signal transducer and activator of transcription 3 (STAT3), forkhead box class O3a (FOXO3a), and proviral integration site for Moloney murine leukemia virus-1 (Pim-1) kinase, using a Food and Drug Administration (FDA)-approved anticancer drug library. Two compounds, namely, radotinib and capmatinib, were identified as top compounds using molecular docking. Among the two compounds, radotinib exhibited significant binding values towards the targeted proteins and their heterodimers. Furthermore, in vitro experiments involving 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), live/dead, 4',6-diamidino-2-phenylindole, and clonogenic assays were performed to evaluate the effect of radotinib in human hepatoblastoma cell line/hepatocellular carcinoma cells. The gene expression data indicated reduced expression of FOXO3a and Pim-1, but no basal-level alteration of STAT3. The Western blot analysis assay showed that the phosphorylation level of STAT3 was significantly decreased upon radotinib treatment. Taken together, our findings suggest that radotinib, which is currently used in the treatment of chronic myeloid leukemia (CML), could be considered as a potential candidate for repurposing in the treatment of HCC.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | | | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| |
Collapse
|
22
|
Chou SP, Chuang YJ, Chen BS. Systems Biology Methods via Genome-Wide RNA Sequences to Investigate Pathogenic Mechanisms for Identifying Biomarkers and Constructing a DNN-Based Drug-Target Interaction Model to Predict Potential Molecular Drugs for Treating Atopic Dermatitis. Int J Mol Sci 2024; 25:10691. [PMID: 39409019 PMCID: PMC11477013 DOI: 10.3390/ijms251910691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
This study aimed to construct genome-wide genetic and epigenetic networks (GWGENs) of atopic dermatitis (AD) and healthy controls through systems biology methods based on genome-wide microarray data. Subsequently, the core GWGENs of AD and healthy controls were extracted from their real GWGENs by the principal network projection (PNP) method for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation. Then, we identified the abnormal signaling pathways by comparing the core signaling pathways of AD and healthy controls to investigate the pathogenesis of AD. Then, IL-1β, GATA3, Akt, and NF-κB were selected as biomarkers for their important roles in the abnormal regulation of downstream genes, leading to cellular dysfunctions in AD patients. Next, a deep neural network (DNN)-based drug-target interaction (DTI) model was pre-trained on DTI databases to predict molecular drugs that interact with these biomarkers. Finally, we screened the candidate molecular drugs based on drug toxicity, sensitivity, and regulatory ability as drug design specifications to select potential molecular drugs for these biomarkers to treat AD, including metformin, allantoin, and U-0126, which have shown potential for therapeutic treatment by regulating abnormal immune responses and restoring the pathogenic signaling pathways of AD.
Collapse
Affiliation(s)
- Sheng-Ping Chou
- Laboratory of Automatic Control, Signal Processing and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Yung-Jen Chuang
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Bor-Sen Chen
- Laboratory of Automatic Control, Signal Processing and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan;
| |
Collapse
|
23
|
Chavda V, Dodiya P, Apostolopoulos V. Adverse drug reactions associated with COVID-19 management. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7353-7376. [PMID: 38743117 DOI: 10.1007/s00210-024-03137-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024]
Abstract
The emergence of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) outbreak, which causes COVID-19, had a devastating impact on both people's lives and the global economy. During the course of the pandemic, the lack of specific drugs or treatments tailored for COVID-19 led to extensive repurposing of existing drugs in the pursuit of effective treatments. Some drug molecules demonstrated efficacy, while others proved ineffective. In this context, the approach of drug repurposing emerged as a novel strategy for combating COVID-19. Repurposed drugs and biologics have shown effectiveness, leading to improved clinical outcomes among patients with COVID-19. Similarly, It is equally important to assess the risk-benefit ratio associated with drugs and biologics adapted for COVID-19 treatment. Herein, we primarily focus on evaluating adverse drug events linked to repurposed COVID-19 medications, repurposed biologics, and COVID-specific drug molecules.
Collapse
Affiliation(s)
- Vivek Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India.
| | - Payal Dodiya
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia.
- Australian Institute for Musculoskeletal Science, Melbourne, VIC, Australia.
| |
Collapse
|
24
|
Alduaij OK, Hussein RK, Abu Alrub S, Zidan SAH. Antimicrobial activities of Diltiazem Hydrochloride: drug repurposing approach. PeerJ 2024; 12:e17809. [PMID: 39329140 PMCID: PMC11426324 DOI: 10.7717/peerj.17809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/03/2024] [Indexed: 09/28/2024] Open
Abstract
Background The growing concern of antibiotic-resistant microbial strains worldwide has prompted the need for alternative methods to combat microbial resistance. Biofilm formation poses a significant challenge to antibiotic efficiency due to the difficulty of penetrating antibiotics through the sticky microbial aggregates. Drug repurposing is an innovative technique that aims to expand the use of non-antibiotic medications to address this issue. The primary objective of this study was to evaluate the antimicrobial properties of Diltiazem HCl, a 1,5-benzothiazepine Ca2 + channel blocker commonly used as an antihypertensive agent, against four pathogenic bacteria and three pathogenic yeasts, as well as its antiviral activity against the Coxsackie B4 virus (CoxB4). Methods To assess the antifungal and antibacterial activities of Diltiazem HCl, the well diffusion method was employed, while crystal violet staining was used to determine the anti-biofilm activity. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) colorimetric assay was utilized to evaluate the antiviral activity of Diltiazem HCl against the CoxB4 virus. Results This study revealed that Diltiazem HCl exhibited noticeable antimicrobial properties against Gram-positive bacteria, demonstrating the highest inhibition of Staphylococcus epidermidis, followed by Staphylococcus aureus. It effectively reduced the formation of biofilms by 95.1% and 90.7% for S. epidermidis, and S. aureus, respectively. Additionally, the antiviral activity of Diltiazem HCl was found to be potent against the CoxB4 virus, with an IC50 of 35.8 ± 0.54 μg mL-1 compared to the reference antiviral Acyclovir (IC50 42.71 ± 0.43 μg mL-1). Conclusion This study suggests that Diltiazem HCl, in addition to its antihypertensive effect, may also be a potential treatment option for infections caused by Gram-positive bacteria and the CoxB4 viruses, providing an additional off-target effect for Diltiazem HCl.
Collapse
Affiliation(s)
- Omar K. Alduaij
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Rageh K. Hussein
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sharif Abu Alrub
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sabry A. H. Zidan
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Assiut-Branch, Assiut, Egypt
| |
Collapse
|
25
|
Poonia N, Jadhav NV, Mamatha D, Garg M, Kabra A, Bhatia A, Ojha S, Lather V, Pandita D. Nanotechnology-assisted combination drug delivery: a progressive approach for the treatment of acute myeloid leukemia. Ther Deliv 2024; 15:893-910. [PMID: 39268925 PMCID: PMC11497954 DOI: 10.1080/20415990.2024.2394012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Acute myeloid leukemia (AML), a heterogeneous hematopoietic cancer prevalent in adults, has been a leading cause of leukemia-associated deaths for decades. Despite advancements in understanding its pathology and pharmacological targets, therapeutic strategies have seen minimal change. The standard treatment, combining cytarabine and anthracycline, has persisted, accompanied by challenges such as pharmacokinetic issues and non-specific drug delivery, leading to severe side effects. Nanotechnology offers a promising solution through combination drug delivery. FDA-approved CPX351 (VYXEOS™) a liposomal formulation delivering doxorubicin and cytarabine, exemplifies enhanced therapeutic efficacy. Ongoing research explores various nanocarriers for delivering multiple bioactives, addressing drug targeting, pharmacokinetics and chemoresistance. This review highlights nanotechnology-based combination therapies for the effective management of AML, presenting a potential breakthrough in leukemia.
Collapse
Affiliation(s)
- Neelam Poonia
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Nikita Vijay Jadhav
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
| | - Davuluri Mamatha
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector-125, Noida, 201313, India
| | - Atul Kabra
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University (Govt of Punjab), Dabwali Road, Bathinda, Punjab, 151001, India
| | - Shreesh Ojha
- Pharmacology, College of Medicine & Health Sciences, P.O. Box 15551, Al Ain, UAE
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University, Noida, 201313, Uttar Pradesh, India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
- Centre for Advanced Formulation & Technology (CAFT), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
| |
Collapse
|
26
|
Olalekan SO, Obakachi VA, Badeji AA, Akinsipo (Oyelaja) OB, Familoni O, Asekun OT, Oladipo SD, Osinubi AD. Exploring the therapeutic potential of prolinamides as multi-targeted agents for Alzheimer's disease treatment: molecular docking and molecular dynamic simulation studies. In Silico Pharmacol 2024; 12:80. [PMID: 39224128 PMCID: PMC11365881 DOI: 10.1007/s40203-024-00250-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease (AD) presents a significant global health challenge, with its prevalence expected to rise sharply in the coming years. Despite extensive research, effective treatments addressing the multifaceted pathophysiology of AD remain elusive. This study investigates the therapeutic potential of twenty-seven prolinamides (P1 - P27), with the focus on their interactions with key proteins implicated in AD pathogenesis. Four of the compounds, namely; 10-((4-nitrophenyl)prolyl)-10 H-phenothiazine (P14), 2-((4-nitrophenyl)prolyl)isoindoline (P19), 1-(4-formylphenyl)-N-(p-tolyl)pyrrolidine-2-carboxamide (P22), and N,1-bis(4-nitrophenyl)pyrrolidine-2-carboxamide (P27) showed promising potential as Alzheimer's drug. In-silico approaches including molecular docking, molecular dynamic (MD) simulation, post md study, physicochemical and drug-likeness parameters were employed to ascertain the potential of these compounds as inhibitors of certain proteins implicated in the pathophysiology of Alzheimer's disease. Molecular docking and dynamics simulations demonstrated that P14, P19, P22 and P27 exhibited promising binding affinities towards crucial AD-associated proteins, including Beta-Secretase 1 (BACE1), Butyrylcholinesterase (BuChE), and Tau-tubulin kinase 2 (TTBK2). Structural stability analyses revealed that prolinamides, particularly P22 and P27 for BACE1 and P14 and P19 for BuChE, exhibited greater stability than their reference ligands, indicated by lower RMSD, RoG, and RMSF values. For BuChE, Rivastigmine had a docking score of -7.0 kcal/mol, a binding free energy (ΔGbind) of -22.19 ± 2.44 kcal/mol, RMSD of 1.361 ± 0.162 Å, RMSF of 9.357 ± 3.212 Å, and RoG of 22.919 ± 0.064 Å, whereas P19 exhibited a superior docking score of -10.3 kcal/mol, a significantly better ΔGbind of -33.74 ± 2.84 kcal/mol, RMSD of 1.347 ± 0.132 Å, RMSF of 8.164 ± 2.748 Å, and RoG of 22.868 ± 0.070 Å. Physicochemical and pharmacokinetic assessments affirmed the drug-likeness and bioavailability of P19 notably capable of penetrating the blood-brain barrier. Compounds P19 and P22, emerged as multi-targeted ligands, offering the potential for simultaneous modulation of multiple AD-related pathways. These findings highlight the possibilities of these compounds to be explored as novel therapeutic agents for AD. They also highlight the need for further experimental validation to confirm their efficacy and safety profiles, advancing them toward clinical application in AD management. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00250-z.
Collapse
Affiliation(s)
- Samuel O. Olalekan
- Department of Physiology, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State Nigeria
| | - Vincent A. Obakachi
- Department of Chemical Sciences, University of Johannesburg, Doornfontein Campus, P.O. Box 17011, Johannesburg, 2028 South Africa
| | - Abosede A. Badeji
- Department of Chemical Sciences, Tai Solarin University of Education, Ijagun, P.M.B. 2118, Ijebu Ode, Ogun State Nigeria
| | | | - Oluwole Familoni
- Drug Design Research Group, Department of Chemistry, University of Lagos, Akoka-Yaba, Lagos, 101245 Nigeria
| | - Olayinka T. Asekun
- Drug Design Research Group, Department of Chemistry, University of Lagos, Akoka-Yaba, Lagos, 101245 Nigeria
| | - Segun D. Oladipo
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602 South Africa
- Department of Chemical Sciences, Olabisi Onabanjo University, P.M.B 2002, Ago-Iwoye, Nigeria
| | - Adejoke D. Osinubi
- Department of Chemical Sciences, Tai Solarin University of Education, Ijagun, P.M.B. 2118, Ijebu Ode, Ogun State Nigeria
- Drug Design Research Group, Department of Chemistry, University of Lagos, Akoka-Yaba, Lagos, 101245 Nigeria
| |
Collapse
|
27
|
Zheng X, He Y, Xia B, Tang W, Zhang C, Wang D, Tang H, Zhao P, Peng H, Liu Y. Etravirine Prevents West Nile Virus and Chikungunya Virus Infection Both In Vitro and In Vivo by Inhibiting Viral Replication. Pharmaceutics 2024; 16:1111. [PMID: 39339151 PMCID: PMC11435157 DOI: 10.3390/pharmaceutics16091111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Diseases transmitted by arthropod-borne viruses such as West Nile virus (WNV) and chikungunya virus (CHIKV) pose threat to global public health. Unfortunately, to date, there is no available approved drug for severe symptoms caused by both viruses. It has been reported that reverse transcriptase inhibitors can effectively inhibit RNA polymerase activity of RNA viruses. We screened the anti-WNV activity of the FDA-approved reverse transcriptase inhibitor library and found that 4 out of 27 compounds showed significant antiviral activity. Among the candidates, etravirine markedly inhibited WNV infection in both Huh 7 and SH-SY5Y cells. Further assays revealed that etravirine inhibited the infection of multiple arboviruses, including yellow fever virus (YFV), tick-borne encephalitis virus (TBEV), and CHIKV. A deeper study at the phase of action showed that the drug works primarily during the viral replication process. This was supported by the strong interaction potential between etravirine and the RNA-dependent RNA polymerase (RdRp) of WNV and alphaviruses, as evaluated using molecular docking. In vivo, etravirine significantly rescued mice from WNV infection-induced weight loss, severe neurological symptoms, and death, as well as reduced the viral load and inflammatory cytokines in target tissues. Etravirine showed antiviral effects in both arthrophlogosis and lethal mouse models of CHIKV infection. This study revealed that etravirine is an effective anti-WNV and CHIKV arbovirus agent both in vitro and in vivo due to the inhibition of viral replication, providing promising candidates for clinical application.
Collapse
Affiliation(s)
- Xu Zheng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Yanhua He
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Wanda Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Congcong Zhang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Dawei Wang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Haoran Peng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Yangang Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
28
|
Shafiq N, Jannat A, Munir H, Rashid M, Parveen S. Exploring the potential of FDA approved anti-diabetic drugs for repurposing against COVID-19: a core combination of multiple computational strategies and integrated artificial intelligence. J Biomol Struct Dyn 2024; 42:6556-6576. [PMID: 37455488 DOI: 10.1080/07391102.2023.2234993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
The latest variant of coronavirus is omicron. The World Health Organization (WHO) designated variation 'B.1.1.529' named omicron as a variant of concern (VOC) on 26 November 2021. By September 2020, it will have infected over 16 million patients and killed over 600,000 people over the world. This very infectious viral illness still poses a danger to world health; it has also become the greatest problem the world is facing and become the main area of research. The development of vaccines is insufficient to stop their spread and serious effects. Despite several reputable pharmaceutical firms claiming to have developed a cure for COVID-19. For that purpose, the field-based 3D-QSAR model has been used to analyze a series of anti-diabetic drugs to repurpose them against COVID-19. The LOO verified partial least square (PLS) model generates satisfactory q2 (0.4) and r2 (0.5) values. By using this model 10 compounds were screened out of 55 FDA approved anti-diabetic drugs (built-up library). Additionally, these substances were examined using molecular docking screening and ADMET. Finally, the drugs L8, and L23 were discovered to be the lead drugs. Density functional theory at the B3LYP/6-311G* technique was used to examine structural geometries, electronic characteristics, and molecular electrostatic potential (MEP). This work will greatly assist in the detection and development of leads for early drug development to control COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nusrat Shafiq
- Synthetic and Natural Product Drug Discovery Laboratory, Department of Chemistry, Government College Women University, Faisalabad, Pakistan
| | - Aqsa Jannat
- Synthetic and Natural Product Drug Discovery Laboratory, Department of Chemistry, Government College Women University, Faisalabad, Pakistan
| | - Huma Munir
- Green Chemistry Lab., Department of Chemistry, Government College Women University, Faisalabad, Pakistan
| | - Maryam Rashid
- Synthetic and Natural Product Drug Discovery Laboratory, Department of Chemistry, Government College Women University, Faisalabad, Pakistan
| | - Shagufta Parveen
- Synthetic and Natural Product Drug Discovery Laboratory, Department of Chemistry, Government College Women University, Faisalabad, Pakistan
- Department of Applied Chemistry, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
29
|
D'Souza SE, Khan K, Jalal K, Hassam M, Uddin R. The Gene Network Correlation Analysis of Obesity to Type 1 Diabetes and Cardiovascular Disorders: An Interactome-Based Bioinformatics Approach. Mol Biotechnol 2024; 66:2123-2143. [PMID: 37606877 DOI: 10.1007/s12033-023-00845-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/29/2023] [Indexed: 08/23/2023]
Abstract
The current study focuses on the importance of Protein-Protein Interactions (PPIs) in biological processes and the potential of targeting PPIs as a new treatment strategy for diseases. Specifically, the study explores the cross-links of PPIs network associated with obesity, type 1 diabetes mellitus (T1DM), and cardiac disease (CD), which is an unexplored area of research. The research aimed to understand the role of highly connected proteins in the network and their potential as drug targets. The methodology for this research involves retrieving genes from the NCBI online gene database, intersecting genes among three diseases (type 1 diabetes, obesity, and cardiovascular) using Interactivenn, determining suitable drug molecules using NetworkAnalyst, and performing various bioinformatics analyses such as Generic Protein-Protein Interactions, topological properties analysis, function enrichment analysis in terms of GO, and Kyoto Encyclopedia of Genes and Genomes (KEGG), gene co-expression network, and protein drug as well as protein chemical interaction network. The study focuses on human subjects. The results of this study identified 12 genes [VEGFA (Vascular Endothelial Growth Factor A), IL6 (Interleukin 6), MTHFR (Methylenetetrahydrofolate reductase), NPPB (Natriuretic Peptide B), RAC1 (Rac Family Small GTPase 1), LMNA (Lamin A/C), UGT1A1 (UDP-glucuronosyltransferase family 1 membrane A1), RETN (Resistin), GCG (Glucagon), NPPA (Natriuretic Peptide A), RYR2 (Ryanodine receptor 2), and PRKAG2 (Protein Kinase AMP-Activated Non-Catalytic Subunit Gamma 2)] that were shared across the three diseases and could be used as key proteins for protein-drug/chemical interaction. Additionally, the study provides an in-depth understanding of the complex molecular and biological relationships between the three diseases and the cellular mechanisms that lead to their development. Potentially significant implications for the therapy and management of various disorders are highlighted by the findings of this study by improving treatment efficacy, simplifying treatment regimens, cost-effectiveness, better understanding of the underlying mechanism of these diseases, early diagnosis, and introducing personalized medicine. In conclusion, the current study provides new insights into the cross-links of PPIs network associated with obesity, T1DM, and CD, and highlights the potential of targeting PPIs as a new treatment strategy for these prevalent diseases.
Collapse
Affiliation(s)
- Sharon Elaine D'Souza
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Lab 103 PCMD Ext., Karachi, 75270, Pakistan
| | - Kanwal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Lab 103 PCMD Ext., Karachi, 75270, Pakistan
| | - Khurshid Jalal
- HEJ Research Institute of Chemistry International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Muhammad Hassam
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Lab 103 PCMD Ext., Karachi, 75270, Pakistan
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Lab 103 PCMD Ext., Karachi, 75270, Pakistan.
| |
Collapse
|
30
|
Abisheva S, Rutskaya-Moroshan K, Nuranova G, Batyrkhan T, Abisheva A. Antimalarial Drugs at the Intersection of SARS-CoV-2 and Rheumatic Diseases: What Are the Potential Opportunities? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1171. [PMID: 39064600 PMCID: PMC11279047 DOI: 10.3390/medicina60071171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: The coronavirus disease of 2019 (COVID-19) pandemic has posed a serious threat to humanity and is considered a global health emergency. Antimalarial drugs (ADs) have been used in the treatment of immuno-inflammatory arthritis (IIA) and coronavirus infection (COVID-19). The aim of this review is to analyze the current knowledge about the immunomodulatory and antiviral mechanisms of action, characteristics of use, and side effects of antimalarial drugs. Material and Methods: A literature search was carried out using PubMed, MEDLINE, SCOPUS, and Google Scholar databases. The inclusion criteria were the results of randomized and cohort studies, meta-analyses, systematic reviews, and original full-text manuscripts in the English language containing statistically confirmed conclusions. The exclusion criteria were summary reports, newspaper articles, and personal messages. Qualitative methods were used for theoretical knowledge on antimalarial drug usage in AIRDs and SARS-CoV-2 such as a summarization of the literature and a comparison of the treatment methods. Results: The ADs were considered a "candidate" for the therapy of a new coronavirus infection due to mechanisms of antiviral activity, such as interactions with endocytic pathways, the prevention of glycosylation of the ACE2 receptors, blocking sialic acid receptors, and reducing the manifestations of cytokine storms. The majority of clinical trials suggest no role of antimalarial drugs in COVID-19 treatment or prevention. These circumstances do not allow for their use in the treatment and prevention of COVID-19. Conclusions: The mechanisms of hydroxychloroquine are related to potential cardiotoxic manifestations and demonstrate potential adverse effects when used for COVID-19. Furthermore, the need for high doses in the treatment of viral infections increases the likelihood of gastrointestinal side effects, the prolongation of QT, and retinopathy. Large randomized clinical trials (RCTs) have refuted the fact that there is a positive effect on the course and results of COVID-19.
Collapse
Affiliation(s)
- Saule Abisheva
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| | - Kristina Rutskaya-Moroshan
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| | - Gulnaz Nuranova
- Department of Children’s Diseases with Courses in Pulmonology and Nephrology, NJSC “Astana Medical University”, Astana 010000, Kazakhstan;
| | - Tansholpan Batyrkhan
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| | - Anilim Abisheva
- Department of Family Medicine №1, NJSC “Astana Medical University”, Astana 010000, Kazakhstan; (S.A.); (T.B.); (A.A.)
| |
Collapse
|
31
|
Kombo DC, Stepp JD, Lim S, Elshorst B, Li Y, Cato L, Shomali M, Fink D, LaMarche MJ. Predictions of Colloidal Molecular Aggregation Using AI/ML Models. ACS OMEGA 2024; 9:28691-28706. [PMID: 38973835 PMCID: PMC11223200 DOI: 10.1021/acsomega.4c02886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
To facilitate the triage of hits from small molecule screens, we have used various AI/ML techniques and experimentally observed data sets to build models aimed at predicting colloidal aggregation of small organic molecules in aqueous solution. We have found that Naïve Bayesian and deep neural networks outperform logistic regression, recursive partitioning tree, support vector machine, and random forest techniques by having the lowest balanced error rate (BER) for the test set. Derived predictive classification models consistently and successfully discriminated aggregator molecules from nonaggregator hits. An analysis of molecular descriptors in favor of colloidal aggregation confirms previous observations (hydrophobicity, molecular weight, and solubility) in addition to undescribed molecular descriptors such as the fraction of sp3 carbon atoms (Fsp3), and electrotopological state of hydroxyl groups (ES_Sum_sOH). Naïve Bayesian modeling and scaffold tree analysis have revealed chemical features/scaffolds contributing the most to colloidal aggregation and nonaggregation, respectively. These results highlight the importance of scaffolds with high Fsp3 values in promoting nonaggregation. Matched molecular pair analysis (MMPA) has also deciphered context-dependent substitutions, which can be used to design nonaggregator molecules. We found that most matched molecular pairs have a neutral effect on aggregation propensity. We have prospectively applied our predictive models to assist in chemical library triage for optimal plate selection diversity and purchase for high throughput screening (HTS) in drug discovery projects.
Collapse
Affiliation(s)
- David C. Kombo
- Integrated
Drug Discovery, Sanofi, 350 Water St., Cambridge, Massachusetts 02141, United States
| | - J. David Stepp
- Integrated
Drug Discovery, Sanofi, 350 Water St., Cambridge, Massachusetts 02141, United States
| | - Sungtaek Lim
- Integrated
Drug Discovery, Sanofi, 350 Water St., Cambridge, Massachusetts 02141, United States
| | - Bettina Elshorst
- CMC
Synthetics Early Development Analytics, Sanofi, Industriepark Hochst, Frankfurt 65926, Germany
| | - Yi Li
- Integrated
Drug Discovery, Sanofi, 350 Water St., Cambridge, Massachusetts 02141, United States
| | - Laura Cato
- Molecular
Oncology, Sanofi, 350
Water St., Cambridge, Massachusetts 02141, United States
| | - Maysoun Shomali
- Molecular
Oncology, Sanofi, 350
Water St., Cambridge, Massachusetts 02141, United States
| | - David Fink
- Integrated
Drug Discovery, Sanofi, 350 Water St., Cambridge, Massachusetts 02141, United States
| | - Matthew J. LaMarche
- Integrated
Drug Discovery, Sanofi, 350 Water St., Cambridge, Massachusetts 02141, United States
| |
Collapse
|
32
|
Hershan AA. Pathogenesis of COVID19 and the applications of US FDA-approved repurposed antiviral drugs to combat SARS-CoV-2 in Saudi Arabia: A recent update by review of literature. Saudi J Biol Sci 2024; 31:104023. [PMID: 38799719 PMCID: PMC11127266 DOI: 10.1016/j.sjbs.2024.104023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/05/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Still, there is no cure for the highly contagious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-caused coronavirus disease 2019 (COVID19). The COVID19 pandemic caused health emergencies which resulted in enormous medical and financial consequences worldwide including Saudi Arabia. Saudi Arabia is the largest Arab country of the Middle East. The urban setting of Saudi Arabia makes it vulnerable towards SARS-CoV-2 (SCV-2). Religious areas of this country are visited by millions of pilgrims every year for the Umrah and Hajj pilgrimage, which contributes to the potential COVID19 epidemic risk. COVID19 throws various challenges to healthcare professionals to choose the right drugs or therapy in clinical settings because of the lack of availability of newer drugs. Current drug development and discovery is an expensive, complex, and long process, which involves a high failure rate in clinical trials. While repurposing of United States Food and Drug Administration (US FDA)-approved antiviral drugs offers numerous benefits including complete pharmacokinetic and safety profiles, which significantly shorten drug development cycles and reduce costs. A range of repurposed US FDA-approved antiviral drugs including ribavirin, lopinavir/ritonavir combination, oseltamivir, darunavir, remdesivir, nirmatrelvir/ritonavir combination, and molnupiravir showed encouraging results in clinical trials in COVID19 treatment. In this article, several COVID19-related discussions have been provided including emerging variants of concern of, COVID19 pathogenesis, COVID19 pandemic scenario in Saudi Arabia, drug repurposing strategies against SCV-2, as well as repurposing of US FDA-approved antiviral drugs that might be considered to combat SCV-2 in Saudi Arabia. Moreover, drug repurposing in the context of COVID19 management along with its limitations and future perspectives have been summarized.
Collapse
Affiliation(s)
- Almonther Abdullah Hershan
- The University of Jeddah, College of Medicine, Department of Medical microbiology and parasitology, Jeddah, Saudi Arabia
| |
Collapse
|
33
|
Gharizadeh A, Abbasi K, Ghareyazi A, Mofrad MRK, Rabiee HR. HGTDR: Advancing drug repurposing with heterogeneous graph transformers. Bioinformatics 2024; 40:btae349. [PMID: 38913860 PMCID: PMC11223801 DOI: 10.1093/bioinformatics/btae349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 04/20/2024] [Accepted: 06/23/2024] [Indexed: 06/26/2024] Open
Abstract
MOTIVATION Drug repurposing is a viable solution for reducing the time and cost associated with drug development. However, thus far, the proposed drug repurposing approaches still need to meet expectations. Therefore, it is crucial to offer a systematic approach for drug repurposing to achieve cost savings and enhance human lives. In recent years, using biological network-based methods for drug repurposing has generated promising results. Nevertheless, these methods have limitations. Primarily, the scope of these methods is generally limited concerning the size and variety of data they can effectively handle. Another issue arises from the treatment of heterogeneous data, which needs to be addressed or converted into homogeneous data, leading to a loss of information. A significant drawback is that most of these approaches lack end-to-end functionality, necessitating manual implementation and expert knowledge in certain stages. RESULTS We propose a new solution, Heterogeneous Graph Transformer for Drug Repurposing (HGTDR), to address the challenges associated with drug repurposing. HGTDR is a three-step approach for knowledge graph-based drug repurposing: (1) constructing a heterogeneous knowledge graph, (2) utilizing a heterogeneous graph transformer network, and (3) computing relationship scores using a fully connected network. By leveraging HGTDR, users gain the ability to manipulate input graphs, extract information from diverse entities, and obtain their desired output. In the evaluation step, we demonstrate that HGTDR performs comparably to previous methods. Furthermore, we review medical studies to validate our method's top 10 drug repurposing suggestions, which have exhibited promising results. We also demonstrated HGTDR's capability to predict other types of relations through numerical and experimental validation, such as drug-protein and disease-protein inter-relations. AVAILABILITY AND IMPLEMENTATION The source code and data are available at https://github.com/bcb-sut/HGTDR and http://git.dml.ir/BCB/HGTDR.
Collapse
Affiliation(s)
- Ali Gharizadeh
- Department of Computer Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9517, Iran
| | - Karim Abbasi
- Department of Computer Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9517, Iran
| | - Amin Ghareyazi
- Department of Computer Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9517, Iran
| | - Mohammad R K Mofrad
- Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, P.O. Box 94720-1740, United States
| | - Hamid R Rabiee
- Department of Computer Engineering, Sharif University of Technology, Tehran, P.O. Box 11155-9517, Iran
| |
Collapse
|
34
|
Abbaspour Kasgari H, Moradi S, Alikhani A, Ahmadian N. Effectiveness of dolutegravir in moderate severity COVID-19 patients: A single-center, randomized, double-blind, placebo-controlled trial. BIOIMPACTS : BI 2024; 15:29952. [PMID: 40161930 PMCID: PMC11954744 DOI: 10.34172/bi.29952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 03/18/2025]
Abstract
INTRODUCTION Drug repurposing as a low-cost, time-saving, and often less risky strategy has been attractive for the treatment of coronavirus disease 2019 (COVID-19) during the pandemic. This trial aimed to evaluate the effectiveness of dolutegravir, an HIV-1 integrase inhibitor, in admitted patients with moderate COVID-19. METHODS This study was a randomized, double-blind, placebo-controlled clinical trial assessing the efficacy of dolutegravir in adults admitted to a hospital in Ghaemshahr, Mazandaran Province, Iran. Patients aged 18-80 years with early symptoms of moderate COVID-19, which was confirmed based on reverse transcription polymerase chain reaction (RT-PCR) and/or chest computed tomography (CT) scan, were considered to be included in this study. Patients were randomly assigned in a 1:1 ratio to receive 50 mg dolutegravir plus the standard treatment regimen or the same value of placebo plus the standard treatment regimen, daily for 7 days. The standard treatment regimen was remdesivir 200 mg on day 1 followed by 100 mg for five days or until discharge. The primary endpoint was recovery 10 days after the beginning of the study. RESULTS Between August 22 and October 23, 2021, of 120 patients who were enrolled, 93 patients were randomly assigned to receive 50 mg dolutegravir (n=46) or the placebo regimen (n=47). No significant difference was observed between the two intervention groups based on the obtained results including frequency of respiratory modes during the first five days of admission, respiratory rate, and O2 saturation during six time periods. CONCLUSION The results showed that in adult patients admitted to the hospital with moderate COVID-19, treatment with dolutegravir was not associated with improvement in clinical recovery. Larger randomized trials are required to provide more robust evidence about the effectiveness of dolutegravir.
Collapse
Affiliation(s)
- Hamideh Abbaspour Kasgari
- Department of Clinical Pharmacy, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Siavash Moradi
- Education Development Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Alikhani
- Antimicrobial Resistance Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasim Ahmadian
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
35
|
Ma MT, Jiang Q, Chen CH, Badeti S, Wang X, Zeng C, Evans D, Bodnar B, Marras SAE, Tyagi S, Bharaj P, Yehia G, Romanienko P, Hu W, Liu SL, Shi L, Liu D. S309-CAR-NK cells bind the Omicron variants in vitro and reduce SARS-CoV-2 viral loads in humanized ACE2-NSG mice. J Virol 2024; 98:e0003824. [PMID: 38767356 PMCID: PMC11237809 DOI: 10.1128/jvi.00038-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/11/2024] [Indexed: 05/22/2024] Open
Abstract
Recent progress on chimeric antigen receptor (CAR)-NK cells has shown promising results in treating CD19-positive lymphoid tumors with minimal toxicities [including graft versus host disease (GvHD) and cytokine release syndrome (CRS) in clinical trials. Nevertheless, the use of CAR-NK cells in combating viral infections has not yet been fully explored. Previous studies have shown that CAR-NK cells expressing S309 single-chain fragment variable (scFv), hereinafter S309-CAR-NK cells, can bind to SARS-CoV-2 wildtype pseudotyped virus (PV) and effectively kill cells expressing wild-type spike protein in vitro. In this study, we further demonstrate that the S309-CAR-NK cells can bind to different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants in vitro. We also show that S309-CAR-NK cells reduce virus loads in the NOD/SCID gamma (NSG) mice expressing the human angiotensin-converting enzyme 2 (hACE2) receptor challenged with SARS-CoV-2 wild-type (strain USA/WA1/2020). Our study demonstrates the potential use of S309-CAR-NK cells for inhibiting infection by SARS-CoV-2 and for the potential treatment of COVID-19 patients unresponsive to otherwise currently available therapeutics. IMPORTANCE Chimeric antigen receptor (CAR)-NK cells can be "off-the-shelf" products that treat various diseases, including cancer, infections, and autoimmune diseases. In this study, we engineered natural killer (NK) cells to express S309 single-chain fragment variable (scFv), to target the Spike protein of SARS-CoV-2, hereinafter S309-CAR-NK cells. Our study shows that S309-CAR-NK cells are effective against different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants. The S309-CAR-NK cells can (i) directly bind to SARS-CoV-2 pseudotyped virus (PV), (ii) competitively bind to SARS-CoV-2 PV with 293T cells expressing the human angiotensin-converting enzyme 2 (hACE2) receptor (293T-hACE2 cells), (iii) specifically target and lyse A549 cells expressing the spike protein, and (iv) significantly reduce the viral loads of SARS-CoV-2 wild-type (strain USA/WA1/2020) in the lungs of NOD/SCID gamma (NSG) mice expressing hACE2 (hACE2-NSG mice). Altogether, the current study demonstrates the potential use of S309-CAR-NK immunotherapy as an alternative treatment for COVID-19 patients.
Collapse
Affiliation(s)
- Minh Tuyet Ma
- Department of Pathology, Immunology, and Laboratory Medicine, South Orange Avenue, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, USA
| | - Qingkui Jiang
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Chih-Hsiung Chen
- Department of Pathology, Immunology, and Laboratory Medicine, South Orange Avenue, Newark, New Jersey, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, USA
| | - Saiaditya Badeti
- Department of Pathology, Immunology, and Laboratory Medicine, South Orange Avenue, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, USA
| | - Xuening Wang
- Department of Pathology, Immunology, and Laboratory Medicine, South Orange Avenue, Newark, New Jersey, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, USA
| | - Cong Zeng
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, Ohio, USA
| | - Deborah Evans
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Brittany Bodnar
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Salvatore A E Marras
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Sanjay Tyagi
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Preeti Bharaj
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Ghassan Yehia
- Genome Editing Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Peter Romanienko
- Genome Editing Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Wenhui Hu
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shan-Lu Liu
- Center for Retrovirus Research, The Ohio State University, Columbus, Ohio, USA
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Lanbo Shi
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Dongfang Liu
- Department of Pathology, Immunology, and Laboratory Medicine, South Orange Avenue, Newark, New Jersey, USA
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, New Jersey, USA
| |
Collapse
|
36
|
Rohweder R, Pereira NG, Micheletti BH, Mosello J, Campos JRM, Pereira MG, Santos CN, Simões NL, Matielo RLB, Bernardes LS, Oppermann MLR, Wender MCO, Lupattelli A, Nordeng H, Schuler-Faccini L. Medication Use Among Pregnant Women With SARS-CoV-2 Infection and Risk of Hospitalization-A Study in Two Brazilian Hospitals. J Pregnancy 2024; 2024:8915166. [PMID: 39021875 PMCID: PMC11254464 DOI: 10.1155/2024/8915166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 07/20/2024] Open
Abstract
There is limited evidence about the use of medications among pregnant women with COVID-19, as well as risk factors for hospitalization due to COVID-19 in pregnancy. We aimed to describe the use of medications among SARS-CoV-2-positive pregnant women at the time around infection and identify predictors for hospitalization due to COVID-19 in two hospitals in Brazil. This is a hospital record-based study among pregnant women with positive SARS-CoV-2 tests between March 2020 and August 2022 from two Brazilian hospitals. Characteristics of sociodemographic, obstetrical, and COVID-19 symptoms were extracted retrospectively. The prevalence use of medications was based on self-reported use, and this was administered at the hospital. Logistic regression was used to estimate predictors of hospitalization due to COVID-19. There were 278 pregnant women included in the study, of which 41 (14.7%) required hospitalization due to COVID-19. The remaining 237 (85.3%) had mild symptoms or were asymptomatic. Most of the women had the infection in the third trimester (n = 149; 53.6%). The most prevalent medications used across all trimesters were analgesics (2.4% to 20.0%), antibacterials (15.0% to 23.1%), and corticosteroids (7.2% to 10.4%). Pre- or gestational hypertensive disorder (odds ratio (OR) 4.94, 95% confidence interval (CI) 1.65, 14.87) and having at least one dose of vaccine against SARS-CoV-2 (OR 0.13, 95% CI 0.04, 0.39) were associated with hospitalization due to COVID-19. Analgesics, antibacterials, and corticosteroids were the most frequently used medications among pregnant women with COVID-19. Women with hypertensive disorders have almost a five-fold increased risk of hospitalization due to COVID-19. Vaccination was the strongest protective factor for severe COVID-19. The COVID-19 vaccination among pregnant women should be promoted, and pregnant women diagnosed with COVID-19 who have hypertensive disorders should be closely monitored.
Collapse
Affiliation(s)
- Ricardo Rohweder
- Graduate Program in Genetics and Molecular BiologyDepartment of GeneticsUniversidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Natálya G. Pereira
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Bruna H. Micheletti
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Jéssica Mosello
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Júlia R. M. Campos
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Matheus G. Pereira
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Cristina N. Santos
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Natália L. Simões
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Regina L. B. Matielo
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
| | - Lisandra S. Bernardes
- Department of Obstetrics and NeonatologyHospital e Maternidade SEPACO, São Paulo, Brazil
- Center for Klinisk Forskning and Afdeling for Kvindesygdomme, Graviditet og FødselNorth Denmark Regional Hospital, Hjørring, Denmark
| | - Maria L. R. Oppermann
- Gynecology and Obstetrics ServiceHospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Maria C. O. Wender
- Gynecology and Obstetrics ServiceHospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Angela Lupattelli
- PharmacoEpidemiology and Drug Safety Research GroupDepartment of PharmacyUniversity of Oslo, Oslo, Norway
| | - Hedvig Nordeng
- PharmacoEpidemiology and Drug Safety Research GroupDepartment of PharmacyUniversity of Oslo, Oslo, Norway
| | - Lavinia Schuler-Faccini
- Graduate Program in Genetics and Molecular BiologyDepartment of GeneticsUniversidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
37
|
Mizuno Y, Nakasone W, Nakamura M, Otaki JM. In Silico and In Vitro Evaluation of the Molecular Mimicry of the SARS-CoV-2 Spike Protein by Common Short Constituent Sequences (cSCSs) in the Human Proteome: Toward Safer Epitope Design for Vaccine Development. Vaccines (Basel) 2024; 12:539. [PMID: 38793790 PMCID: PMC11125730 DOI: 10.3390/vaccines12050539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/12/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Spike protein sequences in SARS-CoV-2 have been employed for vaccine epitopes, but many short constituent sequences (SCSs) in the spike protein are present in the human proteome, suggesting that some anti-spike antibodies induced by infection or vaccination may be autoantibodies against human proteins. To evaluate this possibility of "molecular mimicry" in silico and in vitro, we exhaustively identified common SCSs (cSCSs) found both in spike and human proteins bioinformatically. The commonality of SCSs between the two systems seemed to be coincidental, and only some cSCSs were likely to be relevant to potential self-epitopes based on three-dimensional information. Among three antibodies raised against cSCS-containing spike peptides, only the antibody against EPLDVL showed high affinity for the spike protein and reacted with an EPLDVL-containing peptide from the human unc-80 homolog protein. Western blot analysis revealed that this antibody also reacted with several human proteins expressed mainly in the small intestine, ovary, and stomach. Taken together, these results showed that most cSCSs are likely incapable of inducing autoantibodies but that at least EPLDVL functions as a self-epitope, suggesting a serious possibility of infection-induced or vaccine-induced autoantibodies in humans. High-risk cSCSs, including EPLDVL, should be excluded from vaccine epitopes to prevent potential autoimmune disorders.
Collapse
Affiliation(s)
- Yuya Mizuno
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru, Nishihara 903-0213, Okinawa, Japan
| | - Wataru Nakasone
- Computer Science and Intelligent Systems Unit, Department of Engineering, Faculty of Engineering, University of the Ryukyus, Senbaru, Nishihara 903-0213, Okinawa, Japan
| | - Morikazu Nakamura
- Computer Science and Intelligent Systems Unit, Department of Engineering, Faculty of Engineering, University of the Ryukyus, Senbaru, Nishihara 903-0213, Okinawa, Japan
| | - Joji M. Otaki
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru, Nishihara 903-0213, Okinawa, Japan
| |
Collapse
|
38
|
Mekonen ZT, Fenta TG, Nadeem SP, Cho DJ. Global Health Commodities Supply Chain in the Era of COVID-19 Pandemic: Challenges, Impacts, and Prospects: A Systematic Review. J Multidiscip Healthc 2024; 17:1523-1539. [PMID: 38623396 PMCID: PMC11018129 DOI: 10.2147/jmdh.s448654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/04/2024] [Indexed: 04/17/2024] Open
Abstract
Background The COVID-19 pandemic led to the most substantial health crisis in the 21st Century. This pandemic interrupted the supply of essential commodities for human beings. Among the essential commodities for human survival, disruption of the supply of essential health commodities has become a global concern. Objective The study aimed to systematically analyze published articles on the challenges, impacts, and prospects of the global health commodities' supply chain in the era of the COVID-19 pandemic. Methods A standard searching strategy was conducted in seven research databases to retrieve pertinent articles. Finally, 459 articles were retrieved for further screening, and only 13 articles were selected for final synthesis. Results Almost 38.5% of the studies targeted the supply chain of health commodities used to treat HIV, TB, and malaria. Lockdown policies, travel restrictions, lack of transportation, low manufacturing capacity, and rising costs were the significant challenges indicated for the supply interruption of essential health commodities and COVID-19 vaccines. Findings indicated that the supply interruption of essential health commodities leads to a devastating impact on global health. Conclusion Global medicine shortages due to the pandemic crisis can have a devastatingly harmful impact on patient outcomes and might result in a devastatingly long-lasting effect on the health of the world community. Supply-related challenges of the COVID-19 vaccine affect countries' ambitions for achieving herd immunity quickly. Monitoring the pandemic's effect on the health commodities' supply system and designing a short-term and long-term resilient health supply chain system that can cope with current and future health catastrophes is pivotal.
Collapse
Affiliation(s)
- Zelalem Tilahun Mekonen
- Department of Pharmaceutics and Social Pharmacy, School of Pharmacy, Addis Ababa University, Addis Ababa, Ethiopia
| | - Teferi Gedif Fenta
- Department of Pharmaceutics and Social Pharmacy, School of Pharmacy, Addis Ababa University, Addis Ababa, Ethiopia
| | | | - Denny J Cho
- Logistics Department, Kyrgyz State Technical University, Bishkek, Kyrgyzstan
| |
Collapse
|
39
|
Rani N, Kaushik A, Kardam S, Kag S, Raj VS, Ambasta RK, Kumar P. Reimagining old drugs with new tricks: Mechanisms, strategies and notable success stories in drug repurposing for neurological diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:23-70. [PMID: 38789181 DOI: 10.1016/bs.pmbts.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Recent evolution in drug repurposing has brought new anticipation, especially in the conflict against neurodegenerative diseases (NDDs). The traditional approach to developing novel drugs for these complex disorders is laborious, time-consuming, and often abortive. However, drug reprofiling which is the implementation of illuminating novel therapeutic applications of existing approved drugs, has shown potential as a promising strategy to accelerate the hunt for therapeutics. The advancement of computational approaches and artificial intelligence has expedited drug repurposing. These progressive technologies have enabled scientists to analyse extensive datasets and predict potential drug-disease interactions. By prospecting into the existing pharmacological knowledge, scientists can recognise potential therapeutic candidates for reprofiling, saving precious time and resources. Preclinical models have also played a pivotal role in this field, confirming the effectiveness and mechanisms of action of repurposed drugs. Several studies have occurred in recent years, including the discovery of available drugs that demonstrate significant protective effects in NDDs, relieve debilitating symptoms, or slow down the progression of the disease. These findings highlight the potential of repurposed drugs to change the landscape of NDD treatment. Here, we present an overview of recent developments and major advances in drug repurposing intending to provide an in-depth analysis of traditional drug discovery and the strategies, approaches and technologies that have contributed to drug repositioning. In addition, this chapter attempts to highlight successful case studies of drug repositioning in various therapeutic areas related to NDDs and explore the clinical trials, challenges and limitations faced by researchers in the field. Finally, the importance of drug repositioning in drug discovery and development and its potential to address discontented medical needs is also highlighted.
Collapse
Affiliation(s)
- Neetu Rani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Aastha Kaushik
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Shefali Kardam
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Sonika Kag
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - V Samuel Raj
- Department of Biotechnology and Microbiology, SRM University, Sonepat, Haryana, India
| | - Rashmi K Ambasta
- Department of Biotechnology and Microbiology, SRM University, Sonepat, Haryana, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India.
| |
Collapse
|
40
|
Sertbas M, Ulgen KO. Uncovering the Effect of SARS-CoV-2 on Liver Metabolism via Genome-Scale Metabolic Modeling for Reprogramming and Therapeutic Strategies. ACS OMEGA 2024; 9:15535-15546. [PMID: 38585079 PMCID: PMC10993323 DOI: 10.1021/acsomega.4c00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 04/09/2024]
Abstract
Genome-scale metabolic models (GEMs) are promising computational tools that contribute to elucidating host-virus interactions at the system level and developing therapeutic strategies against viral infection. In this study, the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on liver metabolism was investigated using integrated GEMs of human hepatocytes and SARS-CoV-2. They were generated for uninfected and infected hepatocytes using transcriptome data. Reporter metabolite analysis resulted in significant transcriptional changes around several metabolites involved in xenobiotics, drugs, arachidonic acid, and leukotriene metabolisms due to SARS-CoV-2 infection. Flux balance analysis and minimization of metabolic adjustment approaches unraveled possible virus-induced hepatocellular reprogramming in fatty acid, glycerophospholipid, sphingolipid cholesterol, and folate metabolisms, bile acid biosynthesis, and carnitine shuttle among others. Reaction knockout analysis provided critical reactions in glycolysis, oxidative phosphorylation, purine metabolism, and reactive oxygen species detoxification subsystems. Computational analysis also showed that administration of dopamine, glucosamine, D-xylose, cysteine, and (R)-3-hydroxybutanoate contributes to alleviating viral infection. In essence, the reconstructed host-virus GEM helps us understand metabolic programming and develop therapeutic strategies to battle SARS-CoV-2.
Collapse
Affiliation(s)
- Mustafa Sertbas
- Department of Chemical Engineering, Bogazici University, 34342 Istanbul, Turkey
| | - Kutlu O. Ulgen
- Department of Chemical Engineering, Bogazici University, 34342 Istanbul, Turkey
| |
Collapse
|
41
|
Brady DK, Gurijala AR, Huang L, Hussain AA, Lingan AL, Pembridge OG, Ratangee BA, Sealy TT, Vallone KT, Clements TP. A guide to COVID-19 antiviral therapeutics: a summary and perspective of the antiviral weapons against SARS-CoV-2 infection. FEBS J 2024; 291:1632-1662. [PMID: 36266238 PMCID: PMC9874604 DOI: 10.1111/febs.16662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/11/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Antiviral therapies are integral in the fight against SARS-CoV-2 (i.e. severe acute respiratory syndrome coronavirus 2), the causative agent of COVID-19. Antiviral therapeutics can be divided into categories based on how they combat the virus, including viral entry into the host cell, viral replication, protein trafficking, post-translational processing, and immune response regulation. Drugs that target how the virus enters the cell include: Evusheld, REGEN-COV, bamlanivimab and etesevimab, bebtelovimab, sotrovimab, Arbidol, nitazoxanide, and chloroquine. Drugs that prevent the virus from replicating include: Paxlovid, remdesivir, molnupiravir, favipiravir, ribavirin, and Kaletra. Drugs that interfere with protein trafficking and post-translational processing include nitazoxanide and ivermectin. Lastly, drugs that target immune response regulation include interferons and the use of anti-inflammatory drugs such as dexamethasone. Antiviral therapies offer an alternative solution for those unable or unwilling to be vaccinated and are a vital weapon in the battle against the global pandemic. Learning more about these therapies helps raise awareness in the general population about the options available to them with respect to aiding in the reduction of the severity of COVID-19 infection. In this 'A Guide To' article, we provide an in-depth insight into the development of antiviral therapeutics against SARS-CoV-2 and their ability to help fight COVID-19.
Collapse
Affiliation(s)
- Drugan K. Brady
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Aashi R. Gurijala
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Liyu Huang
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Ali A. Hussain
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Audrey L. Lingan
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | | | - Brina A. Ratangee
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Tristan T. Sealy
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Kyle T. Vallone
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | | |
Collapse
|
42
|
Giri-Rachman EA, Effendy VV, Azmi MHS, Yamahoki N, Stephanie R, Agustiyanti DF, Wisnuwardhani PH, Angelina M, Rubiyana Y, Aditama R, Ningrum RA, Wardiana A, Fibriani A. The SARS-CoV-2 M pro Dimer-Based Screening System: A Synthetic Biology Tool for Identifying Compounds with Dimerization Inhibitory Potential. ACS Synth Biol 2024; 13:509-520. [PMID: 38316139 PMCID: PMC10877612 DOI: 10.1021/acssynbio.3c00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 02/07/2024]
Abstract
The COVID-19 endemic remains a global concern. The search for effective antiviral candidates is still needed to reduce disease risk. However, the availability of high biosafety level laboratory facilities for drug screening is limited in number. To address this issue, a screening system that could be utilized at lower biosafety levels remains essential. This study aimed to develop a novel SARS-CoV-2 main protease (Mpro) dimer-based screening system (DBSS) utilizing synthetic biology in Escherichia coli BL21(DE3). We linked the SARS-CoV-2 Mpro with the DNA-binding domain of AraC regulatory protein, which regulates the reporter gene expression. Protein modeling and molecular docking showed that saquinavir could bind to AraC-Mpro both in its monomer and dimer forms. The constructed DBSS assay indicated the screening system could detect saquinavir inhibitory activity at a concentration range of 4-10 μg/mL compared to the untreated control (P ≤ 0.05). The Vero E6 cell assay validated the DBSS result that saquinavir at 4-10 μg/mL exhibited antiviral activity against SARS-CoV-2. Our DBSS could be used for preliminary screening of numerous drug candidates that possess a dimerization inhibitor activity of SARS-CoV-2 Mpro and also minimize the use of a high biosafety level laboratory.
Collapse
Affiliation(s)
| | - Vergio V. Effendy
- School
of Life Sciences and Technology, Institut
Teknologi Bandung, Bandung 40132, Indonesia
| | - Muhammad H. S. Azmi
- School
of Life Sciences and Technology, Institut
Teknologi Bandung, Bandung 40132, Indonesia
| | - Nicholas Yamahoki
- School
of Life Sciences and Technology, Institut
Teknologi Bandung, Bandung 40132, Indonesia
| | - Rebecca Stephanie
- School
of Life Sciences and Technology, Institut
Teknologi Bandung, Bandung 40132, Indonesia
| | - Dian F. Agustiyanti
- Research
Center for Genetic Engineering, Indonesian
National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Popi H. Wisnuwardhani
- Research
Center for Genetic Engineering, Indonesian
National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Marissa Angelina
- Research
Center for Pharmaceutical Ingredients and Traditional Medicine, Indonesian National Research and Innovation Agency
(BRIN), Serpong 15314, Indonesia
| | - Yana Rubiyana
- Research
Center for Genetic Engineering, Indonesian
National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Reza Aditama
- Biochemistry
Research Group, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Bandung 40132, Indonesia
| | - Ratih A. Ningrum
- Research
Center for Genetic Engineering, Indonesian
National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Andri Wardiana
- Research
Center for Genetic Engineering, Indonesian
National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Azzania Fibriani
- School
of Life Sciences and Technology, Institut
Teknologi Bandung, Bandung 40132, Indonesia
| |
Collapse
|
43
|
Saini R, Kumar V, Patel CN, Sourirajan A, Dev K. Synergistic antibacterial activity of Phyllanthus emblica fruits and its phytocompounds with ampicillin: a computational and experimental study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:857-871. [PMID: 37522914 DOI: 10.1007/s00210-023-02624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Phyllanthus emblica L. (syn. Emblica officinalis), popularly known as amla, Indian gooseberry, or the King of Rasyana, is a member of Phyllanthaceae family and is traditionally used in Ayurveda as an immunity booster. The present study aimed to investigate the synergistic interaction of Phyllanthus emblica (FPE) fruits and its selected phytocompounds with ampicillin against selected bacteria. Further, an in silico technique was used to find if major phytocompounds of FPE could bind to proteins responsible for antibiotic resistance in bacterial pathogens and enhance the bioactivity of ampicillin. FPE and all the selected phytocompounds were found to have synergistic antibacterial activity with ampicillin against tested bacteria in different combinations. However, ellagic acid and quercetin interactions with ampicillin resulted in maximum bioactivity enhancement of 32-128 folds and 16-277 folds, respectively. In silico analysis revealed strong ellagic acid, quercetin, and rutin binding with penicillin-binding protein (PBP-) 3, further supported by MD simulations. Ellagic acid and quercetin also fulfill Lipinski's rule, showing similar toxicity characteristics to ampicillin. FPE showed synergistic interaction with ampicillin, possibly due to the presence of phytocompounds such as gallic acid, ellagic acid, quercetin, and rutin. Molecular docking and MD simulations showed the strong interaction of ellagic acid and quercetin with PBP-3 protein. Therefore, these compounds can be explored as potential non-toxic drug candidates to combat bacterial antimicrobial resistance.
Collapse
Affiliation(s)
- Rakshandha Saini
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, PO Sultanpur, Distt. Solan-173229 HP, Bajhol, India
| | - Vikas Kumar
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab, 140413, India.
| | - Chirag N Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, University School of Science, Gujarat University, Ahmedabad, Gujarat, 380009, India
- Biotechnology Research Center, Technology Innovation Institute, Masdar, Abu Dhabi, 9639, United Arab Emirates
| | - Anuradha Sourirajan
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, PO Sultanpur, Distt. Solan-173229 HP, Bajhol, India
| | - Kamal Dev
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, PO Sultanpur, Distt. Solan-173229 HP, Bajhol, India.
- Department of Pharmacology and Toxicology, Wright State University, Dayton, OH, 45435, USA.
| |
Collapse
|
44
|
Saha T, Lyons N, Yue Yung DB, Quiñones-Mateu ME, Pletzer D, Das SC. Repurposing ebselen as an inhalable dry powder to treat respiratory tract infections. Eur J Pharm Biopharm 2024; 195:114170. [PMID: 38128743 DOI: 10.1016/j.ejpb.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 12/23/2023]
Abstract
Respiratory tract infections (RTIs) are one of the leading causes of death globally, lately exacerbated by the increasing prevalence of antimicrobial resistance. While antimicrobial resistance could be overcome by developing new antimicrobial agents, the use of a safe repurposed agent having potent antimicrobial activity against various RTIs can be an efficient and cost-effective alternative to overcome the long and complex process of developing and testing new drugs. Ebselen, a synthetic organoselenium drug originally developed to treat noise-inducing hearing problems, has shown promising antimicrobial activity in vitro against several respiratory pathogens including viruses (e.g., SARS-CoV-2, influenza A virus) and bacteria (e.g., Mycobacterium tuberculosis, Streptococcus pneumoniae, and Staphylococcus aureus). Inhaled drug delivery is considered a promising approach for treating RTIs, as it can ensure effective drug concentrations at a lower dose, thereby minimizing the side effects that are often encountered by using oral or injectable drugs. In this study, we developed inhalable ebselen dry powder formulations using a spray-drying technique. The amino acids leucine, methionine, and tryptophan were incorporated with ebselen to enhance the yield and aerosolization of the dry powders. The amino acid-containing ebselen dry powders showed a better yield (37-56.4 %) than the amino acid-free formulation (30.9 %). All dry powders were crystalline in nature. The mass median aerodynamic diameter (MMAD) was less than 5 µm for amino acids containing dry powders (3-4 µm) and slightly higher (5.4 µm) for amino acid free dry powder indicating their suitability for inhalation. The aerosol performance was higher when amino acids were used, and the leucine-containing ebselen dry powder showed the highest emitted dose (84 %) and fine particle fraction (68 %). All amino acid formulations had similar cytotoxicity as raw ebselen, tested in respiratory cell line (A549 cells), with half-maximal inhibitory concentrations (IC50) between 100 and 250 μg/mL. Raw ebselen and amino acid-containing dry powders showed similar potent antibacterial activity against the Gram-positive bacteria S. aureus and S. pneumoniae with minimum inhibitory concentrations of 0.31 μg/mL and 0.16 μg/mL, respectively. On the other hand, raw ebselen and the formulations showed limited antimicrobial activity against the Gram-negative pathogens Pseudomonas aeruginosa and Klebsiella pneumoniae. In summary, in this study we were able to develop amino-acid-containing inhalable dry powders of ebselen that could be used against different respiratory pathogens, especially Gram-positive bacteria, which could ensure more drug deposition in the respiratory tract, including the lungs. DPIs are generally used to treat lung (lower respiratory tract) diseases. However, DPIs can also be used to treat both upper and lower RTIs. The deposition of the dry powder in the respiratory tract is dependent on its physicochemical properties and this properties can be modulated to target the intended site of infection (upper and/or lower respiratory tract). Further studies will allow the development of similar formulations of individual and/or combination of antimicrobials that could be used to inhibit a number of respiratory pathogens.
Collapse
Affiliation(s)
- Tushar Saha
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Nikita Lyons
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Deborah Bow Yue Yung
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Miguel E Quiñones-Mateu
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
45
|
Waseem W, Zafar R, Jan MS, Alomar TS, Almasoud N, Rauf A, Khattak H. Drug repurposing of FDA-approved anti-viral drugs via computational screening against novel 6M03 SARS-COVID-19. Ir J Med Sci 2024; 193:73-83. [PMID: 37515684 DOI: 10.1007/s11845-023-03473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE The COVID-19 pandemic has been recognized as severe acute respiratory syndrome, one of the worst and disastrous infectious diseases in human history. Until now, there is no cure to this contagious infection although some multinational pharmaceutical companies have synthesized the vaccines and injecting them into humans, but a drug treatment regimen is yet to come. AIM Among the multiple areas of SARS-CoV-2 that can be targeted, protease protein has significant values due to its essential role in viral replication and life. The repurposing of FDA-approved drugs for the treatment of COVID-19 has been a critical strategy during the pandemic due to the urgency of effective therapies. The novelty in this work refers to the innovative use of existing drugs with greater safety, speed, cost-effectiveness, broad availability, and diversity in the mechanism of action that have been approved and developed for other medical conditions. METHODS In this research work, we have engaged drug reprofiling or drug repurposing to recognize possible inhibitors of protease protein 6M03 in an instantaneous approach through computational docking studies. RESULTS We screened 16 FDA-approved anti-viral drugs that were known for different viral infections to be tested against this contagious novel strain. Through these reprofiling studies, we come up with 5 drugs, namely, Delavirdine, Fosamprenavir, Imiquimod, Stavudine, and Zanamivir, showing excellent results with the negative binding energies in Kcal/mol as - 8.5, - 7.0, - 6.8, - 6.8, and - 6.6, respectively, in the best binding posture. In silico studies allowed us to demonstrate the potential role of these drugs against COVID-19. CONCLUSION In our study, we also observed the nucleotide sequence of protease protein consisting of 316 amino acid residues and the influence of these pronouncing drugs over these sequences. The outcome of this research work provides researchers with a track record for carrying out further investigational procedures by applying docking simulations and in vitro and in vivo experimentation with these reprofile drugs so that a better drug can be formulated against coronavirus.
Collapse
Affiliation(s)
- Wajeeha Waseem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, 54000, Pakistan
| | - Rehman Zafar
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Muhammad Saeed Jan
- Department of Pharmacy, Bacha Khan University Charsadda, Charsadda, 24420, KP, Pakistan.
| | - Taghrid S Alomar
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84427, 11671, Riyadh, Saudi Arabia.
| | - Najla Almasoud
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84427, 11671, Riyadh, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, 23430, Anbar, Pakistan.
| | - Humayoon Khattak
- Department of Pharmacy, Bacha Khan University Charsadda, Charsadda, 24420, KP, Pakistan
| |
Collapse
|
46
|
Li Y, Ma H. Drug repurposing: insights into the antimicrobial effects of AKBA against MRSA. AMB Express 2024; 14:5. [PMID: 38184513 PMCID: PMC10771487 DOI: 10.1186/s13568-024-01660-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024] Open
Abstract
Staphylococcus aureus is a major threat in infectious diseases due to its varied infection types and increased resistance. S. aureus could form persister cells under certain condition and could also attach on medical apparatus to form biofilms, which exhibited extremely high resistance to antibiotics. 3-Acetyl-11-keto-beta-boswellic acid (AKBA) is a well-studied anti-tumor and antioxidant drug. This study is aimed to determine the antimicrobial effects of AKBA against S. aureus and its persister cells and biofilms. The in vitro antimicrobial susceptibility of AKBA was assessed by micro-dilution assay, disc diffusion assay and time-killing assay. Drug combination between AKBA and conventional antibiotics was detected by checkerboard assay. And the antibiofilm effects of AKBA against S. aureus were explored by crystal violet staining combined with SYTO/PI probes staining. Next, RBC lysis activity and CCK-8 kit were used to determine the cytotoxicity of AKBA. In addition, murine subcutaneous abscess model was used to assess the antimicrobial effects of AKBA in vivo. Our results revealed that AKBA was found to show effective antimicrobial activity against methicillin-resistant S. aureus (MRSA) with the minimal inhibitory concentration of 4-8 µg/mL with undetectable cytotoxicity. And no resistant mutation was induced by AKBA after 20 days of consecutive passage. Further, we found that AKBA could be synergy with gentamycin or amikacin against S. aureus and its clinical isolates. By crystal violet and SYTO9/PI staining, AKBA exhibited strong biofilm inhibitory and eradication effects at the concentration of 1 ~ 4 µg/mL. In addition, the effective antimicrobial effect was verified in vivo in a mouse model. And no detectable in vivo toxicity was found. These results indicated that AKBA has great potential to development as an alternative treatment for the refractory S. aureus infections.
Collapse
Affiliation(s)
- Yingjia Li
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Hongbing Ma
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
47
|
Pellegrini M. Advances in Network-Based Drug Repositioning. LECTURE NOTES IN COMPUTER SCIENCE 2024:99-114. [DOI: 10.1007/978-3-031-55248-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
48
|
Mandal SK, Rehman MDMU, Katyal A, Rajvanshi K, Kannan M, Garg M, Murugesan S, Deepa P. In silico anti-viral assessment of phytoconstituents in a traditional (Siddha Medicine) polyherbal formulation - Targeting Mpro and pan-coronavirus post-fusion Spike protein. J Tradit Complement Med 2024; 14:55-69. [PMID: 38223813 PMCID: PMC10785248 DOI: 10.1016/j.jtcme.2023.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 01/16/2024] Open
Abstract
Background and aim Novel nature of the viral pathogen SARS-CoV-2 and the absence of standard drugs for treatment, have been a major challenge to combat this deadly infection. Natural products offer safe and effective remedy, for which traditional ethnic medicine can provide leads. An indigenous poly-herbal formulation, Kabasura Kudineer from Siddha system of medicine was evaluated here using a combination of computational approaches, to identify potential inhibitors against two anti-SARS-CoV-2 targets - post-fusion Spike protein (structural protein) and main protease (Mpro, non-structural protein). Experimental procedure We docked 32 phytochemicals from the poly-herbal formulation against viral post-fusion Spike glycoprotein and Mpro followed by molecular dynamics using Schrodinger software. Drug-likeness analysis was performed using machine learning (ML) approach and pkCSM. Results The binding affinity of the phytochemicals in Kabasura Kudineer revealed the following top-five bioactives: Quercetin > Luteolin > Chrysoeriol > 5-Hydroxy-7,8-Dimethoxyflavone > Scutellarein against Mpro target, and Gallic acid > Piperlonguminine > Chrysoeriol > Elemol > Piperine against post-fusion Spike protein target. Quercetin and Gallic acid exhibited binding stability in complexation with their respective viral-targets and favourable free energy change as revealed by the molecular dynamics simulations and MM-PBSA analysis. In silico predicted pharmacokinetic profiling of these ligands revealed appropriate drug-likeness properties. Conclusion These outcomes provide: (a) potential mechanism for the anti-viral efficacy of the indigenous Siddha formulation, targeting Mpro and post-fusion Spike protein (b) top bioactive lead-molecules that may be developed as natural product-based anti-viral pharmacotherapy and their pleiotropic protective effects may be leveraged to manage co-morbidities associated with COVID-19.
Collapse
Affiliation(s)
- Sumit Kumar Mandal
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - MD Muzaffar-Ur Rehman
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Ashish Katyal
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Kanishk Rajvanshi
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Manoj Kannan
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
- Plaksha University, SAS Nagar, Mohali, 140306, Punjab, India
| | - Mohit Garg
- Department of Chemical Engineering, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - P.R. Deepa
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS Pilani), Pilani Campus, Pilani, 333031, Rajasthan, India
| |
Collapse
|
49
|
Ahmad SS, Khalid M. Evaluations of FDA-approved Drugs Targeting 3CLP of SARS-CoV-2 Employing a Repurposing Strategy. Comb Chem High Throughput Screen 2024; 27:2805-2815. [PMID: 35975855 DOI: 10.2174/1386207325666220816125639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/16/2022] [Accepted: 04/21/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The SARS-CoV-2 coronavirus (COVID-19) has raised innumerable global concerns, and few effective treatment strategies have yet been permitted by the FDA to lighten the disease burden. SARS-CoV-2 3C-like proteinase (3CLP) is a crucial protease and plays a key role in the viral life cycle, as it controls replication, and thus, it is viewed as a target for drug design. METHODS In this study, we performed structure-based virtual screening of FDA drugs approved during 2015-2019 (a total of 220 drugs) for interaction with the active site of 3CLP (PDB ID 6LU7) using AutoDock 4.2. We report the top ten drugs that outperform the reported drugs against 3CLP (Elbasvir and Nelfinavir), particularly Cefiderocol, having the highest affinity among the compounds tested, with a binding energy of -9.97 kcal/mol. H-bond (LYS102:HZ2-ligand: O49), hydrophobic (ligand-VAL104), and electrostatic (LYS102:NZ-ligand: O50) interactions were observed in the cefiderocol-3CLP complex. The docked complex was subjected to a 50 ns molecular dynamics study to check its stability, and stable RMSD and RMSF graphs were observed. RESULTS Accordingly, we suggest cefiderocol might be effective against SARS-CoV-2 and urge that experimental validation be performed to determine the antiviral efficacy of cefiderocol against SARS-CoV-2. DISCUSSION Along with these, cefiderocol is effective for treating respiratory tract pathogens and a wide range of gram-negative bacteria for whom there are limited therapeutic alternatives. CONCLUSION This article aimed to explore the FDA-approved drugs as a repurposing study against 3CLP for COVID-19 management.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Mohammad Khalid
- College of Pharmacy, Department of Pharmacognosy, Prince Sattam Bin Abdul Aziz University, Alkharj 16278, Riyadh, Saudi Arabia
| |
Collapse
|
50
|
Dinata R, Nisa N, Arati C, Rasmita B, Uditraj C, Siddhartha R, Bhanushree B, Saeed-Ahmed L, Manikandan B, Bidanchi RM, Abinash G, Pori B, Khushboo M, Roy VK, Gurusubramanian G. Repurposing immune boosting and anti-viral efficacy of Parkia bioactive entities as multi-target directed therapeutic approach for SARS-CoV-2: exploration of lead drugs by drug likeness, molecular docking and molecular dynamics simulation methods. J Biomol Struct Dyn 2024; 42:43-81. [PMID: 37021347 DOI: 10.1080/07391102.2023.2192797] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/10/2023] [Indexed: 04/07/2023]
Abstract
The COVID-19 pandemic has caused adverse health (severe respiratory, enteric and systemic infections) and environmental impacts that have threatened public health and the economy worldwide. Drug repurposing and small molecule multi-target directed herbal medicine therapeutic approaches are the most appropriate exploration strategies for SARS-CoV-2 drug discovery. This study identified potential multi-target-directed Parkia bioactive entities against SARS-CoV-2 receptors (S-protein, ACE2, TMPRSS2, RBD/ACE2, RdRp, MPro, and PLPro) using ADMET, drug-likeness, molecular docking (AutoDock, FireDock and HDOCK), molecular dynamics simulation and MM-PBSA tools. One thousand Parkia bioactive entities were screened out by virtual screening and forty-five bioactive phytomolecules were selected based on favorable binding affinity and acceptable pharmacokinetic and pharmacodynamics properties. The binding affinity values of Parkia phyto-ligands (AutoDock: -6.00--10.40 kcal/mol; FireDock: -31.00--62.02 kcal/mol; and HDOCK: -150.0--294.93 kcal/mol) were observed to be higher than the reference antiviral drugs (AutoDock: -5.90--9.10 kcal/mol; FireDock: -35.64--59.35 kcal/mol; and HDOCK: -132.82--211.87 kcal/mol), suggesting a potent modulatory action of Parkia bioactive entities against the SARS-CoV-2. Didymin, rutin, epigallocatechin gallate, epicatechin-3-0-gallate, hyperin, ursolic acid, lupeol, stigmasta-5,24(28)-diene-3-ol, ellagic acid, apigenin, stigmasterol, and campesterol strongly bound with the multiple targets of the SARS-CoV-2 receptors, inhibiting viral entry, attachment, binding, replication, transcription, maturation, packaging and spread. Furthermore, ACE2, TMPRSS2, and MPro receptors possess significant molecular dynamic properties, including stability, compactness, flexibility and total binding energy. Residues GLU-589, and LEU-95 of ACE2, GLN-350, HIS-186, and ASP-257 of TMPRSS2, and GLU-14, MET-49, and GLN-189 of MPro receptors contributed to the formation of hydrogen bonds and binding interactions, playing vital roles in inhibiting the activity of the receptors. Promising results were achieved by developing multi-targeted antiviral Parkia bioactive entities as lead and prospective candidates under a small molecule strategy against SARS-CoV-2 pathogenesis. The antiviral activity of Parkia bioactive entities needs to be further validated by pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Roy Dinata
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | - Nisekhoto Nisa
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | - Chettri Arati
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | | | - Chetia Uditraj
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | | | | | | | - Bose Manikandan
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | | | - Giri Abinash
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | - Buragohain Pori
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | - Maurya Khushboo
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | | |
Collapse
|