1
|
Elsisi DM, Mohamed AM, Seadawy MG, Ahmed A, Abou-Amra ES. One pot multi-component synthesis of novel functionalized pyrazolo furan-2(5H)-one derivatives: in vitro, DFT, molecular docking, and pharmacophore studies, as coronavirus inhibitors. Mol Divers 2025; 29:965-989. [PMID: 39168959 PMCID: PMC11909067 DOI: 10.1007/s11030-024-10885-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: 03/06/2024] [Accepted: 04/23/2024] [Indexed: 08/23/2024]
Abstract
New and facile one-pot approach for the syntheses of 12 derivatives of 3,5-disubstituted furane-2(5H)-one (4a-l) from easily available starting materials. The suitable synthetic procedures for selective synthesis of diverse furane-2(5H)-one derivatives were achieved via multi-component condensation of 1,3-diphenyl-1H-pyrazole-4-carbaldehyde (1), pyruvic acid and different aromatic amines 3a-l in good to high yields and short reaction time by refluxing in acetic acid as well as obtained by another method (method B) when unsaturated arylidene pyruvic acid 6 was refluxed with different aromatic amines in acetic acid but in smaller yield than method A. Structures of the prepared compounds were elucidated by elemental analysis and spectral data as mass, IR, 1H-NMR and 13C-NMR spectroscopy. The antiviral efficacy of compounds 4a-l against SARS-CoV-2 was evaluated using the MTT assay. It was demonstrated that synthetic compounds 4c-e and 4h-j have a potent and selective inhibitory effect on SARS-CoV-2, a strain obtained from Egyptian patients. We utilized density-functional theory (DFT) analyses to deduce the molecular structures and topologies of the more energetic molecules. Molecular docking studies were performed against the SARS-CoV-2 main protease (PDB ID: 6Y84) and the SARS-CoV-2 Nsp9 RNA binding protein (PDB ID: 6W4B) to study the binding mechanism, non-bonding interactions, and binding affinity. Lastly, a hypothetical pharmacophore model was constructed by applying the Molecular Operating Environment (MOE) tool and eleven pharmaceuticals with proven antiviral activity.
Collapse
Affiliation(s)
- Doaa M Elsisi
- Department of Chemistry, Faculty of Science (Girl's Branch), Al-Azhar University, Yousef Abbas Street, Cairo, 11754, Nasr City, Egypt
| | - Ashraf M Mohamed
- Applied Organic Chemistry Department, National Research Centre, Dokki, 12622, Giza, Egypt.
| | - Mohamed G Seadawy
- Biological Prevention Department, Chemical Warfare, Cairo, 11351, Egypt
| | - Aya Ahmed
- Faculty of Nanotechnology for Postgraduate Studies, Cairo University, El-Sheikh Zayed, Cairo, 12588, Egypt
| | - Eman S Abou-Amra
- Department of Chemistry, Faculty of Science (Girl's Branch), Al-Azhar University, Yousef Abbas Street, Cairo, 11754, Nasr City, Egypt.
| |
Collapse
|
2
|
Kautsar AP, Sinuraya RK, van der Schans J, Postma MJ, Suwantika AA. Exploring Public Sentiment on the Repurposing of Ivermectin for COVID-19 Treatment: Cross-Sectional Study Using Twitter Data. JMIR Form Res 2025; 9:e50536. [PMID: 40146987 DOI: 10.2196/50536] [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: 07/05/2023] [Revised: 02/12/2024] [Accepted: 09/09/2024] [Indexed: 03/29/2025] Open
Abstract
A sentiment analysis of 5051 Twitter posts from January 2022 found that 53.4% of them expressed positive views on ivermectin as a COVID-19 treatment, 35.6% of them were neutral, and 11% of them were negative, highlighting the polarized public perception and the need for careful interpretation of social media data in health communication.
Collapse
Affiliation(s)
- Angga Prawira Kautsar
- Unit of Global Health, Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| | - Rano Kurnia Sinuraya
- Unit of Global Health, Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Jurjen van der Schans
- Unit of Global Health, Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Economics, Econometrics and Finance, Faculty of Economics and Business, University of Groningen, Groningen, Indonesia
| | - Maarten Jacobus Postma
- Unit of Global Health, Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Department of Economics, Econometrics and Finance, Faculty of Economics and Business, University of Groningen, Groningen, Indonesia
| | - Auliya A Suwantika
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center for Health Technology Assessment, Universitas Padjadjaran, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
3
|
Almulhim M, Ghasemian A, Memariani M, Karami F, Yassen ASA, Alexiou A, Papadakis M, Batiha GES. Drug repositioning as a promising approach for the eradication of emerging and re-emerging viral agents. Mol Divers 2025:10.1007/s11030-025-11131-8. [PMID: 40100484 DOI: 10.1007/s11030-025-11131-8] [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: 12/02/2024] [Accepted: 02/08/2025] [Indexed: 03/20/2025]
Abstract
The global impact of emerging and re-emerging viral agents during epidemics and pandemics leads to serious health and economic burdens. Among the major emerging or re-emerging viruses include SARS-CoV-2, Ebola virus (EBOV), Monkeypox virus (Mpox), Hepatitis viruses, Zika virus, Avian flu, Influenza virus, Chikungunya virus (CHIKV), Dengue fever virus (DENV), West Nile virus, Rhabdovirus, Sandfly fever virus, Crimean-Congo hemorrhagic fever (CCHF) virus, and Rift Valley fever virus (RVFV). A comprehensive literature search was performed to identify existing studies, clinical trials, and reviews that discuss drug repositioning strategies for the treatment of emerging and re-emerging viral infections using databases, such as PubMed, Scholar Google, Scopus, and Web of Science. By utilizing drug repositioning, pharmaceutical companies can take advantage of a cost-effective, accelerated, and effective strategy, which in turn leads to the discovery of innovative treatment options for patients. In light of antiviral drug resistance and the high costs of developing novel antivirals, drug repositioning holds great promise for more rapid substitution of approved drugs. Main repositioned drugs have included chloroquine, ivermectin, dexamethasone, Baricitinib, tocilizumab, Mab114 (Ebanga™), ZMapp (pharming), Artesunate, imiquimod, saquinavir, capmatinib, naldemedine, Trametinib, statins, celecoxib, naproxen, metformin, ruxolitinib, nitazoxanide, gemcitabine, Dorzolamide, Midodrine, Diltiazem, zinc acetate, suramin, 5-fluorouracil, quinine, minocycline, trifluoperazine, paracetamol, berbamine, Nifedipine, and chlorpromazine. This succinct review will delve into the topic of repositioned drugs that have been utilized to combat emerging and re-emerging viral pathogens.
Collapse
Affiliation(s)
- Marwa Almulhim
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| | - Mojtaba Memariani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Farnaz Karami
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Asmaa S A Yassen
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
4
|
Neblea IE, Iordache TV, Sarbu A, Chiriac AL, Gavrila AM, Trica B, Biru IE, Caras I, Teodorescu M, Perrin FX, Zaharia A. Biomimetic Molecularly Imprinted Nanogels for the Recognition of Spike Glycoproteins. ACS APPLIED BIO MATERIALS 2025; 8:2215-2228. [PMID: 39961804 DOI: 10.1021/acsabm.4c01757] [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: 03/18/2025]
Abstract
The rapid spread and mutation of SARS-CoV-2, the virus responsible for COVID-19, has set the foundation for extensive research into next-generation therapeutic strategies. A critical component of SARS-CoV-2 is the trimeric Spike (S) glycoprotein, which facilitates viral entry into host cells by interacting with the receptor-binding domain (RBD). To inhibit and block viral entry, we designed and developed molecularly imprinted synthetic nanogel antibodies (MIP-SNAs) that cap the Spike S1 RBD. This aims to provide a versatile, biosecure, and effective therapeutic tool for the prevention and treatment of SARS-CoV-2 infection. Herein, we employed reverse miniemulsion polymerization to synthesize MIP-SNAs using poly(ethylene glycol) diacrylate (PEGDA), a nontoxic, nonimmunogenic and FDA-approved polymer, able to interact noncovalently with the functional groups of template Spike S1 RBD. In addition, the formulation of MIP-SNAs was based on a preliminary investigation of protein conformation by circular dichroism. Characterization of the SNAs was conducted using several techniques to investigate the chemical structure, thermal stability, size, and morphology. Under optimal conditions, the MIP-SNAs exhibited high specificity, with rebinding capacities up to 6-fold higher compared to the control nonimprinted synthetic nanogel antibodies. MIP-SNAs also demonstrated notable selectivity toward the SARS-CoV-2 Spike S1 RBD protein compared to the structural resembling Spike proteins of Bat-CoV, while cytocompatibility assays confirmed the biocompatible character of the SNAs. Given the excellent features of the recently developed MIP-SNAs, we are one step closer to finding efficient but also patient-friendly prevention and treatment solutions for SARS-CoV-2 infection. Beyond immediate applications, this technology offers the potential for broader diagnostic and therapeutic uses against related viral pathogens.
Collapse
Affiliation(s)
- Iulia Elena Neblea
- Advanced Polymer Materials and Polymer Recycling Group, National Institute of Research & Development in Chemistry and Petrochemistry ICECHIM, Splaiul Independentei no. 202, 6 District Bucharest 060021, Romania
- Department of Bioresources and Polymer Science, Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 1-7 Gh. Polizu Street, 1 District, Bucharest 011061, Romania
| | - Tanţa-Verona Iordache
- Advanced Polymer Materials and Polymer Recycling Group, National Institute of Research & Development in Chemistry and Petrochemistry ICECHIM, Splaiul Independentei no. 202, 6 District Bucharest 060021, Romania
| | - Andrei Sarbu
- Advanced Polymer Materials and Polymer Recycling Group, National Institute of Research & Development in Chemistry and Petrochemistry ICECHIM, Splaiul Independentei no. 202, 6 District Bucharest 060021, Romania
| | - Anita-Laura Chiriac
- Advanced Polymer Materials and Polymer Recycling Group, National Institute of Research & Development in Chemistry and Petrochemistry ICECHIM, Splaiul Independentei no. 202, 6 District Bucharest 060021, Romania
| | - Ana-Mihaela Gavrila
- Advanced Polymer Materials and Polymer Recycling Group, National Institute of Research & Development in Chemistry and Petrochemistry ICECHIM, Splaiul Independentei no. 202, 6 District Bucharest 060021, Romania
| | - Bogdan Trica
- Advanced Polymer Materials and Polymer Recycling Group, National Institute of Research & Development in Chemistry and Petrochemistry ICECHIM, Splaiul Independentei no. 202, 6 District Bucharest 060021, Romania
| | - Iuliana Elena Biru
- Department of Bioresources and Polymer Science, Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 1-7 Gh. Polizu Street, 1 District, Bucharest 011061, Romania
| | - Iuliana Caras
- National Institute for Medico-Military Research and Development "Cantacuzino", Spl. Independentei 103, fifth District, Bucharest 050096, Romania
| | - Mircea Teodorescu
- Department of Bioresources and Polymer Science, Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 1-7 Gh. Polizu Street, 1 District, Bucharest 011061, Romania
| | - François-Xavier Perrin
- Laboratoire Matériaux Polymères Interfaces et Environnement Marin-MAPIEM EA 4323 SeaTech-Ecole d'ingénieurs, Université de Toulon, BP 20132, La Garde, Toulon Cedex 83957, France
| | - Anamaria Zaharia
- Advanced Polymer Materials and Polymer Recycling Group, National Institute of Research & Development in Chemistry and Petrochemistry ICECHIM, Splaiul Independentei no. 202, 6 District Bucharest 060021, Romania
| |
Collapse
|
5
|
Chen E, Xi L. Cardiovascular adverse effects of antiviral therapies for COVID-19: Evidence and plausible mechanisms. Acta Pharmacol Sin 2025; 46:554-564. [PMID: 39251859 PMCID: PMC11845466 DOI: 10.1038/s41401-024-01382-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024]
Abstract
Antiviral therapeutics have made a critical contribution in mitigating the symptoms and clinical outcomes of the coronavirus disease of 2019 (COVID-19), in which a single-stranded RNA viral pathogen, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causes multi-organ injuries. Several antivirals were widely prescribed to treat COVID-19, either through the emergency use authorization (EUA) by the governmental regulatory agencies (i.e., remdesivir, paxlovid, molnupiravir, and the SARS-CoV-2-targeted monoclonal antibodies - tixagevimab and cilgavimab), as well as the repurposed use of the existing antiviral or antimalarial drugs (e.g., hydroxychloroquine, chloroquine, and ivermectin). Despite their efficacy in ameliorating COVID-19 symptoms, some adverse side-effects of the antivirals were also reported during the COVID-19 pandemic. Our current review has aimed to gather and extrapolate the recently published information concerning cardiovascular adverse effects caused by each of the antivirals. We also provide further discussion on the potential cellular mechanisms underlying the cardiovascular adverse effects of the selected antiviral drugs, which should be carefully considered when evaluating risk factors in managing patients with COVID-19 or similar infectious diseases. It is foreseeable that future antiviral drug development assisted with the newest artificial intelligence platform may improve the accuracy to predict the structures of biomolecules of antivirals and therefore to mitigate their associated cardiovascular adversities.
Collapse
Affiliation(s)
- Eileen Chen
- Virginia Commonwealth University School of Medicine (M.D. Class 2027), Richmond, VA, 23298, USA
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA.
| |
Collapse
|
6
|
Siddesh M, Sharanya R, Spoorthy L, Bhat D, Udaya Kumar AH, Mahesha, Hema MK, Lokanath NK. Investigation of the molecular basis of halogenated Schiff base derivative by combined crystallographic and computational studies. J Biomol Struct Dyn 2025; 43:2479-2490. [PMID: 38189357 DOI: 10.1080/07391102.2023.2301512] [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: 04/01/2023] [Accepted: 10/07/2023] [Indexed: 01/09/2024]
Abstract
Halogenated Schiff base derivatives are gaining more popularity in supramolecular chemistry due to the synergistic effect of hydrogen and halogen-based noncovalent interactions, which helps to design novel therapeutic materials. In this work, we have examined the nature of molecular interactions to investigate the structure-functional relationship of a halogen-based derivative. The FTIR, HRMS and NMR spectroscopic techniques confirmed the formation of the desired novel Schiff base compound. Further, crystal structure studies showed an infinite 1D supramolecular chain formed by type-I halogen…halogen interaction. The Hirshfeld surface and enrichment ratio analyses were performed to visualize and assess the role of diverse interactions involved in crystal packing. The QTAIM, NCI, LOL and ELF studies were conducted extensively to comprehend the strength of interaction constructed based on electron density distribution. The global and local reactive indices were determined using DFT studies to analyze the molecular properties of the compound. Antibacterial activity against MRSA bacteria was performed and showed a good zone of inhibition. The docking analysis was performed for 1mwt protein and validated. The in silico molecular docking studies of the halogenated Schiff base structure with the penicillin-binding protein showed a good docking affinity of -7.5 kcal/mol and supported by in vitro studies. The ligand binding stability within the protein's active site was further demonstrated by molecular dynamics (MD) simulation studies for the Schiff base molecule.
Collapse
Affiliation(s)
- M Siddesh
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
| | - R Sharanya
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
| | - L Spoorthy
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
| | - Dhruva Bhat
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
| | - A H Udaya Kumar
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
| | - Mahesha
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
- Department of Physics, SJCE, JSS Science and Technology University, Mysuru, India
| | - M K Hema
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
| | - N K Lokanath
- Department of Studies in Physics, University of Mysore, Mysuru, Karnataka, India
| |
Collapse
|
7
|
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
|
8
|
Rosa RJ, Ferezin LP, de Campos MCT, Moura HSD, Berra TZ, Ribeiro NM, Teibo TKA, Vinci ALT, Ramos ACV, do Nascimento MC, Fuentealba Torres MÁ, Arcêncio RA. Perception of Risk for Developing Severe Illness or Complications from COVID-19 in Brazil: Focus on Factors Linked to Socially Vulnerable Populations, 2020-2023. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:251. [PMID: 40003477 PMCID: PMC11855750 DOI: 10.3390/ijerph22020251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025]
Abstract
In this study, the aim was to comparatively examine the perception of risk for developing severe illness or complications due to COVID-19 among the general population and socially vulnerable populations in Brazil, focusing on uncovering the associated factors that disproportionately impacted people experiencing homelessness and slum dwellers. This study is part of the project "Social Thermometer-COVID-19 in Brazil", which employed a hybrid approach, combining a national online survey with field research in state capitals and the Federal District. Data collection took place from August 2020 to October 2023, and the data were analyzed using descriptive statistics and logistic regression. A total of 5094 participants were included in this study, with 2363 from the general population and 2731 from the socially vulnerable population. Among the general population, the majority of participants were women, white individuals, those with higher incomes, formal employment, and higher education levels. Concerning the vulnerable population, most were men, Black individuals, those with lower incomes, unemployment, and lower education levels. It was observed that 87% of the general population perceived a risk of severe COVID-19, compared to 74% of the vulnerable population. Slum dwellers who received emergency aid (ORa = 1.39; 95% CI: 1.02-1.91), adhered to mask-wearing practices (ORa = 1.93; 95% CI: 1.39-2.66), used COVID-19-related medications (ORa = 2.13; 95% CI: 1.31-3. 64), and those with pre-existing conditions, such as high blood pressure (ORa = 1.86; 95% CI: 1.20-2.98), demonstrated a heightened perception of risk for severe COVID-19 complications. Among the homeless population, individuals who wore masks (ORa = 1.67; 95% CI: 1.26-2.20 and had been vaccinated (ORa = 1.44; 95% CI: 1.04-1.98) were also more likely to perceive a high risk. In conclusion, in this study, significant disparities are revealed in the perception of COVID-19 risk between the general and socially vulnerable populations in Brazil. Factors such as receiving emergency aid, adherence to mask-wearing, use of COVID-19-related medications, and pre-existing health conditions were associated with increased risk perception. Despite facing greater socioeconomic challenges, vulnerable groups, particularly those experiencing homelessness and slum dwellers, showed a lower perception of the risk for severe COVID-19 complications.
Collapse
Affiliation(s)
- Rander Junior Rosa
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| | - Letícia Perticarrara Ferezin
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| | - Mônica Chiodi Toscano de Campos
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
- Nursing Departament, Unversity of Brasília, Brasília 70910-900, Brazil
| | - Heriederson Sávio Dias Moura
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| | - Thaís Zamboni Berra
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| | - Natacha Martins Ribeiro
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| | - Titilade Kehinde Ayandeyi Teibo
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| | - André Luiz Teixeira Vinci
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| | | | | | | | - Ricardo Alexandre Arcêncio
- School of Nursing, University of São Paulo at Ribeirão Preto, Ribeirão Preto 14040-902, Brazil; (L.P.F.); (M.C.T.d.C.); (H.S.D.M.); (T.Z.B.); (N.M.R.); (T.K.A.T.); (A.L.T.V.); (R.A.A.)
| |
Collapse
|
9
|
Chen Y, Huang X, Guo Z, Zhang J, Zhang L, Dai R. Study of Pharmacokinetics for Ivermectin B1a from Beagle Dogs. J Chromatogr Sci 2025; 63:bmad092. [PMID: 38134186 DOI: 10.1093/chromsci/bmad092] [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: 06/16/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023]
Abstract
Ivermectin has been widely used for antiparasitic drug, and has recently shown a broad-spectrum antiviral activity, including anti-Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the pharmacokinetic property of ivermectin has not been fully investigated yet. During the plasma preparation, ~32-46% of ivermectin was found in the precipitation. An Liquid Chromatograph-Mass Spectrometer (LC-MS/MS) method for ivermectin in the whole blood samples from beagle dogs was developed and validated. The specificity, accuracy, precision (intra-day and inter-day), matrix effect, recovery and stability of analyte reported here are satisfied with the criteria of Food and Drug Administration (FDA)-Bioanalysis guideline. The oral administrations pharmacokinetics of ivermectin in beagle dogs under fasting and after high-fat meal were studied, and the following parameters were obtained: fasting Cmax, 104 ± 35 μg·L-1; area under the concentration-time curve (AUC0-∞), 2,555 ± 941 h·μg·L-1; and high-fat meal Cmax, 147 ± 35 μg·L-1; AUC0-∞, 4,198 ± 1,279 h·μg·L-1. When the P-gp inhibitor curcumin was also coadministrated orally, Cmax and AUC0-∞ were found to be 177 ± 57 and 4,213 ± 948 h·μg·L-1, respectively. With the comparison to fasting treatment, coadministration of P-gp inhibitor curcumin resulted in increase of the exposure of ivermectin by 1.6-fold, while the exposure after the high-fat diet versus fasting was increased approximately in 1.4-fold, indicating that alternative absorption might play an important role for increasing the exposure of ivermectin for future clinic applications.
Collapse
Affiliation(s)
- Yuyang Chen
- School of Pharmacy, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou, Guangdong 511436, China
| | - Xiaofang Huang
- Guangdong Ruigu Biotech Corporation, 18 Chuangxing Road, High-tech Zone, Qingyuan, Guangdong 511517, China
| | - Zizheng Guo
- Guangdong Ruigu Biotech Corporation, 18 Chuangxing Road, High-tech Zone, Qingyuan, Guangdong 511517, China
| | - Jingyu Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Xuhui District, Shanghai 200237, China
| | - Lixin Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Xuhui District, Shanghai 200237, China
| | - Renke Dai
- School of Pharmacy, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou, Guangdong 511436, China
- Guangdong Ruigu Biotech Corporation, 18 Chuangxing Road, High-tech Zone, Qingyuan, Guangdong 511517, China
| |
Collapse
|
10
|
Alvisi G, Manaresi E, Pavan S, Jans DA, Wagstaff KM, Gallinella G. Avermectins Inhibit Replication of Parvovirus B19 by Disrupting the Interaction Between Importin α and Non-Structural Protein 1. Viruses 2025; 17:220. [PMID: 40006975 PMCID: PMC11860776 DOI: 10.3390/v17020220] [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: 01/03/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Human parvovirus B19 (B19V) is a major human pathogen in which the ssDNA genome is replicated within the nucleus of infected human erythroid progenitor cells (EPCs) through a process involving both cellular and viral proteins, including the non-structural protein (NS)1. We previously characterized the interaction between NS1 classical nuclear localization signal (cNLS: GACHAKKPRIT-182) and host cell importin (IMP)α and proposed it as a potential target for antiviral drug development. Here, we further extend on such findings. First, we demonstrate that NS1 nuclear localization is required for viral production since introducing the K177T substitution in a cloned, infectious viral genome resulted in a non-viable virus. Secondly, we demonstrate that the antiparasitic drug ivermectin (IVM), known to inhibit the IMPα/β dependent nuclear import pathway, could impair the NS1-NLS:IMPα interaction and suppress viral replication in UT7/EpoS1 cells in a dose-dependent manner. We also show that a panel of structurally related avermectins (AVMs) can dissociate the NS1-NLS:IMPα complex with half-maximal inhibitory concentrations in the nanomolar range. Among them, Eprinomectin emerged as the most selective inhibitor of B19V replication, with a selectivity index of c. 5.0. However, when tested in EPCs generated from peripheral blood mononuclear cells, which constitute a cellular population close to the natural target cells in bone marrow, the inhibitory effect of IVM and Eprinomectin was demonstrated to a lesser extent, and both compounds exhibited high toxicity, thus highlighting the need for more specific inhibitors of the NS1-NLS:IMPα interaction.
Collapse
Affiliation(s)
- Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy;
| | - Elisabetta Manaresi
- Department of Pharmacy and Biotechnology, University of Bologna, 40138 Bologna, Italy;
| | - Silvia Pavan
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy;
| | - David A. Jans
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute Monash University, Clayton, VIC 3800, Australia; (D.A.J.); (K.M.W.)
| | - Kylie M. Wagstaff
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute Monash University, Clayton, VIC 3800, Australia; (D.A.J.); (K.M.W.)
| | - Giorgio Gallinella
- Department of Pharmacy and Biotechnology, University of Bologna, 40138 Bologna, Italy;
| |
Collapse
|
11
|
Sai Yengu N, Raheem A, Pons AG, Ho WL, Ali SMS, Haseeb A, Fadlalla Ahmad TK, Mustafa MS. The impact of ivermectin on COVID-19 outcomes: a systematic review and meta-analysis. Ann Med Surg (Lond) 2025; 87:809-829. [PMID: 40110299 PMCID: PMC11918548 DOI: 10.1097/ms9.0000000000002762] [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: 06/24/2024] [Revised: 10/13/2024] [Accepted: 11/11/2024] [Indexed: 03/22/2025] Open
Abstract
Background The COVID-19 pandemic, resulting in approximately seven million deaths globally, underscores the urgency for effective treatments. Ivermectin, among several repurposed drugs, garnered interest due to its antiviral properties. However, conflicting evidence from observational studies and randomized controlled trials raised questions about its efficacy and safety. Method This systematic review and meta-analysis followed MOOSE and PRISMA guidelines. Comprehensive searches were conducted in databases including Scopus, Embase, PubMed, and Web of Science up to April 2024. Data were extracted independently by two reviewers and analyzed using Comprehensive Meta-Analysis V3 software. Results Across 33 studies encompassing 15,376 participants, ivermectin showed no significant impact on critical outcomes such as mortality [risk ratio (RR) 0.911, 95% confidence intervals (CI) 0.732-1.135], mechanical ventilation (RR 0.727, 95% CI 0.521-1.016), polymerase chain reaction conversion (RR 1.024, 95% CI 0.936-1.120), ICU admissions (RR 0.712, 95% CI 0.274-1.850), or hospitalization rates (RR 0.735, 95% CI 0.464-1.165) compared to controls. However, it significantly reduced time to symptom alleviation (standardized mean difference -0.302, 95% CI -0.587 to -0.018) and sustained symptom relief (RR 0.897, 95% CI 0.873-0.921). Adverse event (AE) rates were similar between the ivermectin and control groups (RR 0.896, 95% CI 0.797-1.007). Meta-regression indicated older age and diabetes as predictors of AEs. Conclusion Despite its observed benefits in symptom management, ivermectin did not significantly influence critical clinical outcomes in COVID-19 patients. These findings highlight the importance of continued research to identify effective treatments for COVID-19, emphasizing the need for high-quality studies with robust methodology to inform clinical practice and public health policy effectively.
Collapse
Affiliation(s)
- Nithin Sai Yengu
- Dr Pinnamaneni Siddhartha Institute of Medical Sciences, Vijayawada, India
| | | | | | - Wing Lam Ho
- Saint George's University School of Medicine University Center, Grenada
| | | | - Abdul Haseeb
- Jinnah Sindh Medical University, Karachi, Pakistan
| | | | | |
Collapse
|
12
|
Alyami MH, Alyami HS, Abdo AM, A. Sabry S, Mansour SMG, El-Nahas HM, Ayoub MM. Experimental Investigation into the Design, Optimization, Toxicity, and Anti-Viral Efficacy of Proliposomes Loaded with Ivermectin Against Infectious Bronchitis Virus Using an Embryonated Chicken Egg Model. Pharmaceutics 2025; 17:165. [PMID: 40006532 PMCID: PMC11859826 DOI: 10.3390/pharmaceutics17020165] [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: 11/21/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Infectious bronchitis virus (IBV) causes a significant illness in birds, making it a leading source of financial loss in the poultry business. The objective of this study was to assess the effectiveness of proliposomes (PLs) containing ivermectin (IVM) against IBV using embryonated chicken eggs (ECEs). Methods: A three-factor, two-level (23) full factorial design was employed; carrier/lipid phase ratio (A), stearyl glycyrrhetinate amount (B), and phospholipid type (C) were studied as independent variables, while product yield (PY), entrapment efficiency (EE), particle size (PS), polydispersity index (PDI), zeta potential (ZP), and cumulative percentage of drug released after 6 h (Q6h) were characterized. The selected formulations (PL2 and PL6) were subjected to further characterizations, including IVM toxicity and anti-viral activity. Results: The PY% ranged from 88.6 ± 2.19% to 98.8 ± 0.45%, EE% was from 71.8 ± 2.01% to 96.1 ± 0.51%, PS was from 330.1 ± 55.65 nm to 1801.6 ± 45.61 nm, PDI was from 0.205 ± 0.06 to 0.603 ± 0.03, ZP was from -18.2 ± 0.60 mV to -50.1 ± 1.80 mV, and Q6h was from 80.95 ± 1.36% to 88.79 ± 2.03%. IVM-loaded PLs had lower toxicity in ECEs than pure IVM; the mortality rate was substantially reduced in IBV-infected ECEs injected with PL2 rather than pure IVM. As further evidence of IVM's anti-viral action against IBV, quantitative real-time polymerase chain reaction (qRT-PCR) revealed that the PL2-treated group exhibited further reduction in IBV's copies in comparison with the pure IVM-treated group. Conclusions: PLs loaded with IVM are an innovative and potentially effective way to inhibit IBV.
Collapse
Affiliation(s)
- Mohammad H. Alyami
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia
| | - Hamad S. Alyami
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia
| | - Asmaa M. Abdo
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Shereen A. Sabry
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Shimaa M. G. Mansour
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Hanan M. El-Nahas
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Margrit M. Ayoub
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
13
|
Kumar M, Baig MS, Bhardwaj K. Advancements in the development of antivirals against SARS-Coronavirus. Front Cell Infect Microbiol 2025; 15:1520811. [PMID: 39917633 PMCID: PMC11798951 DOI: 10.3389/fcimb.2025.1520811] [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: 10/31/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) caused an outbreak in 2002-2003, spreading to 29 countries with a mortality rate of about 10%. Strict quarantine and infection control methods quickly stopped the spread of the disease. Later research showed that SARS-CoV came from animals (zoonosis) and stressed the possibility of a similar spread from host to human, which was clearly shown by the COVID-19 outbreak. The COVID-19 pandemic, instigated by SARS-CoV-2, has affected 776 million confirmed cases and more than seven million deaths globally as of Sept 15, 2024. The existence of animal reservoirs of coronaviruses continues to pose a risk of re-emergence with improved fitness and virulence. Given the high death rate (up to 70 percent) and the high rate of severe sickness (up to 68.7 percent in long-COVID patients), it is even more critical to identify new therapies as soon as possible. This study combines research on antivirals that target SARS coronaviruses that have been conducted over the course of more than twenty years. It is a beneficial resource that might be useful in directing future studies.
Collapse
Affiliation(s)
- Mrityunjay Kumar
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, India
| | - Mirza Sarwar Baig
- Centre for Virology, School of Interdisciplinary Science and Technology, Jamia Hamdard, New Delhi, India
| | - Kanchan Bhardwaj
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| |
Collapse
|
14
|
Adha SA, Afifah NN, Latarissa IR, Iftinan GN, Kusuma ASW, Febriyanti RM, Barliana MI, Lestari K. Herbal Medicines as Complementary Therapy for Managing Complications in COVID-19 Patients with Diabetes Mellitus. Diabetes Metab Syndr Obes 2025; 18:135-146. [PMID: 39840393 PMCID: PMC11746946 DOI: 10.2147/dmso.s498774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/26/2024] [Indexed: 01/23/2025] Open
Abstract
Diabetes mellitus (DM) is recognized and classified as a group of conditions marked by persistent high blood glucose levels. It is also an inflammatory condition that may influence concurrent disease states, including Coronavirus Disease 2019 (COVID-19). Currently, no effective drug has been found to treat COVID-19, especially in DM patients. Many herbal medicines, such as the well-known Andrographis paniculata, have been explored as drugs and complementary therapies due to their antidiabetic, antibacterial, antiviral, anti-inflammatory, and immunomodulatory effects. This study aimed to examine the potential of herbal medicines as complementary therapy in DM patients with COVID-19 complications, drawing from in-vitro and in-vivo investigations. This study analyzed articles published within the last 15 years using keywords including "herbal medicines", "COVID-19", "Diabetes Mellitus", "antidiabetics", "antiviral", and "anti-inflammatory". The results showed that several herbal medicines could serve as complementary therapy for DM patients with COVID-19 complications. These include Andrographis paniculata, Ageratum conyzoides, Artocarpus altilis, Centella asiatica, Momordica charantia, Persea gratissima, Phyllanthus urinaria, Physalis angulata, Tinospora cordifolia, and Zingiber zerumbet. Herbal medicines may serve as a complementary therapy for DM patients with COVID-19, but these claims need experimental validation in infection models and among affected patients.
Collapse
Affiliation(s)
- Syah Akbarul Adha
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Nadiya Nurul Afifah
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - 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
| | - Ghina Nadhifah Iftinan
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Arif Satria Wira Kusuma
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - Raden Maya Febriyanti
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 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
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
15
|
Tripathi A, Chauhan S, Khasa R. A Comprehensive Review of the Development and Therapeutic Use of Antivirals in Flavivirus Infection. Viruses 2025; 17:74. [PMID: 39861863 PMCID: PMC11769230 DOI: 10.3390/v17010074] [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: 11/09/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Flaviviruses are a diverse group of viruses primarily transmitted through hematophagous insects like mosquitoes and ticks. Significant expansion in the geographic range, prevalence, and vectors of flavivirus over the last 50 years has led to a dramatic increase in infections that can manifest as hemorrhagic fever or encephalitis, leading to prolonged morbidity and mortality. Millions of infections every year pose a serious threat to worldwide public health, encouraging scientists to develop a better understanding of the pathophysiology and immune evasion mechanisms of these viruses for vaccine development and antiviral therapy. Extensive research has been conducted in developing effective antivirals for flavivirus. Various approaches have been extensively utilized in clinical trials for antiviral development, targeting virus entry, replication, polyprotein synthesis and processing, and egress pathways exploiting virus as well as host proteins. However, to date, no licensed antiviral drug exists to treat the diseases caused by these viruses. Understanding the mechanisms of host-pathogen interaction, host immunity, viral immune evasion, and disease pathogenesis is highly warranted to foster the development of antivirals. This review provides an extensively detailed summary of the most recent advances in the development of antiviral drugs to combat diseases.
Collapse
Affiliation(s)
- Aarti Tripathi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA;
- Galveston National Laboratory, Galveston, TX 77555, USA
| | - Shailendra Chauhan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA;
- Galveston National Laboratory, Galveston, TX 77555, USA
| | - Renu Khasa
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami/UHealth, Miami, FL 33136, USA
| |
Collapse
|
16
|
Walunj SB, Mishra G, Wagstaff KM, Patankar S, Jans DA. The Ivermectin Related Compound Moxidectin Can Target Apicomplexan Importin α and Limit Growth of Malarial Parasites. Cells 2025; 14:39. [PMID: 39791740 PMCID: PMC11720742 DOI: 10.3390/cells14010039] [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: 11/16/2024] [Revised: 12/22/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025] Open
Abstract
Signal-dependent transport into and out of the nucleus mediated by members of the importin (IMP) superfamily is crucial for eukaryotic function, with inhibitors targeting IMPα being of key interest as anti-infectious agents, including against the apicomplexan Plasmodium species and Toxoplasma gondii, causative agents of malaria and toxoplasmosis, respectively. We recently showed that the FDA-approved macrocyclic lactone ivermectin, as well as several other different small molecule inhibitors, can specifically bind to and inhibit P. falciparum and T. gondii IMPα functions, as well as limit parasite growth. Here we focus on the FDA-approved antiparasitic moxidectin, a structural analogue of ivermectin, for its IMPα-targeting and anti-apicomplexan properties for the first time. We use circular dichroism and intrinsic tryptophan fluorescence measurements to show that moxidectin can bind directly to apicomplexan IMPαs, thereby inhibiting their key binding functions at low μM concentrations, as well as possessing anti-parasitic activity against P. falciparum in culture. The results imply a class effect in terms of IMPα's ability to be targeted by macrocyclic lactone compounds. Importantly, in the face of rising global emergence of resistance to approved anti-parasitic agents, the findings highlight the potential of moxidectin and possibly other macrocyclic lactone compounds as antimalarial agents.
Collapse
Affiliation(s)
- Sujata B. Walunj
- Nuclear Signaling Laboratory, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; (S.B.W.); (K.M.W.)
- Molecular Parasitology Laboratory, Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India; (G.M.); (S.P.)
| | - Geetanjali Mishra
- Molecular Parasitology Laboratory, Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India; (G.M.); (S.P.)
| | - Kylie M. Wagstaff
- Nuclear Signaling Laboratory, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; (S.B.W.); (K.M.W.)
| | - Swati Patankar
- Molecular Parasitology Laboratory, Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India; (G.M.); (S.P.)
| | - David A. Jans
- Nuclear Signaling Laboratory, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; (S.B.W.); (K.M.W.)
| |
Collapse
|
17
|
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
|
18
|
Angius F, Puxeddu S, Zaimi S, Canton S, Nematollahzadeh S, Pibiri A, Delogu I, Alvisi G, Moi ML, Manzin A. SARS-CoV-2 Evolution: Implications for Diagnosis, Treatment, Vaccine Effectiveness and Development. Vaccines (Basel) 2024; 13:17. [PMID: 39852796 PMCID: PMC11769326 DOI: 10.3390/vaccines13010017] [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: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, driven by the rapid evolution of the SARS-CoV-2 virus, presents ongoing challenges to global public health. SARS-CoV-2 is characterized by rapidly evolving mutations, especially in (but not limited to) the spike protein, complicating predictions about its evolutionary trajectory. These mutations have significantly affected transmissibility, immune evasion, and vaccine efficacy, leading to multiple pandemic waves with over half a billion cases and seven million deaths globally. Despite several strategies, from rapid vaccine development and administration to the design and availability of antivirals, including monoclonal antibodies, already having been employed, the persistent circulation of the virus and the emergence of new variants continue to result in high case numbers and fatalities. In the past four years, immense research efforts have contributed much to our understanding of the viral pathogenesis mechanism, the COVID-19 syndrome, and the host-microbe interactions, leading to the development of effective vaccines, diagnostic tools, and treatments. The focus of this review is to provide a comprehensive analysis of the functional impact of mutations on diagnosis, treatments, and vaccine effectiveness. We further discuss vaccine safety in pregnancy and the implications of hybrid immunity on long-term protection against infection, as well as the latest developments on a pan-coronavirus vaccine and nasal formulations, emphasizing the need for continued surveillance, research, and adaptive public health strategies in response to the ongoing SARS-CoV-2 evolution race.
Collapse
Affiliation(s)
- Fabrizio Angius
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvia Puxeddu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvio Zaimi
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Serena Canton
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Sepehr Nematollahzadeh
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Andrea Pibiri
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Ilenia Delogu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| |
Collapse
|
19
|
Khan ZA, Yadav MK, Lim DW, Kim H, Wang JH, Ansari A. Viral-host molecular interactions and metabolic modulation: Strategies to inhibit flaviviruses pathogenesis. World J Virol 2024; 13:99110. [PMID: 39722758 PMCID: PMC11551686 DOI: 10.5501/wjv.v13.i4.99110] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 10/18/2024] Open
Abstract
Flaviviruses, which include globally impactful pathogens, such as West Nile virus, yellow fever virus, Zika virus, Japanese encephalitis virus, and dengue virus, contribute significantly to human infections. Despite the ongoing emergence and resurgence of flavivirus-mediated pathogenesis, the absence of specific therapeutic options remains a challenge in the prevention and treatment of flaviviral infections. Through the intricate processes of fusion, transcription, replication, and maturation, the complex interplay of viral and host metabolic interactions affects pathophysiology. Crucial interactions involve metabolic molecules, such as amino acids, glucose, fatty acids, and nucleotides, each playing a pivotal role in the replication and maturation of flaviviruses. These viral-host metabolic molecular interactions hijack and modulate the molecular mechanisms of host metabolism. A comprehensive understanding of these intricate metabolic pathways offers valuable insights, potentially unveiling novel targets for therapeutic interventions against flaviviral pathogenesis. This review emphasizes promising avenues for the development of therapeutic agents that target specific metabolic molecules, such as amino acids, glucose, fatty acids, and nucleotides, which interact with flavivirus replication and are closely linked to the modulation of host metabolism. The clinical limitations of current drugs have prompted the development of new inhibitory strategies for flaviviruses based on an understanding of the molecular interactions between the virus and the host.
Collapse
Affiliation(s)
- Zeeshan Ahmad Khan
- Biohealth Products Research Center (BPRC), Research Center for Aged-life Redesign (RCAR), Department of Physical Therapy, INJE University, Gimhae 5084, South Korea
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda 151401, India
| | - Dong-Woo Lim
- Department of Diagnostics, College of Korean Medicine, Dongguk University, Goyang 10326, South Korea
| | - Hojun Kim
- Division of Rehabilitation Medicine of Korean Medicine, Department of Oriental Rehabilitation Medicine, Dongguk University, Ilsan Hospital, Goyang 10326, South Korea
| | - Jing-Hua Wang
- Institute of Oriental Medicine, Dongguk University, Goyang 10326, South Korea
| | - AbuZar Ansari
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul 07985, South Korea
| |
Collapse
|
20
|
Aronson JK. When I use a word . . . Academic felonies and misdemeanours-adverse effects. BMJ 2024; 387:q2787. [PMID: 39662995 DOI: 10.1136/bmj.q2787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Affiliation(s)
- Jeffrey K Aronson
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
- Follow Jeffrey on X: @JKAronson
| |
Collapse
|
21
|
Aydın Aksoy E, Güçiz Doğan B, Yalçın SS. Nutrient Supplements for Young Children and Mothers' Self Medication with Over-the-Counter Drugs During the COVID-19 Pandemic. Nutrients 2024; 16:4182. [PMID: 39683575 DOI: 10.3390/nu16234182] [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: 11/12/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The restriction of access to health services during the COVID-19 pandemic has led to an increase in self-medication. This study aims to examine mothers' use of nutrient supplements with over-the-counter (OTC) medications for their children, including instances of self-medication for themselves. The study also explores maternal characteristics associated with this behavior, the specific medications used, and the reasons for use. METHOD In this descriptive study, 450 mothers with children aged 2 to 6 years in Türkiye were recruited through social media platforms. Questions focused on whether mothers used supplements for themselves and their children, types of products, frequency, and reasons for use. Multivariable binary logistic regression was conducted to examine the factors associated with OTC medication use for children. RESULT Nearly half of the mothers reported administering OTC medications to their children. Factors associated with this practice included the child's age (specifically 48-72 months), attendance at nursery, perceived underweight status, and regular health visits with a pediatrician. Additionally, mothers who frequently used medications without a doctor's recommendation were 5.8 times more likely to give OTC drugs to their children. Maternal self-medication was significantly associated with an increased likelihood of OTC medication use for children (OR = 12.1). The most commonly used supplements included vitamin D, fish oil, multivitamins, vitamin C, immune boosters, zinc, probiotics, herbal teas, oral/nasal sprays, throat lozenges, and aspirin, with the primary purposes being prevention and treatment. CONCLUSIONS The administration of OTC medications in young children, who rely heavily on maternal care should be more closely monitored to ensure their safety and well-being, especially during epidemics.
Collapse
Affiliation(s)
- Esin Aydın Aksoy
- Department of Social Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06230, Türkiye
- Department of Pediatrics, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul 34418, Türkiye
| | - Bahar Güçiz Doğan
- Public Health Institute, Hacettepe University, Ankara 06230, Türkiye
| | - Sıddıka Songül Yalçın
- Department of Social Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06230, Türkiye
- Division of Social Pediatrics, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye
- Department of Vaccine Studies, Institude of Vaccine, Hacettepe University, Ankara 06230, Türkiye
| |
Collapse
|
22
|
Kota S, Nelapati AK, Govada VR. Plant resources for immunonutrients and immunomodulators to combat infectious respiratory viral diseases: a review. 3 Biotech 2024; 14:302. [PMID: 39554986 PMCID: PMC11568085 DOI: 10.1007/s13205-024-04143-y] [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: 06/05/2024] [Accepted: 10/26/2024] [Indexed: 11/19/2024] Open
Abstract
Boosting the immune system has become a crucial aspect in the global battle against the COVID-19 pandemic and other similar infections to protect oneself against symptoms, especially in the prevention of viral infections of the lower respiratory tract. The importance of conducting more studies to create successful herbal formulations as infection prevention measures is emphasized in this review, which looks at the function of immune-boosting nutrients, medicinal plants, and herbal treatments. We reviewed and analyzed 207 studies published from 1946 to the present using reputable databases like Google Scholar, PubMed, and NCBI. The review examined 115 plant species in total and identified 12 key nutrients, including vitamins A, D, C, omega-3 fatty acids, iron, and zinc, while noting that four plant families, Rosaceae, Asteraceae, Amaryllidaceae, and Acanthaceae, show potential against respiratory infections like influenza, RSV, and SARS-CoV. To lower the risk of infection, it is recommended to consume nutritious meals that have immune-modulating qualities. Information on the bioactive components of medicinal herbs, spices, and plants that have been effective in treating respiratory viral infections and related conditions is compiled in this review, which highlights phytoactive substances with antibacterial and antiviral activity as effective modulators to lower the risk of infections. Furthermore, it is highlighted that ancient knowledge systems, like Ayurveda and Naturopathy, should be integrated to help develop new herbal formulations. To improve immunity and lessen vulnerability to serious respiratory infections, the results highlight the need for including immune-modulating foods and plant-based medicines into everyday routines.
Collapse
Affiliation(s)
- Sobha Kota
- Department of Chemical Engineering, RVR & JC College of Engineering, Guntur, Andhra Pradesh 522 019 India
| | - Anand Kumar Nelapati
- Department of Biotechnology, Vignan’s Foundation for Science, Technology and Research, Vadlamudi, Guntur, Andhra Pradesh 522 213 India
| | - Vayunandana Rao Govada
- Department of Chemical Engineering, RVR & JC College of Engineering, Guntur, Andhra Pradesh 522 019 India
| |
Collapse
|
23
|
Osati EFO, Shayo GA, Sangeda RZ, Nagu TJ, Moshiro C, Adams N, Ramadhani A, Wajanga B, Muniko A, Seni J, Nicholaus MA, Nyaisonga G, Mbije C, Meda JR, Rainer D, Nkya ME, Mhame P, Samwel L, Vumilia L, Shekalaghe S, Kilonzo KG, Makubi A. Clinical manifestations and treatment outcomes among hospitalised COVID-19 patients in tertiary hospitals in Tanzania, 2021-2022: a retrospective cohort study. BMJ PUBLIC HEALTH 2024; 2:e000881. [PMID: 40018602 PMCID: PMC11816690 DOI: 10.1136/bmjph-2023-000881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/25/2024] [Indexed: 03/01/2025]
Abstract
Background There have been differential mortality rates from COVID-19 in different parts of the world. It is not clear whether the clinical presentation does also differ, thus the need for this study in a sub-Saharan African setting. The aim of this study was to describe the clinical manifestations and outcomes of patients diagnosed with COVID-19 in selected tertiary hospitals in Tanzania. Methods This was a retrospective analysis of hospitalised adults confirmed SAR-COV-2 infection in five tertiary-level hospitals in Tanzania. Data collected and analysed included sociodemographic, radiological and clinical characteristics of the patients as well as the outcome of the admission (discharge vs death). Results Out of 1387 COVID-19 patients, 52% were males. The median age was 60 years ((IQR)=(19-102)). The most common symptoms were dyspnoea (943,68%), cough (889, 64%), fever (597,43%) and fatigue (570, 41%). In-hospital mortality was (476, 34%). Mortality significantly increased with increasing age, being the most in age >90 years (aHR (95% CI)=4.4 (2.52 to 28.82), p=0.02). Other predictors of mortality were not possessing a health insurance, (aHR (95% CI)=3.7 (1.09 to 14.25), p=0.04); chest pain, (aHR (95% CI)=2.27 (1.36 to 4.13), p=0.03); HIV positivity, (aHR (95% CI)=3.9 (1.46 to 8.15), p=0.03); neutrophilia, (aHR (95% CI)=1.12 (1.01 to 2.65), p=0.03); no use of ivermectin, (aHR (95% CI)=1.21 (1.04 to 1.57), p=0.04) and non-use of steroids, (aHR (95% CI)=1.36 (1.18 to 2.78), p=0.04). The retrospective nature of this study which based on documented patients' records, with a large number of patients left out of the analysis due to missed data, this might in a way affect the results of the present study. Conclusion In-hospital mortality was 34%. The independent predictors of mortality were advanced age, HIV infection, no possession of a health insurance, chest pain, neutrophilia and no use of steroids or ivermectin.
Collapse
Affiliation(s)
- Elisha Fred Otieno Osati
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Grace Ambrose Shayo
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Raphael Z Sangeda
- Department of Pharmaceutical Microbiology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
| | - Tumaini Joseph Nagu
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Candida Moshiro
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
| | - Naveeda Adams
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Athumani Ramadhani
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Bahati Wajanga
- Department of Internal Medicine, Bugando Medical Centre, Mwanza, Tanzania, United Republic of
| | - Albert Muniko
- Department of Internal Medicine, Bugando Medical Centre, Mwanza, Tanzania, United Republic of
| | - Jeremiah Seni
- Department of Internal Medicine, Catholic University of Health and Allied Sciences Bugando, Mwanza, Tanzania, United Republic of
| | - Mary A Nicholaus
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre, Moshi, Kilimanjaro, Tanzania, United Republic of
| | - Gervas Nyaisonga
- Department of Internal Medicine, Mbeya Zonal Referral Hospital, Mbeya, Tanzania, United Republic of
| | - Christian Mbije
- Department of Internal Medicine, Mbeya Zonal Referral Hospital, Mbeya, Tanzania, United Republic of
| | - John Robson Meda
- Department of Internal Medicine, University of Dodoma, Dodoma, Tanzania, United Republic of
| | - Denis Rainer
- Department of Internal Medicine, Benjamin Mkapa Hospital, Dodoma, Tanzania, United Republic of
| | - Martha Elisande Nkya
- Community, Management and Development for Health, Dar es Salaam, Tanzania, United Republic of
| | - Paulo Mhame
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Lucy Samwel
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Liggyle Vumilia
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Seif Shekalaghe
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Kajiru G Kilonzo
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre, Moshi, Kilimanjaro, Tanzania, United Republic of
| | - Abel Makubi
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| |
Collapse
|
24
|
Lefebvre M, Chahinian H, La Scola B, Fantini J. Characterization and Fluctuations of an Ivermectin Binding Site at the Lipid Raft Interface of the N-Terminal Domain (NTD) of the Spike Protein of SARS-CoV-2 Variants. Viruses 2024; 16:1836. [PMID: 39772146 PMCID: PMC11680242 DOI: 10.3390/v16121836] [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: 10/14/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Most studies on the docking of ivermectin on the spike protein of SARS-CoV-2 concern the receptor binding domain (RBD) and, more precisely, the RBD interface recognized by the ACE2 receptor. The N-terminal domain (NTD), which controls the initial attachment of the virus to lipid raft gangliosides, has not received the attention it deserves. In this study, we combined molecular modeling and physicochemical approaches to analyze the mode of interaction of ivermectin with the interface of the NTD-facing lipid rafts of the host cell membrane. We characterize a binding area that presents point mutations and deletions in successive SARS-CoV-2 variants from the initial strain to omicron KP.3 circulating in many countries in 2024. We show that ivermectin has exceptional flexibility, allowing the drug to bind to the spike protein of all variants tested. The energy of interaction is specific to each variant, allowing a classification according to their affinity for ivermectin in the following ascending order: Omicron KP.3 < Delta < Omicron BA.5 < Alpha < Wuhan (B.1) < Omicron BA.1. The binding site of ivermectin is subject to important variations of the NTD, including the Y144 deletion. It overlaps with the ganglioside binding domain of the NTD, as demonstrated by docking and physicochemical studies. These results suggest a new mechanism of antiviral action for ivermectin based on competitive inhibition for initial virus attachment to lipid rafts. The current KP.3 variant is still recognized by ivermectin, although with an affinity slightly lower than the Wuhan strain.
Collapse
Affiliation(s)
- Marine Lefebvre
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France; (M.L.); (B.L.S.)
- Microbes Evolution Phylogeny and Infections (MEPHI), Aix-Marseille Université, 27 Boulevard Jean Moulin, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille (AP-HM), 264 Rue Saint-Pierre, 13005 Marseille, France
- Department of Biology, Faculty of Medicine, Aix-Marseille University, INSERM UA16, 13015 Marseille, France;
| | - Henri Chahinian
- Department of Biology, Faculty of Medicine, Aix-Marseille University, INSERM UA16, 13015 Marseille, France;
| | - Bernard La Scola
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France; (M.L.); (B.L.S.)
- Microbes Evolution Phylogeny and Infections (MEPHI), Aix-Marseille Université, 27 Boulevard Jean Moulin, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille (AP-HM), 264 Rue Saint-Pierre, 13005 Marseille, France
| | - Jacques Fantini
- Department of Biology, Faculty of Medicine, Aix-Marseille University, INSERM UA16, 13015 Marseille, France;
| |
Collapse
|
25
|
Steenekamp EM, Liebenberg W, Lemmer HJR, Gerber M. Formulation and Ex Vivo Evaluation of Ivermectin Within Different Nano-Drug Delivery Vehicles for Transdermal Drug Delivery. Pharmaceutics 2024; 16:1466. [PMID: 39598589 PMCID: PMC11597838 DOI: 10.3390/pharmaceutics16111466] [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: 10/15/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Ivermectin gained widespread attention as the "miracle drug" during the coronavirus disease 2019 (COVID-19) pandemic. Its inclusion in the 21st World Health Organization (WHO) List of Essential Medicines is attributed to its targeted anti-helminthic response, high efficacy, cost-effectiveness and favorable safety profile. Since the late 2000s, this bio-inspired active pharmaceutical ingredient (API) gained renewed interest for its diverse therapeutic capabilities. However, producing ivermectin formulations does remain challenging due to its poor water solubility, resulting in low bioavailability after oral administration. Therefore, the transdermal drug delivery of ivermectin was considered to overcome these challenges, which are observed after oral administration. Methods: Ivermectin was incorporated in a nano-emulsion, nano-emulgel and a colloidal suspension as ivermectin-loaded nanoparticles. The nano-drug delivery vehicles were optimized, characterized and evaluated through in vitro membrane release studies, ex vivo skin diffusion studies and tape-stripping to determine whether ivermectin was successfully released from its vehicle and delivered transdermally and/or topically throughout the skin. This study concluded with cytotoxicity tests using the methyl thiazolyl tetrazolium (MTT) and neutral red (NR) assays on both human immortalized epidermal keratinocytes (HaCaT) and human immortalized dermal fibroblasts (BJ-5ta). Results: Ivermectin was successfully released from each vehicle, delivered transdermally and topically throughout the skin and demonstrated little to no cytotoxicity at concentrations that diffused through the skin. Conclusions: The type of nano-drug delivery vehicle used to incorporate ivermectin influences its delivery both topically and transdermally, highlighting the dynamic equilibrium between the vehicle, the API and the skin.
Collapse
Affiliation(s)
| | | | | | - Minja Gerber
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), North-West University, Potchefstroom 2531, South Africa; (E.M.S.); (W.L.); (H.J.R.L.)
| |
Collapse
|
26
|
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
|
27
|
Zhu S, Yoshida N, Matsushita R, Rahman MS, Kimura K. Circulation of COVID-19-Related Medicines on Japanese Websites during the COVID-19 Pandemic and Their Quality and Authenticity. Am J Trop Med Hyg 2024; 111:1097-1106. [PMID: 39288767 PMCID: PMC11542516 DOI: 10.4269/ajtmh.23-0710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 06/21/2024] [Indexed: 09/19/2024] Open
Abstract
Substandard and falsified medical products for treating COVID-19 have spread worldwide. These medicines have entered Japan through personal importation of products purchased via the Internet. In this study, we investigated the circulation of 19 COVID-19-related medicines on the Internet in Japan and evaluated the pharmaceutical quality and authenticity of 2 medicines (dexamethasone tablets and ivermectin tablets) obtained online. We purchased 23 samples of 0.5-mg dexamethasone tablets and 13 samples of 3-mg ivermectin tablets from the Internet in December 2020 and July 2022. We investigated the quality and authenticity of the obtained samples through visual observation and tested their authenticity. We conducted pharmacopoeia compliance testing (quantitative assay, content uniformity tests, and dissolution tests) using the high-performance liquid chromatography-photodiode array detector method. No prescription was ever required at the time of purchase. Visual observation revealed that most samples lacked a package insert and some samples had packaging deficiencies. In terms of authenticity, eight ivermectin samples were genuine; the authenticity of the other samples remained uncertain. Four dexamethasone samples and three ivermectin samples failed quality testing based on pharmacopeia validation standards. Our findings illustrate that dexamethasone and ivermectin tablets of poor quality are available online. It is important to increase consumer awareness and provide information about these medicines to prevent the purchase of substandard medicines via the Internet.
Collapse
Affiliation(s)
- Shu Zhu
- Medi-Quality Security Institute, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- Medicine Security Workshop, 4F Venture Business Laboratory, Kanazawa University, Kanazawa, Japan
- AI Hospital/Macro Signal Dynamics Research and Development Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Naoko Yoshida
- Medicine Security Workshop, 4F Venture Business Laboratory, Kanazawa University, Kanazawa, Japan
- AI Hospital/Macro Signal Dynamics Research and Development Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ryo Matsushita
- Clinical Pharmacy and Healthcare Sciences, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Mohammad Sofiqur Rahman
- Medi-Quality Security Institute, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kazuko Kimura
- Medi-Quality Security Institute, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- Medicine Security Workshop, 4F Venture Business Laboratory, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
28
|
Chandra H, Yadav A, Prasad R, Sagar K, Bhardwaj N, Kumar Gupta K, Singh Thakur G, Nigam M, Pezzani R, Paulo Martins de Lima J, Douglas Melo Coutinho H, Prakash Mishra A. COVID 19: Prevention and treatment through the Indian perspective. Cytokine 2024; 183:156756. [PMID: 39284260 DOI: 10.1016/j.cyto.2024.156756] [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: 07/26/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 11/20/2024]
Abstract
The most destructive period the world has experienced seems to be behind us. Not a single nation was spared by this disease, and many continue to struggle today. Even after recovering from COVID, patient may continue to experience some post-COVID effects, such as heart irregularities or a decline in lung vitality. In the past three years (2019-2022), the world has witnessed the power of a small entity, a single peculiar virus. Science initially appeared to be helpless in this regard, but due to the emergence of disease, pharmaceutics (the development of anti-covid drugs), immunology (the rapid antigen test), microbiology (the isolation of viruses from infected people), biotechnology (the development of recombinant vaccines), biochemistry (the blood profile, the D-dimer test), and biochemistry (blood profile, D-dimer test), biophysics (PCR, RT-PCR, CT Scan, MRI) had worked together to fight the disease. The results of these efforts are the development of new diagnostic techniques, possible treatment and finally the availability of vaccines against COVID-19. However, it is not proven that the treatment through the traditional medical system is directly active on SARS-CoV-2 but is instead indirectly acting on SARS-CoV-2 effects by improving symptoms derived from the viral disease. In India, the traditional system of medicine and tradition knowledge together worked in the pandemic and proved effective strategies in prevention and treatment of SARS-CoV-2. The use of effective masks, PPE kits, plasma therapy, yoga, lockdowns and social seclusion, use of modern antiviral drugs, monoclonal antibodies, herbal remedies, homoeopathy, hygienic practice, as well as the willpower of people, are all contributing to the fight against COVID. Which methods or practices will be effective against COVID nobody is aware since medical professionals who wear PPE kits do not live longer, and some people in India who remained unprotected and roamed freely were not susceptible to infection. The focus of this review is on the mode of transmission, diagnosis, preventive measures, vaccines currently under development, modern medicine developed against SARS-CoV-2, ayurvedic medicine used during pandemic, homoeopathic medicine used during pandemic, and specific yoga poses that can be used to lessen COVID-related symptoms.
Collapse
Affiliation(s)
- Harish Chandra
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India; School of Agriculture, Uttaranchal University, Dehradun 248007, Uttarakhand, India.
| | - Archana Yadav
- Department of Microbiology, Institute of Biosciences and Biotechnology, C.S.J.M. University, Kanpur 208024, Uttar Pradesh, India.
| | - Rajendra Prasad
- School of Agriculture, Uttaranchal University, Dehradun 248007, Uttarakhand, India.
| | - Kalpana Sagar
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India
| | - Nitin Bhardwaj
- Department of Zoology and Environmental Sciences, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India.
| | - Kartikey Kumar Gupta
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India.
| | - Ghanshyam Singh Thakur
- Department of Naturopathy & Yoga, H. N. B. Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand, India.
| | - Manisha Nigam
- Department of Biochemistry, H. N. B. Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand, India.
| | - Raffaele Pezzani
- Phytotherapy Lab (PhT-Lab), Endocrinology Unit, Department of Medicine (DIMED), University of Padova, via Ospedale 105, Padova 35128, Italy; AIROB, Associazione Italiana per la Ricerca Oncologica di Base, Padova, Italy.
| | | | | | - Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, Bloemfontein 9300, South Africa.
| |
Collapse
|
29
|
García-Méndez JO, Fernández-Garza LE, Vallejo-Oviedo K, Gómez-Curiel DI, Barrera-Barrera SA, Ordaz-Cuellar R, Sosa-García JO, García-Torrentera RA, Cervera E, Barrera-Saldaña HA. Clinical Experience With Ivermectin and Nitazoxanide in the Management of COVID-19 Among Mexican Out- and Inpatients. Cureus 2024; 16:e74513. [PMID: 39726468 PMCID: PMC11670897 DOI: 10.7759/cureus.74513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background and objective The use of ivermectin and nitazoxanide in the treatment of coronavirus disease 2019 (COVID-19) has been a subject of controversy. In this study, we aimed to describe our clinical experience in treating COVID-19 patients with these drugs in Mexico. Material and methods The study involved out- and inpatient clinical assessments of COVID-19 patients conducted in Mexico City from September 2020 to November 2021. Outpatients were treated with either ivermectin, nitazoxanide, or both drugs, while all inpatients received both. Clinical and laboratory analyses were used to assess the results. Results Of the 228 subjects in the outpatient group, 26.8% received ivermectin, 25.4% nitazoxanide, and 47.8% both. The proportion of negative polymerase chain reaction (PCR) was highest in patients treated late with ivermectin (≥5 days after symptom onset; p=0.004), followed by those receiving late treatment with nitazoxanide, and those with the combination at any time. The inpatient group had 179 subjects. A significant increase was seen in neutrophil, lymphocyte, monocyte, ferritin, and D-dimer levels, while an opposite trend was observed for C-reactive protein (CRP) and fibrinogen levels. Mechanical ventilation requirement was 15.5%, and 5% died during hospitalization. Conclusions Despite the limitations of our study, based on its findings, ivermectin and nitazoxanide could be useful in reducing the viral load, the requirement for mechanical ventilation, proinflammatory and procoagulant parameters, and the fatality rate in COVID-19 patients. Controlled clinical trials evaluating this combination should be carried out to determine its true usefulness and safety profile.
Collapse
Affiliation(s)
| | - Luis E Fernández-Garza
- Internal Medicine, General Hospital of Zone No. 2, Mexican Institute of Social Security, Monterrey, MEX
| | | | | | | | | | | | | | - Eduardo Cervera
- Hematology, Instituto Nacional de Cancerología, Mexico City, MEX
| | | |
Collapse
|
30
|
Mawazi SM, Fathima N, Mahmood S, Al-Mahmood SMA. Antiviral therapy for COVID-19 virus: A narrative review and bibliometric analysis. Am J Emerg Med 2024; 85:98-107. [PMID: 39244809 DOI: 10.1016/j.ajem.2024.09.001] [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: 05/25/2024] [Revised: 07/28/2024] [Accepted: 09/01/2024] [Indexed: 09/10/2024] Open
Abstract
The COVID-19 epidemic has become a major international health emergency. Millions of people have died as a result of this phenomenon since it began. Has there been any successful pharmacological treatment for COVID-19 since the initial report on the virus? How many searches are undertaken to address the impact of the infection? What is the number of drugs that have undergone investigation? What are the mechanisms of action and adverse effects associated with the investigated pharmaceuticals used to treat COVID-19? Has the Food and Drug Administration (FDA) approved any medication to treat COVID-19? To date, our understanding is based on a restricted corpus of published investigations into the treatment of COVID-19. It is important to note that no single study comprehensively encompasses all pharmacological interventions for COVID-19. This paper provides an introductory summary of a bibliometric analysis conducted on the data about COVID-19, sourced explicitly from two platforms, namely PubMed and ScienceDirect. The analysis encompasses the period spanning from 2019 to 2022. Furthermore, this study examines the published literature about the pharmacological interventions for the novel coronavirus disease 2019 (COVID-19), explicitly focusing on the safety and effectiveness of different medications such as Remdesivir (marketed as Veklury®), Lopinavir/Ritonavir (commercially known as Kaletra® or Aluvia®), Ribavirin, Favipiravir (marketed as Avigan®), Ivermectin, Casirivimab and Imdevimab (branded as Ronapreve®), Sotrovimab (marketed as Xevudy®), Anakinra, Molnupiravir, Nirmatrelvir/Ritonavir (marketed as Paxlovid®), and Galidesivir. Findings indicate that while Remdesivir and Nirmatrelvir/Ritonavir show significant efficacy in reducing hospitalization and severe outcomes, drugs like Lopinavir/Ritonavir and Ivermectin have inconsistent results. Our insights suggest a multifaceted approach incorporating these therapies can significantly improve patient outcomes. Repurposing drugs has been critical in rapidly responding to COVID-19, allowing existing medications to be used in new ways to combat the virus. Combination therapies and further research are essential to optimize treatment strategies.
Collapse
Affiliation(s)
- Saeid Mezail Mawazi
- School of Pharmacy, Management & Science University (MSU), Section 13, 40100 Shah Alam, Selangor, Malaysia.
| | - Nousheen Fathima
- Department of Pharmacology, Global College of Pharmacy, Jawaharlal Technology University, Hyderabad (Jntuh) 501504, India
| | - Syed Mahmood
- Faculty of Pharmacy, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | |
Collapse
|
31
|
Barzola FN, Laiolo J, Cotelo C, Joray MB, Volpini X, Rivero MR, Rópolo AS, Touz MC, Feliziani C. Cytotoxic effects of ivermectin on Giardia lamblia: induction of apoptosis and cell cycle arrest. Front Microbiol 2024; 15:1484805. [PMID: 39545240 PMCID: PMC11560887 DOI: 10.3389/fmicb.2024.1484805] [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: 08/22/2024] [Accepted: 10/17/2024] [Indexed: 11/17/2024] Open
Abstract
Introduction Giardia lamblia is a flagellated protozoan parasite causing giardiasis, a common intestinal infection characterized by diarrhea, abdominal cramps, and nausea. Treatments employed to combat this parasitic infection have remained unchanged for the past 40 years, leading to the emergence of resistant strains and prompting the search for new therapeutic agents. Methods This study investigated the cytotoxic effects of ivermectin (IVM) on G. lamblia trophozoites. We conducted dose-response experiments to assess IVM-induced cytotoxicity. We utilized various biochemical and ultrastructural analyses to explore the underlying mechanisms of cell death, including reactive oxygen species (ROS) production, DNA fragmentation, cell cycle arrest, and apoptosis markers. Results Our findings demonstrate that IVM induces dose-dependent cytotoxicity and triggers cell death pathways. We found that IVM treatment generates elevated levels of reactive oxygen species (ROS), DNA fragmentation, and arrests of trophozoites in the cell cycle's S phase. Additionally, ultrastructural analysis reveals morphological alterations consistent with apoptosis, such as cytoplasmic vacuolization, chromatin condensation, and tubulin distribution. Discussion The insights gained from this study may contribute to developing new therapeutic strategies against giardiasis, addressing the challenge posed by drug-resistant strains.
Collapse
Affiliation(s)
- Florencia Nicole Barzola
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Jerónimo Laiolo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
- Facultad de Ciencias de la Salud, Universidad Católica De Córdoba, Córdoba, Argentina
| | - Camilo Cotelo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Belén Joray
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE), Consejo Nacional de Investigaciones Cientí-ficas y Técnicas (CONICET)/Universidad Católica de Córdoba (UCC), Córdoba, Argentina
| | - Ximena Volpini
- Centro de Investigaciones en Bioquímica Clínica e Inmunología – Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - María Romina Rivero
- Instituto De Desarrollo Agroindustrial y De La Salud (IDAS-CONCIET), Universidad Nacional De Rio Cuarto, Rio Cuarto, Argentina
| | - Andrea Silvana Rópolo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Carolina Touz
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza Feliziani
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
32
|
Badary HA, Hashem MB, El-Kassas M. Drug-induced liver injury during the era of COVID-19 polypharmacy: a statement of account, lessons learned, and a proposed approach. EGYPTIAN LIVER JOURNAL 2024; 14:75. [DOI: 10.1186/s43066-024-00381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 10/07/2024] [Indexed: 01/03/2025] Open
Abstract
AbstractThe coronavirus disease 2019 (COVID-19) causes a systemic illness that can result in various manifestations. In addition to severe acute respiratory syndrome, patients often exhibit complications unrelated to the respiratory system. Potential liver damage can occur in 14.8 to 53.0% of the affected patients. Liver impairment in COVID-19 can also occur because of the use of polypharmacy during disease management. It is essential to be aware of drug-induced liver injury (DILI) in patients diagnosed with COVID-19, especially when considering the off-label usage of medications in both preventative and therapeutic regimens used on a wide scale. This review aims to give pertinent information regarding drugs utilized thus far in COVID-19 patients and their potential toxicity to the liver. We also present a suggested management approach to DILI in COVID-19 patients and lessons learned from the pharmacological management of this pandemic.
Collapse
|
33
|
Garcia-Atutxa I, Mondragon-Teran P, Huerta-Saquero A, Villanueva-Flores F. Advancements in monkeypox vaccines development: a critical review of emerging technologies. Front Immunol 2024; 15:1456060. [PMID: 39464881 PMCID: PMC11502315 DOI: 10.3389/fimmu.2024.1456060] [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: 06/27/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Monkeypox (mpox) is a zoonotic illness caused by the monkeypox virus (MPXV), with higher health concerns among people who are pregnant, children, and persons who are immunocompromised, including people with untreated and advanced HIV disease. Significant progress has been made in developing vaccines against mpox, yet critical challenges and limitations persist in ensuring their effectiveness, safety, and accessibility. The pertinence of this review is highlighted by the World Health Organization's declaration of a global health emergency on August 14, 2024, due to the recent mpox outbreak, underscoring the critical necessity for effective vaccine solutions in the face of a rapidly evolving virus. Here, we comprehensively analyze various vaccine platforms utilized in mpox prevention, including attenuated and non-replicating virus vaccines, viral vector-based vaccines, recombinant protein vaccines, and DNA and mRNA vaccines. We evaluate the advantages and limitations of each platform, highlighting the urgent need for ongoing research and innovation to enhance vaccine efficacy and safety. Recent advancements, such as incorporating immunostimulatory sequences, improved delivery systems, and developing polyvalent vaccines, are explored for their potential to offer broader protection against diverse orthopoxvirus strains. This work underscores the need to optimize currently available vaccines and investigate novel vaccination strategies to address future public health emergencies effectively. By focusing on these advanced methodologies, we aim to contribute to the development of robust and adaptable vaccine solutions for mpox and other related viral threats.
Collapse
Affiliation(s)
- Igor Garcia-Atutxa
- Computer Science Department, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Paul Mondragon-Teran
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| | - Alejandro Huerta-Saquero
- Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Mexico
| | - Francisca Villanueva-Flores
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| |
Collapse
|
34
|
Fei X, Li Z, Pan Z, Liang Y, Tan C, Cheng D, Yang Q. Avermectin B1 mediates antitumor activity and induces autophagy in osteosarcoma through the AMPK/ULK1 signaling pathway. Cancer Chemother Pharmacol 2024; 94:599-613. [PMID: 39235611 PMCID: PMC11438708 DOI: 10.1007/s00280-024-04695-z] [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: 12/07/2023] [Accepted: 06/30/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Osteosarcoma is the most common malignant bone tumor in children and adolescents. Conventional chemotherapy remains unsatisfactory due to drug toxicity and resistance issues. Therefore, there is an urgent need to develop more effective treatments for advanced osteosarcoma. In the current study, we focused on evaluating the anticancer efficacy of avermectin B1, a novel avermectin analog, against osteosarcoma cells. METHODS The half-inhibitory concentration of avermectin B1 was calculated in three osteosarcoma cell lines. Then, functional experiments were conducted to evaluate the effects of avermectin B1 on cell proliferation, the cell cycle, apoptosis and autophagy. Moreover, the AMPK/ULK1 signaling pathway was detected by Western blot assay. Finally, the in vivo effect of avermectin B1 on tumor growth and metastasis was investigated using the xenograft mouse model. To examine the role of the AMPK/ULK1 pathway, an AMPK-specific inhibitor (dorsomorphin) was used in combination with avermectin B1. RESULTS Avermectin B1 inhibited the proliferation of osteosarcoma cells in a dose-dependent manner based on CCK8 and colony formation assays. Then, it was found to inhibit migration and invasion by wound healing assay and cell migration and invasion assay. In addition, avermectin B1 induced osteosarcoma cell apoptosis and autophagy. In vivo, avermectin B1 effectively inhibited osteosarcoma cell growth and pulmonary metastasis. Mechanistically, avermectin B1 activated the AMPK/ULK1 pathway to exert antitumor activity in vitro and in vivo. Dorsomorphin significantly attenuated the Avermectin B1-induced antitumor activities. CONCLUSION Our study suggests that avermectin B1 is a potential agent to treat osteosarcoma cells through the AMPK/ULK1 signaling pathway.
Collapse
Affiliation(s)
- Xiang Fei
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 600, Yishan Road, Shanghai, 200233, China
| | - Zhaohui Li
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 600, Yishan Road, Shanghai, 200233, China
| | - Zhen Pan
- Department of Orthopedics, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yonghui Liang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 600, Yishan Road, Shanghai, 200233, China
| | - Chen Tan
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 600, Yishan Road, Shanghai, 200233, China
| | - Dongdong Cheng
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 600, Yishan Road, Shanghai, 200233, China.
| | - Qingcheng Yang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 600, Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
35
|
Jhanwar A, Sharma D, Das U. Unraveling the structural and functional dimensions of SARS-CoV2 proteins in the context of COVID-19 pathogenesis and therapeutics. Int J Biol Macromol 2024; 278:134850. [PMID: 39168210 DOI: 10.1016/j.ijbiomac.2024.134850] [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: 01/12/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) has emerged as the causative agent behind the global pandemic of Coronavirus Disease 2019 (COVID-19). As the scientific community strives to comprehend the intricate workings of this virus, a fundamental aspect lies in deciphering the myriad proteins it expresses. This knowledge is pivotal in unraveling the complexities of the viral machinery and devising targeted therapeutic interventions. The proteomic landscape of SARS-CoV2 encompasses structural, non-structural, and open-reading frame proteins, each playing crucial roles in viral replication, host interactions, and the pathogenesis of COVID-19. This comprehensive review aims to provide an updated and detailed examination of the structural and functional attributes of SARS-CoV2 proteins. By exploring the intricate molecular architecture, we have highlighted the significance of these proteins in viral biology. Insights into their roles and interplay contribute to a deeper understanding of the virus's mechanisms, thereby paving the way for the development of effective therapeutic strategies. As the global scientific community strives to combat the ongoing pandemic, this synthesis of knowledge on SARS-CoV2 proteins serves as a valuable resource, fostering informed approaches toward mitigating the impact of COVID-19 and advancing the frontier of antiviral research.
Collapse
Affiliation(s)
- Aniruddh Jhanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Dipika Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Uddipan Das
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
36
|
Sansone NMS, Boschiero MN, Marson FAL. Efficacy of Ivermectin, Chloroquine/Hydroxychloroquine, and Azithromycin in Managing COVID-19: A Systematic Review of Phase III Clinical Trials. Biomedicines 2024; 12:2206. [PMID: 39457519 PMCID: PMC11505156 DOI: 10.3390/biomedicines12102206] [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: 08/23/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/28/2024] Open
Abstract
Background: During the coronavirus disease (COVID)-19 pandemic several drugs were used to manage the patients mainly those with a severe phenotype. Potential drugs were used off-label and major concerns arose from their applicability to managing the health crisis highlighting the importance of clinical trials. In this context, we described the mechanisms of the three repurposed drugs [Ivermectin-antiparasitic drug, Chloroquine/Hydroxychloroquine-antimalarial drugs, and Azithromycin-antimicrobial drug]; and, based on this description, the study evaluated the clinical efficacy of those drugs published in clinical trials. The use of these drugs reflects the period of uncertainty that marked the beginning of the COVID-19 pandemic, which made them a possible treatment for COVID-19. Methods: In our review, we evaluated phase III randomized controlled clinical trials (RCTs) that analyzed the efficacy of these drugs published from the COVID-19 pandemic onset to 2023. We included eight RCTs published for Ivermectin, 11 RCTs for Chloroquine/Hydroxychloroquine, and three RCTs for Azithromycin. The research question (PICOT) accounted for P-hospitalized patients with confirmed or suspected COVID-19; I-use of oral or intravenous Ivermectin OR Chloroquine/Hydroxychloroquine OR Azithromycin; C-placebo or no placebo (standard of care); O-mortality OR hospitalization OR viral clearance OR need for mechanical ventilation OR clinical improvement; and T-phase III RCTs. Results: While studying these drugs' respective mechanisms of action, the reasons for which they were thought to be useful became apparent and are as follows: Ivermectin binds to insulin-like growth factor and prevents nuclear transportation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), therefore preventing cell entrance, induces apoptosis, and osmotic cell death and disrupts viral replication. Chloroquine/Hydroxychloroquine blocks the movement of SARS-CoV-2 from early endosomes to lysosomes inside the cell, also, this drug blocks the binding between SARS-CoV-2 and Angiotensin-Converting Enzyme (ACE)-2 inhibiting the interaction between the virus spike proteins and the cell membrane and this drug can also inhibit SARS-CoV-2 viral replication causing, ultimately, the reduction in viral infection as well as the potential to progression for a higher severity phenotype culminating with a higher chance of death. Azithromycin exerts a down-regulating effect on the inflammatory cascade, attenuating the excessive production of cytokines and inducing phagocytic activity, and acts interfering with the viral replication cycle. Ivermectin, when compared to standard care or placebo, did not reduce the disease severity, need for mechanical ventilation, need for intensive care unit, or in-hospital mortality. Only one study demonstrated that Ivermectin may improve viral clearance compared to placebo. Individuals who received Chloroquine/Hydroxychloroquine did not present a lower incidence of death, improved clinical status, or higher chance of respiratory deterioration compared to those who received usual care or placebo. Also, some studies demonstrated that Chloroquine/Hydroxychloroquine resulted in worse outcomes and side-effects included severe ones. Adding Azithromycin to a standard of care did not result in clinical improvement in hospitalized COVID-19 participants. In brief, COVID-19 was one of the deadliest pandemics in modern human history. Due to the potential health catastrophe caused by SARS-CoV-2, a global effort was made to evaluate treatments for COVID-19 to attenuate its impact on the human species. Unfortunately, several countries prematurely justified the emergency use of drugs that showed only in vitro effects against SARS-CoV-2, with a dearth of evidence supporting efficacy in humans. In this context, we reviewed the mechanisms of several drugs proposed to treat COVID-19, including Ivermectin, Chloroquine/Hydroxychloroquine, and Azithromycin, as well as the phase III clinical trials that evaluated the efficacy of these drugs for treating patients with this respiratory disease. Conclusions: As the main finding, although Ivermectin, Chloroquine/Hydroxychloroquine, and Azithromycin might have mechanistic effects against SARS-CoV-2 infection, most phase III clinical trials observed no treatment benefit in patients with COVID-19, underscoring the need for robust phase III clinical trials.
Collapse
Affiliation(s)
- Nathália Mariana Santos Sansone
- Laboratory of Molecular Biology and Genetics, Laboratory of Clinical and Molecular Microbiology, LunGuardian Research Group—Epidemiology of Respiratory and Infectious Diseases, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (N.M.S.S.); (M.N.B.)
| | - Matheus Negri Boschiero
- Laboratory of Molecular Biology and Genetics, Laboratory of Clinical and Molecular Microbiology, LunGuardian Research Group—Epidemiology of Respiratory and Infectious Diseases, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (N.M.S.S.); (M.N.B.)
- São Paulo Hospital, Federal University of São Paulo, São Paulo 04023-062, SP, Brazil
| | - Fernando Augusto Lima Marson
- Laboratory of Molecular Biology and Genetics, Laboratory of Clinical and Molecular Microbiology, LunGuardian Research Group—Epidemiology of Respiratory and Infectious Diseases, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (N.M.S.S.); (M.N.B.)
| |
Collapse
|
37
|
Nor Rashid N, Amrani L, Alwan A, Mohamed Z, Yusof R, Rothan H. Angiotensin-Converting Enzyme-2 (ACE2) Downregulation During Coronavirus Infection. Mol Biotechnol 2024:10.1007/s12033-024-01277-5. [PMID: 39266903 DOI: 10.1007/s12033-024-01277-5] [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: 10/09/2023] [Accepted: 08/29/2024] [Indexed: 09/14/2024]
Abstract
Angiotensin-converting enzyme-2 (ACE2) downregulation represents a detrimental factor in people with a baseline ACE2 deficiency associated with older age, hypertension, diabetes, and cardiovascular diseases. Human coronaviruses, including HCoV-NL63, SARS-CoV-1, and SARS CoV-2 infect target cells via binding of viral spike (S) glycoprotein to the ACE2, resulting in ACE2 downregulation through yet unidentified mechanisms. This downregulation disrupts the enzymatic activity of ACE2, essential in protecting against organ injury by cleaving and disposing of Angiotensin-II (Ang II), leading to the formation of Ang 1-7, thereby exacerbating the accumulation of Ang II. This accumulation activates the Angiotensin II type 1 receptor (AT1R) receptor, leading to leukocyte recruitment and increased proinflammatory cytokines, contributing to organ injury. The biological impacts and underlying mechanisms of ACE2 downregulation during SARS-CoV-2 infection have not been well defined. Therefore, there is an urgent need to establish a solid theoretical and experimental understanding of the mechanisms of ACE2 downregulation during SARS-CoV-2 entry and replication in the host cells. This review aims to discuss the physiological impact of ACE2 downregulation during coronavirus infection, the relationship between ACE2 decline and virus pathogenicity, and the possible mechanisms of ACE2 degradation, along with the therapeutic approaches.
Collapse
Affiliation(s)
- Nurshamimi Nor Rashid
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lina Amrani
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | | | - Zulqarnain Mohamed
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Rohana Yusof
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia.
| | - Hussin Rothan
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
- Pfizer, Pearl River, NY, USA.
| |
Collapse
|
38
|
Devi TL, Devi MM, Okram M, Singh OM. Repurposed Drugs during the Outbreak of Pandemic COVID-19: A Mini-Review on Their Molecular Structures and Hit-and-Trial Results. ACS OMEGA 2024; 9:36858-36864. [PMID: 39246499 PMCID: PMC11375728 DOI: 10.1021/acsomega.4c05357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024]
Abstract
One of the most significant threats to global public health in the 21st century is the novel coronavirus disease (COVID-19) caused by SARS-CoV-2. It rapidly turned into a global pandemic after it was identified in late 2019, and the World Health Organization announced the end of the pandemic on May 5, 2023. Current strategies for managing this disease include vaccination and repurposing antimalarial and antibiotic medications to alleviate symptoms like fever and throat pain, which are associated with acute respiratory distress syndrome (ARDS). Antiviral drugs such as chloroquine, hydroxychloroquine, azithromycin, remdesivir, and favipiravir have been repurposed for the treatment of COVID-19. They were previously recommended for treating SARS-CoV and MERS-CoV. However, the inefficacy and adverse side effects of these repurposed drugs led to a decrease in their widespread use in treating COVID-19 patients. The lack of approved drugs for combating this coronavirus and its unpredictable variants remains a significant challenge.
Collapse
Affiliation(s)
- Thangjam Linda Devi
- Department of Chemistry, Manipur University, Canchipur, Imphal 795003, India
| | | | - Monika Okram
- Department of Chemistry, Chandigarh University, Mohali, Punjab 160036, India
| | | |
Collapse
|
39
|
Vanhee C, Jacobs B, Kamugisha A, Canfyn M, Van Der Meersch H, Ceyssens B, Deconinck E, Van Hoorde K, Willocx M. Substandard and falsified ivermectin tablets obtained for self-medication during the COVID-19 pandemic as a source of potential harm. Drug Test Anal 2024; 16:957-967. [PMID: 38043940 DOI: 10.1002/dta.3618] [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: 10/11/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 12/05/2023]
Abstract
In 2019, a global viral pandemic, due to the SARS-CoV-2 virus, broke out. Soon after, the search for a vaccine and/or antiviral medicine began. One of the candidate antiviral medicines tested was ivermectin. Although several health authorities warned the public against the use of this medicine outside clinical trials, the drug was widely used at the end of 2020 and in 2021. Simultaneously, several reports started to emerge demonstrating serious adverse effects after self-medicating with ivermectin. It stands to reason that the self-administration of substandard or falsified (SF) medicines bearing harmful quality deficiencies have contributed to this phenomenon. In order to have a better view on the nature of these harmful quality deficiencies, SF ivermectin samples, intercepted in large quantities by the Belgian regulatory agencies during the period 2021-2022, were analyzed in our official medicines control laboratory. None of the samples (n = 19) were compliant to the quality criteria applicable to medicinal products. These SF products either suffered from a systematic underdosing of the active pharmaceutical ingredient or were severely contaminated with bacteria, two of which were contaminated with known pathogens that cause gastrointestinal illness upon oral intake. In addition to the direct risks of self-medicating with such a product, the improper usage and dosage of ivermectin medication might also facilitate ivermectin tolerance or resistance in parasites. This may have detrimental consequences on a global scale, certainly as the number of newly developed active pharmaceutical ingredients that can safely be used to combat parasites is rather scarce.
Collapse
Affiliation(s)
- Celine Vanhee
- Service Medicines and Health Products, Scientific Direction of Chemical and Physical Health Risks, Sciensano, Brussels, Belgium
| | - Bram Jacobs
- Service of Foodborne Pathogen, Scientific Direction of Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Angélique Kamugisha
- Service Medicines and Health Products, Scientific Direction of Chemical and Physical Health Risks, Sciensano, Brussels, Belgium
| | - Michael Canfyn
- Service Medicines and Health Products, Scientific Direction of Chemical and Physical Health Risks, Sciensano, Brussels, Belgium
| | | | - Bart Ceyssens
- Federal Agency for Medicine and Health Care Products, Brussels, Belgium
| | - Eric Deconinck
- Service Medicines and Health Products, Scientific Direction of Chemical and Physical Health Risks, Sciensano, Brussels, Belgium
| | - Koenraad Van Hoorde
- Service of Foodborne Pathogen, Scientific Direction of Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | - Marie Willocx
- Service Medicines and Health Products, Scientific Direction of Chemical and Physical Health Risks, Sciensano, Brussels, Belgium
| |
Collapse
|
40
|
Wimalawansa SJ. Unlocking insights: Navigating COVID-19 challenges and Emulating future pandemic Resilience strategies with strengthening natural immunity. Heliyon 2024; 10:e34691. [PMID: 39166024 PMCID: PMC11334859 DOI: 10.1016/j.heliyon.2024.e34691] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/22/2024] Open
Abstract
The original COVID-19 vaccines, developed against SARS-CoV-2, initially mitigated hospitalizations. Bivalent vaccine boosters were used widely during 2022-23, but the outbreaks persisted. Despite this, hospitalizations, mortality, and outbreaks involving dominant mutants like Alpha and Delta increased during winters when the population's vitamin D levels were at their lowest. Notably, 75 % of human immune cell/system functions, including post-vaccination adaptive immunity, rely on adequate circulatory vitamin D levels. Consequently, hypovitaminosis compromises innate and adaptive immune responses, heightening susceptibility to infections and complications. COVID-19 vaccines primarily target SARS-CoV-2 Spike proteins, thus offering only a limited protection through antibodies. mRNA vaccines, such as those for COVID-19, fail to generate secretory/mucosal immunity-like IgG responses, rendering them ineffective in halting viral spread. Additionally, mutations in the SARS-CoV-2 binding domain reduce immune recognition by vaccine-derived antibodies, leading to immune evasion by mutant viruses like Omicron variants. Meanwhile, the repeated administration of bivalent boosters intended to enhance efficacy resulted in the immunoparesis of recipients. As a result, relying solely on vaccines for outbreak prevention, it became less effective. Dominant variants exhibit increased affinity to angiotensin-converting enzyme receptor-2, enhancing infectivity but reducing virulence. Meanwhile, spike protein-related viral mutations do not impact the potency of widely available, repurposed early therapies, like vitamin D and ivermectin. With the re-emergence of COVID-19 and impending coronaviral pandemics, regulators and health organizations should proactively consider approval and strategic use of cost-effective adjunct therapies mentioned above to counter the loss of vaccine efficacy against emerging variants and novel coronaviruses and eliminate vaccine- and anti-viral agents-related serious adverse effects. Timely implementation of these strategies could reduce morbidity, mortality, and healthcare costs and provide a rational approach to address future epidemics and pandemics. This perspective critically reviews relevant literature, providing insights, justifications, and viewpoints into how the scientific community and health authorities can leverage this knowledge cost-effectively.
Collapse
Affiliation(s)
- Sunil J. Wimalawansa
- Medicine, Endocrinology, and Nutrition, B14 G2, De Soyza Flats, Moratuwa, Sri Lanka
| |
Collapse
|
41
|
Annamalai Subramani P, Tipthara P, Kolli SK, Nicholas J, Barnes SJ, Ogbondah MM, Kobylinski KC, Tarning J, Adams JH. Efficacy of ivermectin and its metabolites against Plasmodium falciparum liver stages in primary human hepatocytes. Antimicrob Agents Chemother 2024; 68:e0127223. [PMID: 38904389 PMCID: PMC11304735 DOI: 10.1128/aac.01272-23] [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: 10/03/2023] [Accepted: 06/01/2024] [Indexed: 06/22/2024] Open
Abstract
Ivermectin, a broad-spectrum anti-parasitic drug, has been proposed as a novel vector control tool to reduce malaria transmission by mass drug administration. Ivermectin and some metabolites have mosquito-lethal effect, reducing Anopheles mosquito survival. Ivermectin inhibits liver stage development in a rodent malaria model, but no inhibition was observed in a primate malaria model or in a human malaria challenge trial. In the liver, cytochrome P450 3A4 and 3A5 enzymes metabolize ivermectin, which may impact drug efficacy. Thus, understanding ivermectin metabolism and assessing this impact on Plasmodium liver stage development is critical. Using primary human hepatocytes (PHHs), we characterized ivermectin metabolism and evaluated the efficacy of ivermectin and its primary metabolites M1 (3″-O-demethyl ivermectin) and M3 (4-hydroxymethyl ivermectin) against Plasmodium falciparum liver stages. Two different modes of ivermectin exposure were evaluated: prophylactic mode (days 0-3 post-infection) and curative mode (days 3-5 post-infection). We used two different PHH donors and modes to determine the inhibitory concentration (IC50) of ivermectin, M1, M3, and the known anti-malarial drug pyrimethamine, with IC50 values ranging from 1.391 to 14.44, 9.95-23.71, 4.767-8.384, and 0.9073-5.416 µM, respectively. In our PHH model, ivermectin and metabolites M1 and M3 demonstrated inhibitory activity against P. falciparum liver stages in curative treatment mode (days 3-5) and marginal activity in prophylactic treatment mode (days 0-3). Ivermectin had improved efficacy when co-administered with ketoconazole, a specific inhibitor of cytochrome P450 3A4 enzyme. Further studies should be performed to examine ivermectin liver stage efficacy when co-administered with CYP3A4 inhibitors and anti-malarial drugs to understand the pharmacokinetic and pharmacodynamic drug-drug interactions that enhance efficacy against human malaria parasites in vitro.
Collapse
Affiliation(s)
- Pradeep Annamalai Subramani
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Phornpimon Tipthara
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Samantha J. Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Madison M. Ogbondah
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Kevin C. Kobylinski
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - John H. Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
42
|
Kocas M, Comoglu T, Ozkul A. Development and in vitro antiviral activity of ivermectin liposomes as a potential drug carrier system. Arch Pharm (Weinheim) 2024; 357:e2300708. [PMID: 38702288 DOI: 10.1002/ardp.202300708] [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: 12/04/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
This study aimed to assess and compare diverse formulations of ivermectin-loaded liposomes, employing lipid film hydration and ethanol injection methods. Three lipids (DOPC, SPC, and DSPC) were used in predetermined molar ratios. A total of 18 formulations were created, and a factorial design determined the optimal formulation based on particle size, polydispersity index (PDI), zeta potential, and encapsulation efficiency. The average mean particle size, PDI and zeta potential of the selected formulations (F1, F2, F7, F9, and F11) was, respectively, 196.40 ± 44.60 nm, 0.39 ± 0.09, and -40.24 ± 9.17. The encapsulation efficiency exceeded 80%, with a mean loading capacity of 4.00 ± 1.70%. In vitro studies included transmission electron microscopy, Fourier transform infrared spectroscopy, drug release, and antiviral activity assessments against SARS-CoV-2. The liposomal formulations demonstrated superior antiviral activity compared to free ivermectin, as indicated by lower IC50 values. The results of this study emphasize the effectiveness of ivermectin-loaded liposomes in inhibiting viral activity, highlighting their potential as promising candidates for antiviral therapy. The findings suggest that the strategic use of liposomes as drug carriers can significantly modulate and improve the antiviral properties of ivermectin, offering a novel approach to harnessing its full therapeutic potential. Collectively, these results provide a robust foundation for further exploration of ivermectin as a viral protection tool and optimization of its delivery mechanisms.
Collapse
Affiliation(s)
- Meryem Kocas
- Department of Pharmaceutical Technology, Selcuk University Faculty of Pharmacy, Konya, Turkey
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
- Department of Pharmaceutical Technology, Ankara University Faculty of Pharmacy, Ankara, Turkey
| | - Tansel Comoglu
- Department of Pharmaceutical Technology, Ankara University Faculty of Pharmacy, Ankara, Turkey
| | - Aykut Ozkul
- Department of Virology, Ankara University Faculty of Veterinary Medicine, Ankara, Turkey
| |
Collapse
|
43
|
Sellers RS, Dormitzer PR. Toxicologic Pathology Forum: mRNA Vaccine Safety-Separating Fact From Fiction. Toxicol Pathol 2024; 52:333-342. [PMID: 39254115 PMCID: PMC11528946 DOI: 10.1177/01926233241278298] [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: 09/11/2024]
Abstract
SARS-CoV-2 spread rapidly across the globe, contributing to the death of millions of individuals from 2019 to 2023, and has continued to be a major cause of morbidity and mortality after the pandemic. At the start of the pandemic, no vaccines or anti-viral treatments were available to reduce the burden of disease associated with this virus, as it was a novel SARS coronavirus. Because of the tremendous need, the development of vaccines to protect against COVID-19 was critically important. The flexibility and ease of manufacture of nucleic acid-based vaccines, specifically mRNA-based products, allowed the accelerated development of COVID-19 vaccines. Although mRNA-based vaccines and therapeutics had been in clinical trials for over a decade, there were no licensed mRNA vaccines on the market at the start of the pandemic. The rapid development of mRNA-based COVID-19 vaccines reduced serious complications and death from the virus but also engendered significant public concerns, which continue now, years after emergency-use authorization and subsequent licensure of these vaccines. This article summarizes and addresses some of the safety concerns that continue to be expressed about these vaccines and their underlying technology.
Collapse
Affiliation(s)
- Rani S. Sellers
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
44
|
Dela Cruz J, Lammel D, Kim SW, Bi M, Rillig M. COVID-19 pandemic-related drugs and microplastics from mask fibers jointly affect soil functions and processes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:50630-50641. [PMID: 39102138 PMCID: PMC11364614 DOI: 10.1007/s11356-024-34587-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/28/2024] [Indexed: 08/06/2024]
Abstract
The COVID-19 pandemic has led to an unprecedented increase in pharmaceutical drug consumption and plastic waste disposal from personal protective equipment. Most drugs consumed during the COVID-19 pandemic were used to treat other human and animal diseases. Hence, their nearly ubiquitous presence in the soil and the sharp increase in the last 3 years led us to investigate their potential impact on the environment. Similarly, the compulsory use of face masks has led to an enormous amount of plastic waste. Our study aims to investigate the combined effects of COVID-19 drugs and microplastics from FFP2 face masks on important soil processes using soil microcosm experiments. We used three null models (additive, multiplicative, and dominative models) to indicate potential interactions among different pharmaceutical drugs and mask MP. We found that the multiple-factor treatments tend to affect soil respiration and FDA hydrolysis more strongly than the individual treatments. We also found that mask microplastics when combined with pharmaceuticals caused greater negative effects on soil. Additionally, null model predictions show that combinations of high concentrations of pharmaceuticals and mask MP have antagonistic interactions on soil enzyme activities, while the joint effects of low concentrations of pharmaceuticals (with or without MP) on soil enzyme activities are mostly explained by null model predictions. Our study underscores the need for more attention on the environmental side effects of pharmaceutical contamination and their potential interactions with other anthropogenic global change factors.
Collapse
Affiliation(s)
- Jeane Dela Cruz
- Institute of Biology, Freie Universität Berlin, 14195, Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research, 14195, Berlin, Germany
| | - Daniel Lammel
- Institute of Biology, Freie Universität Berlin, 14195, Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research, 14195, Berlin, Germany
| | - Shin Woong Kim
- Institute of Biology, Freie Universität Berlin, 14195, Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research, 14195, Berlin, Germany
| | - Mohan Bi
- Institute of Biology, Freie Universität Berlin, 14195, Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research, 14195, Berlin, Germany
| | - Matthias Rillig
- Institute of Biology, Freie Universität Berlin, 14195, Berlin, Germany.
- Berlin-Brandenburg Institute of Advanced Biodiversity Research, 14195, Berlin, Germany.
| |
Collapse
|
45
|
Wijewickrema A, Banneheke H, Pathmeswaran A, Refai FW, Kauranaratne M, Malavige N, Jeewandara C, Ekanayake M, Samaraweera D, Thambavita D, Galappatthy P. Efficacy and safety of oral ivermectin in the treatment of mild to moderate Covid-19 patients: a multi-centre double-blind randomized controlled clinical trial. BMC Infect Dis 2024; 24:719. [PMID: 39039459 PMCID: PMC11264372 DOI: 10.1186/s12879-024-09563-y] [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: 06/28/2023] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Evidence on ivermectin as a treatment for Covid-19 is controversial. A Cochrane review concluded that the efficacy and safety of ivermectin is uncertain (evidence up to April 2022) and WHO recommended its use only in the setting of clinical trials. This study aimed to assess the efficacy and safety of oral ivermectin in hospitalized patients with mild to moderate Covid-19. TRIAL DESIGN AND METHODS A double-blind, randomized placebo-controlled clinical trial was conducted among RT-PCR-confirmed, adults, hospitalised within the first four days of symptoms. Patients received oral ivermectin 24 mg or placebo daily for five days. RT-PCR was repeated on days five and ten. Clinical progression was monitored using the World Health Organization Clinical Progression Scale. Serum ivermectin levels were measured on days three, five, and seven. The primary outcome was the difference in the viral load between day zero and ten in the two groups. RESULTS Out of 1699 patients screened, 249 underwent randomization and 127 received ivermectin, and 122 placebo. D10 median viral load for E gene (IQR) was 2,000 copies/mL (100 - 20,500) with ivermectin (n = 80) and 4,100 copies/mL (1,000-65,600) with placebo (n = 81, p = 0.028), per protocol analysis. The difference in Log viral load between day zero and ten between ivermectin and placebo was 3.72 and 2.97 respectively (p = 0.022). There was no significant difference in the WHO clinical progression scale or the adverse effects. Ivermectin blood levels taken before or with meals were not significantly different. Only 7 and 17 patients achieved blood levels above 160ng/ML and 100ng/ML respectively and they did not achieve a significantly lower viral load. CONCLUSION Although ivermectin resulted in statistically significant lower viral load in patients with mild to moderate Covid-19, it had no significant effect on clinical symptoms. TRIAL REGISTRATION NUMBER SLCTR/2021/020, Sri Lanka Clinical Trials Registry. 19/07/2021.
Collapse
Affiliation(s)
| | - Hasini Banneheke
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka.
- North Wales Medical School, Bangor University, Bangor, UK.
| | | | | | | | - Neelika Malavige
- Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Chandima Jeewandara
- Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | | | - Dhanusha Thambavita
- Department of Pharmacology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | | |
Collapse
|
46
|
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
|
47
|
Bhimraj A, Morgan RL, Shumaker AH, Baden L, Cheng VCC, Edwards KM, Gallagher JC, Gandhi RT, Muller WJ, Nakamura MM, O’Horo JC, Shafer RW, Shoham S, Murad MH, Mustafa RA, Sultan S, Falck-Ytter Y. Infectious Diseases Society of America Guidelines on the Treatment and Management of Patients With COVID-19 (September 2022). Clin Infect Dis 2024; 78:e250-e349. [PMID: 36063397 PMCID: PMC9494372 DOI: 10.1093/cid/ciac724] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 02/07/2023] Open
Abstract
There are many pharmacologic therapies that are being used or considered for treatment of coronavirus disease 2019 (COVID-19), with rapidly changing efficacy and safety evidence from trials. The objective was to develop evidence-based, rapid, living guidelines intended to support patients, clinicians, and other healthcare professionals in their decisions about treatment and management of patients with COVID-19. In March 2020, the Infectious Diseases Society of America (IDSA) formed a multidisciplinary guideline panel of infectious disease clinicians, pharmacists, and methodologists with varied areas of expertise to regularly review the evidence and make recommendations about the treatment and management of persons with COVID-19. The process used a living guideline approach and followed a rapid recommendation development checklist. The panel prioritized questions and outcomes. A systematic review of the peer-reviewed and grey literature was conducted at regular intervals. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach was used to assess the certainty of evidence and make recommendations. Based on the most recent search conducted on 31 May 2022, the IDSA guideline panel has made 32 recommendations for the treatment and management of the following groups/populations: pre- and postexposure prophylaxis, ambulatory with mild-to-moderate disease, and hospitalized with mild-to-moderate, severe but not critical, and critical disease. As these are living guidelines, the most recent recommendations can be found online at: https://idsociety.org/COVID19guidelines. At the inception of its work, the panel has expressed the overarching goal that patients be recruited into ongoing trials. Since then, many trials were conducted that provided much-needed evidence for COVID-19 therapies. There still remain many unanswered questions as the pandemic evolved, which we hope future trials can answer.
Collapse
Affiliation(s)
- Adarsh Bhimraj
- Division of Infectious Diseases, Houston Methodist Hospital, Houston, Texas
| | - Rebecca L Morgan
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Case Western Reserve University, School of Medicine, Cleveland, Ohio
| | - Amy Hirsch Shumaker
- Department of Medicine, Case Western Reserve University, School of Medicine, Cleveland, Ohio
- VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | | | - Vincent Chi Chung Cheng
- Queen Mary Hospital, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kathryn M Edwards
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center,Nashville, Tennessee
| | - Jason C Gallagher
- Department of Pharmacy Practice, Temple University, Philadelphia, Pennsylvania
| | - Rajesh T Gandhi
- Infectious Diseases Division, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts
| | - William J Muller
- Division of Pediatric Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago and Northwestern University, Chicago, Illinois
| | - Mari M Nakamura
- Antimicrobial Stewardship Program and Division of Infectious Diseases, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - John C O’Horo
- Division of Infectious Diseases, Joint Appointment Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota
| | - Robert W Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Palo Alto, California
| | - Shmuel Shoham
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - M Hassan Murad
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, Minnesota
| | - Reem A Mustafa
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Shahnaz Sultan
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis VA Healthcare System, Minneapolis, Minnesota
| | - Yngve Falck-Ytter
- Department of Medicine, Case Western Reserve University, School of Medicine, Cleveland, Ohio
- VA Northeast Ohio Healthcare System, Cleveland, Ohio
| |
Collapse
|
48
|
Mikamo H, Takahashi S, Yamagishi Y, Hirakawa A, Harada T, Nagashima H, Noguchi C, Masuko K, Maekawa H, Kashii T, Ohbayashi H, Hosokawa S, Maejima K, Yamato M, Manosuthi W, Paiboonpol S, Suganami H, Tanigawa R, Kawamura H. Efficacy and safety of ivermectin in patients with mild COVID-19 in Japan and Thailand. J Infect Chemother 2024; 30:536-543. [PMID: 38154616 DOI: 10.1016/j.jiac.2023.12.012] [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: 10/24/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Ivermectin is an antiparasitic drug administered to hundreds of millions of people worldwide. Fundamental research suggests that ivermectin is effective against coronavirus disease 2019 (COVID-19); therefore, we investigated the efficacy and safety of ivermectin as a COVID-19 treatment option. METHODS This multi-regional (Japan and Thailand), multicenter, placebo-controlled, randomized, double-blind, parallel-group, Phase III study evaluated the efficacy and safety of ivermectin in patients with mild COVID-19 (IVERMILCO Study). The participants took a specified number of the investigational product (ivermectin or placebo) tablets of, adjusted to a dose of 0.3-0.4 mg/kg, orally on an empty stomach once daily for three days. The primary efficacy endpoint was the time at which clinical symptoms first showed an improving trend by 168 h after investigational product administration. RESULTS A total of 1030 eligible participants were assigned to receive the investigational product; 502 participants received ivermectin and 527 participants received a placebo. The primary efficacy endpoint was approximately 96 h (approximately four days) for both ivermectin and placebo groups, which did not show statistically significant difference (stratified log-rank test, p = 0.61). The incidence of adverse events and adverse drug reactions did not show statistically significant differences between the ivermectin and placebo groups (chi-square test, p = 0.97, p = 0.59). CONCLUSIONS The results show that ivermectin (0.3-0.4 mg/kg), as a treatment for patients with mild COVID-19, is ineffective; however, its safety has been confirmed for participants, including minor participants of 12 years or older (IVERMILCO Study ClinicalTrials.gov number, NCT05056883.).
Collapse
Affiliation(s)
- Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University, 1-1, Yazakokarimata Nagakute-shi, Aichi, 480-1195, Japan.
| | - Satoshi Takahashi
- Department of Infection Control and Laboratory Medicine, Sapporo Medical University School of Medicine, 16-291, Minami1-jonishi, Chuo-ku, Sapporo-shi, Hokkaido, 060-8543, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Kochi Medical School, Kochi University, 185-1, Okocho-Kohasu, Nankoku-shi, Kochi, 783-8505, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Toshiyuki Harada
- Center for Respiratory Diseases, Department of Pulmonary Medicine, Japan Community Healthcare Organization Hokkaido Hospital, 1-8-3-18, Nakanoshima, Toyohira-ku, Sapporo, Hokkaido, 062-8618, Japan
| | | | - Chiaki Noguchi
- Sumida General Clinic, 3-4-8, Taihei, Sumida-ku, Tokyo, 130-0012, Japan
| | - Kentaro Masuko
- Shimamura Memorial Hospital, 2-4-1, Sekimachi-Kita, Nerima-ku, Tokyo, 177-0051, Japan
| | - Hiromitsu Maekawa
- Maekawa Medical Clinic, 1-1-8, Kitasaiwai, Nishi-ku, Yokohama-shi, Kanagawa, 220-0004, Japan
| | - Tatsuhiko Kashii
- Department of Oncology, Japan Organization of Occupational Health and Safety Toyama Rosai Hospital, 992, Rokuromaru, Uozu-shi, Toyama, 937-0042, Japan
| | - Hiroyuki Ohbayashi
- Department of Allergy and Respiratory Medicine, Tohno Chuo Clinic, 1-14-1, Matsugasecho, Mizunami-shi, Gifu, 509-6134, Japan
| | - Shinichiro Hosokawa
- Hosokawa Surgical Clinic, 1-75-2, Nishikomenocho, Nakamura-ku, Nagoya-shi, Aichi, 453-0812, Japan
| | - Katsuyuki Maejima
- Department of Internal Medicine, Diabetes Medicine, Maejima Clinic, 1-15-1, Midoricho, Showa-ku, Nagoya-shi, Aichi, 466-0013, Japan
| | - Masaya Yamato
- Department of General Internal Medicine, Infectious Disease, Rinku General Medical Center, 2-23, Rinku-Oraikita, Izumisano-shi, Osaka, 598-8577, Japan
| | - Weerawat Manosuthi
- Bamrasnaradura Infectious Diseases Institute, Ministry of Public Health, 38 Moo 4, Talat Khwan, Tiwanon Road, Mueang District, Nonthaburi 11000, Thailand
| | - Supachai Paiboonpol
- Department of Medicine, Ratchaburi Hospital, 85 Somboonkul Road, Na Mueang Subdistrict Muang District, Ratchaburi Provinc 70000, Thailand
| | - Hideki Suganami
- Global Data Science Center, Kowa Company, Ltd., 4-14, 3-Chome, Nihonbashi-Honcho Chuo-ku, Tokyo, 103-8433, Japan
| | - Ryohei Tanigawa
- Global Clinical Development Department, Kowa Company, Ltd., 4-14, 3-Chome, Nihonbashi-Honcho Chuo-ku, Tokyo, 103-8433, Japan
| | - Hitoshi Kawamura
- Medical Writing Department, Kowa Company, Ltd., 4-14, 3-Chome, Nihonbashi-Honcho Chuo-ku, Tokyo, 103-8433, Japan
| |
Collapse
|
49
|
Israr J, Alam S, Kumar A. Drug repurposing for respiratory infections. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:207-230. [PMID: 38942538 DOI: 10.1016/bs.pmbts.2024.03.033] [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: 06/30/2024]
Abstract
Respiratory infections such as Coronavirus disease 2019 are a substantial worldwide health challenge, frequently resulting in severe sickness and death, especially in susceptible groups. Conventional drug development for respiratory infections faces obstacles such as extended timescales, substantial expenses, and the rise of resistance to current treatments. Drug repurposing is a potential method that has evolved to quickly find and reuse existing medications for treating respiratory infections. Drug repurposing utilizes medications previously approved for different purposes, providing a cost-effective and time-efficient method to tackle pressing medical needs. This chapter summarizes current progress and obstacles in repurposing medications for respiratory infections, focusing on notable examples of repurposed pharmaceuticals and their probable modes of action. The text also explores the significance of computational approaches, high-throughput screening, and preclinical investigations in identifying potential candidates for repurposing. The text delves into the significance of regulatory factors, clinical trial structure, and actual data in confirming the effectiveness and safety of repurposed medications for respiratory infections. Drug repurposing is a valuable technique for quickly increasing the range of treatments for respiratory infections, leading to better patient outcomes and decreasing the worldwide disease burden.
Collapse
Affiliation(s)
- Juveriya Israr
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India; Department of Biotechnology, Era University, Lucknow, Uttar Pradesh, India
| | - Shabroz Alam
- Department of Biotechnology, Era University, Lucknow, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Mandhana, Kanpur, Uttar Pradesh, India.
| |
Collapse
|
50
|
Pérez-Jacoiste Asín MA, De Castro M, De Dios B, Pérez-Ayala A, Lalueza A, García-Reyne A, Losada I, Herrero-Martínez JM, Jiménez PH, Lumbreras C, Lizasoain M, López Medrano F. Enhancing screening adherence for strongyloides infection in latinx inpatients with COVID-19: a local protocol implementation study. Pathog Glob Health 2024; 118:232-240. [PMID: 37525476 PMCID: PMC11221480 DOI: 10.1080/20477724.2023.2240088] [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: 08/02/2023] Open
Abstract
Strongyloides stercoralis hyperinfection syndrome has been observed in immunosuppressed coronavirus disease 2019 (COVID-19) patients. Detecting and treating asymptomatic Strongyloides infection in individuals from endemic areas can effectively prevent hyperinfection. Unfortunately, many clinicians are unaware of this neglected infection. Therefore, we aimed to evaluate whether including Strongyloides screening in COVID-19 management protocols would encourage this practice. To accomplish this, we conducted a retrospective single-center study at 'Hospital Universitario 12 de Octubre' in Madrid, Spain, comparing two consecutive cohorts. The first cohort comprised all Latinx patients over 18 years old who were admitted for COVID-19 between March 1st and April 30th, 2020. The second cohort consisted of Latinx patients admitted between July 1st and December 31st, 2020, following an amendment to the COVID-19 management protocol that recommended screening for strongyloidiasis in at-risk patients. We identified 559 and 795 patients in the first and second periods, respectively. The percentage of individuals screened increased significantly from 8.8% to 51.6% after the screening recommendation was included in the protocol (odds ratio [OR] 11.08, 95% confidence interval [CI] 8.01-15.33). In both periods, the screening rate was significantly higher among those receiving immunosuppression than those who did not receive steroids and/or tocilizumab. No other factors influenced the screening rate. In conclusion, including strongyloidiasis screening recommendations in COVID-19 management protocols led to its increased implementation. However, the overall screening rate remained low, emphasizing the need for further efforts to enhance screening practices.
Collapse
Affiliation(s)
- María Asunción Pérez-Jacoiste Asín
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Medicine, Universidad Complutense, Madrid, Spain
| | - Marta De Castro
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Begoña De Dios
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ana Pérez-Ayala
- Department of Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Antonio Lalueza
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Medicine, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Infecciosas (CIBERINFEC; CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana García-Reyne
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Irene Losada
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- Barcelona Institute for Global Health, ISGlobal-Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Juan María Herrero-Martínez
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Medicine, Universidad Complutense, Madrid, Spain
| | | | - Carlos Lumbreras
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Medicine, Universidad Complutense, Madrid, Spain
| | - Manuel Lizasoain
- Unit of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Francisco López Medrano
- School of Medicine, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Infecciosas (CIBERINFEC; CB21/13/00009), Instituto de Salud Carlos III, Madrid, Spain
- Unit of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|