1
|
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
|
2
|
Fu Q, Liu Y, Peng C, Muluh TA, Anayyat U, Liang L. Recent Advancement in Inhaled Nano-drug Delivery for Pulmonary, Nasal, and Nose-to-brain Diseases. Curr Drug Deliv 2025; 22:3-14. [PMID: 38275044 DOI: 10.2174/0115672018268047231207105652] [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: 08/25/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 01/27/2024]
Abstract
Pulmonary, nasal, and nose-to-brain diseases involve clinical approaches, such as bronchodilators, inhaled steroids, oxygen therapy, antibiotics, antihistamines, nasal steroids, decongestants, intranasal drug delivery, neurostimulation, and surgery to treat patients. However, systemic medicines have serious adverse effects, necessitating the development of inhaled formulations that allow precise drug delivery to the airways with minimum systemic drug exposure. Particle size, surface charge, biocompatibility, drug capacity, and mucoadhesive are unique chemical and physical features that must be considered for pulmonary and nasal delivery routes due to anatomical and permeability considerations. The traditional management of numerous chronic diseases has a variety of drawbacks. As a result, targeted medicine delivery systems that employ nanotechnology enhancer drug efficiency and optimize the overall outcome are created. The pulmonary route is one of the most essential targeted drug delivery systems because it allows the administering of drugs locally and systemically to the lungs, nasal cavity, and brain. Furthermore, the lungs' beneficial characteristics, such as their ability to inhibit first-pass metabolism and their thin epithelial layer, help treat several health complications. The potential to serve as noninvasive self-administration delivery sites of the lung and nasal routes is discussed in this script. New methods for treating respiratory and some systemic diseases with inhalation have been explored and highlight particular attention to using specialized nanocarriers for delivering various drugs via the nasal and pulmonary pathways. The design and development of inhaled nanomedicine for pulmonary, nasal, and respiratory medicine applications is a potential approach for clinical translation.
Collapse
Affiliation(s)
- Qiuxia Fu
- Department of General Medicine, Luzhou People's Hospital, Luzhou 646000, Sichuan, China, (PRC)
| | - Yangjie Liu
- Department of General Medicine, Luzhou People's Hospital, Luzhou 646000, Sichuan, China, (PRC)
| | - Cao Peng
- Department of General Medicine, Luzhou People's Hospital, Luzhou 646000, Sichuan, China, (PRC)
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Umer Anayyat
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Liu Liang
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan, China PRC
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Qi Q, Pang J, Chen Y, Tang Y, Wang H, Gul S, Sun Y, Tang W, Sheng M. Targeted Drug Screening Leveraging Senescence-Induced T-Cell Exhaustion Signatures in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:11232. [PMID: 39457014 PMCID: PMC11508728 DOI: 10.3390/ijms252011232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most prevalent cancer and a leading cause of cancer-related mortality globally, with most patients diagnosed at advanced stages and facing limited early treatment options. This study aimed to identify characteristic genes associated with T-cell exhaustion due to senescence in hepatocellular carcinoma patients, elucidating the interplay between senescence and T-cell exhaustion. We constructed prognostic models based on five signature genes (ENO1, STMN1, PRDX1, RAN, and RANBP1) linked to T-cell exhaustion, utilizing elastic net regression. The findings indicate that increased expression of ENO1 in T cells may contribute to T-cell exhaustion and Treg infiltration in hepatocellular carcinoma. Furthermore, molecular docking was employed to screen small molecule compounds that target the anti-tumor effects of these exhaustion-related genes. This study provides crucial insights into the diagnosis and treatment of hepatocellular carcinoma, establishing a strong foundation for the development of predictive biomarkers and therapeutic targets for affected patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wenru Tang
- Laboratory of Molecular Genetics of Aging & Tumor, Medicine School, Kunming University of Science and Technology, Kunming 650500, China; (Q.Q.); (J.P.); (Y.C.); (Y.T.); (H.W.); (S.G.); (Y.S.)
| | - Miaomiao Sheng
- Laboratory of Molecular Genetics of Aging & Tumor, Medicine School, Kunming University of Science and Technology, Kunming 650500, China; (Q.Q.); (J.P.); (Y.C.); (Y.T.); (H.W.); (S.G.); (Y.S.)
| |
Collapse
|
6
|
Jiang X, Li D, Maghsoudloo M, Zhang X, Ma W, Fu J. Targeting furin, a cellular proprotein convertase, for COVID-19 prevention and therapeutics. Drug Discov Today 2024; 29:104026. [PMID: 38762086 DOI: 10.1016/j.drudis.2024.104026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
SARS-CoV-2 has triggered an international outbreak of the highly contagious acute respiratory disease known as COVID-19. Identifying key targets in the virus infection lifecycle is crucial for developing effective prevention and therapeutic strategies against it. Furin is a serine endoprotease that belongs to the family of proprotein convertases and plays a critical role in the entry of host cells by SARS-CoV-2. Furin can cleave a specific S1/S2 site, PRRAR, on the spike protein of SARS-CoV-2, which promotes viral transmission by facilitating membrane fusion. Hence, targeting furin could hold clinical implications for the prevention and treatment of COVID-19. This review offers an overview of furin's structure, substrates, function, and inhibitors, with a focus on its potential role in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xia Jiang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China; Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau
| | - Dabing Li
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China; School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Xinghai Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Wenzhe Ma
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau.
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China; Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
7
|
Abou-Elnour FS, El-Habashy SE, Essawy MM, Abdallah OY. Codelivery of ivermectin and methyl dihydrojasmonate in nanostructured lipid carrier for synergistic antileukemia therapy. Int J Pharm 2024; 656:124086. [PMID: 38580074 DOI: 10.1016/j.ijpharm.2024.124086] [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: 02/03/2024] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Chronic myeloid leukemia is a life-threatening blood-cancer prevalent among children and adolescents. Research for innovative therapeutics combine drug-repurposing, phytotherapeutics and nanodrug-delivery. Ivermectin (Ivn) is a potent anthelmintic, repurposed for antileukemic-activity. However, Ivn exerts off-target toxicity. Methyl-dihydrojasmonate (MJ) is a phytochemical of known antileukemic potential. Herein, we developed for the first-time Ivn/MJ-coloaded nanostructured-lipid-carrier (Ivn@MJ-NLC) for leveraging the antileukemic-activity of the novel Ivn/MJ-combination while ameliorating possible adverse-effects. The developed Ivn@MJ-NLC possessed optimum-nanosize (97 ± 12.70 nm), PDI (0.33 ± 0.02), entrapment for Ivn (97.48 ± 1.48 %) and MJ (99.48 ± 0.57 %) and controlled-release of Ivn (83 % after 140 h) and MJ (80.98 ± 2.45 % after 48 h). In-vitro K562 studies verified Ivn@MJ-NLC prominent cytotoxicity (IC50 = 35.01 ± 2.23 µg/mL) with pronounced Ivn/MJ-synergism (combination-index = 0.59) at low-concentrations (5-10 µg/mL Ivn). Superior Ivn@MJ-NLC cytocompatibility was established on oral-epithelial-cells (OEC) with high OEC/K562 viability-ratio (1.49-1.85). The innovative Ivn@MJ-NLC enhanced K562-nuclear-fragmentation and afforded upregulation of caspase-3 and BAX (1.71 ± 0.07 and 1.45 ± 0.07-fold-increase, respectively) compared to control. Ex-vivo hemocompatibility and in-vivo-biocompatibility of parenteral-Ivn@MJ-NLC, compared to Ivn-solution, was verified via biochemical-blood analysis, histological and histomorphometric studies of liver and kidney tissues. Our findings highlight Ivn@MJ-NLC as an Ivn/MJ synergistic antileukemic platform, ameliorating possible adverse-effects.
Collapse
Affiliation(s)
- Fatma S Abou-Elnour
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Salma E El-Habashy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Marwa M Essawy
- Department of Oral Pathology, Faculty of Dentistry, Alexandria University, Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
8
|
Aboras SI, Megahed AA, El-Yazbi F, Maher HM. Utility of sustainable ratio derivative spectrophotometry for the concurrent assay of synergistic repurposed drugs for COVID-19 infections; Insilico pharmacokinetics proof. BMC Chem 2024; 18:50. [PMID: 38454503 PMCID: PMC10921645 DOI: 10.1186/s13065-024-01147-w] [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: 10/12/2023] [Accepted: 02/19/2024] [Indexed: 03/09/2024] Open
Abstract
The cutting-edge combination of fluvoxamine (FVM) and ivermectin (IVM) has been presented as a proposed dosage form for the treatment of COVID-19 infections in early diagnosed patients. The main objective of this work is to develop simple, sensitive, and efficient methods for the synchronous quantification of FVM and IVM without any prior separation. Four green UV-methods were employed for the synchronous quantification, namely: Fourier functions convolution of absorption spectra, FFAS, Fourier functions convolution of derivative spectra of absorption curves, FFDS, Fourier function convolution of ratio spectra of absorption curves, FFRS and the dual-wavelength method, DWM. FFRS and DWM approaches can be able to reconcile the two components' significantly interfering spectrum presented in this commixture. Good linearity was checked in the range of 5-40, and 2.5-25 μg/mL for the FVM, and IVM, respectively. All approaches developed have been recommended in compliance with ICH principles. Furthermore, the approaches' greenness was predestined by "National Environmental Method Index" (NEMI), "Analytical GREEnness metric (AGREE)", the "Analytical Eco-Scale", and the "Green Analytical Procedure Index" (GAPI). In addition, spider diagram was utilized for the assessment of the greenness index of the solvent used. Beside greenness, the sustainability of our methods was investigated using the HEXAGON tool. Continuing the constant pursuit of greenness, drug-drug interactions (DDIs) between FVM & IVM were predicted by insilico tools to ensure the safety of the suggested mixture as a preliminary step before invitro and in vivo studies. Because they were deemed sustainable, affordable, and successful, the suggested UV-methods may be used for routine quality control investigations of the indicated formulations FVM & IVM.
Collapse
Affiliation(s)
- Sara I Aboras
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, University of Alexandria, Al-mesallah, Alexandria, 21521, Egypt.
| | - Ahmed A Megahed
- Al-Basra Health Unit, Alamriya Medical Area, Ministry of Health, Alexandria, Egypt
| | - Fawzy El-Yazbi
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, University of Alexandria, Al-mesallah, Alexandria, 21521, Egypt
| | - Hadir M Maher
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, University of Alexandria, Al-mesallah, Alexandria, 21521, Egypt
| |
Collapse
|
9
|
Zure D, David Kuo HW, Drizo A. Insights of phytoremediation mechanisms for viruses based on in-vitro, in-vivo and in-silico assessments of selected herbal plants. CHEMOSPHERE 2024; 351:141101. [PMID: 38171396 DOI: 10.1016/j.chemosphere.2023.141101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/24/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
Waterborne pathogenic viruses present unrelenting challenges to the global health and wastewater treatment industry. Phytoremediation offers promising solutions for wastewater treatment through plant-based technologies. This study investigated antiviral mechanisms in-vivo using bacteriophages MS2 and T4 as surrogates for effective herbs screened in-vitro from three embryophytes (Ocimum basilicum, Mentha sp., Plectranthus amboinicus), two macrophytes (Eichhornia crassipes, Pistia stratiotes) and a perennial grass (Cyperus rotundas). In-silico virtual screening predicted antiviral phytochemicals for further antiviral potency assessment. Results suggested in-vitro antiviral activities of embryophytes and macrophytes were higher (43-62%) than grass (21-26%). O. basilicum (OB, 57-62%) and P. stratiotes (PS, 59-60%) exhibited the highest antiviral activities. In-vivo tests showed notable virus reduction (>60%) in culture solution, attributed to rhizofiltration (66-74%) and phytoinactivation/phytodegradation (63-84%). In-silico analysis identified rutin as a primary antiviral phytochemical for MS2 (-9.7 kcal/mol) and T4 (-10.9 kcal/mol), correlating with dose-response inactivation (∼58-62%). In-vivo tests suggested additional phytocompounds may contribute to viral inactivation, presenting new opportunities for herb-based wastewater treatment solutions. Consequently, this study not only demonstrates the antiviral capabilities of OB and PS but also introduces an innovative approach for addressing viral contaminants in water.
Collapse
Affiliation(s)
- Diaiti Zure
- Department of Environmental Science and Engineering
| | | | - Aleksandra Drizo
- Sustainable Science and Management Program, Tunghai University, Taiwan
| |
Collapse
|
10
|
Kamal MF, Youssef RM, Morshedy S, El-Sayed NW. Green and Smart Quantitative Quality Control for Veterinary Mixture of Ivermectin and Clorsulon: Ecological Evaluation of Spectral Analyses via Analytical Eco-Scale, Green Analytical Procedure Index, and Analytical GREEnness Metric Approaches. J AOAC Int 2023; 106:1455-1463. [PMID: 37647651 DOI: 10.1093/jaoacint/qsad098] [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: 06/30/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND The global financial market is still highly threatened by bovine fasciolosis, a parasitic infection that targets cattle, mainly in tropical regions. Binary combination of ivermectin (IVER) and clorsulon (CLO), in challenging concentration ratios, is typically indicated for treatment and control of fasciolosis. OBJECTIVE The present study aims at smart simultaneous spectrophotometric assay of both compounds at their high ratio in marketed formulation and synthetic mixtures, without any prior separation. Furthermore, their greenness profile was evaluated and compared with previous reported assay methods, including the official one. METHODS Mathematical-based proposed methods are the dual-wavelength, induced dual-wavelength, and first derivative ratio methods. Each is developed, optimized, and applied to determine simultaneously IVER and CLO at linear ranges of 1-30 and 5-40 μg/mL, respectively. They have been validated according to ICH guidelines. Statistical Student t-tests and F-tests compared the proposed methods with a USP chromatographic technique. Ecological appraisal is accomplished using three independent metrics: Analytical Eco-Scale (AES), Green Analytical Procedure Index (GAPI), and Analytical GREEnness Metric Approach (AGREE). RESULTS Satisfactory recoveries, ICH compliance, and adherence of proposed methods to the ecological safety margin are achieved. CONCLUSIONS Developed methods are eco-friendly and cost-effective and can accomplish a routine quantitative quality control for concurrent determination of both drugs. HIGHLIGHTS Veterinary antimicrobials need analytical quality control using safer and green methodologies. Data manipulated spectral analyses of IVER and CLO, in a ratio of 1:10% (v/v), are developed and optimized. AES, GAPI, and AGREE approaches illustrate the high green compliance in respect to assays reported in the literature. Furthermore, the United States Pharmacopeia (USP) assay for IVER and CLO in injectable dosage form depends on analysis of each drug separately in the presence of the other drug, but it cannot determine both drugs simultaneously.
Collapse
Affiliation(s)
- Miranda F Kamal
- Damanhour University, Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, Beheira 25511, Egypt
| | - Rasha M Youssef
- Alexandria University, Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, El-Messalah, Alexandria 21521, Egypt
| | - Samir Morshedy
- Damanhour University, Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, Beheira 25511, Egypt
| | - Nehal W El-Sayed
- College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Pharmaceutical Sciences (Pharmaceutical Chemistry) Department, Alexandria 1029, Egypt
| |
Collapse
|
11
|
Liu H, Chen J, Shao W, Yan S, Ding S. Efficacy and Safety of Novel Oral Antivirals in Hospitalized COVID-19 Patients: A Network Meta-Analysis of Randomized Clinical Trials. Clin Epidemiol 2023; 15:1041-1053. [PMID: 37933389 PMCID: PMC10625770 DOI: 10.2147/clep.s422386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/26/2023] [Indexed: 11/08/2023] Open
Abstract
OBJECTIVE Numerous pharmacological interventions are now under investigation for the treatment of the 2019 coronavirus pandemic (COVID-19), and the evidence is rapidly evolving. Our aim is to evaluate the comparative efficacy and safety of these drugs. METHODS We searched for randomized clinical trials (RCTs) on the efficacy and safety of novel oral antivirals for the treatment of hospitalized COVID-19 patients until November 30, 2022, including baricitinib, ivermectin (IVM), favipiravir (FVP), chloroquine (CQ), lopinavir and ritonavir (LPV/RTV), hydroxychloroquine (HCQ), and hydroxychloroquine plus azithromycin (HCQ+AZT). The main outcomes of this network meta-analysis (NMA) were in-hospital mortality, adverse event (AE), recovery time, and improvement in peripheral capillary oxygen saturation (SpO2). For dichotomous results, the odds ratio (OR) was used, and the 95% confidence interval (CI) was determined. We also used meta-regression to explore whether different treatments affected efficacy and safety. STATA 15.0 was used to conduct the NMA. The research protocol was registered with PROSPERO (#CRD 42023415743). RESULTS Thirty-six RCTs, with 33,555 hospitalized COVID-19 patients, were included in this analysis. First, we compared the efficacy of different novel oral antivirals. Baricitinib (OR 0.56, 95% CI: 0.35 to 0.90) showed the highest probability of being the optimal probiotic species in reducing in-hospital mortality and suggested that none of the interventions reduced AE better than placebo. In terms of safety outcomes, IVM ranked first in improving the recovery time of hospitalized COVID-19 patients (mean difference (MD) -1.36, 95% CI: -2.32 to -0.39). In addition, patients were most likely to increase SpO2 (OR 1.77, 95% CI: 0.09 to 3.45). The meta-regression revealed no significant differences between participants using different novel oral antivirals in all outcomes in hospitalized COVID-19 patients. CONCLUSION Currently, baricitinib has reduced in-hospital mortality in hospitalized COVID-19 patients, with moderate certainty of evidence. IVM appeared to be a safer option than placebo in improving recovery time, while FVP was associated with increased SpO2 safety outcomes. These preliminary evidence-based observations should guide clinical practice until more data are made public.
Collapse
Affiliation(s)
- Haoshuang Liu
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Jingfeng Chen
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Weihao Shao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Su Yan
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Suying Ding
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| |
Collapse
|
12
|
Wang C, Chen Y, Chen X, Hu C, Chen J, Guo A. Evaluation of Antiviral Activity of Ivermectin against Infectious Bovine Rhinotracheitis Virus in Rabbit Model. Animals (Basel) 2023; 13:3164. [PMID: 37893888 PMCID: PMC10603647 DOI: 10.3390/ani13203164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Infectious bovine rhinotracheitis (IBR) caused by bovine herpes virus 1 (BoHV-1) can lead to enormous economic losses in the cattle industry. Vaccine immunization is preferentially used to decrease its transmission speed and resultant clinical signs, rather than to completely stop viral infection. Therefore, a drug effective in treating IBR is urgently needed. Our previous work demonstrated that ivermectin significantly inhibited viral replication in a cell infection model. This study aimed to investigate its antiviral effects in vivo by using a rabbit infection model. The viral inhibition assay was first used to confirm that ivermectin at low concentrations (6-25 nM) could reduce viral titers (TCID50) significantly (p < 0.001) at 24 h post-infection. In rabbits, ivermectin was administrated with one to three doses, based on the recommended anti-parasite treatment dosage (0.2 mg/kg bodyweight) through subcutaneous injection at different days post-infection in the treated IBRV infection groups, while non-treated infection group was used as the control. The infected rabbits showed hyperthermia and other clinical signs, but the number of high-fever rabbits in the ivermectin treatment groups was significantly lower than that in the non-treated infection group. Furthermore, in ivermectin treatment groups, the cumulative clinical scores correlated negatively with drug doses and positively with delay of administration time post-infection. The overall nasal shedding time in ivermectin-treated groups was two days shorter than the non-treated challenge group. At the same time point, the titer of neutralizing antibodies in the treatment group with triple doses was higher than the other two-dose groups, but the difference between the treatment groups decreased with the delay of drug administration. Correspondingly, the serious extent of lung lesions was negatively related to the dosage, but positively related to the delay of drug administration. The qPCR with tissue homogenates showed that the virus was present in both the lung tissues and trigeminals of the infected rabbits. In conclusion, ivermectin treatment had therapeutic effect by decreasing clinical signs and viral shedding, but could not stop virus proliferation in lung tissues and trigeminals.
Collapse
Affiliation(s)
- Chen Wang
- The National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (C.W.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ruminant Bio-Products, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Yingyu Chen
- The National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (C.W.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ruminant Bio-Products, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Xi Chen
- The National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (C.W.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ruminant Bio-Products, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Changmin Hu
- The National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (C.W.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ruminant Bio-Products, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianguo Chen
- The National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (C.W.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ruminant Bio-Products, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Aizhen Guo
- The National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (C.W.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ruminant Bio-Products, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
13
|
Xie Y, Jin C, Sang H, Liu W, Wang J. Ivermectin Protects Against Experimental Autoimmune Encephalomyelitis in Mice by Modulating the Th17/Treg Balance Involved in the IL-2/STAT5 Pathway. Inflammation 2023; 46:1626-1638. [PMID: 37227550 PMCID: PMC10209955 DOI: 10.1007/s10753-023-01829-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/26/2023]
Abstract
Multiple sclerosis (MS), a T-cell-mediated autoimmune disease that affects the central nervous system (CNS), is characterized by white matter demyelination, axon destruction, and oligodendrocyte degeneration. Ivermectin, an anti-parasitic drug, has anti-inflammatory, anti-tumor, and antiviral properties. However, to date, there are no in-depth studies on the effect of ivermectin on the function effector of T cells in murine experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Here, we conducted in vitro experiments and found that ivermectin inhibited the proliferation of total T cells (CD3+) and their subsets (CD4+ and CD8+ T cells) as well as T cells secreting the pro-inflammatory cytokines IFN-γ and IL-17A; ivermectin also increased IL-2 production and IL-2Rα (CD25) expression, which was accompanied by an increase in the frequency of CD4+CD25+Foxp3+ regulatory T cells (Treg). Importantly, ivermectin administration reduced the clinical symptoms of EAE mice by preventing the infiltration of inflammatory cells into the CNS. Additional mechanisms showed that ivermectin promoted Treg cells while inhibiting pro-inflammatory Th1 and Th17 cells and their IFN-γ and IL-17 secretion; ivermectin also upregulated IL-2 production from MOG35-55-stimulated peripheral lymphocytes. Finally, ivermectin decreased IFN-γ and IL-17A production and increased IL-2 level, CD25 expression, and STAT5 phosphorylation in the CNS. These results reveal a previously unknown etiopathophysiological mechanism by which ivermectin attenuates the pathogenesis of EAE, indicating that it may be a promising option for T-cell-mediated autoimmune diseases such as MS.
Collapse
Affiliation(s)
- Yu Xie
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, 115 Ximen Street, Kaifeng, 475000, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chaolei Jin
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, 115 Ximen Street, Kaifeng, 475000, China
| | - Hongzhen Sang
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, 115 Ximen Street, Kaifeng, 475000, China
| | - Wenhua Liu
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, 115 Ximen Street, Kaifeng, 475000, China
| | - Junpeng Wang
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, 115 Ximen Street, Kaifeng, 475000, China.
| |
Collapse
|
14
|
Zhao H, Wang L, Yan Y, Zhao QH, He J, Jiang R, Luo CJ, Qiu HL, Miao YQ, Gong SG, Yuan P, Wu WH. Identification of the shared gene signatures between pulmonary fibrosis and pulmonary hypertension using bioinformatics analysis. Front Immunol 2023; 14:1197752. [PMID: 37731513 PMCID: PMC10507338 DOI: 10.3389/fimmu.2023.1197752] [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: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023] Open
Abstract
Pulmonary fibrosis (PF) and pulmonary hypertension (PH) have common pathophysiological features, such as the significant remodeling of pulmonary parenchyma and vascular wall. There is no effective specific drug in clinical treatment for these two diseases, resulting in a worse prognosis and higher mortality. This study aimed to screen the common key genes and immune characteristics of PF and PH by means of bioinformatics to find new common therapeutic targets. Expression profiles are downloaded from the Gene Expression Database. Weighted gene co-expression network analysis is used to identify the co-expression modules related to PF and PH. We used the ClueGO software to enrich and analyze the common genes in PF and PH and obtained the protein-protein interaction (PPI) network. Then, the differential genes were screened out in another cohort of PF and PH, and the shared genes were crossed. Finally, RT-PCR verification and immune infiltration analysis were performed on the intersection genes. In the result, the positive correlation module with the highest correlation between PF and PH was determined, and it was found that lymphocyte activation is a common feature of the pathophysiology of PF and PH. Eight common characteristic genes (ACTR2, COL5A2, COL6A3, CYSLTR1, IGF1, RSPO3, SCARNA17 and SEL1L) were gained. Immune infiltration showed that compared with the control group, resting CD4 memory T cells were upregulated in PF and PH. Combining the results of crossing characteristic genes in ImmPort database and RT-PCR, the important gene IGF1 was obtained. Knocking down IGF1 could significantly reduce the proliferation and apoptosis resistance in pulmonary microvascular endothelial cells, pulmonary smooth muscle cells, and fibroblasts induced by hypoxia, platelet-derived growth factor-BB (PDGF-BB), and transforming growth factor-β1 (TGF-β1), respectively. Our work identified the common biomarkers of PF and PH and provided a new candidate gene for the potential therapeutic targets of PF and PH in the future.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Materials and Chemistry & Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Yan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin-Hua Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing He
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ci-Jun Luo
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hong-Ling Qiu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu-Qing Miao
- School of Materials and Chemistry & Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China
| | - Su-Gang Gong
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen-Hui Wu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
15
|
Arshad S, Raza S, Rafique R, Altaf I, Sattar A. Lack of antiviral activity of ivermectin against foot-and-mouth disease virus serotype O in BALB/c mice. Microb Pathog 2023; 182:106245. [PMID: 37422171 DOI: 10.1016/j.micpath.2023.106245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/10/2023]
Abstract
Ivermectin is an FDA approved drug and showed in vitro antiviral activity against different serotypes of Foot-and-mouth disease virus (FMDV). We here assessed the effect of ivermectin in 12 day old female BALB/c mice infected with 50LD50 FMDV serotype O intraperitoneally. Initially FMDV was adopted on 3-day old BALB/c mice by blind passages. After successful adaptation of virus mice showed hind limb paralysis. Mice were divided in 6 different groups and each group has 6 mice. Ivermectin was given at clinically prescribed dose of 500 μg/kg subcutaneously at different time interval. Ivermectin was given at 0 h post infection (hpi) and 12 hpi. Moreover we compared commercially available ivermectin with purified ivermectin preparation in sterilized DMSO. Viral load was evaluated through RT-qPCR and ELISA in different groups. Results showed that positive control and negative control has CT-value 26.28 and 38 respectively. Treated groups at 0hpi, 12hpi, purified ivermectin and pre-post treatment group has CT values 24.89, 29.44, 27.26 and 26.69 respectively that showed there was no significant reduction in virus load in treated groups as compare to positive control. In histopathology of lung tissue perialveolar capillaries were congested and alveoli were altelactic. Some emphysema was seen in alveoli and mild thickening in the alveolar wall was observed. In the alveolar epithelium mononuclear cells infiltration was seen. There was discoloration haemorrhages and enlargement of heart. Degeneration, fragmentation and loss of sarcoplasm were seen in the cardiac muscle fibers. Above results showed that ivermectin did not lessen lung and heart viral load. This study contributes that ivermectin does not have a significant antiviral effect when used in mice against FMDV serotype O, according to a growing body of research.
Collapse
Affiliation(s)
- Sheeza Arshad
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Sohail Raza
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan.
| | - Rehan Rafique
- Foot-and-mouth disease Research Center, Lahore, Pakistan
| | - Imran Altaf
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Adeel Sattar
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| |
Collapse
|
16
|
Cardoso de Souza Z, Humberto Xavier Júnior F, Oliveira Pinheiro I, de Souza Rebouças J, Oliveira de Abreu B, Roberto Ribeiro Mesquita P, de Medeiros Rodrigues F, Costa Quadros H, Manuel Fernandes Mendes T, Nguewa P, Marques Alegretti S, Paiva Farias L, Rocha Formiga F. Ameliorating the antiparasitic activity of the multifaceted drug ivermectin through a polymer nanocapsule formulation. Int J Pharm 2023; 639:122965. [PMID: 37084836 DOI: 10.1016/j.ijpharm.2023.122965] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 03/30/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023]
Abstract
Ivermectin (IVM) is a potent antiparasitic widely used in human and veterinary medicine. However, the low oral bioavailability of IVM restricts its therapeutic potential in many parasitic infections, highlighting the need for novel formulation approaches. In this study, poly(ε-caprolactone) (PCL) nanocapsules containing IVM were successfully developed using the nanoprecipitation method. Pumpkin seed oil (PSO) was used as an oily core in the developed nanocapsules. Previously, PSO was chemically analyzed by headspace solid-phase microextraction coupled to gas chromatography/mass spectrometry (HS-SPME/GC-MS). The solubility of IVM in PSO was found to be 4,266.5 ± 38.6 μg/mL. In addition, the partition coefficient of IVM in PSO/water presented a logP of 2.44. A number of nanocapsule batches were produced by factorial design resulting in an optimized formulation. Negatively charged nanocapsules measuring around 400 nm demonstrated unimodal size distribution, and presented regular spherical morphology under transmission electron microscopy. High encapsulation efficiency (98-100%) was determined by HPLC. IVM-loaded capsules were found to be stable in nanosuspensions at 4°C and 25°C, with no significant variations in particle size observed over a period of 150 days. Nanoencapsulated IVM (0.3 mM) presented reduced toxicity to J774 macrophages and L929 fibroblasts compared to free IVM. Moreover, IVM-loaded nanocapsules also demonstrated enhanced in vitro anthelmintic activity against Strongyloides venezuelensis in comparison to free IVM. Collectively, the present findings demonstrate the promising potential of PCL-PSO nanocapsules to improve the antiparasitic effects exerted by IVM.
Collapse
Affiliation(s)
- Zilyane Cardoso de Souza
- Graduate Program in Applied Cellular and Molecular Biology, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | | | - Irapuan Oliveira Pinheiro
- Graduate Program in Applied Cellular and Molecular Biology, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil
| | | | - Brenda Oliveira de Abreu
- Graduate Program in Health Sciences, University of Pernambuco (UPE), 50100-130 Recife, PE, Brazil
| | | | | | - Helenita Costa Quadros
- Gonçalo Moniz Institute (IGM), Oswaldo Cruz Foundation (FIOCRUZ), 40296-710 Salvador, BA, Brazil
| | | | - Paul Nguewa
- University of Navarra, ISTUN Institute of Tropical Health, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), 31009, Pamplona, Spain
| | - Silmara Marques Alegretti
- Departament of Animal Biology, State University of Campinas (UNICAMP), 13083-862, Campinas, SP, Brazil
| | - Leonardo Paiva Farias
- Gonçalo Moniz Institute (IGM), Oswaldo Cruz Foundation (FIOCRUZ), 40296-710 Salvador, BA, Brazil
| | - Fabio Rocha Formiga
- Graduate Program in Applied Cellular and Molecular Biology, University of Pernambuco (UPE), 50100-130, Recife, PE, Brazil; Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), 50670-420 Recife, PE, Brazil.
| |
Collapse
|
17
|
Vojdani A, Vojdani E, Saidara E, Maes M. Persistent SARS-CoV-2 Infection, EBV, HHV-6 and Other Factors May Contribute to Inflammation and Autoimmunity in Long COVID. Viruses 2023; 15:v15020400. [PMID: 36851614 PMCID: PMC9967513 DOI: 10.3390/v15020400] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
A novel syndrome called long-haul COVID or long COVID is increasingly recognized in a significant percentage of individuals within a few months after infection with SARS-CoV-2. This disorder is characterized by a wide range of persisting, returning or even new but related symptoms that involve different tissues and organs, including respiratory, cardiac, vascular, gastrointestinal, musculo-skeletal, neurological, endocrine and systemic. Some overlapping symptomatologies exist between long COVID and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Very much like with long ME/CFS, infections with herpes family viruses, immune dysregulation, and the persistence of inflammation have been reported as the most common pattern for the development of long COVID. This review describes several factors and determinants of long COVID that have been proposed, elaborating mainly on viral persistence, reactivation of latent viruses such as Epstein-Barr virus and human herpesvirus 6 which are also associated with the pathology of ME/CFS, viral superantigen activation of the immune system, disturbance in the gut microbiome, and multiple tissue damage and autoimmunity. Based on these factors, we propose diagnostic strategies such as the measurement of IgG and IgM antibodies against SARS-CoV-2, EBV, HHV-6, viral superantigens, gut microbiota, and biomarkers of autoimmunity to better understand and manage this multi-factorial disorder that continues to affect millions of people in the world.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab, Inc., Los Angeles, CA 90035, USA
- Cyrex Laboratories, LLC, Phoenix, AZ 85034, USA
- Correspondence: ; Tel.: +1-310-657-1077
| | | | - Evan Saidara
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
18
|
Ivermectin Affects Neutrophil-Induced Inflammation through Inhibition of Hydroxylysine but Stimulation of Cathepsin G and Phenylalanine Secretion. Biomedicines 2022; 10:biomedicines10123284. [PMID: 36552040 PMCID: PMC9775137 DOI: 10.3390/biomedicines10123284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The invasion and integrin-dependent adhesion of neutrophils to lung tissues and their secretion lead to the development of pneumonia in various pulmonary pathologies, including acute respiratory distress syndrome in coronavirus disease. We studied the effect of ivermectin, a possible therapeutic agent for inflammation and cancer, on integrin-dependent neutrophil adhesion to fibronectin and the concomitant secretion. Ivermectin did not affect the attachment of neutrophils to the substrate and the reactive oxygen species production but sharply inhibited the adhesion-induced release of hydroxylysine and stimulated the release of phenylalanine and cathepsin G. Hydroxylysine is a product of lysyl hydroxylase, which is overexpressed in tumor cells with an increased ability to invade and metastasize. The inhibition of hydroxylysine release by ivermectin, by analogy, may indicate the suppression of neutrophil invasion into tissue. The increase in the release of phenylalanine in our experiments coincided with the secretion of cathepsin G, which indicates the possible role of this enzyme in the cleavage of phenylalanine. What is the substrate in such a reaction is unknown. We demonstrated that exogenously added angiotensin II (1-8) can serve as a substrate for phenylalanine cleavage. Mass spectrometry revealed the formation of angiotensin II (1-7) in the secretion of neutrophils, which attached to fibronectin in the presence of ivermectin and exogenous angiotensin II (1-8), indicating a possible involvement of ivermectin in the inactivation of angiotensin II.
Collapse
|
19
|
Ivermectin Attenuates CCl 4-Induced Liver Fibrosis in Mice by Suppressing Hepatic Stellate Cell Activation. Int J Mol Sci 2022; 23:ijms232416043. [PMID: 36555680 PMCID: PMC9782196 DOI: 10.3390/ijms232416043] [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] [Received: 09/26/2022] [Revised: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Liver fibrosis, a common liver dysfunction with high morbidity and mortality rates, is the leading cause of cirrhosis and hepatocellular carcinoma, for which there are no effective therapies. Ivermectin is an antiparasitic drug that also has been showing therapeutic actions in many other diseases, including antiviral and anticancer actions, as well as treating metabolic diseases. Herein, we evaluated the function of ivermectin in regulating liver fibrosis. Firstly, carbon tetrachloride (CCl4)-injected Balb/c mice were used to assess the antifibrosis effects of ivermectin in vivo. Further, CFSC, a rat hepatic stellate cell (HSC) line, was used to explore the function of ivermectin in HSC activation in vitro. The in vivo data showed that ivermectin administration alleviated histopathological changes, improved liver function, reduced collagen deposition, and downregulated the expression of profibrotic genes. Mechanistically, the ivermectin treatment inhibited intrahepatic macrophage accumulation and suppressed the production of proinflammatory factors. Importantly, the ivermectin administration significantly decreased the protein levels of α-smooth muscle actin (α-SMA) both in vivo and in vitro, suggesting that the antifibrotic effects of ivermectin are mainly due to the promotion of HSC deactivation. The present study demonstrates that ivermectin may be a potential therapeutic agent for the prevention of hepatic fibrosis.
Collapse
|
20
|
Extracellular binding sites of positive and negative allosteric P2X4 receptor modulators. Life Sci 2022; 311:121143. [DOI: 10.1016/j.lfs.2022.121143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022]
|
21
|
Xu B, Nikolaienko O, Levchenko V, Choubey AS, Isaeva E, Staruschenko A, Palygin O. Modulation of P2X 4 receptor activity by ivermectin and 5-BDBD has no effect on the development of ARPKD in PCK rats. Physiol Rep 2022; 10:e15510. [PMID: 36353932 PMCID: PMC9647406 DOI: 10.14814/phy2.15510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 06/16/2023] Open
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is an inherited pathology caused mainly by mutations of the polycystic kidney and hepatic disease 1 (PKHD1) gene, which usually leads to end-stage renal disease. Previous studies suggested that the P2X purinoreceptor 4 (P2X4 R) may play an important role in the progression of ARPKD. To test this hypothesis, we assessed the chronic effects of ivermectin (P2X4 R allosteric modulator) and 5-BDBD (P2X4 R antagonist) on the development of ARPKD in PCK/CrljCrl-Pkhd1pck/CRL (PCK) rats. Our data indicated that activation of ATP-mediated P2X4 R signaling with ivermectin for 6 weeks in high dose (50 mg/L; water supplementation) decreased the total body weight of PCK rats while the heart and kidney weight remained unaffected. Smaller doses of ivermectin (0.5 or 5 mg/L, 6 weeks) or the inhibition of P2X4 R signaling with 5-BDBD (18 mg/kg/day, food supplement for 8 weeks) showed no effect on electrolyte balance or the basic physiological parameters. Furthermore, cystic index analysis for kidneys and liver revealed no effect of smaller doses of ivermectin (0.5 or 5 mg/L) and 5-BDBD on the cyst development of PCK rats. We observed a slight increase in the cystic liver index on high ivermectin dose, possibly due to the cytotoxicity of the drug. In conclusion, this study revealed that pharmacological modulation of P2X4 R by ivermectin or 5-BDBD does not affect the development of ARPKD in PCK rats, which may provide insights for future studies on investigating the therapeutic potential of adenosine triphosphate (ATP)-P2 signaling in PKD diseases.
Collapse
Affiliation(s)
- Biyang Xu
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
| | - Oksana Nikolaienko
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Bogomoletz Institute of PhysiologyDepartment of Cellular MembranologyKyivUkraine
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
| | | | - Elena Isaeva
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and PhysiologyUniversity of South FloridaTampaFloridaUSA
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Hypertension and Kidney Research CenterUniversity of South FloridaTampaFloridaUSA
- The James A. Haley Veterans HospitalTampaFloridaUSA
| | - Oleg Palygin
- Department of PhysiologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Division of Nephrology, Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
22
|
Cheng Q, Zhao G, Chen J, Jia Q, Fang Z. Comparative efficacy and safety of pharmacological interventions for severe COVID-19 patients: An updated network meta-analysis of 48 randomized controlled trials. Medicine (Baltimore) 2022; 101:e30998. [PMID: 36254081 PMCID: PMC9575403 DOI: 10.1097/md.0000000000030998] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/06/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To date, there has been little agreement on what drug is the "best" drug for treating severe COVID-19 patients. This study aimed to assess the efficacy and safety of different medications available at present for severe COVID-19. METHODS We searched databases for randomized controlled trials (RCTs) published up to February 28, 2022, with no language restrictions, of medications recommended for patients (aged 16 years or older) with severe COVID-19 infection. We extracted data on trials and patient characteristics, and the following primary outcomes: all-cause mortality (ACM), and treatment-emergent adverse events (TEAEs). RESULTS We identified 4021 abstracts and of these included 48 RCTs comprising 9147 participants through database searches and other sources. For decrease in ACM, we found that ivermectin/doxycycline, C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), methylprednisolone, interferon-beta/standard-of-care (SOC), interferon-beta-1b, convalescent plasma, remdesivir, lopinavir/ritonavir, immunoglobulin gamma, high dosage sarilumab (HS), auxora, and imatinib were effective when compared with placebo or SOC group. We found that colchicine and interferon-beta/SOC were only associated with the TEAEs of severe COVID-19 patients. CONCLUSION This study suggested that ivermectin/doxycycline, C-IVIG, methylprednisolone, interferon-beta/SOC, interferon-beta-1b, convalescent plasma (CP), remdesivir, lopinavir/ritonavir, immunoglobulin gamma, HS, auxora, and imatinib were efficacious for treating severe COVID-19 patients. We found that most medications were safe in treating severe COVID-19. More large-scale RCTs are still needed to confirm the results of this study.
Collapse
Affiliation(s)
- Qinglin Cheng
- Hangzhou Center for Disease Control and Prevention, Hangzhou, China
- School of Medicine, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Qinglin Cheng, Division of Infectious Diseases, Hangzhou Center for Disease Control and Prevention, 568 Mingshi Road, Hangzhou 310021, China (e-mail: )
| | - Gang Zhao
- Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Junfang Chen
- Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Qingjun Jia
- Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Zijian Fang
- Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
23
|
Manomaipiboon A, Pholtawornkulchai K, Poopipatpab S, Suraamornkul S, Maneerit J, Ruksakul W, Phumisantiphong U, Trakarnvanich T. Efficacy and safety of ivermectin in the treatment of mild to moderate COVID-19 infection: a randomized, double-blind, placebo-controlled trial. Trials 2022; 23:714. [PMID: 36028897 PMCID: PMC9412770 DOI: 10.1186/s13063-022-06649-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/07/2022] [Indexed: 12/23/2022] Open
Abstract
Background The emergent outbreak of coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has emphasized the requirement for therapeutic opportunities to overcome this pandemic. Ivermectin is an antiparasitic drug that has shown effectiveness against various agents, including SARS-CoV-2. This study aimed to assess the efficacy of ivermectin treatment compared with the standard of care (SOC) among people with mild to moderate COVID-19 symptoms. Methods In this randomized, double-blind, placebo-controlled, single-center, parallel-arm, superiority trial among adult hospitalized patients with mild to moderate COVID-19, 72 patients (mean age 48.57 ± 14.80 years) were randomly assigned to either the ivermectin (n=36) or placebo (n=36) group, along with receiving standard care. We aimed to compare the negativity of reverse transcription polymerase chain reaction (RT-PCR) result at days 7 and 14 of enrolment as the primary outcome. The secondary outcomes were duration of hospitalization, frequency of clinical worsening, survival on day 28, and adverse events. Results At days 7 and 14, no differences were observed in the proportion of PCR-positive patients (RR 0.97 at day 7 (p=0.759) and 0.95 at day 14 (p=0.813). No significant differences were found between the groups for any of the secondary endpoints, and no adverse events were reported. Conclusion No difference was found in the proportion of PCR-positive cases after treatment with ivermectin compared with standard care among patients with mild to moderate COVID-19 symptoms. However, early symptomatic recovery was observed without side effects. Trial registration ClinicalTrials.gov NCT05076253. Registered on 8 October 2021, prospectively. Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06649-3.
Collapse
Affiliation(s)
- Anan Manomaipiboon
- Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, Thailand
| | | | - Sujaree Poopipatpab
- Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, Thailand
| | - Swangjit Suraamornkul
- Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, Thailand
| | - Jakravoot Maneerit
- Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, Thailand
| | - Wiroj Ruksakul
- Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, Thailand
| | | | - Thananda Trakarnvanich
- Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Dusit, Bangkok, Thailand.
| |
Collapse
|
24
|
Barati N, Motavallihaghi S, Nikfar B, Chaichian S, Momtazi-Borojeni AA. Potential therapeutic effects of Ivermectin in COVID-19. Exp Biol Med (Maywood) 2022; 247:1388-1396. [PMID: 35686662 PMCID: PMC9442455 DOI: 10.1177/15353702221099579] [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] [Indexed: 02/03/2023] Open
Abstract
COVID-19 is a critical pandemic that affected communities around the world, and there is currently no specific drug treatment for it. The virus enters the human cells via spikes and induces cytokine production and finally arrests the cell cycle. Ivermectin shows therapeutic potential for treating COVID-19 infection based on in vitro studies. Docking studies have shown a strong affinity between Ivermectin and some virulence factors of COVID-19. Notably, clinical evidence has demonstrated that Ivermectin with usual doses is effective by both the prophylactic and therapeutic approaches in all phases of the disease. Ivermectin inhibits both the adhesion and replication of the virus. Local therapy of the lung with Ivermectin or combination therapy may get better results and decrease the dose of the drug.
Collapse
Affiliation(s)
- Nastaran Barati
- Research Center For Molecular
Medicine, Hamadan University of Medical Sciences, Hamadan 9174223425,
Iran
- Medicinal Plants and Natural
Products Research Center, Hamadan University of Medical Sciences, Hamadan
9174223425, Iran
| | | | - Banafsheh Nikfar
- Pars Advanced and Minimally
Invasive Medical Manners Research Center, Pars Hospital, Iran University of
Medical Sciences, Tehran 1415944911, Iran
| | - Shahla Chaichian
- Pars Advanced and Minimally
Invasive Medical Manners Research Center, Pars Hospital, Iran University of
Medical Sciences, Tehran 1415944911, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical
Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences,
Mashhad 8167994434, Iran
| |
Collapse
|
25
|
Singh VK, Chaurasia H, Mishra R, Srivastava R, Yadav AK, Dwivedi J, Singh P, Singh RK. COVID-19: Pathophysiology, transmission, and drug development for therapeutic treatment and vaccination strategies. Curr Pharm Des 2022; 28:2211-2233. [PMID: 35909276 DOI: 10.2174/1381612828666220729093340] [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: 12/17/2021] [Accepted: 04/04/2022] [Indexed: 11/22/2022]
Abstract
COVID-19, a dreaded and highly contagious pandemic, is flagrantly known for its rapid prevalence across the world. Till date, none of the treatments are distinctly accessible for this life-threatening disease. Under the prevailing conditions of medical emergency, one creative strategy for the identification of novel and potential antiviral agents gaining momentum in research institutions and progressively being leveraged by pharmaceutical companies is target-based drug repositioning/repurposing. A continuous monitoring and recording of results offer an anticipation that this strategy may help to reveal new medications for viral infections. This review recapitulates the neoteric illation of COVID-19, its genomic dispensation, molecular evolution via phylogenetic assessment, drug targets, the most frequently worldwide used repurposed drugs and their therapeutic applications, and a recent update on vaccine management strategies. The available data from solidarity trials exposed that the treatment with several known drugs, viz. lopinavir-ritonavir, chloroquine, hydroxychloroquine, etc had displayed various antagonistic effects along with no impactful result in diminution of mortality rate. The drugs like remdesivir, favipiravir, and ribavirin proved to be quite safer therapeutic options for treatment against COVID-19. Similarly, dexamethasone, convalescent plasma therapy and oral administration of 2DG are expected to reduce the mortality rate of COVID-19 patients.
Collapse
Affiliation(s)
- Vishal Kumar Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Himani Chaurasia
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Richa Mishra
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Ritika Srivastava
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Aditya K Yadav
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Jayati Dwivedi
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Prashant Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Ramendra K Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| |
Collapse
|
26
|
Yang S, Shen S, Hou N. Is Ivermectin Effective in Treating COVID-19? Front Pharmacol 2022; 13:858693. [PMID: 35800451 PMCID: PMC9253511 DOI: 10.3389/fphar.2022.858693] [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: 01/21/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 was first discovered in December 2019 and subsequently became a global pandemic with serious political, economic, and social implications worldwide. We urgently need to find drugs that can be effective against COVID-19. Among the many observational studies, ivermectin has attracted the attention of many countries. Ivermectin is a broad-spectrum antiparasitic drug that also has some antiviral effects. We reviewed studies related to ivermectin for the treatment of COVID-19 over the last 2 years (2019.12-2022.03) via search engines such as PubMed, Web of Science, and EBSCOhost. Seven studies showed a lower mortality rate in the ivermectin group than in the control group, six studies found that the ivermectin group had a significantly fewer length of hospitalization than the control group, and eight studies showed better negative RT-PCR responses in the IVM group than in the control group. Our systematic review indicated that ivermectin may be effective for mildly to moderately ill patients. There is no clear evidence or guidelines to recommend ivermectin as a therapeutic agent for COVID-19, so physicians should use it with caution in the absence of better alternatives in the clinical setting, and self-medication is not recommended for patients.
Collapse
Affiliation(s)
- Shuangshuang Yang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Graduate Department, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China
| | - Shan Shen
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ning Hou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
27
|
Zhang Y, Sun T, Li M, Lin Y, Liu Y, Tang S, Dai C. Ivermectin-Induced Apoptotic Cell Death in Human SH-SY5Y Cells Involves the Activation of Oxidative Stress and Mitochondrial Pathway and Akt/mTOR-Pathway-Mediated Autophagy. Antioxidants (Basel) 2022; 11:antiox11050908. [PMID: 35624772 PMCID: PMC9137967 DOI: 10.3390/antiox11050908] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 12/19/2022] Open
Abstract
Ivermectin (IVM) could cause potential neurotoxicity; however, the precise molecular mechanisms remain unclear. This study explores the cytotoxicity of IVM in human neuroblastoma (SH-SY5Y) cells and the underlying molecular mechanisms. The results show that IVM treatment (2.5–15 μM) for 24 h could induce dose-dependent cell death in SH-SY5Y cells. Compared to the control, IVM treatment significantly promoted the production of ROS, mitochondrial dysfunction, and cell apoptosis. IVM treatment also promoted mitophagy and autophagy, which were charactered by the decreased expression of phosphorylation (p)-Akt and p-mTOR proteins, increased expression of LC3II, Beclin1, ATG5, PINK, and Pakin1 proteins and autophagosome formation. N-acetylcysteine treatment significantly inhibited the IVM-induced production of ROS and cell death in SH-SY5Y cells. Autophagy inhibitor (e.g., 3-methyladenine) treatment significantly inhibited IVM-induced autophagy, oxidative stress, and cell apoptosis. Taken together, our results reveal that IVM could induce autophagy and apoptotic cell death in SH-SY5Y cells, which involved the production of ROS, activation of mitochondrial pathway, and inhibition of Akt/mTOR pathway. Autophagy inhibition improved IVM-induced oxidative stress and apoptotic cell death in SH-SY5Y cells. This current study provides new insights into understanding the molecular mechanism of IVM-induced neurotoxicity and facilitates the discovery of potential neuroprotective agents.
Collapse
Affiliation(s)
- Yuan Zhang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Tun Sun
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Meng Li
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Yanling Lin
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Yue Liu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
| | - Shusheng Tang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Correspondence: (S.T.); (C.D.)
| | - Chongshan Dai
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; (Y.Z.); (T.S.); (M.L.); (Y.L.); (Y.L.)
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- Correspondence: (S.T.); (C.D.)
| |
Collapse
|
28
|
Zheng J, Lu C, Ding Y, Zhang J, Tan F, Liu J, Yang G, Wang Y, Li Z, Yang M, Yang Y, Gong W, Gao C. Red blood cell-hitchhiking mediated pulmonary delivery of ivermectin: Effects of nanoparticle properties. Int J Pharm 2022; 619:121719. [PMID: 35390488 PMCID: PMC8978457 DOI: 10.1016/j.ijpharm.2022.121719] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 12/21/2022]
Abstract
Recent studies have demonstrated that ivermectin (IVM) exhibits antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative virus of coronavirus disease 2019 (COVID-19). However, the repurposing of IVM for the treatment of COVID-19 has presented challenges primarily due to the low IVM plasma concentration after oral administration, which was well below IC50. Here, a red blood cell (RBC)-hitchhiking strategy was used for the targeted delivery of IVM-loaded nanoparticles (NPs) to the lung. IVM-loaded poly (lactic-co-glycolic acid) (PLGA) NPs (IVM-PNPs) and chitosan-coating IVM-PNPs (IVM-CSPNPs) were prepared and adsorbed onto RBCs. Both RBC-hitchhiked IVM-PNPs and IVM-CSPNPs could significantly enhance IVM delivery to lungs, improve IVM accumulation in lung tissue, inhibit the inflammatory responses and finally significantly alleviate the progression of acute lung injury. Specifically, the redistribution and circulation effects were related to the properties of NPs. RBC-hitchhiked cationic IVM-CSPNPs showed a longer circulation time, slower accumulation and elimination rates, and higher anti-inflammatory activities than RBC-hitchhiked anionic IVM-PNPs. Therefore, RBC-hitchhiking provides an alternative strategy to improve IVM pharmacokinetics and bioavailability for repurposing of IVM to treat COVID-19. Furthermore, according to different redistribution effects of different NPs, RBC-hitchhiked NPs may achieve various accumulation rates and circulation times for different requirements of drug delivery.
Collapse
Affiliation(s)
- Jinpeng Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Caihong Lu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China; School of Pharmacy, Guangxi Medical University, Nanning 530021, PR China
| | - Yaning Ding
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China; School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Benxi 117004, PR China
| | - Jinbang Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China; College of Pharmacy, Henan University, Kaifeng 475000, PR China
| | - Fangyun Tan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China; School of Pharmacy, Guangxi Medical University, Nanning 530021, PR China
| | - Jingzhou Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Guobao Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China.
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China.
| |
Collapse
|
29
|
Reynolds DL, Simpson EB. Evaluation of ivermectin antiviral activity against avian infectious bronchitis virus using a chicken embryo model. Int J Vet Sci Med 2022; 10:19-24. [PMID: 35382155 PMCID: PMC8959520 DOI: 10.1080/23144599.2022.2050077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Ivermectin is widely used in both animals and humans as an FDA-approved parasiticide. Ivermectin has also been reported to have antiviral activity against several viruses including coronaviruses. There are reports that indicate ivermectin may have some role in diminishing the disease caused by SARS-CoV-2, but the evidence is inconclusive. The objective of this study was to determine if ivermectin was efficacious in inhibiting avian infectious bronchitis virus (IBV, a coronavirus) replication in chicken embryos. Briefly, our approach was to use the Massachusetts vaccine strain of IBV in combination with various doses of ivermectin and then inoculate these preparations into chicken embryos to determine if IBV replication was inhibited. The embryos were examined for IBV lesions and samples of chorioallantoic fluid were collected for IBV RT-PCR analysis. Several trials were performed, and the results of our study indicate that ivermectin did not inhibit IBV replication in chicken embryos.
Collapse
Affiliation(s)
- Donald L. Reynolds
- School of Veterinary Medicine and Biomedical Sciences University of Nebraska – Lincoln, Lincoln, NE, USA
| | - E. Barry Simpson
- School of Veterinary Medicine and Biomedical Sciences University of Nebraska – Lincoln, Lincoln, NE, USA
| |
Collapse
|
30
|
Osorio C, Sfera A, Anton JJ, Thomas KG, Andronescu CV, Li E, Yahia RW, Avalos AG, Kozlakidis Z. Virus-Induced Membrane Fusion in Neurodegenerative Disorders. Front Cell Infect Microbiol 2022; 12:845580. [PMID: 35531328 PMCID: PMC9070112 DOI: 10.3389/fcimb.2022.845580] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
A growing body of epidemiological and research data has associated neurotropic viruses with accelerated brain aging and increased risk of neurodegenerative disorders. Many viruses replicate optimally in senescent cells, as they offer a hospitable microenvironment with persistently elevated cytosolic calcium, abundant intracellular iron, and low interferon type I. As cell-cell fusion is a major driver of cellular senescence, many viruses have developed the ability to promote this phenotype by forming syncytia. Cell-cell fusion is associated with immunosuppression mediated by phosphatidylserine externalization that enable viruses to evade host defenses. In hosts, virus-induced immune dysfunction and premature cellular senescence may predispose to neurodegenerative disorders. This concept is supported by novel studies that found postinfectious cognitive dysfunction in several viral illnesses, including human immunodeficiency virus-1, herpes simplex virus-1, and SARS-CoV-2. Virus-induced pathological syncytia may provide a unified framework for conceptualizing neuronal cell cycle reentry, aneuploidy, somatic mosaicism, viral spreading of pathological Tau and elimination of viable synapses and neurons by neurotoxic astrocytes and microglia. In this narrative review, we take a closer look at cell-cell fusion and vesicular merger in the pathogenesis of neurodegenerative disorders. We present a "decentralized" information processing model that conceptualizes neurodegeneration as a systemic illness, triggered by cytoskeletal pathology. We also discuss strategies for reversing cell-cell fusion, including, TMEM16F inhibitors, calcium channel blockers, senolytics, and tubulin stabilizing agents. Finally, going beyond neurodegeneration, we examine the potential benefit of harnessing fusion as a therapeutic strategy in regenerative medicine.
Collapse
Affiliation(s)
- Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Adonis Sfera
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Jonathan J. Anton
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Karina G. Thomas
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Christina V. Andronescu
- Medical Anthropology – Department of Anthropology, Stanford University, Stanford, CA, United States
| | - Erica Li
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Rayan W. Yahia
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Andrea García Avalos
- Universidad Nacional Autónoma de México (UNAM), Facultad de Medicina Campus, Ciudad de Mexico, Mexico
| | - Zisis Kozlakidis
- International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
31
|
Souri M, Chiani M, Farhangi A, Mehrabi MR, Nourouzian D, Raahemifar K, Soltani M. Anti-COVID-19 Nanomaterials: Directions to Improve Prevention, Diagnosis, and Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:783. [PMID: 35269270 PMCID: PMC8912597 DOI: 10.3390/nano12050783] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023]
Abstract
Following the announcement of the outbreak of COVID-19 by the World Health Organization, unprecedented efforts were made by researchers around the world to combat the disease. So far, various methods have been developed to combat this "virus" nano enemy, in close collaboration with the clinical and scientific communities. Nanotechnology based on modifiable engineering materials and useful physicochemical properties has demonstrated several methods in the fight against SARS-CoV-2. Here, based on what has been clarified so far from the life cycle of SARS-CoV-2, through an interdisciplinary perspective based on computational science, engineering, pharmacology, medicine, biology, and virology, the role of nano-tools in the trio of prevention, diagnosis, and treatment is highlighted. The special properties of different nanomaterials have led to their widespread use in the development of personal protective equipment, anti-viral nano-coats, and disinfectants in the fight against SARS-CoV-2 out-body. The development of nano-based vaccines acts as a strong shield in-body. In addition, fast detection with high efficiency of SARS-CoV-2 by nanomaterial-based point-of-care devices is another nanotechnology capability. Finally, nanotechnology can play an effective role as an agents carrier, such as agents for blocking angiotensin-converting enzyme 2 (ACE2) receptors, gene editing agents, and therapeutic agents. As a general conclusion, it can be said that nanoparticles can be widely used in disinfection applications outside in vivo. However, in in vivo applications, although it has provided promising results, it still needs to be evaluated for possible unintended immunotoxicity. Reviews like these can be important documents for future unwanted pandemics.
Collapse
Affiliation(s)
- Mohammad Souri
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran
| | - Mohsen Chiani
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Ali Farhangi
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Mohammad Reza Mehrabi
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Dariush Nourouzian
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran 13169-43551, Iran; (M.S.); (M.C.); (A.F.)
| | - Kaamran Raahemifar
- Data Science and Artificial Intelligence Program, College of Information Sciences and Technology (IST), Penn State University, State College, PA 16801, USA;
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
- School of Optometry and Vision Science, Faculty of Science, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19967-15433, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology, Tehran 14176-14411, Iran
| |
Collapse
|
32
|
JIA Y, HAN B, WANG R. Ivermectin induces mitophagy in H9c2 cells via activation of the PINK1/Parkin pathway. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.65621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | - Rene WANG
- Jinan First People’s Hospital, China
| |
Collapse
|
33
|
Gupta SD, Nandy M, Song DG, Pan CH. Present therapeutic and diagnostic approaches for SARS-CoV-2 infection. COMPUTATIONAL APPROACHES FOR NOVEL THERAPEUTIC AND DIAGNOSTIC DESIGNING TO MITIGATE SARS-COV-2 INFECTION 2022. [PMCID: PMC9300475 DOI: 10.1016/b978-0-323-91172-6.00025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The novel Coronavirus (nCoV), severe acute respiratory syndrome-Coronavirus-2 (SARS-CoV-2), has shaken the whole world and posed significant challenges to the global healthcare system for more than a year. The scientific community across the globe is trying to combat this virus by developing a safe vaccine that can provide long-term immunity against the virus. The other means of overcoming its pathogenicity is to treat the infected people with available drugs and/or novel therapeutic strategies. The available drugs were previously designed to combat viral infections and come with tested safety. This appears to be the most practical approach as a quick response to the highly infectious pandemic with high morbidity and mortality. Although many repurposed drugs like favipiravir and hydroxychloroquine have been tried, they have been proven toxic and/or less efficacious. This has led the world to find urgent therapeutic interventions (traditional and novel), to help decrease the severity of COVID-19 infection and aim towards recovery. This chapter of the book will discuss the most up-to-date published data with respect to prevention and treatment of COVID-19 infection. Diagnosis also plays an important part in controlling the pandemic caused by the virus. A cheap, accurate and fast identification test for the virus is the need of the hour. This chapter will also throw light on the various diagnostic procedures available for the identification of SARS-CoV-2, till date, along with their advantages and disadvantages.
Collapse
|
34
|
Hashemi B, Akram FA, Amirazad H, Dadashpour M, Sheervalilou M, Nasrabadi D, Ahmadi M, Sheervalilou R, Ameri Shah Reza M, Ghazi F, Roshangar L. Emerging importance of nanotechnology-based approaches to control the COVID-19 pandemic; focus on nanomedicine iterance in diagnosis and treatment of COVID-19 patients. J Drug Deliv Sci Technol 2022; 67:102967. [PMID: 34777586 PMCID: PMC8576597 DOI: 10.1016/j.jddst.2021.102967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023]
Abstract
The ongoing outbreak of the newly emerged coronavirus disease 2019, which has tremendously concerned global health safety, is the result of infection with severe acute respiratory syndrome of coronavirus 2 with high morbidity and mortality. Because of the coronavirus has no specific treatment, so it is necessary to early detection and produce antiviral agents and efficacious vaccines in order to prevent the contagion of coronavirus. Due to the unique properties of nanomaterials, nanotechnology appears to be a highly relevant discipline in this global emergency, providing expansive chemical functionalization to develop advanced biomedical tools. Fascinatingly, nanomedicine as a hopeful approach for the treatment and diagnosis of diseases, could efficiently help success the fight among coronavirus and host cells. In this review, we will critically discuss how nanomedicine can play an indispensable role in creating useful treatments and diagnostics for coronavirus.
Collapse
Affiliation(s)
- Behnam Hashemi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Firouzi-Amandi Akram
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Amirazad
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Dadashpour
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Biotechnology Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Milad Sheervalilou
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davood Nasrabadi
- Department of Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Biotechnology Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Farhood Ghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Ding Y, Lv B, Zheng J, Lu C, Liu J, Lei Y, Yang M, Wang Y, Li Z, Yang Y, Gong W, Han J, Gao C. RBC-hitchhiking chitosan nanoparticles loading methylprednisolone for lung-targeting delivery. J Control Release 2021; 341:702-715. [PMID: 34933051 PMCID: PMC8684098 DOI: 10.1016/j.jconrel.2021.12.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/24/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022]
Abstract
Hyper-inflammation associated with cytokine storm syndrome causes high mortality in patients with COVID-19. Glucocorticoids, such as methylprednisolone sodium succinate (MPSS), effectively inhibit this inflammatory response. However, frequent and chronic administration of glucocorticoids at high doses leads to hormone dependence and serious side effects. The aim of the present study was to combine nanoparticles with erythrocytes for the targeted delivery of MPSS to the lungs. Chitosan nanoparticles loading MPSS (MPSS-CSNPs) were prepared and adsorbed on the surface of red blood cells (RBC-MPSS-CSNPs) by non-covalent interaction. In vivo pharmacokinetic study indicated that RBC-hitchhiking could significantly reduce the plasma concentration of the drug and prolong the circulation time. The mean residence time (MRT) and area under the curve (AUC) of the RBC-MPSS-CSNPs group were significantly higher than those of the MPSS-CSNPs group and the MPSS injection group. Moreover, in vivo imaging and tissue distribution indicated that RBC-hitchhiking facilitated the accumulation of nanoparticles loading fluorescein in the lung, preventing uptake of these nanoparticles by the liver. Furthermore, compared with the MPSS-CSNPs and MPSS treatment groups, treatment with RBC-MPSS-CSNPs considerably inhibited the production of inflammatory cytokines such as TNF-α and IL-6, and consequently attenuated lung injury induced by lipopolysaccharide in rats. Therefore, RBC-hitchhiking is a potentially effective strategy for the delivery of nanoparticles to the lungs for the treatment of acute lung injury and acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Yaning Ding
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110017, China; State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Bai Lv
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; School of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Jinpeng Zheng
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Caihong Lu
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jingzhou Liu
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yaran Lei
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110017, China; State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Meiyan Yang
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yuli Wang
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zhiping Li
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Yang
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wei Gong
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Jing Han
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Chunsheng Gao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110017, China; State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
36
|
Ferreira G, Santander A, Savio F, Guirado M, Sobrevia L, Nicolson GL. SARS-CoV-2, Zika viruses and mycoplasma: Structure, pathogenesis and some treatment options in these emerging viral and bacterial infectious diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166264. [PMID: 34481867 PMCID: PMC8413106 DOI: 10.1016/j.bbadis.2021.166264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/22/2021] [Accepted: 08/30/2021] [Indexed: 01/28/2023]
Abstract
The molecular evolution of life on earth along with changing environmental, conditions has rendered mankind susceptible to endemic and pandemic emerging infectious diseases. The effects of certain systemic viral and bacterial infections on morbidity and mortality are considered as examples of recent emerging infections. Here we will focus on three examples of infections that are important in pregnancy and early childhood: SARS-CoV-2 virus, Zika virus, and Mycoplasma species. The basic structural characteristics of these infectious agents will be examined, along with their general pathogenic mechanisms. Coronavirus infections, such as caused by the SARS-CoV-2 virus, likely evolved from zoonotic bat viruses to infect humans and cause a pandemic that has been the biggest challenge for humanity since the Spanish Flu pandemic of the early 20th century. In contrast, Zika Virus infections represent an expanding infectious threat in the context of global climate change. The relationship of these infections to pregnancy, the vertical transmission and neurological sequels make these viruses highly relevant to the topics of this special issue. Finally, mycoplasmal infections have been present before mankind evolved, but they were rarely identified as human pathogens until recently, and they are now recognized as important coinfections that are able to modify the course and prognosis of various infectious diseases and other chronic illnesses. The infectious processes caused by these intracellular microorganisms are examined as well as some general aspects of their pathogeneses, clinical presentations, and diagnoses. We will finally consider examples of treatments that have been used to reduce morbidity and mortality of these infections and discuss briefly the current status of vaccines, in particular, against the SARS-CoV-2 virus. It is important to understand some of the basic features of these emerging infectious diseases and the pathogens involved in order to better appreciate the contributions of this special issue on how infectious diseases can affect human pregnancy, fetuses and neonates.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay.
| | - Axel Santander
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Florencia Savio
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Mariana Guirado
- Department of Infectious Diseases, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaeology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Medical School (Faculty of Medicine), São Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (UMCG), 9713GZ Groningen, the Netherlands
| | - Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
37
|
Santos GC, Martins LM, Bregadiolli BA, Moreno VF, Silva‐Filho LC, Silva BHST. Heterocyclic compounds as antiviral drugs: Synthesis, structure–activity relationship and traditional applications. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | | | | | - Vitor Fernandes Moreno
- School of Sciences, Department of Chemistry São Paulo State University (UNESP) Bauru Brazil
| | | | | |
Collapse
|
38
|
Theodoros K, Sharma M, Anton P, Hugo C, Ellen O, Hultgren NW, Ritou E, Williams DS, Orian S S, Srinivasa T R. The ApoA-I mimetic peptide 4F attenuates in vitro replication of SARS-CoV-2, associated apoptosis, oxidative stress and inflammation in epithelial cells. Virulence 2021; 12:2214-2227. [PMID: 34494942 PMCID: PMC8437485 DOI: 10.1080/21505594.2021.1964329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/12/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
An oral antiviral against SARS-CoV-2 that also attenuates inflammatory instigators of severe COVID-19 is not available to date. Herein, we show that the apoA-I mimetic peptide 4 F inhibits Spike mediated viral entry and has antiviral activity against SARS-CoV-2 in human lung epithelial Calu3 and Vero-E6 cells. In SARS-CoV-2 infected Calu3 cells, 4 F upregulated inducers of the interferon pathway such as MX-1 and Heme oxygenase 1 (HO-1) and downregulated mitochondrial reactive oxygen species (mito-ROS) and CD147, a host protein that mediates viral entry. 4 F also reduced associated cellular apoptosis and secretion of IL-6 in both SARS-CoV-2 infected Vero-E6 and Calu3 cells. Thus, 4 F attenuates in vitro SARS-CoV-2 replication, associated apoptosis in epithelial cells and secretion of IL-6, a major cytokine related to COVID-19 morbidity. Given established safety of 4 F in humans, clinical studies are warranted to establish 4 F as therapy for COVID-19.
Collapse
Affiliation(s)
- Kelesidis Theodoros
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Madhav Sharma
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Petcherski Anton
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Cristelle Hugo
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - O’Connor Ellen
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
| | - Nan W Hultgren
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Eleni Ritou
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - David S Williams
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Shirihai Orian S
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Reddy Srinivasa T
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
39
|
Pereira CVM, Mastandrea GRA, Medeiros ACCDS, Gryschek RCB, de Paula FM, Corral MA. COVID-19 and strongyloidiasis: what to expect from this coinfection? Clinics (Sao Paulo) 2021; 76:e3528. [PMID: 34817048 PMCID: PMC8579849 DOI: 10.6061/clinics/2021/e3528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
| | | | | | - Ronaldo Cesar Borges Gryschek
- Laboratorio de Investigacao Medica (LIM06-Laboratorio de Imunopatologia da Equistossomose), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Secao de Helmintologia, Instituto de Medicina Tropical (IMT), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Fabiana Martins de Paula
- Laboratorio de Investigacao Medica (LIM06-Laboratorio de Imunopatologia da Equistossomose), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Secao de Helmintologia, Instituto de Medicina Tropical (IMT), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Marcelo Andreetta Corral
- Faculdade de Medicina, Universidade Santo Amaro, Sao Paulo, SP, BR
- Laboratorio de Investigacao Medica (LIM06-Laboratorio de Imunopatologia da Equistossomose), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Secao de Helmintologia, Instituto de Medicina Tropical (IMT), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
40
|
Zaheer T, Pal K, Abbas RZ, Torres MDPR. COVID-19 and Ivermectin: Potential threats associated with human use. J Mol Struct 2021; 1243:130808. [PMID: 34149064 PMCID: PMC8195608 DOI: 10.1016/j.molstruc.2021.130808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/01/2021] [Accepted: 05/29/2021] [Indexed: 12/24/2022]
Abstract
Drugs re-purposing due to COVID-19 virus has declared a number of useful candidates for treatment and prevention of the virus. Ivermectin (IVM) has gained much popularity due to a strong background of magical applications against a broad spectrum of pathogens. The in- vitro studies of ivermectin have shown promise, the thorough clinical trials of its efficacy in the treatment and prevention of SARS-CoV-2 are still warranted. Useful strategies for analyzing projected use of IVM in human coronaviruses might be developed. It may be done by concluding ongoing clinical trials and culturing lessons from IVM usage in veterinary practice. The potential toxicity and careful dosage analyses are urgently required before declaring it as an anti-SARS-CoV-2 drug candidate. This manuscript overviews the background and potential threats associated with the off-label use of IVM as prophylactic drug or treatment option against COVID-19 virus.
Collapse
Affiliation(s)
- Tean Zaheer
- Chemotherapy Laboratory, Department of Parasitology, University of Agriculture, Faisalabad- 38040, Pakistan
| | - Kaushik Pal
- Laboratório de Biopolímeros e Sensores, Instituto de Macromoléculas, Universidade Federal do Rio de Janeiro (LABIOS/IMA/UFRJ), Cidade Universitária, Rio de Janeiro 21941-90, Brazil
| | - Rao Zahid Abbas
- Chemotherapy Laboratory, Department of Parasitology, University of Agriculture, Faisalabad- 38040, Pakistan
| | - María Del Pilar Rodríguez Torres
- Laboratorio de Ondas de Choque (LOCH),Centro de Física Aplicada y Tecnología Avanzada, (CFATA),Universidad Nacional Autónoma de México, Campus UNAM Juriquilla Boulevard, Juriquilla no. 3001, Santiago de Querétaro, Qro., C.P. 76230, Mexico
| |
Collapse
|
41
|
Kamat S, Kumari M, Jayabaskaran C. Nano-engineered tools in the diagnosis, therapeutics, prevention, and mitigation of SARS-CoV-2. J Control Release 2021; 338:813-836. [PMID: 34478750 PMCID: PMC8406542 DOI: 10.1016/j.jconrel.2021.08.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/13/2021] [Accepted: 08/28/2021] [Indexed: 01/07/2023]
Abstract
The recent outbreak of SARS-CoV-2 has forever altered mankind resulting in the COVID-19 pandemic. This respiratory virus further manifests into vital organ damage, resulting in severe post COVID-19 complications. Nanotechnology has been moonlighting in the scientific community to combat several severe diseases. This review highlights the triune of the nano-toolbox in the areas of diagnostics, therapeutics, prevention, and mitigation of SARS-CoV-2. Nanogold test kits have already been on the frontline of rapid detection. Breath tests, magnetic nanoparticle-based nucleic acid detectors, and the use of Raman Spectroscopy present myriads of possibilities in developing point of care biosensors, which will ensure sensitive, affordable, and accessiblemass surveillance. Most of the therapeutics are trying to focus on blocking the viral entry into the cell and fighting with cytokine storm, using nano-enabled drug delivery platforms. Nanobodies and mRNA nanotechnology with lipid nanoparticles (LNPs) as vaccines against S and N protein have regained importance. All the vaccines coming with promising phase 3 clinical trials have used nano-delivery systems for delivery of vaccine-cargo, which are currently administered widely in many countries. The use of chemically diverse metal, carbon and polymeric nanoparticles, nanocages and nanobubbles demonstrate opportunities to develop anti-viral nanomedicine. In order to prevent and mitigate the viral spread, high-performance charged nanofiber filters, spray coating of nanomaterials on surfaces, novel materials for PPE kits and facemasks have been developed that accomplish over 90% capture of airborne SARS-CoV-2. Nano polymer-based disinfectants are being tested to make smart-transport for human activities. Despite the promises of this toolbox, challenges in terms of reproducibility, specificity, efficacy and emergence of new SARS-CoV-2 variants are yet to overcome.
Collapse
Affiliation(s)
- Siya Kamat
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Madhuree Kumari
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India.
| | - C Jayabaskaran
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| |
Collapse
|
42
|
Cruciani M, Pati I, Masiello F, Malena M, Pupella S, De Angelis V. Ivermectin for Prophylaxis and Treatment of COVID-19: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2021; 11:1645. [PMID: 34573986 PMCID: PMC8470309 DOI: 10.3390/diagnostics11091645] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 08/31/2021] [Accepted: 09/06/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Ivermectin has received particular attention as a potential treatment for COVID-19. However, the evidence to support its clinical efficacy is controversial. OBJECTIVES We undertook a new systematic review of ivermectin for the treatment and prophylaxis of COVID-19, including new primary studies, outcomes other than mortality, and grading the quality of the available evidence following the Cochrane guidance for methodology. METHODS We searched electronic databases, repository databases, and clinical trial registries (up to June 2021). The measure of treatment effect was risk difference (RD) with 95% confidence intervals (CIs). The GRADE system was used to assess the certainty of the evidence. RESULTS The review includes 11 RCTs (2436 participants). The certainty of the available evidence was quite low or very low due to risk of bias, inconsistency, and imprecision. When the analysis was limited to patients with baseline mild or moderate disease (8 reports, 1283 patients), there were no differences in mortality between ivermectin and control groups (low level of certainty); in patients with baseline severe diseases (3 reports, 304 patients), the use of ivermectin significantly decreased mortality compared to the controls (RD -0.17; 95% CIs, -0.24/-0.10; p = 0.00001; low level of certainty). In terms of disease progression (to severe pneumonia, admission to intensive care unit, and/or mechanical ventilation), the results were much the same. At day 14, the rate of patients with a negative RT-PCR test was 21% higher (from 5 to 36% higher) for ivermectin recipients than it was for the controls (low quality of evidence). Three studies (736 subjects) indicated that prophylaxis with ivermectin increased the likelihood of preventing COVID-19 compared to controls (low quality of evidence). Serious adverse events were rarely reported. CONCLUSIONS There is limited evidence for the benefit of ivermectin for COVID-19 treatment and prophylaxis, and most of this evidence is of low quality. Further evidence is needed to fine-tune potential indications and optimal treatment protocols for ivermectin as a treatment for COVID-19.
Collapse
Affiliation(s)
- Mario Cruciani
- Italian National Blood Centre, National Institute of Health, 00162 Rome, Italy; (I.P.); (F.M.); (S.P.); (V.D.A.)
- Infectious Diseases Unit, AULSS9 Scaligera, 37100 Verona, Italy;
| | - Ilaria Pati
- Italian National Blood Centre, National Institute of Health, 00162 Rome, Italy; (I.P.); (F.M.); (S.P.); (V.D.A.)
| | - Francesca Masiello
- Italian National Blood Centre, National Institute of Health, 00162 Rome, Italy; (I.P.); (F.M.); (S.P.); (V.D.A.)
| | - Marina Malena
- Infectious Diseases Unit, AULSS9 Scaligera, 37100 Verona, Italy;
| | - Simonetta Pupella
- Italian National Blood Centre, National Institute of Health, 00162 Rome, Italy; (I.P.); (F.M.); (S.P.); (V.D.A.)
| | - Vincenzo De Angelis
- Italian National Blood Centre, National Institute of Health, 00162 Rome, Italy; (I.P.); (F.M.); (S.P.); (V.D.A.)
| |
Collapse
|
43
|
M Mansour S, N Shamma R, A Ahmed K, A Sabry N, Esmat G, A Mahmoud A, Maged A. Safety of inhaled ivermectin as a repurposed direct drug for treatment of COVID-19: A preclinical tolerance study. Int Immunopharmacol 2021; 99:108004. [PMID: 34333358 PMCID: PMC8299187 DOI: 10.1016/j.intimp.2021.108004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Introduction SARS-CoV-2 replication in cell cultures has been shown to be inhibited by ivermectin. However, ivermectin's low aqueous solubility and bioavailability hinders its application in COVID-19 treatment. Also, it has been suggested that best outcomes for this medication can be achieved via direct administration to the lung. Objectives This study aimed at evaluating the safety of a novel ivermectin inhalable formulation in rats as a pre-clinical step. Methods Hydroxy propyl-β-cyclodextrin (HP-β-CD) was used to formulate readily soluble ivermectin lyophilized powder. Adult male rats were used to test lung toxicity for ivermectin-HP-β-CD formulations in doses of 0.05, 0.1, 0.2, 0.4 and 0.8 mg/kg for 3 successive days. Results The X-ray diffraction for lyophilized ivermectin-HP-β-CD revealed its amorphous structure that increased drug aqueous solubility 127-fold and was rapidly dissolved within 5 s in saline. Pulmonary administration of ivermectin-HP-β-CD in doses of 0.2, 0.4 and 0.8 mg/kg showed dose-dependent increase in levels of TNF-α, IL-6, IL-13 and ICAM-1 as well as gene expression of MCP-1, protein expression of PIII-NP and serum levels of SP-D paralleled by reduction in IL-10. Moreover, lungs treated with ivermectin (0.2 mg/kg) revealed mild histopathological alterations, while severe pulmonary damage was seen in rats treated with ivermectin at doses of 0.4 and 0.8 mg/kg. However, ivermectin-HP-β-CD formulation administered in doses of 0.05 and 0.1 mg/kg revealed safety profiles. Conclusion The safety of inhaled ivermectin-HP-β-CD formulation is dose-dependent. Nevertheless, use of low doses (0.05 and 0.1 mg/kg) could be considered as a possible therapeutic regimen in COVID-19 cases.
Collapse
Affiliation(s)
- Suzan M Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Rehab N Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Nirmeen A Sabry
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt
| | - Gamal Esmat
- Department of Endemic Medicine and Hepatogastroenterology, Faculty of Medicine, Cairo University, Egypt
| | - Azza A Mahmoud
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Amr Maged
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt; Pharmaceutical Factory, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| |
Collapse
|
44
|
Azhar A, Hassan N, Singh M, Al-Hosaini K, Kamal MA. Synopsis on Pharmotechnological Approaches in Diagnostic to Management Strategies in Fighting Against COVID-19. Curr Pharm Des 2021; 27:4086-4099. [PMID: 34269664 DOI: 10.2174/1381612827666210715154004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
Nanoparticles (NPs) are projected to play a significant role in fighting against coronavirus disease (COVID-19). The various properties of NPs like magnetic and optical can be exploited to build diagnostic test kits. The unembellished morphological and physiochemical resemblances of SARS-CoV-2 with synthetic NPs make them a potent tool for mediation. Nanoparticles can be analytically functionalized with different proteins, polymers, and functional groups to perform specific inhibitory functions while also serving as delivery vehicles . Moreover, NPs can also be employed to prepare broad-spectrum respiratory drugs and vaccines that can guard seasonal flu and prepare the human race for the pandemic in the future. The present review outlines the role of NPs in detection, diagnostic and therapeutic against members of the coronavirus family. We emphasize nanomaterial-based approaches to address coronaviruses in general and SARS-CoV-2 in particular. We discuss NPs based detection systems like graphene (G-FET), biosensors, and plasmonic photothermal associated sensors. Inorganic, organic virus-like & self-assembly protein (VLP), and photodynamic inactivation of SARS-CoV-2 are also presented as therapeutic approaches exploiting NPs.
Collapse
Affiliation(s)
- Asim Azhar
- Aligarh College of Education, Aligarh Uttar Pradesh, India
| | - Nazia Hassan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Manvi Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Khaled Al-Hosaini
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451. Saudi Arabia
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
45
|
The antiviral activity of iota-, kappa-, and lambda-carrageenan against COVID-19: A critical review. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2021; 12:100826. [PMID: 34222718 PMCID: PMC8240443 DOI: 10.1016/j.cegh.2021.100826] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/22/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022] Open
Abstract
Objective There is no specific antiviral treatment available for coronavirus disease 2019 (COVID-19). Among the possible natural constituents is carrageenan, a polymer derived from marine algae that possesses a variety of antiviral properties. The purpose of this review was to summarize the evidence supporting carrageenan subtypes' antiviral activity against the emerging severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of COVID-19. Methods PubMed/MEDLINE and Google Scholar searches were conducted for publications using the terms 'carrageenan', 'iota carrageenan', 'kappa carrageenan', lambda-carrageenan', 'coronavirus', 'common cold', 'rhinovirus', and 'SARS-CoV-2' search was also done in grey literature to increase our understanding. A search for the word "carrageenan" was also carried out. Most of the publications were discussed in narrative. Results Carrageenan has been shown to have potent antiviral activity against both coronaviruses (coronavirus NL63, SARS-CoV-2) and non-coronaviruses such as dengue virus, herpes simplex virus, cytomegalovirus, vaccinia virus, vesicular stomatitis virus, sindbis virus, human immunodeficiency virus, influenza virus, human papillomavirus, rabies virus, junin virus, tacaribe virus, African swine fever, bovine herpes virus, suid herpes virus, and rhinovirus. No in vivo study has been conducted using carrageenan as an anti-SARS-CoV-2 agent. The majority of the in vivo research was done on influenza, a respiratory virus that causes common cold together with coronavirus. Thus, various clinical trials were conducted to determine the transferability of these in vitro data to clinical effectiveness against SARS-CoV-2. When combined with oral ivermectin, nasally administered iota-carrageenan improved outcome in COVID-19 patients. It is still being tested in clinics for single-dose administration. Conclusion Though the carrageenan exhibited potent antiviral activity against SARS-CoV-2 and was used to treat COVID-19 under emergency protocol in conjunction with oral medications such as ivermectin, there is no solid evidence from clinical trials to support its efficacy. Thus, clinical trials are required to assess its efficacy for COVID-19 treatment prior to broad application.
Collapse
|
46
|
Attia YA, El-Saadony MT, Swelum AA, Qattan SYA, Al-Qurashi AD, Asiry KA, Shafi ME, Elbestawy AR, Gado AR, Khafaga AF, Hussein EOS, Ba-Awadh H, Tiwari R, Dhama K, Alhussaini B, Alyileili SR, El-Tarabily KA, Abd El-Hack ME. COVID-19: pathogenesis, advances in treatment and vaccine development and environmental impact-an updated review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:22241-22264. [PMID: 33733422 PMCID: PMC7969349 DOI: 10.1007/s11356-021-13018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/15/2021] [Indexed: 05/08/2023]
Abstract
Diseases negatively impact the environment, causing many health risks and the spread of pollution and hazards. A novel coronavirus, severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has led to a recent respiratory syndrome epidemic in humans. In December 2019, the sudden emergence of this new coronavirus and the subsequent severe disease it causes created a serious global health threat and hazards. This is in contrast to the two aforementioned coronaviruses, SARS-CoV-2 (in 2002) and middle east respiratory syndrome coronavirus MERS-CoV (in 2012), which were much more easily contained. The World Health Organization (WHO) dubbed this contagious respiratory disease an "epidemic outbreak" in March 2020. More than 80 companies and research institutions worldwide are working together, in cooperation with many governmental agencies, to develop an effective vaccine. To date, six authorized vaccines have been registered. Up till now, no approved drugs and drug scientists are racing from development to clinical trials to find new drugs for COVID-19. Wild animals, such as snakes, bats, and pangolins are the main sources of coronaviruses, as determined by the sequence homology between MERS-CoV and viruses in these animals. Human infection is caused by inhalation of respiratory droplets. To date, the only available treatment protocol for COVID-19 is based on the prevalent clinical signs. This review aims to summarize the current information regarding the origin, evolution, genomic organization, epidemiology, and molecular and cellular characteristics of SARS-CoV-2 as well as the diagnostic and treatment approaches for COVID-19 and its impact on global health, environment, and economy.
Collapse
Affiliation(s)
- Youssef A Attia
- Agriculture Department, Faculty of Environmental Sciences, King Abdulaziz University, P.O. Box 80208, Jeddah, 21589, Saudi Arabia.
- The Strategic Center to Kingdom Vision Realization, King Abdulaziz University, Jeddah, Saudi Arabia.
- Animal and Poultry Production Department, Faculty of Agriculture, Damanhour University, Damanhour, Egypt.
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Ayman A Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia.
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Sharkia, Zagazig, 44519, Egypt.
| | - Shaza Y A Qattan
- Department of Biological Sciences, Microbiology, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Adel D Al-Qurashi
- Agriculture Department, Faculty of Environmental Sciences, King Abdulaziz University, P.O. Box 80208, Jeddah, 21589, Saudi Arabia
| | - Khalid A Asiry
- Agriculture Department, Faculty of Environmental Sciences, King Abdulaziz University, P.O. Box 80208, Jeddah, 21589, Saudi Arabia
| | - Manal E Shafi
- Department of Biological Sciences, Zoology, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Ahmed R Elbestawy
- Poultry and Fish Diseases Department, Faculty of Veterinary Medicine, Damanhour University, Damanhur, 22511, Egypt
| | - Ahmed R Gado
- Poultry and Fish Diseases Department, Faculty of Veterinary Medicine, Damanhour University, Damanhur, 22511, Egypt
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, Alexandria, 22758, Egypt
| | - Elsayed O S Hussein
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Hani Ba-Awadh
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Kuldeep Dhama
- Division of Pathology, Indian Veterinary Research Institute (IVRI), Izatnagar-243, Bareilly, Uttar Pradesh, 122, India
| | - Bakr Alhussaini
- Department of Pediatric, Faculty of Medicine, King Abdualziz University, Jeddah, Saudi Arabia
| | - Salem R Alyileili
- Department of Integrative Agriculture, College of Food and Agriculture, United Arab Emirates University, 15551, Al-Ain, United Arab Emirates
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, 15551, Al-Ain, United Arab Emirates.
- Harry Butler Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia.
| | - Mohamed E Abd El-Hack
- Department of Poultry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
47
|
Choudhury A, Das NC, Patra R, Bhattacharya M, Ghosh P, Patra BC, Mukherjee S. Exploring the binding efficacy of ivermectin against the key proteins of SARS-CoV-2 pathogenesis: an in silico approach. Future Virol 2021. [PMCID: PMC7996102 DOI: 10.2217/fvl-2020-0342] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aim: COVID-19 is currently the biggest threat to mankind. Recently, ivermectin (a US FDA-approved antiparasitic drug) has been explored as an anti-SARS-CoV-2 agent. Herein, we have studied the possible mechanism of action of ivermectin using in silico approaches. Materials & methods: Interaction of ivermectin against the key proteins involved in SARS-CoV-2 pathogenesis were investigated through molecular docking and molecular dynamic simulation. Results: Ivermectin was found as a blocker of viral replicase, protease and human TMPRSS2, which could be the biophysical basis behind its antiviral efficiency. The antiviral action and ADMET profile of ivermectin was on par with the currently used anticorona drugs such as hydroxychloroquine and remdesivir. Conclusion: Our study enlightens the candidature of ivermectin as an effective drug for treating COVID-19.
Collapse
Affiliation(s)
- Abhigyan Choudhury
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, West Bengal, India
| | - Nabarun C Das
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, West Bengal, India
| | - Ritwik Patra
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, West Bengal, India
| | | | - Pratik Ghosh
- Department of Zoology, Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Bidhan C Patra
- Department of Zoology, Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, West Bengal, India
| |
Collapse
|
48
|
Metabolism and interactions of Ivermectin with human cytochrome P450 enzymes and drug transporters, possible adverse and toxic effects. Arch Toxicol 2021; 95:1535-1546. [PMID: 33719007 PMCID: PMC7956433 DOI: 10.1007/s00204-021-03025-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/09/2021] [Indexed: 12/23/2022]
Abstract
The review presents metabolic properties of Ivermectin (IVM) as substrate and inhibitor of human P450 (P450, CYP) enzymes and drug transporters. IVM is metabolized, both in vivo and in vitro, by C-hydroxylation and O-demethylation reactions catalyzed by P450 3A4 as the major enzyme, with a contribution of P450 3A5 and 2C9. In samples from both in vitro and in vivo metabolism, a number of metabolites were detected and as major identified metabolites were 3″-O-demethylated, C4-methyl hydroxylated, C25 isobutyl-/isopropyl-hydroxylated, and products of oxidation reactions. Ivermectin inhibited P450 2C9, 2C19, 2D6, and CYP3A4 with IC50 values ranging from 5.3 μM to no inhibition suggesting that it is no or weak inhibitor of the enzymes. It is suggested that P-gp (MDR1) transporter participate in IVM efflux at low drug concentration with a slow transport rate. At the higher, micromolar concentration range, which saturates MDR1 (P-gp), MRP1, and to a lesser extent, MRP2 and MRP3 participate in IVM transport across physiological barriers. IVM exerts a potent inhibition of P-gp (ABCB1), MRP1 (ABCC1), MRP2 (ABCC2), and BCRP1 (ABCG2), and medium to weak inhibition of OATP1B1 (SLC21A6) and OATP1B3 (SLCOB3) transport activity. The metabolic and transport properties of IVM indicate that when IVM is co-administered with other drugs/chemicals that are potent inhibitors/inducers P4503A4 enzyme and of MDR1 (P-gp), BCRP or MRP transporters, or when polymorphisms of the drug transporters and P450 3A4 exist, drug–drug or drug–toxic chemical interactions might result in suboptimal response to the therapy or to toxic effects.
Collapse
|
49
|
Kinobe RT, Owens L. A systematic review of experimental evidence for antiviral effects of ivermectin and an in silico analysis of ivermectin's possible mode of action against SARS-CoV-2. Fundam Clin Pharmacol 2021; 35:260-276. [PMID: 33427370 PMCID: PMC8013482 DOI: 10.1111/fcp.12644] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 01/02/2023]
Abstract
Viral infections remain a major cause of economic loss with an unmet need for novel therapeutic agents. Ivermectin is a putative antiviral compound; the proposed mechanism is the inhibition of nuclear translocation of viral proteins, facilitated by mammalian host importins, a necessary process for propagation of infections. We systematically reviewed the evidence for the applicability of ivermectin against viral infections including SARS‐CoV‐2 regarding efficacy, mechanisms and selective toxicity. The SARS‐CoV‐2 genome was mined to determine potential nuclear location signals for ivermectin and meta‐analyses for in vivo studies included all comparators over time, dose range and viral replication in multiple organs. Ivermectin inhibited the replication of many viruses including those in Flaviviridae, Circoviridae and Coronaviridae families in vitro. Real and mock nuclear location signals were identified in SARS‐CoV‐2, a potential target for ivermectin and predicting a sequestration bait for importin β, stopping infected cells from reaching a virus‐resistant state. While pharmacokinetic evaluations indicate that ivermectin could be toxic if applied based on in vitro studies, inhibition of viral replication in vivo was shown for Porcine circovirus in piglets and Suid herpesvirus in mice. Overall standardized mean differences and 95% confidence intervals for ivermectin versus controls were −4.43 (−5.81, −3.04), p < 0.00001. Based on current results, the potential for repurposing ivermectin as an antiviral agent is promising. However, further work is needed to reconcile in vitro studies with clinical efficacy. Developing ivermectin as an additional antiviral agent should be pursued with an emphasis on pre‐clinical trials in validated models of infection.
Collapse
Affiliation(s)
- Robert T Kinobe
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Leigh Owens
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
50
|
Kaur H, Shekhar N, Sharma S, Sarma P, Prakash A, Medhi B. Ivermectin as a potential drug for treatment of COVID-19: an in-sync review with clinical and computational attributes. Pharmacol Rep 2021; 73:736-749. [PMID: 33389725 PMCID: PMC7778723 DOI: 10.1007/s43440-020-00195-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION COVID-19 cases are on surge; however, there is no efficient treatment or vaccine that can be used for its management. Numerous clinical trials are being reviewed for use of different drugs, biologics, and vaccines in COVID-19. A much empirical approach will be to repurpose existing drugs for which pharmacokinetic and safety data are available, because this will facilitate the process of drug development. The article discusses the evidence available for the use of Ivermectin, an anti-parasitic drug with antiviral properties, in COVID-19. METHODS A rational review of the drugs was carried out utilizing their clinically significant attributes. A more thorough understanding was met by virtual embodiment of the drug structure and realizable viral targets using artificial intelligence (AI)-based and molecular dynamics (MD)-simulation-based study. CONCLUSION Certain studies have highlighted the significance of ivermectin in COVID-19; however, it requires evidences from more Randomised Controlled Trials (RCTs) and dose- response studies to support its use. In silico-based analysis of ivermectin's molecular interaction specificity using AI and classical mechanics simulation-based methods indicates positive interaction of ivermectin with viral protein targets, which is leading for SARS-CoV 2 N-protein NTD (nucleocapsid protein N-terminal domain).
Collapse
Affiliation(s)
- Harpinder Kaur
- Department of Pharmacology, Post Graduate Institute of Medical Education and Reseasrch (PGIMER), Chandigarh, 160012, India
| | - Nishant Shekhar
- Department of Pharmacology, Post Graduate Institute of Medical Education and Reseasrch (PGIMER), Chandigarh, 160012, India
| | - Saurabh Sharma
- Department of Pharmacology, Post Graduate Institute of Medical Education and Reseasrch (PGIMER), Chandigarh, 160012, India
| | - Phulen Sarma
- Department of Pharmacology, Post Graduate Institute of Medical Education and Reseasrch (PGIMER), Chandigarh, 160012, India
| | - Ajay Prakash
- Department of Pharmacology, Post Graduate Institute of Medical Education and Reseasrch (PGIMER), Chandigarh, 160012, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Reseasrch (PGIMER), Chandigarh, 160012, India.
| |
Collapse
|