1
|
Uehara T, Yotsuyanagi H, Ohmagari N, Doi Y, Yamato M, Imamura T, Sakaguchi H, Fukushi A, Takeda Y, Baba K, Nobori H, Miyamoto T, Arita S, Dodo R, Shimba A, Fukao K, Shishido T, Tsuge Y, Mukae H. Ensitrelvir treatment-emergent amino acid substitutions in SARS-CoV-2 3CL pro detected in the SCORPIO-SR phase 3 trial. Antiviral Res 2025; 236:106097. [PMID: 39892563 DOI: 10.1016/j.antiviral.2025.106097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
The impact of treatment-emergent amino acid substitutions (TEAASs) in severe acute respiratory syndome coronavirus 2 (SARS-CoV-2) 3C-like protease (3CLpro) on clinical and virologic outcomes was evaluated in patients with mild-to-moderate coronavirus disease 2019 (COVID-19) who received ensitrelvir 125 mg in the SCORPIO-SR trial. Individuals were randomised to ensitrelvir or matched placebo once daily for 5 days (first dose <72 h after disease onset). 3CLpro-TEAASs were identified by sequencing nsp5 encoding 3CLpro from pre- and post-treatment nasopharyngeal swabs. Time to resolution of a composite of five characteristic COVID-19 symptoms (TTR) was compared between patients with and without the most common 3CLpro-TEAASs in the ensitrelvir arm. The ensitrelvir and placebo intention-to-treat populations comprised 345 and 341 patients, respectively. 3CLpro-TEAASs were detected in 19/204 (9.3%) ensitrelvir-treated and 3/137 (2.2%) placebo-treated patients with paired sequence data. The most common 3CLpro-TEAASs in the ensitrelvir arm were M49L (n = 12), M49I (n = 3) and S144A (n = 2). In the placebo arm, all 3CLpro-TEAASs occurred in ≤1 patient. Median (95% confidence interval) TTR was comparable between patients with and without those TEAASs (158.8 h [112.1-281.9] vs 189.7 h [151.4-234.4]). Mean viral RNA levels declined more slowly in patients with M49L/I or S144A versus those without. Reductions in viral titre were unaffected by these TEAASs. The characteristics of recombinant SARS-CoV-2 with 3CLpro mutations were explored in vitro. Recombinant viruses with some 3CLpro mutations had reduced susceptibility to ensitrelvir in vitro, with limited effects on viral and competitive fitness. Continued surveillance is warranted to monitor the spread of viruses with 3CLpro mutations.
Collapse
Affiliation(s)
- Takeki Uehara
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan.
| | | | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health, Tokyo, Japan
| | - Yohei Doi
- Departments of Microbiology and Infectious Diseases, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masaya Yamato
- Department of General Medicine and Infectious Diseases, Rinku General Medical Center, Izumisano, Japan
| | - Takumi Imamura
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Hiroki Sakaguchi
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Akimasa Fukushi
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Yosuke Takeda
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Keiko Baba
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Haruaki Nobori
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Tadashi Miyamoto
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Shuhei Arita
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Reiko Dodo
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Alice Shimba
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Keita Fukao
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Takao Shishido
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka, Japan
| | - Yuko Tsuge
- Drug Development and Regulatory Science Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
2
|
Nyström K, Trybala E, Said J, Roth A, Patzi Churqui M, Kärmander A, Cihlar T, Bilello JP, Bergström T, Lagging M. Remdesivir is active in vitro against tick-borne encephalitis virus and selects for resistance mutations in the viral RNA-dependent RNA polymerase. Infect Dis (Lond) 2025:1-8. [PMID: 39973341 DOI: 10.1080/23744235.2025.2468510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Tick-borne encephalitis (TBE) is a neurological disease caused by the tick-borne encephalitis virus (TBEV). Despite available vaccines, breakthrough infections occur, some fatal. OBJECTIVES As no antiviral therapy for TBE is currently approved, this study evaluated the in vitro activity of already licenced remdesivir (RDV) and sofosbuvir (SOF) for possible drug repurposing against TBEV. METHODS TBEV was cultured in A549 cells, and the inhibitory effects of RDV (GS-5734), its parent nucleotide GS-441524, and SOF (GS-7977) were assessed. RESULTS After 78 h, RDV demonstrated significantly lower EC50 values than SOF (0.14 vs. 11 µM) based on TBEV RNA levels measured by RT-qPCR. RDV also had a lower mean EC50 (0.55 µM) compared to GS-441524 and SOF (>8.9 and 13.1 µM, respectively) using crystal violet staining after 5 days. After 11 passages of TBEV in the presence of RDV, emergence of virus with a higher EC50 (1.32 vs. 0.55 µM) was detected with two mutations (L3122F and Y3278F) in NS5, the viral RNA-dependent RNA polymerase (RdRp), and one substitution in envelope (E) protein (E402G). Similarly, SOF resistance appeared after 20 passages, increasing EC50 values (35.5 vs. 10 µM). CONCLUSION RDV exhibits potent in vitro antiviral activity against TBEV via specific targeting of the viral RdRp as confirmed by the emergence of resistance-associated double NS5 substitutions in vitro in the presence of RDV. While the potential in vivo implications of the observed RDV resistance remain to be determined, these in vitro data support further assessment of RDV for the treatment of TBEV infection.
Collapse
Affiliation(s)
- Kristina Nyström
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Edward Trybala
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joanna Said
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anette Roth
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marianela Patzi Churqui
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ambjörn Kärmander
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Tomas Bergström
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martin Lagging
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
3
|
Batool S, Chokkakula S, Jeong JH, Baek YH, Song MS. SARS-CoV-2 drug resistance and therapeutic approaches. Heliyon 2025; 11:e41980. [PMID: 39897928 PMCID: PMC11786845 DOI: 10.1016/j.heliyon.2025.e41980] [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: 05/02/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
In light of the transition of COVID-19 from a pandemic to an endemic phase, there is still a dire need to address challenges associated with drug resistance, particularly among immunocompromised and high-risk populations. This review explores the current state of research on SARS-CoV-2 drug resistance and underscores the ongoing need for effective therapeutic strategies. It critically evaluates existing knowledge on resistance mechanisms and therapeutic options, aiming to consolidate information and highlight areas for future research. By examining the complex interactions between the virus and its host, the review advocates for a multifaceted approach, including combination therapies, targeted drug development, and continuous surveillance of viral mutations. It also emphasizes the impact of evolving viral variants on antiviral efficacy and suggests adaptive treatment protocols. This review aims to enhance our understanding of SARS-CoV-2 drug resistance and contribute to more effective management of COVID-19 through a discussion of promising strategies such as drug repurposing and combination therapies.
Collapse
Affiliation(s)
- Sania Batool
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Santosh Chokkakula
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Ju Hwan Jeong
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Yun Hee Baek
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Min-Suk Song
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| |
Collapse
|
4
|
De Jesús-González LA, León-Juárez M, Lira-Hernández FI, Rivas-Santiago B, Velázquez-Cervantes MA, Méndez-Delgado IM, Macías-Guerrero DI, Hernández-Castillo J, Hernández-Rodríguez X, Calderón-Sandate DN, Mata-Martínez WS, Reyes-Ruíz JM, Osuna-Ramos JF, García-Herrera AC. Advances and Challenges in Antiviral Development for Respiratory Viruses. Pathogens 2024; 14:20. [PMID: 39860981 PMCID: PMC11768830 DOI: 10.3390/pathogens14010020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
The development of antivirals for respiratory viruses has advanced markedly in response to the growing threat of pathogens such as Influenzavirus (IAV), respiratory syncytial virus (RSV), and SARS-CoV-2. This article reviews the advances and challenges in this field, highlighting therapeutic strategies that target critical stages of the viral replication cycle, including inhibitors of viral entry, replication, and assembly. In addition, innovative approaches such as inhibiting host cellular proteins to reduce viral resistance and repurposing existing drugs are explored, using advanced bioinformatics tools that optimize the identification of antiviral candidates. The analysis also covers emerging technologies such as nanomedicine and CRISPR gene editing, which promise to improve the stability and efficacy of treatments. While current antivirals offer valuable options, they face challenges such as viral evolution and the need for accessible treatments for vulnerable populations. This article underscores the importance of continued innovation in biotechnology to overcome these limitations and provide safe and effective treatments. Combining traditional and advanced approaches in developing antivirals is essential in order to address respiratory viral diseases that affect global health.
Collapse
Affiliation(s)
- Luis Adrián De Jesús-González
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
| | - Moisés León-Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México 11000, Mexico;
| | - Flor Itzel Lira-Hernández
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
| | - Bruno Rivas-Santiago
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
| | - Manuel Adrián Velázquez-Cervantes
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Ciudad de México 11000, Mexico;
| | - Iridiana Monserrat Méndez-Delgado
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
- Especialidad en Medicina Familiar, Unidad Académica de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
- Instituto Mexicano del Seguro Social, Unidad de Medicina Familiar # 4, Servicio de Medicina Familiar, Guadalupe, Zacatecas 98618, Mexico
| | - Daniela Itzel Macías-Guerrero
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
- Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Jonathan Hernández-Castillo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | - Ximena Hernández-Rodríguez
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
- Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Daniela Nahomi Calderón-Sandate
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
- Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Willy Salvador Mata-Martínez
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
- Especialidad en Medicina Familiar, Unidad Académica de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
- Instituto Mexicano del Seguro Social, Unidad de Medicina Familiar # 4, Servicio de Medicina Familiar, Guadalupe, Zacatecas 98618, Mexico
| | - José Manuel Reyes-Ruíz
- División de Investigación en Salud, Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS), Veracruz 91897, Mexico;
- Facultad de Medicina, Región Veracruz, Universidad Veracruzana (UV), Veracruz 91700, Mexico
| | | | - Ana Cristina García-Herrera
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico; (F.I.L.-H.); (B.R.-S.); (I.M.M.-D.); (D.I.M.-G.); (X.H.-R.); (D.N.C.-S.); (W.S.M.-M.); (A.C.G.-H.)
| |
Collapse
|
5
|
Jones LP, Bergeron HC, Martin DE, Murray J, Sancilio FD, Tripp RA. Probenecid Inhibits Extracellular Signal-Regulated Kinase and c-Jun N-Terminal Kinase Mitogen-Activated Protein Kinase Pathways in Regulating Respiratory Syncytial Virus Response. Int J Mol Sci 2024; 25:12452. [PMID: 39596517 PMCID: PMC11594929 DOI: 10.3390/ijms252212452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
We examined the effect of probenecid in regulating the ERK and JNK downstream MAPK pathways affecting respiratory syncytial virus replication. BACKGROUND We have previously shown that probenecid inhibits RSV, influenza virus, and SARS-CoV-2 replication in vitro in preclinical animal models and in humans. In a Phase two randomized, placebo-controlled, single-blind, dose range-finding study using probenecid to treat non-hospitalized patients with symptomatic, mild-to-moderate COVID-19, we previously showed that a 1000 mg twice daily treatment for 5 days reduced the median time to viral clearance from 11 to 7 days, and a 500 mg twice daily treatment for 5 days reduced the time to viral clearance from 11 to 9 days more than the placebo. METHODS In this study, we sought to determine the mechanism of action of the probenecid inhibition of RSV replication in human respiratory epithelial (A549) cells. RESULTS We show that probenecid inhibits the RSV-induced phosphorylation of JNKs and ERKs and the downstream phosphorylation of c-jun, a component of the AP-1 transcription complex needed for virus replication. The inhibition of JNKs by probenecid reversed the repression of transcription factor HNF-4. CONCLUSION The probenecid inhibition of JNK and ERK phosphorylation involves the MAPK pathway that precludes virus replication.
Collapse
Affiliation(s)
- Les P. Jones
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
| | - Harrison C. Bergeron
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
| | | | - Jackelyn Murray
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
| | - Fred D. Sancilio
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL 33431, USA;
| | - Ralph A. Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA; (L.P.J.); (H.C.B.); (J.M.)
- TrippBio, Inc., Jacksonville, FL 32256, USA;
| |
Collapse
|
6
|
Gökmen GG, Mirsafi FS, Leißner T, Akan T, Mishra YK, Kışla D. Zinc oxide nanomaterials: Safeguarding food quality and sustainability. Compr Rev Food Sci Food Saf 2024; 23:e70051. [PMID: 39530622 DOI: 10.1111/1541-4337.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/17/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
In this era, where food safety and sustainability are paramount concerns, the utilization of zinc oxide (ZnO) nanoparticles (NPs) is a promising solution to enhance the safety, quality, and sustainability of food products. ZnO NPs in the food industry have evolved significantly over time, reflecting advancements in synthesizing methods, antimicrobial activities, and risk assessment considerations for human health and the environment. This comprehensive review delves into the historical trajectory, current applications, and prospects of ZnO NPs in food-related contexts. Synthesizing methods, ranging from solvothermal and solgel techniques to laser ablation and microfluidic reactors, have facilitated the production of ZnO NPs with tailored properties suited for diverse food applications. The remarkable antimicrobial activity of ZnO NPs against a wide spectrum of pathogens has garnered attention for their potential to enhance food safety and extend shelf-life. Furthermore, comprehensive risk assessment methodologies have been employed to evaluate the potential impacts of ZnO NPs on human health and the environment, regarding toxicity, migration, and ecological implications. By navigating the intricate interplay between synthesis methods, antimicrobial efficacy, inhibitory mechanisms, and risk assessment protocols, by elucidating the multifaceted role of ZnO NPs in shaping the past, present, and future of the food industry, this review offers valuable insights and promising avenues for researchers, policymakers, and industry stakeholders to enhance food safety, quality, and sustainability.
Collapse
Affiliation(s)
- Gökhan Gurur Gökmen
- Department of Food Engineering, Faculty of Engineering, Ege University, Izmir, Bornova, Turkey
| | - Fatemeh Sadat Mirsafi
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Sønderborg, Denmark
| | - Till Leißner
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Sønderborg, Denmark
| | - Tamer Akan
- Department of Physics, Faculty of Science, Eskisehir Osmangazi University, Eskisehir, Odunpazarı, Turkey
| | - Yogendra Kumar Mishra
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Sønderborg, Denmark
| | - Duygu Kışla
- Department of Food Engineering, Faculty of Engineering, Ege University, Izmir, Bornova, Turkey
| |
Collapse
|
7
|
Li J, Shi XH, Fu DD, Du L, Tang B, Ao J, Ma AX, Hou YN, Wang ZG, Liu SL, Pang DW. An Inhalable Nanoshield for Effective Prevention of Influenza Virus Infections. ACS NANO 2024; 18:27327-27339. [PMID: 39315858 DOI: 10.1021/acsnano.4c04631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Influenza virus (IV) infection currently poses a serious and continuing threat to the global public health. Developing effective prevention strategies is important to defend against infection and spread of IV. Here, we developed a triple-protective nanoshield against IV infection in the lungs, formed by self-assembling DSPE-PEG amphiphilic polymers encapsulating the flu-preventive antiviral drug Arbidol internally. The preventive effect of the nanoshield against virus infection includes increasing the viscosity in the surrounding environment to physically defend against viral entry, forming a hydrated layer to block the interaction between viruses and cells, and inhibiting virus replication. Our finding suggested that a single inhalation of the nanoshield provides effective protection against IV infection for at least 8 h. Thus, this nanoshield may be a potential pandemic protection agent against IV, especially in viral environments, where no prophylactic or therapeutic measures are available.
Collapse
Affiliation(s)
- Jing Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Xue-Hui Shi
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Dan-Dan Fu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Lei Du
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Bo Tang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Jian Ao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Ai-Xin Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Yi-Ning Hou
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Dai-Wen Pang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
8
|
Bolinger AA, Li J, Xie X, Li H, Zhou J. Lessons learnt from broad-spectrum coronavirus antiviral drug discovery. Expert Opin Drug Discov 2024; 19:1023-1041. [PMID: 39078037 PMCID: PMC11390334 DOI: 10.1080/17460441.2024.2385598] [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: 02/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Highly pathogenic coronaviruses (CoVs), such as severe acute respiratory syndrome CoV (SARS-CoV), Middle East respiratory syndrome CoV (MERS-CoV), and the most recent SARS-CoV-2 responsible for the COVID-19 pandemic, pose significant threats to human populations over the past two decades. These CoVs have caused a broad spectrum of clinical manifestations ranging from asymptomatic to severe distress syndromes (ARDS), resulting in high morbidity and mortality. AREAS COVERED The accelerated advancements in antiviral drug discovery, spurred by the COVID-19 pandemic, have shed new light on the imperative to develop treatments effective against a broad spectrum of CoVs. This perspective discusses strategies and lessons learnt in targeting viral non-structural proteins, structural proteins, drug repurposing, and combinational approaches for the development of antivirals against CoVs. EXPERT OPINION Drawing lessons from the pandemic, it becomes evident that the absence of efficient broad-spectrum antiviral drugs increases the vulnerability of public health systems to the potential onslaught by highly pathogenic CoVs. The rapid and sustained spread of novel CoVs can have devastating consequences without effective and specifically targeted treatments. Prioritizing the effective development of broad-spectrum antivirals is imperative for bolstering the resilience of public health systems and mitigating the potential impact of future highly pathogenic CoVs.
Collapse
Affiliation(s)
- Andrew A. Bolinger
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jun Li
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
9
|
Wang H. Practical updates in clinical antiviral resistance testing. J Clin Microbiol 2024; 62:e0072823. [PMID: 39051778 PMCID: PMC11323466 DOI: 10.1128/jcm.00728-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
The laboratory diagnosis of antiviral resistance is a quickly changing field due to new drug availability, the sunsetting of older drugs, the development of novel technologies, rapid viral evolution, and the financial/logistic pressures of the clinical laboratory. This mini-review summarizes the current state of clinically available antiviral resistance testing in the United States in 2024, covering the most commonly used test methods, mechanisms, and clinical indications for herpes simplex virus, cytomegalovirus, human immunodeficiency virus, influenza, hepatitis B virus, and hepatitis C virus drug resistance testing. Common themes include the move away from phenotypic to genotypic methods for first-line clinical testing, as well as uncertainty surrounding the clinical meaningfulness of minority variant detection as next-generation sequencing methods have become more commonplace.
Collapse
Affiliation(s)
- Hannah Wang
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
10
|
Bekono BD, Onguéné PA, Simoben CV, Owono LCO, Ntie-Kang F. Computational discovery of dual potential inhibitors of SARS-CoV-2 spike/ACE2 and M pro: 3D-pharmacophore, docking-based virtual screening, quantum mechanics and molecular dynamics. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:277-298. [PMID: 38907013 DOI: 10.1007/s00249-024-01713-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/01/2024] [Accepted: 06/02/2024] [Indexed: 06/23/2024]
Abstract
To find drugs against COVID-19, caused by the SARS-CoV-2, promising targets include the fusion of the viral spike with the human angiotensin-converting enzyme 2 (ACE2) as well as the main protease (Mpro). These proteins are responsible for viral entry and replication, respectively. We combined several state-of-the-art computational methods, including, protein-ligand interaction fingerprint, 3D-pharmacophores, molecular-docking, MM-GBSA, DFT, and MD simulations to explore two databases: ChEMBL and NANPDB to identify molecules that could both block spike/ACE2 fusion and inhibit Mpro. A total of 1,690,649 compounds from the two databases were screened using the pharmacophore model obtained from PLIF analysis. Five recent complexes of Mpro co-crystallized with different ligands were used to generate the pharmacophore model, allowing 4,829 compounds that passed this prefilter. These were then submitted to molecular docking against Mpro. The 5% top-ranked docking hits from docking result having scores < -8.32 kcal mol-1 were selected and then docked against spike/ACE2. Only four compounds: ChEMBL244958, ChEMBL266531, ChEMBL3680003, and 1-methoxy-3-indolymethyl glucosinolate (4) displayed binding energies < - 8.21 kcal mol-1 (for the native ligand) were considered as putative dual-target inhibitors. Furthermore, predictive ADMET, MM-GBSA and DFT/6-311G(d,p) were performed on these compounds and compared with those of well-known antivirals. DFT calculations showed that ChEMBL244958 and compound 4 had significant predicted reactivity values. Molecular dynamics simulations of the docked complexes were run for 100 ns and used to validate the stability docked poses and to confirm that these hits are putative dual binders of the spike/ACE2 and the Mpro.
Collapse
Affiliation(s)
- Boris D Bekono
- Department of Physics, Ecole Normale Supérieure, University of Yaoundé I, P. O. Box 47, Yaoundé, CM-00237, Cameroon.
- Center for Drug Discovery, Faculty of Science, University of Buea, P.O. Box 63, Buea, CM-00237, Cameroon.
| | - Pascal Amoa Onguéné
- Center for Drug Discovery, Faculty of Science, University of Buea, P.O. Box 63, Buea, CM-00237, Cameroon
- Department of Chemistry, University of Yaoundé I Institute of Wood Technology Mbalmayo, University of Yaoundé I, BP 50, Mbalmayo, Cameroon
| | - Conrad V Simoben
- Center for Drug Discovery, Faculty of Science, University of Buea, P.O. Box 63, Buea, CM-00237, Cameroon
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Luc C O Owono
- Department of Physics, Ecole Normale Supérieure, University of Yaoundé I, P. O. Box 47, Yaoundé, CM-00237, Cameroon
- CEPAMOQ, Faculty of Science, University of Douala, CM-00237, Douala, Cameroon
| | - Fidele Ntie-Kang
- Center for Drug Discovery, Faculty of Science, University of Buea, P.O. Box 63, Buea, CM-00237, Cameroon.
- Department of Chemistry, Faculty of Science, University of Buea, CM-00237, Buea, Cameroon.
- Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany.
| |
Collapse
|
11
|
Karahmet Sher E, Alebić M, Marković Boras M, Boškailo E, Karahmet Farhat E, Karahmet A, Pavlović B, Sher F, Lekić L. Nanotechnology in medicine revolutionizing drug delivery for cancer and viral infection treatments. Int J Pharm 2024; 660:124345. [PMID: 38885775 DOI: 10.1016/j.ijpharm.2024.124345] [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: 02/26/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Advancements in nanotechnology were vastly applied in medicine and pharmacy, especially in the field of nano-delivery systems. It took a long time for these systems to ensure precise delivery of very delicate molecules, such as RNA, to cells at concentrations that yield remarkable efficiency, with success rates reaching 95.0% and 94.5%. These days, there are several advantages of using nanotechnological solutions in the prevention and treatment of cancer and viral infections. Its interventions improve treatment outcomes both due to increased effectiveness of the drug at target location and by reducing adverse reactions, thereby increasing patient adherence to the therapy. Based on the current knowledge an updated review was made, and perspective, opportunities and challenges in nanomedicine were discussed. The methods employed include comprehensive examination of existing literature and studies on nanoparticles and nano-delivery systems including both in vitro tests performed on cell cultures and in vivo assessments carried out on appropriate animal models, with a specific emphasis on their applications in oncology and virology. This brings together various aspects including both structure and formation as well as its association with characteristic behaviour in organisms, providing a novel perspective. Furthermore, the practical application of these systems in medicine and pharmacy with a focus on viral diseases and malignancies was explored. This review can serve as a valuable guide for fellow researchers, helping them navigate the abundance of findings in this field. The results indicate that applications of nanotechnological solutions for the delivery of medicinal products improving therapeutic outcomes will continue to expand.
Collapse
Affiliation(s)
- Emina Karahmet Sher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| | - Mirna Alebić
- Department of Pharmacy, University Hospital Centre Zagreb, Zagreb 10000, Croatia
| | - Marijana Marković Boras
- Department of Laboratory Diagnostic, University Clinical Hospital Mostar, Mostar 88000, Bosnia and Herzegovina; International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Emina Boškailo
- International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Esma Karahmet Farhat
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Department of Food and Nutrition, Faculty of Food Technology, Juraj Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Alma Karahmet
- International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Bojan Pavlović
- Faculty of Physical Education and Sports, University of East Sarajevo, Lukavica, Republika Srpska 75327, Bosnia and Herzegovina
| | - Farooq Sher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| | - Lana Lekić
- Faculty of Health Studies, University of Sarajevo, Sarajevo 71000, Bosnia and Herzegovina
| |
Collapse
|
12
|
Gupta YD, Bhandary S. Artificial Intelligence for Understanding Mechanisms of Antimicrobial Resistance and Antimicrobial Discovery. ARTIFICIAL INTELLIGENCE AND MACHINE LEARNING IN DRUG DESIGN AND DEVELOPMENT 2024:117-156. [DOI: 10.1002/9781394234196.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Mukherjee A, Ghosh KK, Chakrabortty S, Gulyás B, Padmanabhan P, Ball WB. Mitochondrial Reactive Oxygen Species in Infection and Immunity. Biomolecules 2024; 14:670. [PMID: 38927073 PMCID: PMC11202257 DOI: 10.3390/biom14060670] [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: 05/15/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Reactive oxygen species (ROS) contain at least one oxygen atom and one or more unpaired electrons and include singlet oxygen, superoxide anion radical, hydroxyl radical, hydroperoxyl radical, and free nitrogen radicals. Intracellular ROS can be formed as a consequence of several factors, including ultra-violet (UV) radiation, electron leakage during aerobic respiration, inflammatory responses mediated by macrophages, and other external stimuli or stress. The enhanced production of ROS is termed oxidative stress and this leads to cellular damage, such as protein carbonylation, lipid peroxidation, deoxyribonucleic acid (DNA) damage, and base modifications. This damage may manifest in various pathological states, including ageing, cancer, neurological diseases, and metabolic disorders like diabetes. On the other hand, the optimum levels of ROS have been implicated in the regulation of many important physiological processes. For example, the ROS generated in the mitochondria (mitochondrial ROS or mt-ROS), as a byproduct of the electron transport chain (ETC), participate in a plethora of physiological functions, which include ageing, cell growth, cell proliferation, and immune response and regulation. In this current review, we will focus on the mechanisms by which mt-ROS regulate different pathways of host immune responses in the context of infection by bacteria, protozoan parasites, viruses, and fungi. We will also discuss how these pathogens, in turn, modulate mt-ROS to evade host immunity. We will conclude by briefly giving an overview of the potential therapeutic approaches involving mt-ROS in infectious diseases.
Collapse
Affiliation(s)
- Arunima Mukherjee
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
| | - Sabyasachi Chakrabortty
- Department of Chemistry, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, 17176 Stockholm, Sweden
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
| | - Writoban Basu Ball
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| |
Collapse
|
14
|
Fayisa ST, Pokharel N, Solomon A, Negash L, Biltibo LA, Bedasso SH, Abebe AT. Navigating diagnostic challenges: imaging strategies for herpes simplex encephalitis in resource limited settings: A case report. Radiol Case Rep 2024; 19:2183-2187. [PMID: 38515771 PMCID: PMC10950604 DOI: 10.1016/j.radcr.2024.02.060] [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: 12/31/2023] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Encephalitis refers to the inflammatory condition affecting the brain parenchyma, leading to various neurological impairments. It can have various causes: infectious, postinfectious, and noninfectious origins. In this case, we present a 76-year-old man who presented to the emergency room with complaints of headache and behavioral changes. Initially, a Computed Tomography (CT) scan raised suspicion of herpes simplex encephalitis and prompted the initiation of treatment. Subsequently, Magnetic Resonance Imaging (MRI) and Cerebrospinal fluid (CSF) culture confirmed the diagnosis. However, despite medical intervention, the patient's condition unexpectedly deteriorated, and he unfortunately passed away after spending 2 weeks in the Intensive Care Unit (ICU). Possible factors contributing to this outcome include delayed presentation to medical care, viral resistance, or the inherent nature of the infection itself, particularly in elderly patients.
Collapse
Affiliation(s)
- Shimalis Tadasa Fayisa
- Department of Radiology, College of Health Science, Mizan Tepi University, Mizan, Ethiopia
| | - Nishma Pokharel
- Departement of medicine, Kathmandu Medical college, Kathmandu, Nepal
| | | | - Lense Negash
- Departement of Medicine, S't Paul's Hospital Millenium Medical Collage hospital, Addis Ababa, Ethiopia
| | - Lidiya Ayele Biltibo
- Departement of Diagnostic Radiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Selamawit Hailu Bedasso
- Departement of Medicine, ALERT Hospital/Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Abel Tsehay Abebe
- Departement of Diagnostic Radiology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
15
|
Palazzotti D, Sguilla M, Manfroni G, Cecchetti V, Astolfi A, Barreca ML. Small Molecule Drugs Targeting Viral Polymerases. Pharmaceuticals (Basel) 2024; 17:661. [PMID: 38794231 PMCID: PMC11124969 DOI: 10.3390/ph17050661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Small molecules that specifically target viral polymerases-crucial enzymes governing viral genome transcription and replication-play a pivotal role in combating viral infections. Presently, approved polymerase inhibitors cover nine human viruses, spanning both DNA and RNA viruses. This review provides a comprehensive analysis of these licensed drugs, encompassing nucleoside/nucleotide inhibitors (NIs), non-nucleoside inhibitors (NNIs), and mutagenic agents. For each compound, we describe the specific targeted virus and related polymerase enzyme, the mechanism of action, and the relevant bioactivity data. This wealth of information serves as a valuable resource for researchers actively engaged in antiviral drug discovery efforts, offering a complete overview of established strategies as well as insights for shaping the development of next-generation antiviral therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Letizia Barreca
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy; (D.P.); (M.S.); (G.M.); (V.C.); (A.A.)
| |
Collapse
|
16
|
Sun M, Manson ML, Guo T, de Lange ECM. CNS Viral Infections-What to Consider for Improving Drug Treatment: A Plea for Using Mathematical Modeling Approaches. CNS Drugs 2024; 38:349-373. [PMID: 38580795 PMCID: PMC11026214 DOI: 10.1007/s40263-024-01082-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
Neurotropic viruses may cause meningitis, myelitis, encephalitis, or meningoencephalitis. These inflammatory conditions of the central nervous system (CNS) may have serious and devastating consequences if not treated adequately. In this review, we first summarize how neurotropic viruses can enter the CNS by (1) crossing the blood-brain barrier or blood-cerebrospinal fluid barrier; (2) invading the nose via the olfactory route; or (3) invading the peripheral nervous system. Neurotropic viruses may then enter the intracellular space of brain cells via endocytosis and/or membrane fusion. Antiviral drugs are currently used for different viral CNS infections, even though their use and dosing regimens within the CNS, with the exception of acyclovir, are minimally supported by clinical evidence. We therefore provide considerations to optimize drug treatment(s) for these neurotropic viruses. Antiviral drugs should cross the blood-brain barrier/blood cerebrospinal fluid barrier and pass the brain cellular membrane to inhibit these viruses inside the brain cells. Some antiviral drugs may also require intracellular conversion into their active metabolite(s). This illustrates the need to better understand these mechanisms because these processes dictate drug exposure within the CNS that ultimately determine the success of antiviral drugs for CNS infections. Finally, we discuss mathematical model-based approaches for optimizing antiviral treatments. Thereby emphasizing the potential of CNS physiologically based pharmacokinetic models because direct measurement of brain intracellular exposure in living humans faces ethical restrictions. Existing physiologically based pharmacokinetic models combined with in vitro pharmacokinetic/pharmacodynamic information can be used to predict drug exposure and evaluate efficacy of antiviral drugs within the CNS, to ultimately optimize the treatments of CNS viral infections.
Collapse
Affiliation(s)
- Ming Sun
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Martijn L Manson
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tingjie Guo
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
17
|
Shen SJ, Feng PC, Wu RC, Kuo YH, Liu SJ, Ito H. Resorbable nanofibrous membranes for local and sustained co-delivery of acyclovir and ketorolac in herpes therapy. Int J Pharm 2024; 654:123988. [PMID: 38467207 DOI: 10.1016/j.ijpharm.2024.123988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/21/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Herpes simplex and herpes zoster are both viral infections caused by members of the herpesvirus family. The former is characterized by painful, fluid-filled blisters or sores on the skin and mucous membranes, while the latter presents as a painful rash with blisters, typically occurring in a single band or patch along one side of the body. The treatment remains a challenge since current antiviral therapy via oral administration may lead to unfavorable side effects such as headaches, nausea, and diarrhea. This study used electrospinning to develop biodegradable nanofibrous poly(lactic-co-glycolic acid) (PLGA) membranes for delivery of both acyclovir and ketorolac. The structure of the spun nanofibers was assessed via scanning electron microscopy (SEM), and the appearance of loaded acyclovir and ketorolac in the nanofibers was confirmed with Fourier-transform infrared spectroscopy (FTIR) and differential scanning calorimetry (DSC). Release profiles of these drugs from the nanofibrous membranes were assessed using in vitro elution studies, high-performance liquid chromatography (HPLC) assays, and in vivo drug release patterns. The electrospun nanofibers had a size range of 283-725 nm in diameter, resembling the extracellular matrix of natural tissue and demonstrated excellent flexibility and extensibility. Notably, the drug-eluting nanofibers exhibited an extended release of high levels of acyclovir and ketorolac over a 21-day period. Thus, biodegradable drug-eluting membranes with a prolonged drug release could be a potential therapeutic approach for treating herpes infections.
Collapse
Affiliation(s)
- Shih-Jyun Shen
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
| | - Pin-Chao Feng
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Ren-Chin Wu
- Department of Anatomic Pathology, Linkou Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Yi-Hua Kuo
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan; Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan.
| | - Hiroshi Ito
- Graduate School of Organic Materials Science, Yamagata University, Yonezawa, Yamagata 992-8510, Japan
| |
Collapse
|
18
|
Hossain MG, Islam M, Araf Y, Paul SK, Akter S, Khan MK, Ahmed MU, Khan S, Akbar SMF, Debnath CR. Comprehensive analysis of antigenic variations and genomic properties of hepatitis B virus in clinical samples in the mid-north east region of Bangladesh. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 119:105572. [PMID: 38367678 DOI: 10.1016/j.meegid.2024.105572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/19/2024]
Abstract
This investigation delineates an exhaustive analysis of the clinical, immunological, and genomic landscapes of hepatitis B virus (HBV) infection across a cohort of 22 verified patients. The demographic analysis unveiled a pronounced male bias (77.27%), with patient ages spanning 20 to 85 years and durations of illness ranging from 10 days to 4 years. Predominant clinical manifestations included fever, fatigue, anorexia, abdominal discomfort, and arthralgia, alongside observed co-morbidities such as chronic renal disorders and hepatocellular carcinoma. Antigenic profiling of the HBV envelope proteins elucidated significant heterogeneity among the infected subjects, particularly highlighted by discordances in the detection capabilities of small and large HBsAg assays, suggesting antigenic diversity. Quantitative assessment of viral loads unveiled a broad spectrum, accompanied by atypical HBeAg reactivity patterns, challenging the reliability of existing serological markers. Correlative studies between viral burden and antigenicity of the envelope proteins unearthed phenomena indicative of diagnostic evasion. Notably, samples demonstrating robust viral replication were paradoxically undetectable by the large HBsAg ELISA kit, advocating for more sophisticated diagnostic methodologies. Genotypic examination of three HBV isolates classified them as genotype D (D2), with phylogenetic alignment to strains from various global origins. Mutational profiling identified pivotal mutations within the basic core promoter and preS2/S1 regions, associated with an augmented risk of hepatocellular carcinoma. Further, mutations discerned in the small HBsAg and RT/overlap regions were recognized as contributors to vaccine and/or diagnostic escape mechanisms. In summation, this scholarly discourse elucidates the intricate interplay of clinical presentations, antigenic diversity, and genomic attributes in HBV infection, accentuating the imperative for ongoing investigative endeavors to refine diagnostic and therapeutic modalities.
Collapse
Affiliation(s)
- Md Golzar Hossain
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh.
| | - Mahfuz Islam
- Department of Microbiology, Mymensingh Medical College, Mymensingh, Bangladesh
| | - Yusha Araf
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Shyamal Kumar Paul
- Department of Microbiology, Mymensingh Medical College, Mymensingh, Bangladesh
| | - Sharmin Akter
- Department of Physiology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | | | - Muzahed Uddin Ahmed
- Department of Medicine, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Sakirul Khan
- Research Center for Global and Local Infectious Diseases, Oita University, Oita, Japan; Department of Microbiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Sheikh Mohammad Fazle Akbar
- Research Center for Global and Local Infectious Diseases, Oita University, Oita, Japan; Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Ehime, Japan; Miyakawa Memorial Research Foundation, Tokyo, Japan
| | | |
Collapse
|
19
|
Borges PHO, Ferreira SB, Silva FP. Recent Advances on Targeting Proteases for Antiviral Development. Viruses 2024; 16:366. [PMID: 38543732 PMCID: PMC10976044 DOI: 10.3390/v16030366] [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: 01/19/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 05/23/2024] Open
Abstract
Viral proteases are an important target for drug development, since they can modulate vital pathways in viral replication, maturation, assembly and cell entry. With the (re)appearance of several new viruses responsible for causing diseases in humans, like the West Nile virus (WNV) and the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), understanding the mechanisms behind blocking viral protease's function is pivotal for the development of new antiviral drugs and therapeutical strategies. Apart from directly inhibiting the target protease, usually by targeting its active site, several new pathways have been explored to impair its activity, such as inducing protein aggregation, targeting allosteric sites or by inducing protein degradation by cellular proteasomes, which can be extremely valuable when considering the emerging drug-resistant strains. In this review, we aim to discuss the recent advances on a broad range of viral proteases inhibitors, therapies and molecular approaches for protein inactivation or degradation, giving an insight on different possible strategies against this important class of antiviral target.
Collapse
Affiliation(s)
- Pedro Henrique Oliveira Borges
- Laboratory of Organic Synthesis and Biological Prospecting, Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil;
- Laboratory of Experimental and Computational Biochemistry of Drugs, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil
| | - Sabrina Baptista Ferreira
- Laboratory of Organic Synthesis and Biological Prospecting, Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil;
| | - Floriano Paes Silva
- Laboratory of Experimental and Computational Biochemistry of Drugs, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
20
|
Padhi AK, Maurya S. Uncovering the secrets of resistance: An introduction to computational methods in infectious disease research. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:173-220. [PMID: 38448135 DOI: 10.1016/bs.apcsb.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Antimicrobial resistance (AMR) is a growing global concern with significant implications for infectious disease control and therapeutics development. This chapter presents a comprehensive overview of computational methods in the study of AMR. We explore the prevalence and statistics of AMR, underscoring its alarming impact on public health. The role of AMR in infectious disease outbreaks and its impact on therapeutics development are discussed, emphasizing the need for novel strategies. Resistance mutations are pivotal in AMR, enabling pathogens to evade antimicrobial treatments. We delve into their importance and contribution to the spread of AMR. Experimental methods for quantitatively evaluating resistance mutations are described, along with their limitations. To address these challenges, computational methods provide promising solutions. We highlight the advantages of computational approaches, including rapid analysis of large datasets and prediction of resistance profiles. A comprehensive overview of computational methods for studying AMR is presented, encompassing genomics, proteomics, structural bioinformatics, network analysis, and machine learning algorithms. The strengths and limitations of each method are briefly outlined. Additionally, we introduce ResScan-design, our own computational method, which employs a protein (re)design protocol to identify potential resistance mutations and adaptation signatures in pathogens. Case studies are discussed to showcase the application of ResScan in elucidating hotspot residues, understanding underlying mechanisms, and guiding the design of effective therapies. In conclusion, we emphasize the value of computational methods in understanding and combating AMR. Integration of experimental and computational approaches can expedite the discovery of innovative antimicrobial treatments and mitigate the threat posed by AMR.
Collapse
Affiliation(s)
- Aditya K Padhi
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India.
| | - Shweata Maurya
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
21
|
Adeosun WB, Loots DT. Medicinal Plants against Viral Infections: A Review of Metabolomics Evidence for the Antiviral Properties and Potentials in Plant Sources. Viruses 2024; 16:218. [PMID: 38399995 PMCID: PMC10892737 DOI: 10.3390/v16020218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Most plants have developed unique mechanisms to cope with harsh environmental conditions to compensate for their lack of mobility. A key part of their coping mechanisms is the synthesis of secondary metabolites. In addition to their role in plants' defense against pathogens, they also possess therapeutic properties against diseases, and their use by humans predates written history. Viruses are a unique class of submicroscopic agents, incapable of independent existence outside a living host. Pathogenic viruses continue to pose a significant threat to global health, leading to innumerable fatalities on a yearly basis. The use of medicinal plants as a natural source of antiviral agents has been widely reported in literature in the past decades. Metabolomics is a powerful research tool for the identification of plant metabolites with antiviral potentials. It can be used to isolate compounds with antiviral capacities in plants and study the biosynthetic pathways involved in viral disease progression. This review discusses the use of medicinal plants as antiviral agents, with a special focus on the metabolomics evidence supporting their efficacy. Suggestions are made for the optimization of various metabolomics methods of characterizing the bioactive compounds in plants and subsequently understanding the mechanisms of their operation.
Collapse
Affiliation(s)
- Wilson Bamise Adeosun
- Human Metabolomics, North-West University, Private Bag X6001, Box 269, Potchefstroom 2531, South Africa;
| | | |
Collapse
|
22
|
Ghosh S, Singha PS, Das LK, Ghosh D. Systematic Review on Major Antiviral Phytocompounds from Common Medicinal Plants against SARS-CoV-2. Med Chem 2024; 20:613-629. [PMID: 38317467 DOI: 10.2174/0115734064262843231120051452] [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: 05/17/2023] [Revised: 08/02/2023] [Accepted: 09/14/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Viral infections are rising around the globe and with evolving virus types and increasing varieties of viral invasions; the human body is developing antimicrobial resistance continuously. This is making the fight of mankind against viruses weak and unsecured. On the other hand, changing lifestyle, globalization and human activities adversely affecting the environment are opening up risks for new viral predominance on human race. In this context the world has witnessed the pandemic of the human Coronavirus disease (COVID-19) recently. The disease is caused by the Coronavirus namely Severe Acute Respiratory Syndrome Coronavirus 2 (SARSCoV- 2). METHODS AND MATERIALS Developing potential and effective vaccine is also time consuming and challenging. The huge resource of plants around us has rich source of potent antiviral compounds. Some of these molecules may serve as tremendously potent lead molecules whose slight structural modifications may give us highly bioactive antiviral derivatives of phytocompounds. Every geographical region is rich in unique plant biodiversity and hence every corner of the world with rich plant biodiversity can serve as abode for potential magical phytocompounds most of which have not been extensively explored for development of antiviral drug formulations against various viruses like the HIV, HPV etc., and the Coronavirus, also known as SARS-CoV-2 which causes the disease COVID-19. RESULTS Several phytocompounds from various medicinal plants have already been screened using in silico tools and some of them have yielded promising results establishing themselves as potent lead molecules for development of drugs against the highly mutating SARS-CoV-2 virus and thus these phytocompounds may be beneficial in treating COVID-19 and help human to win the life threatening battle against the deadly virus. CONCLUSION The best advantage is that these phytocompounds being derived from nature in most of the cases, come with minimum or no side effects compared to that of chemically synthesized conventional bioactive compounds and are indigenously available hence are the source of cost effective drug formulations with strong therapeutic potentials.
Collapse
Affiliation(s)
- Suvendu Ghosh
- Department of Physiology, Hooghly Mohsin College, Chinsura, Hooghly 712 101, West Bengal, India
| | - Partha Sarathi Singha
- Department of Chemistry, Government General Degree College, Kharagpur II, P.O Madpur, Dist, Paschim Medinipur, Pin: 721149, West Bengal, India
| | - Lakshmi Kanta Das
- Department of Chemistry, Government General Degree College, Kharagpur II, P.O Madpur, Dist, Paschim Medinipur, Pin: 721149, West Bengal, India
| | - Debosree Ghosh
- Department of Physiology, Government General Degree College, Kharagpur II, P.O Madpur, Dist, Paschim Medinipur, Pin: 721149, West Bengal, India
| |
Collapse
|
23
|
Havranek B, Demissie R, Lee H, Lan S, Zhang H, Sarafianos SG, Jean-Luc Ayitou A, Islam SM. Discovery of Nirmatrelvir Resistance Mutations in SARS-CoV-2 3CLpro: A Computational-Experimental Approach. J Chem Inf Model 2023; 63:7180-7188. [PMID: 37947496 PMCID: PMC10976418 DOI: 10.1021/acs.jcim.3c01269] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
The COVID-19 pandemic has emphasized the urgency for effective antiviral therapies against SARS-CoV-2. Targeting the main protease (3CLpro) of the virus has emerged as a promising approach, and nirmatrelvir (PF-07321332), the active component of Pfizer's oral drug Paxlovid, has demonstrated remarkable clinical efficacy. However, the emergence of resistance mutations poses a challenge to its continued success. In this study, we employed alchemical free energy perturbation (FEP) alanine scanning to identify nirmatrelvir-resistance mutations within SARS-CoV-2 3CLpro. FEP identified several mutations, which were validated through in vitro IC50 experiments and found to result in 8- and 72-fold increases in nirmatrelvir IC50 values. Additionally, we constructed SARS-CoV-2 omicron replicons containing these mutations, and one of the mutants (S144A/E166A) displayed a 20-fold increase in EC50, confirming the role of FEP in identifying drug-resistance mutations. Our findings suggest that FEP can be a valuable tool in proactively monitoring the emergence of resistant strains and guiding the design of future inhibitors with reduced susceptibility to drug resistance. As nirmatrelvir is currently widely used for treating COVID-19, this research has important implications for surveillance efforts and antiviral development.
Collapse
Affiliation(s)
- Brandon Havranek
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
- ComputePharma, LLC., Chicago, IL, USA, 60607
| | - Robel Demissie
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
- Biophysics Core at Research Resource Center, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Hyun Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
- Biophysics Core at Research Resource Center, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Shuiyun Lan
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Huanchun Zhang
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Stefan G. Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | | | - Shahidul M. Islam
- ComputePharma, LLC., Chicago, IL, USA, 60607
- Department of Chemistry, Delaware State University, Dover, DE, 19901, USA
| |
Collapse
|
24
|
Borase H, Shukla D. The Interplay of Genital Herpes with Cellular Processes: A Pathogenesis and Therapeutic Perspective. Viruses 2023; 15:2195. [PMID: 38005873 PMCID: PMC10675801 DOI: 10.3390/v15112195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Genital herpes, primarily caused by herpes simplex virus-2 (HSV-2), remains a pressing global health concern. Its remarkable ability to intertwine with cellular processes, from harnessing host machinery for replication to subverting antiviral defenses like autophagy and programmed cell death, exemplifies the intricate interplay at the heart of its pathogenesis. While the biomedical community has extensively researched antiviral interventions, the efficiency of these strategies in managing HSV-2 remains suboptimal. Recognizing this, attention has shifted toward leveraging host cellular components to regulate HSV-2 replication and influence the cell cycle. Furthermore, innovative interventional strategies-including drug repurposing, microbivacs, connecting the host microbiome, and exploiting natural secondary metabolites-are emerging as potential game changers. This review summarizes the key steps in HSV-2 pathogenesis and newly discovered cellular interactions, presenting the latest developments in the field, highlighting existing challenges, and offering a fresh perspective on HSV-2's pathogenesis and the potential avenues for its treatment by targeting cellular proteins and pathways.
Collapse
Affiliation(s)
- Hemant Borase
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA;
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
25
|
Cele ZED, Matshe W, Mdlalose L, Setshedi K, Malatji K, Mkhwanazi NP, Ntombela T, Balogun M. Cationic Chitosan Derivatives for the Inactivation of HIV-1 and SARS-CoV-2 Enveloped Viruses. ACS OMEGA 2023; 8:31714-31724. [PMID: 37692209 PMCID: PMC10483524 DOI: 10.1021/acsomega.3c02143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/01/2023] [Indexed: 09/12/2023]
Abstract
Cationic chitosan derivatives have been widely studied as potential antimicrobial agents. However, very little is known about their antiviral activity and mode of action against enveloped viruses. We investigated the ability of hydroxypropanoic acid-grafted chitosan (HPA-CS) and N-(2-hydroxypropyl)-3-trimethylammonium chitosan chloride (HTCC) to inactivate enveloped viruses like the human immunodeficiency virus (HIV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The membrane-disrupting potential of the chitosan derivatives was initially investigated in a hemolysis assay. At 1.0 mg/mL, about 80% hemolysis was observed for the cationic chitosan derivatives, which was significant when compared to almost no membrane-disrupting activity by the unmodified chitosan. Virus inhibition was evaluated using the luciferase-based antiviral assay against the HIV-1 NL4.3 virus (400 TCID). The IC50 of HPA-CS was 4.109 mg/mL, while the HTCC showed a higher antiviral activity at an IC50 = 0.225 mg/mL. For practical application, the antiviral efficacies of the HTCC-coated and uncoated nonmedical masks were evaluated for SARS- CoV-2 virus capture. The coated masks demonstrated an almost excellent performance with nearly 100% viral inhibition compared to less than 60% inhibition by the uncoated masks. Molecular docking predictions suggest that the HTCC polymers interact with the viral spike protein, blocking the coronavirus interaction with the target host cell's angiotensin-converting enzyme 2 cellular receptors.
Collapse
Affiliation(s)
- Zamani E. D. Cele
- Bio-Polymer
Modification and Advanced Therapeutics Laboratory, Centre for Nanostructures
and Advanced Materials, Council for Scientific
and Industrial Research, Pretoria, Gauteng 0001, South Africa
| | - William Matshe
- Bio-Polymer
Modification and Advanced Therapeutics Laboratory, Centre for Nanostructures
and Advanced Materials, Council for Scientific
and Industrial Research, Pretoria, Gauteng 0001, South Africa
| | - Lindani Mdlalose
- Bio-Polymer
Modification and Advanced Therapeutics Laboratory, Centre for Nanostructures
and Advanced Materials, Council for Scientific
and Industrial Research, Pretoria, Gauteng 0001, South Africa
| | - Katlego Setshedi
- Bio-Polymer
Modification and Advanced Therapeutics Laboratory, Centre for Nanostructures
and Advanced Materials, Council for Scientific
and Industrial Research, Pretoria, Gauteng 0001, South Africa
| | - Kanyane Malatji
- Emerging
Research Area Platform, Next Generation Health Cluster, Council for Scientific and Industrial Research, Pretoria, Gauteng 0001, South Africa
| | - Nompumelelo Prudence Mkhwanazi
- College
of Health Science, School of Laboratory Medicine and Medical Science,
HIV Pathogenesis Programme, University of
KwaZulu-Natal, Durban, KwaZulu-Natal 4041 South Africa
| | - Thandokuhle Ntombela
- Faculty
of Science, School of Chemistry, University
of the Witwatersrand, Johannesburg 00000, South Africa
| | - Mohammed Balogun
- Bio-Polymer
Modification and Advanced Therapeutics Laboratory, Centre for Nanostructures
and Advanced Materials, Council for Scientific
and Industrial Research, Pretoria, Gauteng 0001, South Africa
| |
Collapse
|
26
|
Suprewicz Ł, Szczepański A, Lenart M, Piktel E, Fiedoruk K, Barreto-Duran E, Kula-Pacurar A, Savage PB, Milewska A, Bucki R, Pyrć K. Ceragenins exhibit antiviral activity against SARS-CoV-2 by increasing the expression and release of type I interferons upon activation of the host's immune response. Antiviral Res 2023; 217:105676. [PMID: 37481038 DOI: 10.1016/j.antiviral.2023.105676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/24/2023]
Abstract
The COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) heavily burdened the entire world socially and economically. Despite a generation of vaccines and therapeutics to confront infection, it remains a threat. Most available antivirals target viral proteins and block their activity or function. While such an approach is considered effective and safe, finding treatments for specific viruses of concern leaves us unprepared for developed resistance and future viral pandemics of unknown origin. Here, we propose ceragenins (CSAs), synthetic amphipathic molecules designed to mimic the properties of cationic antimicrobial peptides (cAMPs), as potential broad-spectrum antivirals. We show that selected CSAs exhibit antiviral activity against SARS-CoV-2 and low-pathogenic human coronaviruses 229E, OC43, and NL63. The mechanism of action of CSAs against coronaviruses is mainly attributed to the stimulation of antiviral cytokines, such as type I interferons or IL-6. Our study provides insight into a novel immunomodulatory strategy that might play an essential role during the current pandemic and future outbreaks.
Collapse
Affiliation(s)
- Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Bialystok, Poland
| | - Artur Szczepański
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Marzena Lenart
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Bialystok, Bialystok, Poland
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Bialystok, Poland
| | - Emilia Barreto-Duran
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Anna Kula-Pacurar
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Paul B Savage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Aleksandra Milewska
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Bialystok, Poland.
| | - Krzysztof Pyrć
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
27
|
Chowdhary S, Deka R, Panda K, Kumar R, Solomon AD, Das J, Kanoujiya S, Gupta AK, Sinha S, Ruokolainen J, Kesari KK, Gupta PK. Recent Updates on Viral Oncogenesis: Available Preventive and Therapeutic Entities. Mol Pharm 2023; 20:3698-3740. [PMID: 37486263 PMCID: PMC10410670 DOI: 10.1021/acs.molpharmaceut.2c01080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/25/2023]
Abstract
Human viral oncogenesis is a complex phenomenon and a major contributor to the global cancer burden. Several recent findings revealed cellular and molecular pathways that promote the development and initiation of malignancy when viruses cause an infection. Even, antiviral treatment has become an approach to eliminate the viral infections and prevent the activation of oncogenesis. Therefore, for a better understanding, the molecular pathogenesis of various oncogenic viruses like, hepatitis virus, human immunodeficiency viral (HIV), human papillomavirus (HPV), herpes simplex virus (HSV), and Epstein-Barr virus (EBV), could be explored, especially, to expand many potent antivirals that may escalate the apoptosis of infected malignant cells while sparing normal and healthy ones. Moreover, contemporary therapies, such as engineered antibodies antiviral agents targeting signaling pathways and cell biomarkers, could inhibit viral oncogenesis. This review elaborates the recent advancements in both natural and synthetic antivirals to control viral oncogenesis. The study also highlights the challenges and future perspectives of using antivirals in viral oncogenesis.
Collapse
Affiliation(s)
- Shivam Chowdhary
- Department
of Industrial Microbiology, Sam Higginbottom
University of Agriculture, Technology and Sciences, Prayagraj 211007, Uttar Pradesh India
| | - Rahul Deka
- Department
of Bioengineering and Biotechnology, Birla
Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Kingshuk Panda
- Department
of Applied Microbiology, Vellore Institute
of Technology, Vellore 632014, Tamil Nadu, India
| | - Rohit Kumar
- Department
of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, India
| | - Abhishikt David Solomon
- Department
of Molecular & Cellular Engineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, Uttar Pradesh, India
| | - Jimli Das
- Centre
for
Biotechnology and Bioinformatics, Dibrugarh
University, Assam 786004, India
| | - Supriya Kanoujiya
- School
of
Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ashish Kumar Gupta
- Department
of Biophysics, All India Institute of Medical
Sciences, New Delhi 110029, India
| | - Somya Sinha
- Department
of Biotechnology, Graphic Era Deemed to
Be University, Dehradun 248002, Uttarakhand, India
| | - Janne Ruokolainen
- Department
of Applied Physics, School of Science, Aalto
University, 02150 Espoo, Finland
| | - Kavindra Kumar Kesari
- Department
of Applied Physics, School of Science, Aalto
University, 02150 Espoo, Finland
- Division
of Research and Development, Lovely Professional
University, Phagwara 144411, Punjab, India
| | - Piyush Kumar Gupta
- Department
of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- Department
of Biotechnology, Graphic Era Deemed to
Be University, Dehradun 248002, Uttarakhand, India
- Faculty
of Health and Life Sciences, INTI International
University, Nilai 71800, Malaysia
| |
Collapse
|
28
|
Painuli S, Semwal P, Sharma R, Akash S. Superbugs or multidrug resistant microbes: A new threat to the society. Health Sci Rep 2023; 6:e1480. [PMID: 37547359 PMCID: PMC10397562 DOI: 10.1002/hsr2.1480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023] Open
Affiliation(s)
- Sakshi Painuli
- Natural Products Research LaboratoryUttarakhand Council for BiotechnologyDehradunIndia
| | - Prabhakar Semwal
- Department of BiotechnologyGraphic Era (Deemed to be University)DehradunIndia
- Research and Development CellGraphic Era Hill UniversityDehradunIndia
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya KalpanaFaculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu UniversityUttar PradeshVaranasiIndia
| | - Shopnil Akash
- Faculty of Allied Health ScienceDepartment of Pharmacy, Daffodil International UniversityDhakaBangladesh
| |
Collapse
|
29
|
Oey A, McClure M, Symons JA, Chanda S, Fry J, Smith PF, Luciani K, Fayon M, Chokephaibulkit K, Uppala R, Bernatoniene J, Furuno K, Stanley T, Huntjens D, Witek J. Lumicitabine, an orally administered nucleoside analog, in infants hospitalized with respiratory syncytial virus (RSV) infection: Safety, efficacy, and pharmacokinetic results. PLoS One 2023; 18:e0288271. [PMID: 37467213 PMCID: PMC10355467 DOI: 10.1371/journal.pone.0288271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/25/2023] [Indexed: 07/21/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection is the leading cause of infant hospitalizations and mortality. Lumicitabine, an oral nucleoside analog was studied for the treatment of RSV. The phase 1b and phase 2b studies reported here assessed the safety, pharmacokinetics, and pharmacodynamics of lumicitabine in infants/neonates hospitalized with RSV. In the phase 1b study, infants (≥1 to ≤12 months) and neonates (<28 days) received a single-ascending or multiple-ascending doses (single loading dose [LD] then 9 maintenance doses [MD] of lumicitabine, or placebo [3:1]). In the phase 2b study, infants/children (28 days to ≤36 months old) received lumicitabine 40/20 mg/kg, 60/40 mg/kg LD/MD twice-daily or placebo (1:1:1) for 5 days. Safety, pharmacokinetics, and efficacy parameters were assessed over 28 days. Lumicitabine was associated with a dose-related increase in the incidence and severity of reversible neutropenia. Plasma levels of ALS-008112, the active nucleoside analog, were dose-proportional with comparable mean exposure levels at the highest doses in both studies. There were no significant differences between the lumicitabine groups and placebo in reducing viral load, time to viral non-detectability, and symptom resolution. No emergent resistance-associated substitutions were observed at the RSV L-gene positions of interest. In summary, lumicitabine was associated with a dose-related increase in the incidence and severity of reversible neutropenia and failed to demonstrate antiviral activity in RSV-infected hospitalized infants. This contrasts with the findings of the previous RSV-A adult challenge study where significant antiviral activity was noted, without incidence of neutropenia. Trial registration ClinicalTrials.gov Identifier: NCT02202356 (phase 1b); NCT03333317 (phase 2b).
Collapse
Affiliation(s)
- Abbie Oey
- Janssen Research & Development, LLC, South San Francisco, California, United States of America
| | - Matthew McClure
- Janssen Research & Development, LLC, South San Francisco, California, United States of America
| | - Julian A. Symons
- Janssen Research & Development, LLC, South San Francisco, California, United States of America
| | - Sushmita Chanda
- Janssen Research & Development, LLC, South San Francisco, California, United States of America
| | - John Fry
- Janssen Research & Development, LLC, South San Francisco, California, United States of America
| | - Patrick F. Smith
- Certara Strategic Consulting, Parsippany, New Jersey, United States of America
| | - Kathia Luciani
- Department of Infectious Diseases Hospital de Especialidades Pediátricas Omar Torrijos Herrera, Panama City, Panama
| | - Michael Fayon
- CHU de Bordeaux, Pneumologie pédiatrique, CIC 1401 (INSERM), Hôpital Pellegrin-Enfants, Bordeaux Cedex, France
| | - Kulkanya Chokephaibulkit
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rattapon Uppala
- Department of Pediatrics, Khon Kaen University, Khon Kaen, Thailand
| | - Jolanta Bernatoniene
- Paediatric Infectious Disease and Immunology Department, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Kenji Furuno
- General Pediatrics & Interdisciplinary Medicine Fukuoka Children’s Hospital, Fukuoka, Japan
| | - Thorsten Stanley
- Department of Paediatrics, University of Otago, Wellington, New Zealand
| | | | - James Witek
- Janssen Research & Development, LLC, Titusville, New Jersey, United States of America
| | | |
Collapse
|
30
|
von Delft A, Hall MD, Kwong AD, Purcell LA, Saikatendu KS, Schmitz U, Tallarico JA, Lee AA. Accelerating antiviral drug discovery: lessons from COVID-19. Nat Rev Drug Discov 2023; 22:585-603. [PMID: 37173515 PMCID: PMC10176316 DOI: 10.1038/s41573-023-00692-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, a wave of rapid and collaborative drug discovery efforts took place in academia and industry, culminating in several therapeutics being discovered, approved and deployed in a 2-year time frame. This article summarizes the collective experience of several pharmaceutical companies and academic collaborations that were active in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antiviral discovery. We outline our opinions and experiences on key stages in the small-molecule drug discovery process: target selection, medicinal chemistry, antiviral assays, animal efficacy and attempts to pre-empt resistance. We propose strategies that could accelerate future efforts and argue that a key bottleneck is the lack of quality chemical probes around understudied viral targets, which would serve as a starting point for drug discovery. Considering the small size of the viral proteome, comprehensively building an arsenal of probes for proteins in viruses of pandemic concern is a worthwhile and tractable challenge for the community.
Collapse
Affiliation(s)
- Annette von Delft
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Oxford Biomedical Research Centre, National Institute for Health Research, University of Oxford, Oxford, UK.
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | | | | | | | | | | | - Alpha A Lee
- PostEra, Inc., Cambridge, MA, USA.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
31
|
Jana AK, Sharawy M, May ER. Non-equilibrium virus particle dynamics: Microsecond MD simulations of the complete Flock House virus capsid under different conditions. J Struct Biol 2023; 215:107964. [PMID: 37105277 PMCID: PMC10205670 DOI: 10.1016/j.jsb.2023.107964] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/22/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
Flock House virus (FHV) is an animal virus and considered a model system for non-enveloped viruses. It has a small, icosahedral capsid (T=3) and a bipartite positive-sense RNA genome. We present an extensive study of the FHV capsid dynamics from all-atom molecular dynamics simulations of the complete capsid. The simulations explore different biologically relevant conditions (neutral/low pH, with/without RNA in the capsid) using the CHARMM force field. The results show that low pH destabilizes the capsid, causing radial expansion, and RNA stabilizes the capsid. The finding of low pH destabilization is biologically relevant because the capsid is exposed to low pH in the endosome, where conformational changes occur leading to genome release. We also observe structural changes at the fivefold and twofold symmetry axes that likely relate to the externalization of membrane active γ peptides through the fivefold vertex and extrusion of RNA at the twofold axis. Simulations using the Amber force field at neutral pH are also performed and display similar characteristics to the CHARMM simulations.
Collapse
Affiliation(s)
- Asis K Jana
- DepartmentofMolecularandCellBiology, UniversityofConnecticut, Storrs, CT06269-3125, USA; Department of Microbiology and Biotechnology, Sister Nivedita University, New Town, West Bengal 700156, India
| | - Mahmoud Sharawy
- DepartmentofMolecularandCellBiology, UniversityofConnecticut, Storrs, CT06269-3125, USA
| | - Eric R May
- DepartmentofMolecularandCellBiology, UniversityofConnecticut, Storrs, CT06269-3125, USA.
| |
Collapse
|
32
|
Kaushik S, Paliwal SK, Iyer MR, Patil VM. Promising Schiff bases in antiviral drug design and discovery. Med Chem Res 2023; 32:1063-1076. [PMID: 37305208 PMCID: PMC10171175 DOI: 10.1007/s00044-023-03068-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/25/2023] [Indexed: 06/13/2023]
Abstract
Emerging and re-emerging illnesses will probably present a new hazard of infectious diseases and have fostered the urge to research new antiviral agents. Most of the antiviral agents are analogs of nucleosides and only a few are non-nucleoside antiviral agents. There is quite a less percentage of marketed/clinically approved non-nucleoside antiviral medications. Schiff bases are organic compounds that possess a well-demonstrated profile against cancer, viruses, fungus, and bacteria, as well as in the management of diabetes, chemotherapy-resistant cases, and malarial infections. Schiff bases resemble aldehydes or ketones with an imine/azomethine group instead of a carbonyl ring. Schiff bases have a broad application profile not only in therapeutics/medicine but also in industrial applications. Researchers have synthesized and screened various Schiff base analogs for their antiviral potential. Some of the important heterocyclic compounds like istatin, thiosemicarbazide, quinazoline, quinoyl acetohydrazide, etc. have been used to derive novel Schiff base analogs. Keeping in view the outbreak of viral pandemics and epidemics, this manuscript compiles a review of Schiff base analogs concerning their antiviral properties and structural-activity relationship analysis.
Collapse
Affiliation(s)
- Shikha Kaushik
- Department of Pharmaceutical Chemistry, KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, Uttar Pradesh India
- Department of Pharmacy, Banasthali Vidyapith, Tonk, Rajasthan India
| | | | - Malliga R. Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, NIAAA/NIH, Rockville, MD USA
| | - Vaishali M. Patil
- Department of Pharmaceutical Chemistry, KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, Uttar Pradesh India
| |
Collapse
|
33
|
Hessien M, Donia T, Tabll AA, Adly E, Abdelhafez TH, Attia A, Alkafaas SS, Kuna L, Glasnovic M, Cosic V, Smolic R, Smolic M. Mechanistic-Based Classification of Endocytosis-Related Inhibitors: Does It Aid in Assigning Drugs against SARS-CoV-2? Viruses 2023; 15:1040. [PMID: 37243127 PMCID: PMC10222743 DOI: 10.3390/v15051040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) canonically utilizes clathrin-mediated endocytosis (CME) and several other endocytic mechanisms to invade airway epithelial cells. Endocytic inhibitors, particularly those targeting CME-related proteins, have been identified as promising antiviral drugs. Currently, these inhibitors are ambiguously classified as chemical, pharmaceutical, or natural inhibitors. However, their varying mechanisms may suggest a more realistic classification system. Herein, we present a new mechanistic-based classification of endocytosis inhibitors, in which they are segregated among four distinct classes including: (i) inhibitors that disrupt endocytosis-related protein-protein interactions, and assembly or dissociation of complexes; (ii) inhibitors of large dynamin GTPase and/or kinase/phosphatase activities associated with endocytosis; (iii) inhibitors that modulate the structure of subcellular components, especially the plasma membrane, and actin; and (iv) inhibitors that cause physiological or metabolic alterations in the endocytosis niche. Excluding antiviral drugs designed to halt SARS-CoV-2 replication, other drugs, either FDA-approved or suggested through basic research, could be systematically assigned to one of these classes. We observed that many anti-SARS-CoV-2 drugs could be included either in class III or IV as they interfere with the structural or physiological integrity of subcellular components, respectively. This perspective may contribute to our understanding of the relative efficacy of endocytosis-related inhibitors and support the optimization of their individual or combined antiviral potential against SARS-CoV-2. However, their selectivity, combined effects, and possible interactions with non-endocytic cellular targets need more clarification.
Collapse
Affiliation(s)
- Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Thoria Donia
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Ashraf A. Tabll
- National Research Centre, Microbial Biotechnology Department, Biotechnology Research Institute, Giza 12622, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo 11517, Egypt
| | - Eiman Adly
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Tawfeek H. Abdelhafez
- National Research Centre, Microbial Biotechnology Department, Biotechnology Research Institute, Giza 12622, Egypt
| | - Amany Attia
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Lucija Kuna
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Marija Glasnovic
- Department of Medicine, Family Medicine and History of Medicine, Faculty of Medicine Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Vesna Cosic
- Department of Paediatrics and Gynaecology with Obstetrics, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Robert Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
| | - Martina Smolic
- Department of Pharmacology and Biochemistry, Faculty of Dental Medicine and Health Osijek, University of J. J. Strossmayer Osijek, 31000 Osijek, Croatia
- Department of Pharmacology, Faculty of Medicine Osijek, University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
34
|
Targeting Human Proteins for Antiviral Drug Discovery and Repurposing Efforts: A Focus on Protein Kinases. Viruses 2023; 15:v15020568. [PMID: 36851782 PMCID: PMC9966946 DOI: 10.3390/v15020568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Despite the great technological and medical advances in fighting viral diseases, new therapies for most of them are still lacking, and existing antivirals suffer from major limitations regarding drug resistance and a limited spectrum of activity. In fact, most approved antivirals are directly acting antiviral (DAA) drugs, which interfere with viral proteins and confer great selectivity towards their viral targets but suffer from resistance and limited spectrum. Nowadays, host-targeted antivirals (HTAs) are on the rise, in the drug discovery and development pipelines, in academia and in the pharmaceutical industry. These drugs target host proteins involved in the virus life cycle and are considered promising alternatives to DAAs due to their broader spectrum and lower potential for resistance. Herein, we discuss an important class of HTAs that modulate signal transduction pathways by targeting host kinases. Kinases are considered key enzymes that control virus-host interactions. We also provide a synopsis of the antiviral drug discovery and development pipeline detailing antiviral kinase targets, drug types, therapeutic classes for repurposed drugs, and top developing organizations. Furthermore, we detail the drug design and repurposing considerations, as well as the limitations and challenges, for kinase-targeted antivirals, including the choice of the binding sites, physicochemical properties, and drug combinations.
Collapse
|
35
|
Li S, Sun Y, Du M, Shangguan A, Liu Z, Li W, Lina L, Liu W, Zhang S, Han H. Graphene Oxide Nanoparticles Combined with CRISPR/Cas9 System Enable Efficient Inhibition of Pseudorabies Virus. Bioconjug Chem 2023; 34:326-332. [PMID: 36629744 DOI: 10.1021/acs.bioconjchem.2c00570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We describe an application where graphene oxide nanoparticles (GONs) enable combined inhibition of Pseudorabies Virus (PRV) through delivery of a CRISPR/Cas9 system for targeted cleaving of a PRV genome and direct interaction with viral particles. The sheeted GONs could load CRISPR plasmid DNA (pDNA) to form a small sized, near-spheroidal GONs-CRISPR complex, which enables CRISPR pDNA efficient intracellular delivery and transient expression under serum conditions. Cell studies showed that GONs-CRISPR could allow rapid cellular uptake, endolysosomes escape, and nucleus transport within 3 h. Virus studies demonstrated that the pure GONs have antiviral activity and GONs-CRISPR could significantly inhibit PRV replication and result in progeny PRV decreasing by approximately 4000 times in infected host cells. Transmission electron microscopy (TEM) imaging showed that GONs-CRISPR could destroy the PRV structures by directly interacting with viral particles. This GONs-based strategy may extend the advanced application of the CRISPR system for antiviral action.
Collapse
Affiliation(s)
- Shuojun Li
- State Key Laboratory of Agriculture Microbiology, College of life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yumei Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China.,State Key Laboratory of Agriculture Microbiology, School of Animal Science and Technology, School of Animal Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Moqing Du
- State Key Laboratory of Agriculture Microbiology, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Aishao Shangguan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhongzhu Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Wentao Li
- State Key Laboratory of Agriculture Microbiology, School of Animal Science and Technology, School of Animal Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Liu Lina
- Department of Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Wenju Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Shujun Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Heyou Han
- State Key Laboratory of Agriculture Microbiology, College of life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.,State Key Laboratory of Agriculture Microbiology, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
36
|
Tripathi S, Sharma N, Naorem LD, Raghava GPS. ViralVacDB: A manually curated repository of viral vaccines. Drug Discov Today 2023; 28:103523. [PMID: 36764575 DOI: 10.1016/j.drudis.2023.103523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/13/2022] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Over the years, numerous vaccines have been developed against viral infections; however, a complete database that provides comprehensive information on viral vaccines has been lacking. In this review, along with our freely accessible database ViralVacDB, we provide details of the viral vaccines, their type, routes of administration and approving agencies. This repository systematically covers additional information such as disease name, adjuvant, manufacturer, clinical status, age and dosage against 422 viral vaccines, including 145 approved vaccines and 277 in clinical trials. We anticipate that this database will be highly beneficial to researchers and others working in pharmaceuticals and immuno-informatics.
Collapse
Affiliation(s)
- Sadhana Tripathi
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Neelam Sharma
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Leimarembi Devi Naorem
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| |
Collapse
|
37
|
Nag A, Banerjee K, Barman R, Kar J, Sarkar DP, Jana SS, Ghosh S. Direct Correlation between the Secondary Structure of an Amphiphilic Polymer and Its Prominent Antiviral Activity. J Am Chem Soc 2023; 145:579-584. [PMID: 36524964 DOI: 10.1021/jacs.2c11216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
An amphiphilic segmented polyurethane (F-PU-S), with pendant sulfate groups and a flexible hydrocarbon backbone, exhibits intrachain H-bonding-reinforced folding and hierarchical assembly, producing an anionic polymersome with efficient display of sulfate groups at the surface. It shows an excellent antiviral activity against Sendai virus (SV) by inhibiting its entry to the cells. Mechanistic investigation suggests fusion of the SV and the polymersome to produce larger particles in which neither the folded structure of the polymer nor the fusogenic property of the SV exists anymore. In sharp contrast, a structurally similar polymer R-PU-S, in which the chain folding pathway is blocked by replacing the flexible C6 chain with a rigid cyclohexane chain in the backbone, cannot form a similar polymersome structure and hence does not exhibit any antiviral activity. On the other hand, the third polymer (F-PU-C), which is similar to F-PU-S except for the pendant anionic groups (carboxylate instead of sulfate), also fails to exhibit any antiviral activity against SV, confirming the essential role of the chain folding as well as the pendant sulfate groups for the fusion-induced antiviral activity of F-PU-S, which provides an important structural guideline for developing new antiviral polymers.
Collapse
Affiliation(s)
- Atish Nag
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Kumarjeet Banerjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Ranajit Barman
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Joy Kar
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Debi P Sarkar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Siddhartha Sankar Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Suhrit Ghosh
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, 2A and 2B Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| |
Collapse
|
38
|
Huntjens DW, Dijkstra JA, Verwiel LN, Slijkhuis M, Elbers P, Welkers MRA, Veldkamp AI, Kuijvenhoven MA, de Leeuw DC, Abdullah-Koolmees H, Kuipers MT, Bartelink IH. Optimizing Antiviral Dosing for HSV and CMV Treatment in Immunocompromised Patients. Pharmaceutics 2023; 15:pharmaceutics15010163. [PMID: 36678792 PMCID: PMC9863155 DOI: 10.3390/pharmaceutics15010163] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Herpes simplex virus (HSV) and cytomegalovirus (CMV) are DNA viruses that are common among humans. Severely immunocompromised patients are at increased risk of developing HSV or CMV disease due to a weakened immune system. Antiviral therapy can be challenging because these drugs have a narrow therapeutic window and show significant pharmacokinetic variability. Above that, immunocompromised patients have various comorbidities like impaired renal function and are exposed to polypharmacy. This scoping review discusses the current pharmacokinetic (PK) and pharmacodynamic (PD) knowledge of antiviral drugs for HSV and CMV treatment in immunocompromised patients. HSV and CMV treatment guidelines are discussed, and multiple treatment interventions are proposed: early detection of drug resistance; optimization of dose to target concentration by therapeutic drug monitoring (TDM) of nucleoside analogs; the introduction of new antiviral drugs; alternation between compounds with different toxicity profiles; and combinations of synergistic antiviral drugs. This research will also serve as guidance for future research, which should focus on prospective evaluation of the benefit of each of these interventions in randomized controlled trials.
Collapse
Affiliation(s)
- Daan W. Huntjens
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Jacob A. Dijkstra
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-20-444-3524
| | - Lisanne N. Verwiel
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Mirjam Slijkhuis
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Paul Elbers
- Department of Intensive Care Medicine, Laboratory for Critical Care Computational Intelligence (LCCI), Amsterdam Medical Data Science (AMDS), Amsterdam Cardiovascular Science (ACS), Amsterdam Institute for Infection and Immunity (AII), Amsterdam University Medical Centre, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Matthijs R. A. Welkers
- Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Agnes I. Veldkamp
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marianne A. Kuijvenhoven
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - David C. de Leeuw
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Heshu Abdullah-Koolmees
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Postbus 85500, 3508 GA Utrecht, The Netherlands
- Clinical Pharmacy, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Maria T. Kuipers
- Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Imke H. Bartelink
- Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
39
|
Kembou-Ringert JE, Readman J, Smith CM, Breuer J, Standing JF. Applications of the hollow-fibre infection model (HFIM) in viral infection studies. J Antimicrob Chemother 2022; 78:8-20. [PMID: 36411255 PMCID: PMC9780528 DOI: 10.1093/jac/dkac394] [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: 11/23/2022] Open
Abstract
Conventional cell culture systems involve growing cells in stationary cultures in the presence of growth medium containing various types of supplements. At confluency, the cells are divided and further expanded in new culture dishes. This passage from confluent monolayer to sparse cultures does not reflect normal physiological conditions and represents quite a drastic physiological change that may affect the natural cell physiobiology. Hollow-fibre bioreactors were in part developed to overcome these limitations and since their inception, they have widely been used in production of monoclonal antibodies and recombinant proteins. These bioreactors are increasingly used to study antibacterial drug effects via simulation of in vivo pharmacokinetic profiles. The use of the hollow-fibre infection model (HFIM) in viral infection studies is less well developed and in this review we have analysed and summarized the current available literature on the use of these bioreactors, with an emphasis on viruses. Our work has demonstrated that this system can be applied for viral expansion, studies of drug resistance mechanisms, and studies of pharmacokinetic/pharmacodynamic (PK/PD) of antiviral compounds. These platforms could therefore have great applications in large-scale vaccine development, and in studies of mechanisms driving antiviral resistance, since the HFIM could recapitulate the same resistance mechanisms and mutations observed in vivo in clinic. Furthermore, some dosage and spacing regimens evaluated in the HFIM system, as allowing maximal viral suppression, are in line with clinical practice and highlight this 'in vivo-like' system as a powerful tool for experimental validation of in vitro-predicted antiviral activities.
Collapse
Affiliation(s)
- Japhette E Kembou-Ringert
- Department of Infection, Immunity & Inflammation, Great Ormond Street Institute of Child Health (ICH), University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - John Readman
- Department of Infection, Immunity & Inflammation, Great Ormond Street Institute of Child Health (ICH), University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Claire M Smith
- Department of Infection, Immunity & Inflammation, Great Ormond Street Institute of Child Health (ICH), University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Judith Breuer
- Department of Infection, Immunity & Inflammation, Great Ormond Street Institute of Child Health (ICH), University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Joseph F Standing
- Department of Infection, Immunity & Inflammation, Great Ormond Street Institute of Child Health (ICH), University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
40
|
Parigger L, Krassnigg A, Schopper T, Singh A, Tappler K, Köchl K, Hetmann M, Gruber K, Steinkellner G, Gruber CC. Recent changes in the mutational dynamics of the SARS-CoV-2 main protease substantiate the danger of emerging resistance to antiviral drugs. Front Med (Lausanne) 2022; 9:1061142. [PMID: 36590977 PMCID: PMC9794616 DOI: 10.3389/fmed.2022.1061142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction The current coronavirus pandemic is being combated worldwide by nontherapeutic measures and massive vaccination programs. Nevertheless, therapeutic options such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main-protease (Mpro) inhibitors are essential due to the ongoing evolution toward escape from natural or induced immunity. While antiviral strategies are vulnerable to the effects of viral mutation, the relatively conserved Mpro makes an attractive drug target: Nirmatrelvir, an antiviral targeting its active site, has been authorized for conditional or emergency use in several countries since December 2021, and a number of other inhibitors are under clinical evaluation. We analyzed recent SARS-CoV-2 genomic data, since early detection of potential resistances supports a timely counteraction in drug development and deployment, and discovered accelerated mutational dynamics of Mpro since early December 2021. Methods We performed a comparative analysis of 10.5 million SARS-CoV-2 genome sequences available by June 2022 at GISAID to the NCBI reference genome sequence NC_045512.2. Amino-acid exchanges within high-quality regions in 69,878 unique Mpro sequences were identified and time- and in-depth sequence analyses including a structural representation of mutational dynamics were performed using in-house software. Results The analysis showed a significant recent event of mutational dynamics in Mpro. We report a remarkable increase in mutational variability in an eight-residue long consecutive region (R188-G195) near the active site since December 2021. Discussion The increased mutational variability in close proximity to an antiviral-drug binding site as described herein may suggest the onset of the development of antiviral resistance. This emerging diversity urgently needs to be further monitored and considered in ongoing drug development and lead optimization.
Collapse
Affiliation(s)
- Lena Parigger
- Innophore GmbH, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | | | - Amit Singh
- Innophore GmbH, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Katharina Tappler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Michael Hetmann
- Innophore GmbH, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Austrian Centre of Industrial Biotechnology, Graz, Austria
| | - Karl Gruber
- Innophore GmbH, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Austrian Centre of Industrial Biotechnology, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Georg Steinkellner
- Innophore GmbH, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Christian C. Gruber
- Innophore GmbH, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Austrian Centre of Industrial Biotechnology, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| |
Collapse
|
41
|
Kotton CN, Torre-Cisneros J, Aguado JM, Alain S, Baldanti F, Baumann G, Boeken U, de la Calle M, Carbone J, Ciceri F, Comoli P, Couzi L, Danziger-Isakov L, Fernández-Ruiz M, Girmenia C, Grossi PA, Hirsch HH, Humar A, Kamar N, Kotton C, Ljungman P, Malagola M, Mira E, Mueller N, Sester M, Teng CLJ, Torre-Cisneros J, Ussetti P, Westall G, Wolf D, Zamora M. Cytomegalovirus in the transplant setting: Where are we now and what happens next? A report from the International CMV Symposium 2021. Transpl Infect Dis 2022; 24:e13977. [PMID: 36271650 PMCID: PMC10078482 DOI: 10.1111/tid.13977] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 12/24/2022]
Abstract
The CMV Symposium in September 2021 was an international conference dedicated to cytomegalovirus (CMV) infection after solid organ or hematopoietic stem cell transplantation. This review provides an overview of the presentations given by the expert faculty, supplemented with educational clinical cases. Topics discussed include CMV epidemiology and diagnosis, the burden of CMV infection and disease, CMV-specific immunity and management of CMV in transplant settings. Major advances in the prevention and treatment of CMV in the past decade and increased understanding of CMV immunity have led to improved patient outcomes. In the future, management algorithms may be individualized based on the transplant recipient's immune profile, which will mark the start of a new era for patients with CMV.
Collapse
Affiliation(s)
- Camille N Kotton
- Transplant and Immunocompromised Host Infectious Diseases, Infectious Diseases Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julián Torre-Cisneros
- Maimónides Institute for Biomedical Research of Cordoba (IMIBIC)/Reina Sofía University Hospital/University of Cordoba (UCO), Cordoba, Spain.,CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | | | - José Maria Aguado
- University Hospital 12 de Octubre, CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Sophie Alain
- French References Center for Herpes Viruses, Microbiology Department, CHU-Limoges, Limoges, France
| | - Fausto Baldanti
- Università di Pavia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Udo Boeken
- Department of Cardiac Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | | | - Javier Carbone
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Fabio Ciceri
- IRCCS San Raffaele Scientific Institute, University Vita-Salute San Raffaele, Milan, Italy
| | - Patrizia Comoli
- Cell Factory and Center for Advanced Therapies and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lionel Couzi
- Department of Nephrology, Transplantation, Dialysis and Apheresis, CHU Bordeaux CNRS-UMR 5164 ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Lara Danziger-Isakov
- Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, USA
| | | | | | | | | | | | | | | | - Per Ljungman
- Karolinska Hospital and Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | - Dana Wolf
- Hadassah University Medical Center, Jerusalem, Israel
| | - Marty Zamora
- University of Colorado at Denver Anschutz Medical Center, Colorado, USA
| |
Collapse
|
42
|
Omer AAM, Hinkula J, Tran PTH, Melik W, Zattarin E, Aili D, Selegård R, Bengtsson T, Khalaf H. Plantaricin NC8 αβ rapidly and efficiently inhibits flaviviruses and SARS-CoV-2 by disrupting their envelopes. PLoS One 2022; 17:e0278419. [PMID: 36449554 PMCID: PMC9710782 DOI: 10.1371/journal.pone.0278419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
Potent broad-spectrum antiviral agents are urgently needed to combat existing and emerging viral infections. This is particularly important considering that vaccine development is a costly and time consuming process and that viruses constantly mutate and render the vaccine ineffective. Antimicrobial peptides (AMP), such as bacteriocins, are attractive candidates as antiviral agents against enveloped viruses. One of these bacteriocins is PLNC8 αβ, which consists of amphipathic peptides with positive net charges that display high affinity for negatively charged pathogen membrane structures, including phosphatidylserine rich lipid membranes of viral envelopes. Due to the morphological and physiological differences between viral envelopes and host cell plasma membranes, PLNC8 αβ is thought to have high safety profile by specifically targeting viral envelopes without effecting host cell membranes. In this study, we have tested the antiviral effects of PLNC8 αβ against the flaviviruses Langat and Kunjin, coronavirus SARS-CoV-2, influenza A virus (IAV), and human immunodeficiency virus-1 (HIV-1). The concentration of PLNC8 αβ that is required to eliminate all the infective virus particles is in the range of nanomolar (nM) to micromolar (μM), which is surprisingly efficient considering the high content of cholesterol (8-35%) in their lipid envelopes. We found that viruses replicating in the endoplasmic reticulum (ER)/Golgi complex, e.g. SARS-CoV-2 and flaviviruses, are considerably more susceptible to PLNC8 αβ, compared to viruses that acquire their lipid envelope from the plasma membrane, such as IAV and HIV-1. Development of novel broad-spectrum antiviral agents can significantly benefit human health by rapidly and efficiently eliminating infectious virions and thereby limit virus dissemination and spreading between individuals. PLNC8 αβ can potentially be developed into an effective and safe antiviral agent that targets the lipid compartments of viral envelopes of extracellular virions, more or less independent of virus antigenic mutations, which faces many antiviral drugs and vaccines.
Collapse
Affiliation(s)
- Abubakr A. M. Omer
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jorma Hinkula
- Department of Biomedical and Clinical Sciences (BKV), Division of Molecular Medicine and Virology, Mucosa infection och inflammation Center (MIIC), Linköping University, Linköping, Sweden
| | - Pham-Tue-Hung Tran
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Wessam Melik
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Elisa Zattarin
- Laboratory of Molecular Materials, Department of Physics, Chemistry and Biology (IFM), Division of Biophysics and Bioengineering, Linköping University, Linköping, Sweden
| | - Daniel Aili
- Laboratory of Molecular Materials, Department of Physics, Chemistry and Biology (IFM), Division of Biophysics and Bioengineering, Linköping University, Linköping, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Department of Physics, Chemistry and Biology (IFM), Division of Biophysics and Bioengineering, Linköping University, Linköping, Sweden
| | - Torbjörn Bengtsson
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Hazem Khalaf
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
43
|
Abstract
Nanomaterials are prospective candidates for the elimination of viruses due to their multimodal mechanisms of action. Here, we tested the antiviral potential of a largely unexplored nanoparticle of cerium dioxide (CeO2). Two nano-CeO2 with opposing surface charge, (+) and (-), were assessed for their capability to decrease the plaque forming units (PFU) of four enveloped and two non-enveloped viruses during 1-h exposure. Statistically significant antiviral activity towards enveloped coronavirus SARS-CoV-2 and influenza virus was registered already at 20 mg Ce/l. For other two enveloped viruses, transmissible gastroenteritis virus and bacteriophage φ6, antiviral activity was evidenced at 200 mg Ce/l. As expected, the sensitivity of non-enveloped viruses towards nano-CeO2 was significantly lower. EMCV picornavirus showed no decrease in PFU until the highest tested concentration, 2000 mg Ce/l and MS2 bacteriophage showed slight non-monotonic response to high concentrations of nano-CeO2(-). Parallel testing of antiviral activity of Ce3+ ions and SiO2 nanoparticles allows to conclude that nano-CeO2 activity was neither due to released Ce-ions nor nonspecific effects of nanoparticulates. Moreover, we evidenced higher antiviral efficacy of nano-CeO2 compared with Ag nanoparticles. This result along with low antibacterial activity and non-existent cytotoxicity of nano-CeO2 allow us to propose CeO2 nanoparticles for specific antiviral applications.
Collapse
|
44
|
Antiviral Activity of Ficus rubiginosa Leaf Extracts against HSV-1, HCoV-229E and PV-1. Viruses 2022; 14:v14102257. [PMID: 36298811 PMCID: PMC9607631 DOI: 10.3390/v14102257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Ficus rubiginosa plant extract showed antimicrobial activity, but no evidence concerning its antiviral properties was reported. The antiviral activity of the methanolic extract (MeOH) and its n-hexane (H) and ethyl acetate (EA) fractions against Herpes simplex virus-1 (HSV-1), Human coronavirus (HCoV) -229E, and Poliovirus-1 (PV-1) was investigated in the different phases of viral infection in the VERO CCL-81 cell line. To confirm the antiviral efficacy, a qPCR was conducted. The recorded cytotoxic concentration 50% was 513.1, 298.6, and 56.45 µg/mL for MeOH, H, and EA, respectively, assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay after 72 h of treatment. The Ficus rubiginosa leaf extract inhibited the replication of HSV-1 in the early stages of infection, showing a complete inhibition up to 0.62, 0.31, and 1.25 µg/mL. Against HCoV-229E, a total inhibition up to 1.25 µg/mL for MeOH and H as well as 5 µg/mL for EA was observed. Otherwise, no activity was recorded against PV-1. The leaf extract could act directly on the viral envelope, destructuring the lipid membrane and/or directly blocking the enriched proteins on the viral surface. The verified gene inhibition suggested that the treatments with M, H, and EA impaired HSV-1 and HCoV-229E replication, with a greater antiviral efficiency against HSV-1 compared to HCoV-229E, possibly due to a greater affinity of Ficus rubiginosa towards membrane glycoproteins and/or the different lipid envelopes.
Collapse
|
45
|
Adeosun WB, More GK, Steenkamp P, Prinsloo G. Influence of seasonal and geographic variation on the anti-HSV-1 properties and chlorogenic acids content of Helichrysum aureonitens Sch. Bip. Front Mol Biosci 2022; 9:961859. [PMID: 36090044 PMCID: PMC9452954 DOI: 10.3389/fmolb.2022.961859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/20/2022] [Indexed: 12/02/2022] Open
Abstract
Pharmacological studies conducted in the past revealed the potential source of medicinal plants in the development of novel medicines. The phenolic contents of medicinal plants containing chlorogenic acids (CGA) have been linked to a variety of therapeutic effects, especially antiviral activity. Helichrysum aureonitens is a medicinal plant which has been reported to contain chlorogenic acids compounds and has also shown antiviral activities against a number of virus species including Herpes Simplex Virus-1 (HSV-1). In this study, the aim was to determine both the influence of seasonal variation and locality on the antiviral properties of H. aureonitens. Since chlorogenic acids have been reported as potent antiviral compounds, these compounds were targeted to determine the effects of locality and seasonal change on the chlorogenic acid profile, and subsequent antiviral activity. The ultra-performance liquid chromatography-quadrupole time-of-flight mass spectroscopy (UPLC-qTOF-MS) was employed to determine the metabolic profile variations of three derivatives of chlorogenic acids-caffeoylquinic acid (CQA), dicaffeoylquinic acid (DCQA) and tricaffeoylquinic acid (TCQA) in the harvested plants growing in two diverse geographical climates and two different seasons (spring and autumn). Using the cytopathic effect (CPE) reduction approach, twenty-six samples of the plants’ leaves and stems collected during spring and autumn at Telperion nature reserve in Mpumalanga and Wakefield farm, Midlands in KwaZulu-Natal region of South Africa were evaluated for anti-HSV activity. The MTT assay was used for the cytotoxicity evaluation of the extracts prior to antiviral determination. Seventeen (mostly spring collections) of the twenty-six extracts examined were found to have considerable anti-HSV activity as measured by a reduction in tissue culture infectious dose (TCID50) of less than 105. The UPLC-qTOF-MS result revealed that dicaffeoylquinic acid (DCQA) is the most abundant, with higher concentrations in both regions and seasons. 3-CQA was also shown to be the most abundant isomer of caffeoylquinic acid in this investigation.
Collapse
Affiliation(s)
- Wilson Bamise Adeosun
- Department of Agriculture and Animal Health, University of South Africa, Johannesburg, South Africa
- *Correspondence: Wilson Bamise Adeosun,
| | - Garland K. More
- College of Agriculture and Environmental Sciences, University of South Africa, Johannesburg, South Africa
| | - Paul Steenkamp
- Research Centre for Plant Metabolomics, Department of Biochemistry, University of Johannesburg, Johannesburg, South Africa
| | - Gerhard Prinsloo
- Department of Agriculture and Animal Health, University of South Africa, Johannesburg, South Africa
| |
Collapse
|
46
|
A Novel Approach of Antiviral Drugs Targeting Viral Genomes. Microorganisms 2022; 10:microorganisms10081552. [PMID: 36013970 PMCID: PMC9414836 DOI: 10.3390/microorganisms10081552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Outbreaks of viral diseases, which cause morbidity and mortality in animals and humans, are increasing annually worldwide. Vaccines, antiviral drugs, and antibody therapeutics are the most effective tools for combating viral infection. The ongoing coronavirus disease 2019 pandemic, in particular, raises an urgent need for the development of rapid and broad-spectrum therapeutics. Current antiviral drugs and antiviral antibodies, which are mostly specific at protein levels, have encountered difficulties because the rapid evolution of mutant viral strains resulted in drug resistance. Therefore, degrading viral genomes is considered a novel approach for developing antiviral drugs. The current article highlights all potent candidates that exhibit antiviral activity by digesting viral genomes such as RNases, RNA interference, interferon-stimulated genes 20, and CRISPR/Cas systems. Besides that, we introduce a potential single-chain variable fragment (scFv) that presents antiviral activity against various DNA and RNA viruses due to its unique nucleic acid hydrolyzing characteristic, promoting it as a promising candidate for broad-spectrum antiviral therapeutics.
Collapse
|
47
|
Pouyan P, Cherri M, Haag R. Polyglycerols as Multi-Functional Platforms: Synthesis and Biomedical Applications. Polymers (Basel) 2022; 14:polym14132684. [PMID: 35808728 PMCID: PMC9269438 DOI: 10.3390/polym14132684] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 02/07/2023] Open
Abstract
The remarkable and unique characteristics of polyglycerols (PG) have made them an attractive candidate for many applications in the biomedical and pharmaceutical fields. The presence of multiple hydroxy groups on the flexible polyether backbone not only enables the further modification of the PG structure but also makes the polymer highly water-soluble and results in excellent biocompatibility. In this review, the polymerization routes leading to PG with different architectures are discussed. Moreover, we discuss the role of these polymers in different biomedical applications such as drug delivery systems, protein conjugation, and surface modification.
Collapse
|
48
|
Alavi M, Kamarasu P, McClements DJ, Moore MD. Metal and metal oxide-based antiviral nanoparticles: Properties, mechanisms of action, and applications. Adv Colloid Interface Sci 2022; 306:102726. [PMID: 35785596 DOI: 10.1016/j.cis.2022.102726] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/05/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022]
Abstract
Certain types of metal-based nanoparticles are effective antiviral agents when used in their original form ("bare") or after their surfaces have been functionalized ("modified"), including those comprised of metals (e.g., silver) and metal oxides (e.g., zinc oxide, titanium dioxide, or iron dioxide). These nanoparticles can be prepared with different sizes, morphologies, surface chemistries, and charges, which leads to different antiviral activities. They can be used as aqueous dispersions or incorporated into composite materials, such as coatings or packaging materials. In this review, we provide an overview of the design, preparation, and characterization of metal-based nanoparticles. We then discuss their potential mechanisms of action against various kinds of viruses. Finally, the applications of some of the most common metal and metal oxide nanoparticles are discussed, including those fabricated from silver, zinc oxide, iron oxide, and titanium dioxide. In general, the major antiviral mechanisms of metal and metal oxide nanoparticles have been observed to be 1) attachment of nanoparticles to surface moieties of viral particles like spike glycoproteins, that disrupt viral attachment and uncoating in host cells; 2) generation of reactive oxygen species (ROS) that denature viral macromolecules such as nucleic acids, capsid proteins, and/or lipid envelopes; and 3) inactivation of viral glycoproteins by the disruption of the disulfide bonds of viral proteins. Several physicochemical properties of metal and metal oxide nanoparticles including size, shape, zeta potential, stability in physiological conditions, surface modification, and porosity can all impact the antiviral efficacy of the nanoparticles.
Collapse
Affiliation(s)
- Mehran Alavi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Kurdistan, Iran; Nanobiotechnology Laboratory, Biology Department, Faculty of Science, Razi University, Kermanshah, Iran.
| | - Pragathi Kamarasu
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | | | - Matthew D Moore
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
49
|
Nugroho CMH, Kurnia RS, Tarigan S, Silaen OSM, Triwidyaningtyas S, Wibawan IWT, Natalia L, Takdir AK, Soebandrio A. Screening and purification of NanB sialidase from Pasteurella multocida with activity in hydrolyzing sialic acid Neu5Acα(2–6)Gal and Neu5Acα(2–3)Gal. Sci Rep 2022; 12:9425. [PMID: 35676312 PMCID: PMC9177577 DOI: 10.1038/s41598-022-13635-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
Study on sialidases as antiviral agents has been widely performed, but many types of sialidase have not been tested for their antiviral activity. Pasteurella multocida NanB sialidase is one such sialidase that has never been isolated for further research. In this study, the activity of NanB sialidase was investigated in silico by docking the NanB sialidase of Pasteurella multocida to the Neu5Acα(2–6)Gal and Neu5Acα(2–3)Gal ligands. Additionally, some local isolates of Pasteurella multocida, which had the NanB gene were screened, and the proteins were isolated for further testing regarding their activity in hydrolyzing Neu5Acα(2–6)Gal and Neu5Acα(2–3)Gal. Silico studies showed that the NanB sialidase possesses an exceptional affinity towards forming a protein–ligand complex with Neu5Acα(2–6)Gal and Neu5Acα(2–3)Gal. NanB sialidase of Pasteurella multocida B018 at 0.129 U/mL and 0.258 U/mL doses can hydrolyze Neu5Acα(2–6)Gal and Neu5Acα(2–3)Gal better than other doses. In addition, those doses can inhibit effectively H9N2 viral binding to red blood cells. This study suggested that the NanB sialidase of Pasteurella multocida B018 has a potent antiviral activity because can hydrolyze sialic acid on red blood cells surface and inhibit the H9N2 viral binding to the cells.
Collapse
|
50
|
Sharifan A. COVID-19 and the Functionality of Antivirals: The Day After Tomorrow. Health Secur 2022; 20:270-271. [DOI: 10.1089/hs.2022.0073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Amin Sharifan
- Amin Sharifan, PharmD, is a Pharmacist and Researcher, Department of Pharmaceutical Care, Sina Hospital, and Research Center for Rational Use of Drugs; both at the Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|