1
|
Bonomini A, Mercorelli B, Loregian A. Antiviral strategies against influenza virus: an update on approved and innovative therapeutic approaches. Cell Mol Life Sci 2025; 82:75. [PMID: 39945883 PMCID: PMC11825441 DOI: 10.1007/s00018-025-05611-1] [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: 10/01/2024] [Revised: 01/27/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025]
Abstract
Influenza viruses still represent a great concern for Public Health by causing yearly seasonal epidemics and occasionally worldwide pandemics. Moreover, spillover events at the animal-human interface are becoming more frequent nowadays, also involving animal species not previously found as reservoirs. To restrict the effects of influenza virus epidemics, especially in at-risk population, and to prepare a drug arsenal for possible future pandemics, researchers worldwide have been working on the development of antiviral strategies since the 80's of the last century. One of the main obstacles is the considerable genomic variability of influenza viruses, which constantly poses the issues of drug-resistance emergence and immune evasion. This review summarizes the approved therapeutics for clinical management of influenza, promising new anti-flu compounds and monoclonal antibodies currently undergoing clinical evaluation, and molecules with efficacy against influenza virus in preclinical studies. Moreover, we discuss some innovative anti-influenza therapeutic approaches such as combination therapies and targeted protein degradation. Given the limited number of drugs approved for influenza treatment, there is a still strong need for novel potent anti-influenza drugs endowed with a high barrier to drug resistance and broad-spectrum activity against influenza viruses of animal origin that may be responsible of future large outbreaks and pandemics.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- Microbiology and Virology Unit, Padua University Hospital, Padua, Italy.
| |
Collapse
|
2
|
Bonomini A, Zhang J, Ju H, Zago A, Pacetti M, Tabarrini O, Massari S, Liu X, Mercorelli B, Zhan P, Loregian A. Synergistic activity of an RNA polymerase PA-PB1 interaction inhibitor with oseltamivir against human and avian influenza viruses in cell culture and in ovo. Antiviral Res 2024; 230:105980. [PMID: 39117284 DOI: 10.1016/j.antiviral.2024.105980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
In search of novel therapeutic options to treat influenza virus (IV) infections, we previously identified a series of inhibitors that act by disrupting the interactions between the PA and PB1 subunits of the viral RNA polymerase. These compounds showed broad-spectrum antiviral activity against human influenza A and B viruses and a high barrier to the induction of drug resistance in vitro. In this short communication, we investigated the effects of combinations of the PA-PB1 interaction inhibitor 54 with oseltamivir carboxylate (OSC), zanamivir (ZA), favipiravir (FPV), and baloxavir marboxil (BXM) on the inhibition of influenza A and B virus replication in vitro. We observed a synergistic effect of the 54/OSC and 54/ZA combinations and an antagonistic effect when 54 was combined with either FPV or BXM. Moreover, we demonstrated the efficacy of 54 against highly pathogenic avian influenza viruses (HPAIVs) both in cell culture and in the embryonated chicken eggs model. Finally, we observed that 54 enhances OSC protective effect against HPAIV replication in the embryonated eggs model. Our findings represent an advance in the development of alternative therapeutic strategies against both human and avian IV infections.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China
| | - Han Ju
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China
| | - Alessia Zago
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Martina Pacetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China.
| | | | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China.
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
3
|
Li Y, Huo S, Yin Z, Tian Z, Huang F, Liu P, Liu Y, Yu F. Retracted and republished from: "The current state of research on influenza antiviral drug development: drugs in clinical trial and licensed drugs". mBio 2024; 15:e0017524. [PMID: 38551343 PMCID: PMC11077966 DOI: 10.1128/mbio.00175-24] [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: 05/09/2024] Open
Abstract
Influenza viruses (IVs) threaten global human health due to the high morbidity, infection, and mortality rates. Currently, the influenza drugs recommended by the Food and Drug Administration are oseltamivir, zanamivir, peramivir, and baloxavir marboxil. These recommended antivirals are currently effective for major subtypes of IVs as the compounds target conserved domains in neuraminidase or polymerase acidic (PA) protein. However, this trend may gradually change due to the selection of antiviral drugs and the natural evolution of IVs. Therefore, there is an urgent need to develop drugs related to the treatment of influenza to deal with the next pandemic. Here, we summarized the cutting-edge research in mechanism of action, inhibitory activity, and clinical efficacy of drugs that have been approved and drugs that are still in clinical trials for influenza treatment. We hope this review will provide up-to-date and comprehensive information on influenza antivirals and generate hypotheses for screens and development of new broad-spectrum influenza drugs in the near future.
Collapse
Affiliation(s)
- Yanbai Li
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zhe Yin
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zuguang Tian
- Department of High-Tech Development, Baoding City Science and Technology Bureau, Baoding, China
| | - Fang Huang
- Epidemic Prevention Laboratory, Tongzhou District Center For Animal Disease Control and Prevention, Beijing, China
| | - Peng Liu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yue Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| |
Collapse
|
4
|
Chen N, Yao P, Zhang W, Zhang Y, Xin N, Wei H, Zhang T, Zhao C. Selenium nanoparticles: Enhanced nutrition and beyond. Crit Rev Food Sci Nutr 2023; 63:12360-12371. [PMID: 35848122 DOI: 10.1080/10408398.2022.2101093] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Selenium is a trace nutrient that has both nutritional and nutraceutical functions, whereas narrow nutritional range of selenium intake limits its use. Selenium nanoparticles (SeNPs) are less toxic and more bioavailable than traditional forms of selenium, suggesting that SeNPs have the potential to replace traditional selenium in food industries and/or biomedical fields. From the perspective of how SeNPs can be applied in health area, this review comprehensively discusses SeNPs in terms of its preparation, nutritional aspect, detoxification effect of heavy metals, nutraceutical functions and anti-pathogenic microorganism effects. By physical, chemical, or biological methods, inorganic selenium can be transformed into SeNPs which have increased stability and bioavailability as well as low toxicity. SeNPs are more effective than traditional selenium form in synthesizing selenoproteins like glutathione peroxidases. SeNPs can reshape the digestive system to facilitate digestion and absorption of nutrients. SeNPs have shown excellent potential to adjunctively treat cancer patients, enhance immune system, control diabetes, and prevent rheumatoid arthritis. Additionally, SeNPs have good microbial anti-pathogenic effects and can be used with other antimicrobial agents to fight against pathogenic bacteria, fungi, or viruses. Development of novel SeNPs with enhanced functions can greatly benefit the food-, nutraceutical-, and biomedical industries.
Collapse
Affiliation(s)
- Nan Chen
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Peng Yao
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Wei Zhang
- Weihai Baihe Biology Technological Co., Ltd, Rongcheng, Shandong, China
| | - Yutong Zhang
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Naicheng Xin
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Hongdi Wei
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun, China
- Jilin Engineering Technology Research Center for High Value Utilization of Animal By-Products, Jilin University, Changchun, China
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun, China
- Jilin Engineering Technology Research Center for High Value Utilization of Animal By-Products, Jilin University, Changchun, China
| |
Collapse
|
5
|
Li Y, Huo S, Yin Z, Tian Z, Huang F, Liu P, Liu Y, Yu F. The current state of research on influenza antiviral drug development: drugs in clinical trial and licensed drugs. mBio 2023; 14:e0127323. [PMID: 37610204 PMCID: PMC10653855 DOI: 10.1128/mbio.01273-23] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Influenza viruses (IVs) threaten global human health due to the high morbidity, infection, and mortality rates. Currently, the influenza drugs recommended by the FDA are oseltamivir, zanamivir, peramivir, and baloxavir marboxil. Notably, owing to the high variability of IVs, no drug exists that can effectively treat all types and subtypes of IVs. Moreover, the current trend of drug resistance is likely to continue as the viral genome is constantly mutating. Therefore, there is an urgent need to develop drugs related to the treatment of influenza to deal with the next pandemic. Here, we summarized the cutting-edge research in mechanism of action, inhibitory activity, and clinical efficacy of drugs that have been approved and drugs that are still in clinical trials for influenza treatment. We hope this review will provide up-to-date and comprehensive information on influenza antivirals and generate hypotheses for screens and development of new broad-spectrum influenza drugs in the near future.
Collapse
Affiliation(s)
- Yanbai Li
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zhe Yin
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zuguang Tian
- Baoding City Science and Technology Bureau, Baoding, China
| | - Fang Huang
- Tongzhou District Center For Animal Disease Control and Prevention, Beijing, China
| | - Peng Liu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yue Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| |
Collapse
|
6
|
Lotfi B, Mebarka O, Alhatlani BY, Abdallah EM, Kawsar SMA. Pharmacoinformatics and Breed-Based De Novo Hybridization Studies to Develop New Neuraminidase Inhibitors as Potential Anti-Influenza Agents. Molecules 2023; 28:6678. [PMID: 37764457 PMCID: PMC10534564 DOI: 10.3390/molecules28186678] [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: 08/26/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Influenza represents a profoundly transmissible viral ailment primarily afflicting the respiratory system. Neuraminidase inhibitors constitute a class of antiviral therapeutics employed in the management of influenza. These inhibitors impede the liberation of the viral neuraminidase protein, thereby impeding viral dissemination from the infected cell to host cells. As such, neuraminidase has emerged as a pivotal target for mitigating influenza and its associated complications. Here, we apply a de novo hybridization approach based on a breed-centric methodology to elucidate novel neuraminidase inhibitors. The breed technique amalgamates established ligand frameworks with the shared target, neuraminidase, resulting in innovative inhibitor constructs. Molecular docking analysis revealed that the seven synthesized breed molecules (designated Breeds 1-7) formed more robust complexes with the neuraminidase receptor than conventional clinical neuraminidase inhibitors such as zanamivir, oseltamivir, and peramivir. Pharmacokinetic evaluations of the seven breed molecules (Breeds 1-7) demonstrated favorable bioavailability and optimal permeability, all falling within the specified parameters for human application. Molecular dynamics simulations spanning 100 nanoseconds corroborated the stability of these breed molecules within the active site of neuraminidase, shedding light on their structural dynamics. Binding energy assessments, which were conducted through MM-PBSA analysis, substantiated the enduring complexes formed by the seven types of molecules and the neuraminidase receptor. Last, the investigation employed a reaction-based enumeration technique to ascertain the synthetic pathways for the synthesis of the seven breed molecules.
Collapse
Affiliation(s)
- Bourougaa Lotfi
- Group of Computational and Medicinal Chemistry, LMCE Laboratory, University of Biskra, BP 145, Biskra 70700, Algeria;
| | - Ouassaf Mebarka
- Group of Computational and Medicinal Chemistry, LMCE Laboratory, University of Biskra, BP 145, Biskra 70700, Algeria;
| | - Bader Y. Alhatlani
- Unit of Scientific Research, Applied College, Qassim University, Buraydah 52571, Saudi Arabia
| | - Emad M. Abdallah
- Department of Science Laboratories, College of Science and Arts, Qassim University, Ar Rass 51921, Saudi Arabia;
| | - Sarkar M. A. Kawsar
- Laboratory of Carbohydrate and Nucleoside Chemistry, Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong 4331, Bangladesh;
| |
Collapse
|
7
|
Batool S, Chokkakula S, Song MS. Influenza Treatment: Limitations of Antiviral Therapy and Advantages of Drug Combination Therapy. Microorganisms 2023; 11:183. [PMID: 36677475 PMCID: PMC9865513 DOI: 10.3390/microorganisms11010183] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Influenza infection is serious and debilitating for humans and animals. The influenza virus undergoes incessant mutation, segment recombination, and genome reassortment. As a result, new epidemics and pandemics are expected to emerge, making the elimination challenging of the disease. Antiviral therapy has been used for the treatment of influenza since the development of amantadine in the 1960s; however, its use is hampered by the emergence of novel strains and the development of drug resistance. Thus, combinational therapy with two or more antivirals or immunomodulators with different modes of action is the optimal strategy for the effective treatment of influenza infection. In this review, we describe current options for combination therapy, their performance, and constraints imposed by resistance, calling attention to the advantages of combination therapy against severe influenza infections. We also discuss the challenges of influenza therapy and the limitations of approved antiviral drugs.
Collapse
Affiliation(s)
| | | | - Min-Suk Song
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| |
Collapse
|
8
|
An anti-influenza combined therapy assessed by single cell RNA-sequencing. Commun Biol 2022; 5:1075. [PMID: 36216966 PMCID: PMC9549038 DOI: 10.1038/s42003-022-04013-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 09/20/2022] [Indexed: 11/08/2022] Open
Abstract
Influenza makes millions of people ill every year, placing a large burden on the healthcare system and the economy. To develop a treatment against influenza, we combined virucidal sialylated cyclodextrins with interferon lambda and demonstrated, in human airway epithelia, that the two compounds inhibit the replication of a clinical H1N1 strain more efficiently when administered together rather than alone. We investigated the mechanism of action of the combined treatment by single cell RNA-sequencing analysis and found that both the single and combined treatments impair viral replication to different extents across distinct epithelial cell types. We showed that each cell type comprises multiple sub-types, whose proportions are altered by H1N1 infection, and assessed the ability of the treatments to restore them. To the best of our knowledge this is the first study investigating the effectiveness of an antiviral therapy against influenza virus by single cell transcriptomic studies. When combined with interferon lambda, virucidal sialylated cyclodextrins inhibit the replication of a clinical H1N1 influenza strain in ex vivo human airway epithelia more efficiently than when delivered alone.
Collapse
|
9
|
Ashtiwi NM, Sarr D, Nagy T, Reneer ZB, Tripp RA, Rada B. The Hypothiocyanite and Amantadine Combination Treatment Prevents Lethal Influenza A Virus Infection in Mice. Front Immunol 2022; 13:859033. [PMID: 35663985 PMCID: PMC9159274 DOI: 10.3389/fimmu.2022.859033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/12/2022] [Indexed: 11/21/2022] Open
Abstract
The influenza virus has a large clinical burden and is associated with significant mortality and morbidity. The development of effective drugs for the treatment or prevention of influenza is important in order to reduce its impact. Adamantanes and neuraminidase inhibitors are two classes of anti-influenza drugs in which resistance has developed; thus, there is an urgent need to explore new therapeutic options. Boosting antiviral innate immune mechanisms in the airways represents an attractive approach. Hypothiocyanite (OSCN-) is produced by the airway epithelium and is effective in reducing the replication of several influenza A virus strains in vitro. It remains, however, largely unexplored whether OSCN- has such an antiviral effect in vivo. Here we determined the therapeutic potential of OSCN-, alone or in combination with amantadine (AMT), in preventing lethal influenza A virus replication in mice and in vitro. Mice intranasally infected with a lethal dose of A/Puerto Rico/8/1934 (H1N1) or A/Hong Kong/8/1968 (H3N2) were cured by the combination treatment of OSCN- and AMT. Monotherapy with OSCN- or AMT alone did not substantially improve survival outcomes. However, AMT+OSCN- treatment significantly inhibited viral replication, and in vitro treatment inhibited viral entry and nuclear transport of different influenza A virus strains (H1N1 and H3N2) including the AMT-resistant strain A/WSN/33 (H1N1). A triple combination treatment consisting of AMT, oseltamivir, and OSCN- was also tested and further inhibited in vitro viral replication of the AMT-resistant A/WSN/33 strain. These results suggest that OSCN- is a promising anti-influenza treatment option when combined with other antiviral drugs.
Collapse
Affiliation(s)
- Nuha Milad Ashtiwi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Demba Sarr
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Tamás Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Z. Beau Reneer
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
10
|
Liu X, Chen D, Su J, Zheng R, Ning Z, Zhao M, Zhu B, Li Y. Selenium nanoparticles inhibited H1N1 influenza virus-induced apoptosis by ROS-mediated signaling pathways. RSC Adv 2022; 12:3862-3870. [PMID: 35425430 PMCID: PMC8981154 DOI: 10.1039/d1ra08658h] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022] Open
Abstract
Influenza A (H1N1) viruses are distributed around the world and pose a threat to public health. Vaccination is the main treatment strategy to prevent influenza infection, but antiviral drugs also play an important role in controlling seasonal and pandemic influenza. Currently, as influenza viruses may be developing antiviral resistance, new agents with different modes of action are being investigated. Recently, selenium nanoparticles (SeNPs), which have antiviral effects, have attracted increasing attention in biomedical interventions. The appearance of nanotechnology has attracted great attention in the field of nanomedicine. SeNPs constitute an attractive vector platform for delivering a variety of drugs to action targets. SeNPs are being explored for potential therapeutic efficacy in a variety of oxidative stress and inflammation-mediated diseases, such as cancer, arthritis, diabetes, and kidney disease. SeNPs could inhibit infection of Madin-Darby canine kidney (MDCK) cells with H1N1 and prevent chromatin condensation and DNA fragmentation. ROS play a key role in physiological processes for apoptosis. SeNPs significantly inhibited the production of reactive oxygen species (ROS) in MDCK cells. Mechanistic investigation revealed that SeNPs inhibited the apoptosis induced by H1N1 virus infection in MDCK cells by improving the level of GPx1. Our results suggest that SeNPs are an effective selenium source and a promising H1N1 influenza antiviral candidate.
Collapse
Affiliation(s)
- Xia Liu
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| | - Danyang Chen
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| | - Jingyao Su
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| | - Ruilin Zheng
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| | - Zhihui Ning
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| | - Mingqi Zhao
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| | - Bing Zhu
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| | - Yinghua Li
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University No. 318 Renminzhong Road, Yuexiu District Guangzhou 510120 People's Republic of China
| |
Collapse
|
11
|
Chaiqin Qingning Capsule Inhibits Influenza Virus Infection and Inflammation In Vitro and In Vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6640731. [PMID: 34552653 PMCID: PMC8452396 DOI: 10.1155/2021/6640731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 07/21/2021] [Accepted: 08/20/2021] [Indexed: 02/03/2023]
Abstract
Background Chaiqin Qingning Capsule (CQ-C) is a traditional Chinese medicine (TCM) formula commonly used to treat respiratory infectious diseases in China. The aim of this study was to detect the effect and mechanism of CQ-C treated with influenza virus in vitro and vivo. Methods The cytotoxicity and antiviral activity of CQ-C in vitro was determined by methyl thiazolyl tetrazolium (MTT) assay. The regulation of CQ-C on cytokine/chemokine expression was evaluated using RT-qPCR. In addition, the effect of CQ-C on the pathway protein, NF-κB, and its phosphorylation level was verified by western blotting. After virus inoculation, BALB/c mice were administered with CQ-C of different concentrations for 7 days. Body weight, viral titer, lung pathology, and mortality of the mice were measured, and the level of inflammatory cytokines was also examined using real-time RT-qPCR. Results CQ-C inhibited the proliferation of influenza virus of various strains in vitro, with the 50% inhibitory concentration (IC50) ranging from 49 to 59 µg/mL. CQ-C downregulated virus-induced gene expression of IL-6, TNF-α, CXCL8, CXCL10, CCL5, and COX-2 in a dose-dependent manner in A549 cells. Also, CQ-C inhibited the expression of NF-κB protein of the signaling pathway. Moreover, a decrease of the lung index and mortality of mice was observed in the CQ-C (1 g/kg/d) group. The related cytokine/chemokine expression was also decreased in the early stages of infection in the mRNA level. Conclusion As a clinically applied Chinese prescription, our study shows that CQ-C has a wide range of effects on several influenza viruses. Moreover, CQ-C could play an important role in anti-influenza activity and anti-inflammation in vitro and in vivo. Thus, CQ-C may be a promising treatment option for influenza.
Collapse
|
12
|
Medaglia C, Zwygart ACA, Silva PJ, Constant S, Huang S, Stellacci F, Tapparel C. Interferon Lambda Delays the Emergence of Influenza Virus Resistance to Oseltamivir. Microorganisms 2021; 9:1196. [PMID: 34205874 PMCID: PMC8227012 DOI: 10.3390/microorganisms9061196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/28/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022] Open
Abstract
Influenza viruses are a leading cause of morbidity and mortality worldwide. These air-borne pathogens are able to cross the species barrier, leading to regular seasonal epidemics and sporadic pandemics. Influenza viruses also possess a high genetic variability, which allows for the acquisition of resistance mutations to antivirals. Combination therapies with two or more drugs targeting different mechanisms of viral replication have been considered an advantageous option to not only enhance the effectiveness of the individual treatments, but also reduce the likelihood of resistance emergence. Using an in vitro infection model, we assessed the barrier to viral resistance of a combination therapy with the neuraminidase inhibitor oseltamivir and human interferon lambda against the pandemic H1N1 A/Netherlands/602/2009 (H1N1pdm09) virus. We serially passaged the virus in a cell line derived from human bronchial epithelial cells in the presence or absence of increasing concentrations of oseltamivir alone or oseltamivir plus interferon lambda. While the treatment with oseltamivir alone quickly induced the emergence of antiviral resistance through a single mutation in the neuraminidase gene, the co-administration of interferon lambda delayed the emergence of drug-resistant influenza virus variants. Our results suggest a possible clinical application of interferon lambda in combination with oseltamivir to treat influenza.
Collapse
Affiliation(s)
- Chiara Medaglia
- Department of Microbiology and Molecular Medicine, University of Geneva, 1206 Geneva, Switzerland; (C.M.); (A.C.-A.Z.)
| | | | - Paulo Jacob Silva
- Insitute of Materials, Ecole polytechnique fédérale de Lausanne, 1015 Lausanne, Switzerland; (P.J.S.); (F.S.)
| | | | - Song Huang
- Epithelix Sas, 1228 Geneva, Switzerland; (S.C.); (S.H.)
| | - Francesco Stellacci
- Insitute of Materials, Ecole polytechnique fédérale de Lausanne, 1015 Lausanne, Switzerland; (P.J.S.); (F.S.)
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, 1206 Geneva, Switzerland; (C.M.); (A.C.-A.Z.)
| |
Collapse
|
13
|
Ashar HK, Pulavendran S, Rudd JM, Maram P, Achanta M, Chow VTK, Malayer JR, Snider TA, Teluguakula N. Administration of a CXC Chemokine Receptor 2 (CXCR2) Antagonist, SCH527123, Together with Oseltamivir Suppresses NETosis and Protects Mice from Lethal Influenza and Piglets from Swine-Influenza Infection. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:669-685. [PMID: 33453177 PMCID: PMC8027923 DOI: 10.1016/j.ajpath.2020.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Excessive neutrophil influx, their released neutrophil extracellular traps (NETs), and extracellular histones are associated with disease severity in influenza-infected patients. Neutrophil chemokine receptor CXC chemokine receptor 2 (CXCR2) is a critical target for suppressing neutrophilic inflammation. Herein, temporal dynamics of neutrophil activity and NETosis were investigated to determine the optimal timing of treatment with the CXCR2 antagonist, SCH527123 (2-hydroxy-N,N-dimethyl-3-[2-([(R)-1-(5-methyl-furan-2-yl)-propyl]amino)-3,4-dioxo-cyclobut-1-enylamino]-benzamide), and its efficacy together with antiviral agent, oseltamivir, was tested in murine and piglet influenza-pneumonia models. SCH527123 plus oseltamivir markedly improved survival of mice infected with lethal influenza, and diminished lung pathology in swine-influenza-infected piglets. Mechanistically, addition of SCH527123 in the combination treatment attenuated neutrophil influx, NETosis, in both mice and piglets. Furthermore, neutrophils isolated from influenza-infected mice showed greater susceptibility to NETotic death when stimulated with a CXCR2 ligand, IL-8. In addition, CXCR2 stimulation induced nuclear translocation of neutrophil elastase, and enhanced citrullination of histones that triggers chromatin decondensation during NET formation. Studies on temporal dynamics of neutrophils and NETs during influenza thus provide important insights into the optimal timing of CXCR2 antagonist treatment for attenuating neutrophil-mediated lung pathology. These findings reveal that pharmacologic treatment with CXCR2 antagonist together with an antiviral agent could significantly ameliorate morbidity and mortality in virulent and sublethal influenza infections.
Collapse
Affiliation(s)
- Harshini K Ashar
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Sivasami Pulavendran
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Jennifer M Rudd
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Prasanthi Maram
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Mallika Achanta
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Vincent T K Chow
- National University Health System Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, School of Medicine, National University of Singapore, Singapore
| | - Jerry R Malayer
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Timothy A Snider
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | | |
Collapse
|
14
|
Ikram M, Javed B, Raja NI, Mashwani ZUR. Biomedical Potential of Plant-Based Selenium Nanoparticles: A Comprehensive Review on Therapeutic and Mechanistic Aspects. Int J Nanomedicine 2021; 16:249-268. [PMID: 33469285 PMCID: PMC7811472 DOI: 10.2147/ijn.s295053] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Selenium nanoparticles (SeNPs) have advantages over other nanomaterials because of the promising role of selenium in the stabilization of the immune system and activation of the defense response. The use of SeNPs and their supplements not only have pharmacological significance but also boost and prepare the body's immune system to fight the pathogens. This review summarizes the recent progress in the biogenesis of plant-based SeNPs by using various plant species and the role of secondary metabolites on their biocompatible functioning. Phyto-synthesis of SeNPs results in the synthesis of nanomaterials of various, size, shape and biochemical nature and has advantages over other routine physical and chemical methods because of their biocompatibility, eco-friendly nature and in vivo actions. Unfortunately, the plant-based SeNPs failed to attain considerable attention in the pharmaceutical industry. However, a few studies were performed to explore the therapeutic potential of the SeNPs against various cancer cells, microbial pathogens, viral infections, hepatoprotective actions, diabetic management, and antioxidant approaches. Further, some of the selenium-based drug delivery systems are developed by engineering the SeNPs with the functional ligands to deliver drugs to the targeted sites. This review also provides up-to-date information on the mechanistic actions that the SeNPs adopt to achieve their designated tasks as it may help to develop precision medicine with customized treatment and healthcare for the ailing population.
Collapse
Affiliation(s)
- Muhammad Ikram
- Department of Botany, PMAS Arid Agriculture University, Rawalpindi, Punjab 46300, Pakistan
| | - Bilal Javed
- Department of Botany, PMAS Arid Agriculture University, Rawalpindi, Punjab 46300, Pakistan
| | - Naveed Iqbal Raja
- Department of Botany, PMAS Arid Agriculture University, Rawalpindi, Punjab 46300, Pakistan
| | - Zia-Ur-Rehman Mashwani
- Department of Botany, PMAS Arid Agriculture University, Rawalpindi, Punjab 46300, Pakistan
| |
Collapse
|
15
|
Mou Q, Jiang Y, Zhu L, Zhu Z, Ren T. EGCG induces β-defensin 3 against influenza A virus H1N1 by the MAPK signaling pathway. Exp Ther Med 2020; 20:3017-3024. [PMID: 32855668 PMCID: PMC7444400 DOI: 10.3892/etm.2020.9047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/19/2020] [Indexed: 12/22/2022] Open
Abstract
Epigallocatechin gallate (EGCG) is the main component of green tea, which has been proven to inhibit a variety of viruses, including influenza A virus. However, the mechanism of EGCG against influenza virus remains to be further explored. The mechanism of EGCG against influenza virus was studied. The results showed that EGCG significantly increased the levels of HBD3 mRNA and protein, while the levels of phosphorylation of (p)-p38 MAPK, ERK and JNK after EGCG treatment were significantly up-regulated. p38 MAPK, ERK and JNK inhibitors significantly inhibited the expression of HBD3 induced by EGCG. On the other hand, EGCG significantly inhibited the expression of HA and NP proteins in influenza A virus H1N1, but attenuated the anti-influenza A virus effect of EGCG after silencing HBD3. Thus, the anti-influenza virus effect of EGCG is related to the induction of HBD3 expression. In addition, the expression of EGCG-induced HBD3 is related to the p38 MAPK, ERK and JNK signaling pathways. The research data show that EGCG can induce HBD3 expression through p38 MAPK, ERK and JNK signaling pathway to inhibit the replication of influenza A virus H1N1, providing a new and effective candidate drug for influenza virus.
Collapse
Affiliation(s)
- Qiuju Mou
- Department of Blood Transfusion, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou 550014, P.R. China
| | - Yan Jiang
- Department of Microbiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Lili Zhu
- Department of Blood Transfusion, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Zixin Zhu
- School of Basic Medicine Science, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Tingting Ren
- Department of Physiology Chemistry, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
16
|
Laninamivir-Interferon Lambda 1 Combination Treatment Promotes Resistance by Influenza A Virus More Rapidly than Laninamivir Alone. Antimicrob Agents Chemother 2020; 64:AAC.00301-20. [PMID: 32393488 DOI: 10.1128/aac.00301-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
Each year, 5% to 20% of the population of the United States becomes infected with influenza A virus. Combination therapy with two or more antiviral agents has been considered a potential treatment option for influenza virus infection. However, the clinical results derived from combination treatment with two or more antiviral drugs have been variable. We examined the effectiveness of cotreatment with two distinct classes of anti-influenza drugs, i.e., neuraminidase (NA) inhibitor, laninamivir, and interferon lambda 1 (IFN-λ1), against the emergence of drug-resistant virus variants in vitro We serially passaged pandemic A/California/04/09 [A(H1N1)pdm09] influenza virus in a human lung epithelial cell line (Calu-3) in the presence or absence of increasing concentrations of laninamivir or laninamivir plus IFN-λ1. Surprisingly, laninamivir used in combination with IFN-λ1 promoted the emergence of the E119G NA mutation five passages earlier than laninamivir alone (passage 2 versus passage 7, respectively). Acquisition of this mutation resulted in significantly reduced sensitivity to the NA inhibitors laninamivir (∼284-fold) and zanamivir (∼1,024-fold) and decreased NA enzyme catalytic activity (∼5-fold) compared to the parental virus. Moreover, the E119G NA mutation emerged together with concomitant hemagglutinin (HA) mutations (T197A and D222G), which were selected more rapidly by combination treatment with laninamivir plus IFN-λ1 (passages 2 and 3, respectively) than by laninamivir alone (passage 10). Our results show that treatment with laninamivir alone or in combination with IFN-λ1 can lead to the emergence of drug-resistant influenza virus variants. The addition of IFN-λ1 in combination with laninamivir may promote acquisition of drug resistance more rapidly than treatment with laninamivir alone.
Collapse
|
17
|
Lianhua-Qingwen Displays Antiviral and Anti-Inflammatory Activity and Synergistic Effects with Oseltamivir against Influenza B Virus Infection in the Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3196375. [PMID: 32565852 PMCID: PMC7293728 DOI: 10.1155/2020/3196375] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022]
Abstract
Influenza B virus (IBV) is one of the main pathogens of the annual influenza epidemic, and the disease burden is significant, especially among children and young teenagers. In this study, the antiviral and anti-inflammatory effects of a traditional Chinese medicine prescription, the Lianhua-Qingwen capsule, were evaluated. Our results showed that Lianhua-Qingwen capsule can inhibit both Victoria and Yamagata lineages, and the 50% inhibitive concentrations ranged from 0.228 ± 0.150 to 0.754 ± 0.161 mg/mL. The time course results demonstrated that IBV yields were reduced with treatment at 0–4 h after infection, and the mechanistic research verified that Lianhua-Qingwen capsule has hemagglutination inhibition activity against B/Guangzhou/0215/2012 but not A/California/04/2009. In addition to antiviral activity, Lianhua-Qingwen capsule can also inhibit excessive expression of RANTES, IL-6, IL-8, IP-10, TNF-α, MCP-1, MIP-1β, and IFN-λ at the mRNA level and prevent a severe inflammatory response. The in vivo results confirmed that orally administered Lianhua-Qingwen capsule (100–400 mg/kg/day) does not reduce IBV-induced lung viral load and mortality in mice. However, the pathological change in lungs was alleviated, and there were fewer inflammatory cells in the lungs of Lianhua-Qingwen capsule treated mice than those in controls. Further research confirmed that the combination treatment of 200 mg/kg/day of Lianhua-Qingwen capsule with 2 mg/kg/day of oseltamivir significantly reduced IBV infection over the individual administration of either alone in vivo. Our findings prove that Lianhua-Qingwen capsule could be used as an assistant medicine to enhance the effect of oseltamivir against influenza B virus infection.
Collapse
|
18
|
Nile SH, Kim DH, Nile A, Park GS, Gansukh E, Kai G. Probing the effect of quercetin 3-glucoside from Dianthus superbus L against influenza virus infection- In vitro and in silico biochemical and toxicological screening. Food Chem Toxicol 2019; 135:110985. [PMID: 31765700 DOI: 10.1016/j.fct.2019.110985] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022]
Abstract
Investigation of antiviral and cytotoxic effect of quercetin 3-glucoside (Q3G) from Dianthus superbus L over influenza virus infection and replication were studied. Moreover, anti-influenza mechanism was screened by time-dependent antiviral assay, virus-induced symptoms and related gene expressions. The blockade of cap-binding domain of polymerase basic protein subunit were analysed by molecular docking study. The Q3G demonstrated potent antiviral activity showing 4.93, 6.43, 9.94, 8.3, and 7.1 μg/mL of IC50 for A/PR/8/34, A/Victoria/3/75, A/WS/33, B/Maryland/1/59, and B/Lee/40, respectively. The cellular toxicity of Q3G and oseltamivir (control) were tested and >100 μg/mL of CC50 value considered as nontoxic. Influenza A virus infection induced a higher ROS production, however potentially reduced by Q3G treatment and significantly blocked virus infection induced acidic vesicular organelles (AVO). Moreover, Q3G has no inhibitory effect for neuraminidase activity but blocked virus replication through time dependent assay and showed more competitive binding affinity (-8.0 kcal/mal) than GTP (-7.0 kcal/mol) to block polymerase basic protein-2 subunit of influenza virus. Q3G from D. superbus showed potent antiviral activity against influenza A and B viruses with suppressive effect on virus-induced cellular ROS generation and AVO formation. Thus, this study provided a new line of research for Q3G to develop possible natural anti-influenza drug.
Collapse
Affiliation(s)
- Shivraj Hariram Nile
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Doo Hwan Kim
- Department of Bio-resources and Food Science, Konkuk University, Seoul, 143701, South Korea
| | - Arti Nile
- Department of Bio-resources and Food Science, Konkuk University, Seoul, 143701, South Korea
| | - Gyun Seok Park
- Department of Bio-resources and Food Science, Konkuk University, Seoul, 143701, South Korea
| | - Enkhtaivan Gansukh
- Department of Bio-resources and Food Science, Konkuk University, Seoul, 143701, South Korea; Department of Life Science and Biotechnology, Huree University, Ulaanbaatar, Mongolia.
| | - Guoyin Kai
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| |
Collapse
|
19
|
Ghaffari H, Tavakoli A, Moradi A, Tabarraei A, Bokharaei-Salim F, Zahmatkeshan M, Farahmand M, Javanmard D, Kiani SJ, Esghaei M, Pirhajati-Mahabadi V, Monavari SH, Ataei-Pirkooh A. Inhibition of H1N1 influenza virus infection by zinc oxide nanoparticles: another emerging application of nanomedicine. J Biomed Sci 2019; 26:70. [PMID: 31500628 PMCID: PMC6734352 DOI: 10.1186/s12929-019-0563-4] [Citation(s) in RCA: 221] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Currently available anti-influenza drugs are often associated with limitations such as toxicity and the appearance of drug-resistant strains. Therefore, there is a pressing need for the development of novel, safe and more efficient antiviral agents. In this study, we evaluated the antiviral activity of zinc oxide nanoparticles (ZnO-NPs) and PEGylated zinc oxide nanoparticles against H1N1 influenza virus. METHODS The nanoparticles were characterized using the inductively coupled plasma mass spectrometry, x-ray diffraction analysis, and electron microscopy. MTT assay was applied to assess the cytotoxicity of the nanoparticles, and anti-influenza activity was determined by TCID50 and quantitative Real-Time PCR assays. To study the inhibitory impact of nanoparticles on the expression of viral antigens, an indirect immunofluorescence assay was also performed. RESULTS Post-exposure of influenza virus with PEGylated ZnO-NPs and bare ZnO-NPs at the highest non-toxic concentrations could be led to 2.8 and 1.2 log10 TCID50 reduction in virus titer when compared to the virus control, respectively (P < 0.0001). At the highest non-toxic concentrations, the PEGylated and unPEGylated ZnO-NPs led to inhibition rates of 94.6 and 52.2%, respectively, which were calculated based on the viral loads. There was a substantial decrease in fluorescence emission intensity in viral-infected cell treated with PEGylated ZnO-NPs compared to the positive control. CONCLUSIONS Taken together, our study indicated that PEGylated ZnO-NPs could be a novel, effective, and promising antiviral agent against H1N1 influenza virus infection, and future studies can be designed to explore the exact antiviral mechanism of these nanoparticles.
Collapse
Affiliation(s)
- Hadi Ghaffari
- Department of Medical Virology, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Tavakoli
- Department of Medical Virology, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolvahab Moradi
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alijan Tabarraei
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Masoumeh Zahmatkeshan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farahmand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Davod Javanmard
- Department of Medical Virology, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Jalal Kiani
- Department of Medical Virology, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Esghaei
- Department of Medical Virology, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Pirhajati-Mahabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Angila Ataei-Pirkooh
- Department of Medical Virology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Hu CMJ, Chen YT, Fang ZS, Chang WS, Chen HW. Antiviral efficacy of nanoparticulate vacuolar ATPase inhibitors against influenza virus infection. Int J Nanomedicine 2018; 13:8579-8593. [PMID: 30587980 PMCID: PMC6298390 DOI: 10.2147/ijn.s185806] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Influenza virus infections are a major public health concern worldwide. Conventional treatments against the disease are designed to target viral proteins. However, the emergence of viral variants carrying drug-resistant mutations can outpace the development of pathogen-targeting antivirals. Diphyllin and bafilomycin are potent vacuolar ATPase (V-ATPase) inhibitors previously shown to have broad-spectrum antiviral activity. However, their poor water solubility and potential off-target effect limit their clinical application. Methods In this study, we report that nanoparticle encapsulation of diphyllin and bafilomycin improves the drugs’ anti-influenza applicability. Results Using PEG-PLGA diblock copolymers, sub-200 nm diphyllin and bafilomycin nanoparticles were prepared, with encapsulation efficiency of 42% and 100%, respectively. The drug-loaded nanoparticles have sustained drug release kinetics beyond 72 hours and facilitate intracellular drug delivery to two different influenza virus-permissive cell lines. As compared to free drugs, the nanoparticulate V-ATPase inhibitors exhibited lower cytotoxicity and greater in vitro antiviral activity, improving the therapeutic index of diphyllin and bafilomycin by approximately 3 and 5-fold, respectively. In a mouse model of sublethal influenza challenge, treatment with diphyllin nanoparticles resulted in reduced body weight loss and viral titer in the lungs. In addition, following a lethal influenza viral challenge, diphyllin nanoparticle treatment conferred a survival advantage of 33%. Conclusions These results demonstrate the potential of the nanoparticulate V-ATPase inhibitors for host-targeted treatment against influenza.
Collapse
Affiliation(s)
- Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Research Center for Nanotechnology and Infectious Diseases, Taipei, Taiwan,
| | - You-Ting Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan,
| | - Zih-Syun Fang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan,
| | - Wei-Shan Chang
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan,
| | - Hui-Wen Chen
- Research Center for Nanotechnology and Infectious Diseases, Taipei, Taiwan, .,Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan,
| |
Collapse
|
21
|
Melville K, Rodriguez T, Dobrovolny HM. Investigating Different Mechanisms of Action in Combination Therapy for Influenza. Front Pharmacol 2018; 9:1207. [PMID: 30405419 PMCID: PMC6206389 DOI: 10.3389/fphar.2018.01207] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/03/2018] [Indexed: 01/15/2023] Open
Abstract
Combination therapy for influenza can have several benefits, from reducing the emergence of drug resistant virus strains to decreasing the cost of antivirals. However, there are currently only two classes of antivirals approved for use against influenza, limiting the possible combinations that can be considered for treatment. However, new antivirals are being developed that target different parts of the viral replication cycle, and their potential for use in combination therapy should be considered. The role of antiviral mechanism of action in the effectiveness of combination therapy has not yet been systematically investigated to determine whether certain antiviral mechanisms of action pair well in combination. Here, we use a mathematical model of influenza to model combination treatment with antivirals having different mechanisms of action to measure peak viral load, infection duration, and synergy of different drug combinations. We find that antivirals that lower the infection rate and antivirals that increase the duration of the eclipse phase perform poorly in combination with other antivirals.
Collapse
Affiliation(s)
- Kelli Melville
- Physics Department, East Carolina University, Greenville, NC, United States
| | - Thalia Rodriguez
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| | - Hana M. Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| |
Collapse
|
22
|
Zoonotic Influenza and Human Health-Part 2: Clinical Features, Diagnosis, Treatment, and Prevention Strategies. Curr Infect Dis Rep 2018; 20:38. [PMID: 30069787 PMCID: PMC7102074 DOI: 10.1007/s11908-018-0643-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Purpose of Review Zoonotic influenza viruses are those influenza viruses that cross the animal-human barrier and can cause disease in humans, manifesting from minor respiratory illnesses to multiorgan dysfunction. The increasing incidence of infections caused by these viruses worldwide has necessitated focused attention to improve both diagnostic as well as treatment modalities. In this second part of a two-part review, we discuss the clinical features, diagnostic modalities, and treatment of zoonotic influenza, and provide an overview of prevention strategies. Recent Findings Illnesses caused by novel reassortant avian influenza viruses continue to be detected and described; most recently, a human case of avian influenza A(H7N4) has been described from China. We continue to witness increasing rates of A(H7N9) infections, with the latest (fifth) wave, from late 2016 to 2017, being the largest to date. The case fatality rate for A(H7N9) and A(H5N1) infections among humans is much higher than that of seasonal influenza infections. Since the emergence of the A(H1N1) 2009 pandemic, and subsequently A(H7N9), testing and surveillance for novel influenzas have become more effective. Various newer treatment options, including peramivir, favipiravir (T-705), and DAS181, and human or murine monoclonal antibodies have been evaluated in vitro and in animal models. Summary Armed with robust diagnostic modalities, antiviral medications, vaccines, and advanced surveillance systems, we are today better prepared to face a new influenza pandemic and to limit the burden of zoonotic influenza than ever before. Sustained efforts and robust research are necessary to efficiently deal with the highly mutagenic zoonotic influenza viruses.
Collapse
|
23
|
Triple combination of FDA-approved drugs including flufenamic acid, clarithromycin and zanamivir improves survival of severe influenza in mice. Arch Virol 2018; 163:2349-2358. [PMID: 29736671 DOI: 10.1007/s00705-018-3852-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Seasonal influenza virus remains a common cause of mortality despite the use of neuraminidase inhibitors. This study evaluated the efficacy of a triple combination of zanamivir, clarithromycin and flufenamic acid (FFA) in the treatment of influenza virus A(H1N1) infection. An in vitro cell protection assay and a multiple-cycle growth assay showed that the antiviral activity of zanamivir was enhanced when combined with clarithromycin or FFA. A mouse challenge model was used here for the evaluation of the in vivo efficacy of the triple combination treatment. We found that mice receiving the triple combination of FFA, zanamivir, and clarithromycin had a significantly better survival rate than those receiving the double combination of zanamivir and clarithromycin (88% versus 44%, P = 0.0083) or zanamivir monotherapy (88% versus 26%, P = 0.0002). Mice in the FFA-zanamivir-clarithromycin triple combination group also exhibited significantly less body weight loss than those in the zanamivir-clarithromycin double combination group. There was no significant difference in the lung viral titers among the different groups from day 2 to day 6 postinfection. However, the levels of IL-1β, TNF-α and RANTES in the FFA-zanamivir-clarithromycin triple combination group were significantly lower than those in the zanamivir-clarithromycin double combination group, zanamivir monotherapy group, or solvent group on day 2 postinfection. Our findings showed that the FFA-zanamivir-clarithromycin triple combination improved the inflammatory markers and survival of severe influenza A(H1N1) infection in mice.
Collapse
|
24
|
Han J, Perez J, Schafer A, Cheng H, Peet N, Rong L, Manicassamy B. Influenza Virus: Small Molecule Therapeutics and Mechanisms of Antiviral Resistance. Curr Med Chem 2018; 25:5115-5127. [PMID: 28933281 PMCID: PMC8735713 DOI: 10.2174/0929867324666170920165926] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 03/09/2017] [Accepted: 05/26/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Influenza viruses cause severe upper respiratory illness in children and the elderly during seasonal epidemics. Influenza viruses from zoonotic reservoirs can also cause pandemics with significant loss of life in all age groups. Although vaccination is one of the most effective methods to protect against seasonal epidemics, seasonal vaccines vary in efficacy, can be ineffective in the elderly population, and do not provide protection against novel strains. Small molecule therapeutics are a critical part of our antiviral strategies to control influenza virus epidemics and pandemics as well as to ameliorate disease in elderly and immunocompromised individuals. OBJECTIVE This review aims to summarize the existing antiviral strategies for combating influenza viruses, the mechanisms of antiviral resistance for available drugs, and novel therapeutics currently in development. METHODS We systematically evaluated and synthesized the published scientific literature for mechanistic detail into therapeutic strategies against influenza viruses. RESULTS Current IAV strains have developed resistance to neuraminidase inhibitors and nearly complete resistance to M2 ion channel inhibitors, exacerbated by sub-therapeutic dosing used for treatment and chemoprophylaxis. New tactics include novel therapeutics targeting host components and combination therapy, which show potential for fighting influenza virus disease while minimizing viral resistance. CONCLUSION Antiviral drugs are crucial for controlling influenza virus disease burden, but their efficacy is limited by human misuse and the capacity of influenza viruses to circumvent antiviral barriers. To relieve the public health hardship of influenza virus, emerging therapies must be selected for their capacity to impede not only influenza virus disease, but also the development of antiviral resistance.
Collapse
Affiliation(s)
- Julianna Han
- Department of Microbiology, The University of Chicago, Chicago, Illinois 60637, USA
| | - Jasmine Perez
- Department of Microbiology, The University of Chicago, Chicago, Illinois 60637, USA
| | - Adam Schafer
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | - Han Cheng
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | - Balaji Manicassamy
- Department of Microbiology, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
25
|
Ashar HK, Mueller NC, Rudd JM, Snider TA, Achanta M, Prasanthi M, Pulavendran S, Thomas PG, Ramachandran A, Malayer JR, Ritchey JW, Rajasekhar R, Chow VTK, Esmon CT, Teluguakula N. The Role of Extracellular Histones in Influenza Virus Pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:135-148. [PMID: 29107075 PMCID: PMC5745522 DOI: 10.1016/j.ajpath.2017.09.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/31/2017] [Accepted: 09/21/2017] [Indexed: 01/14/2023]
Abstract
Although exaggerated host immune responses have been implicated in influenza-induced lung pathogenesis, the etiologic factors that contribute to these events are not completely understood. We previously demonstrated that neutrophil extracellular traps exacerbate pulmonary injury during influenza pneumonia. Histones are the major protein components of neutrophil extracellular traps and are known to have cytotoxic effects. Here, we examined the role of extracellular histones in lung pathogenesis during influenza. Mice infected with influenza virus displayed high accumulation of extracellular histones, with widespread pulmonary microvascular thrombosis. Occluded pulmonary blood vessels with vascular thrombi often exhibited endothelial necrosis surrounded by hemorrhagic effusions and pulmonary edema. Histones released during influenza induced cytotoxicity and showed strong binding to platelets within thrombi in infected mouse lungs. Nasal wash samples from influenza-infected patients also showed increased accumulation of extracellular histones, suggesting a possible clinical relevance of elevated histones in pulmonary injury. Although histones inhibited influenza growth in vitro, in vivo treatment with histones did not yield antiviral effects and instead exacerbated lung pathology. Blocking with antihistone antibodies caused a marked decrease in lung pathology in lethal influenza-challenged mice and improved protection when administered in combination with the antiviral agent oseltamivir. These findings support the pathogenic effects of extracellular histones in that pulmonary injury during influenza was exacerbated. Targeting histones provides a novel therapeutic approach to influenza pneumonia.
Collapse
Affiliation(s)
- Harshini K Ashar
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Nathan C Mueller
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Jennifer M Rudd
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Timothy A Snider
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Mallika Achanta
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Maram Prasanthi
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Sivasami Pulavendran
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Akhilesh Ramachandran
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Jerry R Malayer
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Jerry W Ritchey
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Rachakatla Rajasekhar
- Department of Anatomy, Arkansas College of Osteopathic Medicine, Fort Smith, Arkansas
| | - Vincent T K Chow
- Department of Microbiology and Immunology, School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, and Department of Pathology, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | | |
Collapse
|
26
|
Carrasco-Hernandez R, Jácome R, López Vidal Y, Ponce de León S. Are RNA Viruses Candidate Agents for the Next Global Pandemic? A Review. ILAR J 2017; 58:343-358. [PMID: 28985316 PMCID: PMC7108571 DOI: 10.1093/ilar/ilx026] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/16/2022] Open
Abstract
Pathogenic RNA viruses are potentially the most important group involved in zoonotic disease transmission, and they represent a challenge for global disease control. Their biological diversity and rapid adaptive rates have proved to be difficult to overcome and to anticipate by modern medical technology. Also, the anthropogenic change of natural ecosystems and the continuous population growth are driving increased rates of interspecies contacts and the interchange of pathogens that can develop into global pandemics. The combination of molecular, epidemiological, and ecological knowledge of RNA viruses is therefore essential towards the proper control of these emergent pathogens. This review outlines, throughout different levels of complexity, the problems posed by RNA viral diseases, covering some of the molecular mechanisms allowing them to adapt to new host species-and to novel pharmaceutical developments-up to the known ecological processes involved in zoonotic transmission.
Collapse
Affiliation(s)
- R Carrasco-Hernandez
- R. Carrasco-Hernandez, PhD, is a postdoctoral research fellow at the Microbiome Laboratory in the Postgraduate Division of the Faculty of Medicine at the Universidad Nacional Autónoma de México, CDMX
| | - Rodrigo Jácome
- Rodrigo Jácome, MD, PhD, is a postdoctoral research fellow at the Microbiome Laboratory in the Postgraduate Division of the Faculty of Medicine at the Universidad Nacional Autónoma de México, CDMX
| | - Yolanda López Vidal
- Yolanda López-Vidal, MD, PhD, is an associate professor “C” and is responsible for the Program of Microbial Molecular Immunology in the Department of Microbiology and Parasitology of the Faculty of Medicine at the Universidad Nacional Autónoma de México, CDMX
| | - Samuel Ponce de León
- Samuel Ponce-de-León, MD, MSc, is an associate professor “C”, is responsible for the Microbiome Laboratory and Coordinator of the University Program for Health Research of the Faculty of Medicine at the Universidad Nacional Autónoma de México, CDMX
| |
Collapse
|
27
|
Vikse J, Klos J, Berg A. A travelling camper with a spiking fever, headache, myalgia, hepatitis, and intracellular inclusions. THE LANCET. INFECTIOUS DISEASES 2017; 17:1318. [PMID: 29173891 PMCID: PMC7150174 DOI: 10.1016/s1473-3099(17)30305-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 11/23/2022]
Affiliation(s)
- Jens Vikse
- Department of Medicine, Stavanger University Hospital, Stavanger, Norway.
| | - Jan Klos
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Aase Berg
- Department of Medicine, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
28
|
Beigel JH, Bao Y, Beeler J, Manosuthi W, Slandzicki A, Dar SM, Panuto J, Beasley RL, Perez-Patrigeon S, Suwanpimolkul G, Losso MH, McClure N, Bozzolo DR, Myers C, Holley HP, Hoopes J, Lane HC, Hughes MD, Davey RT. Oseltamivir, amantadine, and ribavirin combination antiviral therapy versus oseltamivir monotherapy for the treatment of influenza: a multicentre, double-blind, randomised phase 2 trial. THE LANCET. INFECTIOUS DISEASES 2017; 17:1255-1265. [PMID: 28958678 PMCID: PMC5777222 DOI: 10.1016/s1473-3099(17)30476-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/20/2017] [Accepted: 07/26/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Influenza continues to have a substantial socioeconomic and health impact despite a long established vaccination programme and approved antivirals. Preclinical data suggest that combining antivirals might be more effective than administering oseltamivir alone in the treatment of influenza. METHODS We did a randomised, double-blind, multicentre phase 2 trial of a combination of oseltamivir, amantadine, and ribavirin versus oseltamivir monotherapy with matching placebo for the treatment of influenza in 50 sites, consisting of academic medical centre clinics, emergency rooms, and private physician offices in the USA, Thailand, Mexico, Argentina, and Australia. Participants who were aged at least 18 years with influenza and were at increased risk of complications were randomly assigned (1:1) by an online computer-generated randomisation system to receive either oseltamivir (75 mg), amantadine (100 mg), and ribavirin (600 mg) combination therapy or oseltamivir monotherapy twice daily for 5 days, given orally, and participants were followed up for 28 days. Blinded treatment kits were used to achieve masking of patients and staff. The primary endpoint was the percentage of participants with virus detectable by PCR in nasopharyngeal swab at day 3, and was assessed in participants who were randomised, had influenza infection confirmed by the central laboratory on a baseline nasopharyngeal sample, and had received at least one dose of study drug. Safety assessment was done in all patients in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, number NCT01227967. FINDINGS Between March 1, 2011, and April 29, 2016, 633 participants were randomly assigned to receive combination antiviral therapy (n=316) or monotherapy (n=317). Seven participants were excluded from analysis: three were not properly randomised, three withdrew from the study, and one was lost to follow-up. The primary analysis included 394 participants, excluding 47 in the pilot phase, 172 without confirmed influenza, and 13 without an endpoint sample. 80 (40·0%) of 200 participants in the combination group had detectable virus at day 3 compared with 97 (50·0%) of 194 (mean difference 10·0, 95% CI 0·2-19·8, p=0·046) in the monotherapy group. The most common adverse events were gastrointestinal-related disorders, primarily nausea (65 [12%] of 556 reported adverse events in the combination group vs 63 [11%] of 585 reported adverse events in the monotherapy group), diarrhoea (56 [10%] of 556 vs 64 [11%] of 585), and vomiting (39 [7%] of 556 vs 23 [4%] of 585). There was no benefit in multiple clinical secondary endpoints, such as median duration of symptoms (4·5 days in the combination group vs 4·0 days in the monotherapy group; p=0·21). One death occurred in the study in an elderly participant in the monotherapy group who died of cardiovascular failure 13 days after randomisation, judged by the site investigator as not related to study intervention. INTERPRETATION Although combination treatment showed a significant decrease in viral shedding at day 3 relative to monotherapy, this difference was not associated with improved clinical benefit. More work is needed to understand why there was no clinical benefit when a difference in virological outcome was identified. FUNDING National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA.
Collapse
Affiliation(s)
| | - Yajing Bao
- Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Joy Beeler
- Leidos Biomedical Research, Frederick, MD, USA
| | | | | | - Sadia M Dar
- Clinical Research Solutions, Smyrna, TN, USA
| | - John Panuto
- Clinical Research Solutions, Middleburg Heights, OH, USA
| | | | | | - Gompol Suwanpimolkul
- King Chulalongkorn Memorial Hospital, Department of Medicine, Chulalongkorn University, Bangkok, Thailand; HIV-NAT, Thai Red Cross AIDS Research Center, Bangkok, Thailand
| | - Marcelo H Losso
- Hospital General De Agudos J M Ramos Mejía, Buenos Aires, Argentina
| | | | | | | | | | | | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | | | - Richard T Davey
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| |
Collapse
|
29
|
Finding the right combination antiviral therapy for influenza. THE LANCET. INFECTIOUS DISEASES 2017; 17:1221-1222. [PMID: 28958679 DOI: 10.1016/s1473-3099(17)30537-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 08/24/2017] [Indexed: 12/11/2022]
|
30
|
Li Y, Lin Z, Guo M, Xia Y, Zhao M, Wang C, Xu T, Chen T, Zhu B. Inhibitory activity of selenium nanoparticles functionalized with oseltamivir on H1N1 influenza virus. Int J Nanomedicine 2017; 12:5733-5743. [PMID: 28848350 PMCID: PMC5557909 DOI: 10.2147/ijn.s140939] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
As an effective antiviral agent, the clinical application of oseltamivir (OTV) is limited by the appearance of drug-resistant viruses. Due to their low toxicity and excellent activity, the antiviral capabilities of selenium nanoparticles (SeNPs) has attracted increasing attention in recent years. To overcome the limitation of drug resistance, the use of modified NPs with biologics to explore novel anti-influenza drugs is developing rapidly. In this study, OTV surface-modified SeNPs with superior antiviral properties and restriction on drug resistance were synthesized. OTV decoration of SeNPs (Se@OTV) obviously inhibited H1N1 infection and had less toxicity. Se@OTV interfered with the H1N1 influenza virus to host cells through inhibiting the activity of hemagglutinin and neuraminidase. The mechanism was that Se@OTV was able to prevent H1N1 from infecting MDCK cells and block chromatin condensation and DNA fragmentation. Furthermore, Se@OTV inhibited the generation of reactive oxygen species and activation of p53 phosphorylation and Akt. These results demonstrate that Se@OTV is a promising efficient antiviral pharmaceutical for H1N1.
Collapse
Affiliation(s)
- Yinghua Li
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| | - Zhengfang Lin
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| | - Min Guo
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| | - Yu Xia
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| | - Mingqi Zhao
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| | - Changbing Wang
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| | - Tiantian Xu
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, People’s Republic of China
| | - Bing Zhu
- Virus Laboratory, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University
| |
Collapse
|
31
|
Smee DF, Dagley A, Tarbet EB. Combinations of L-N G-monomethyl-arginine and oseltamivir against pandemic influenza A virus infections in mice. Antivir Chem Chemother 2017; 25:11-17. [PMID: 28417640 DOI: 10.1177/2040206617691885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
L-NG-monomethyl-arginine (L-NMMA) is an experimental compound that suppresses nitric oxide production in animals. The compound was combined with oseltamivir to treat lethal influenza A/California/04/2009 (H1N1) pandemic virus infections in mice. Treatments were given twice a day for five days starting 4 h (oseltamivir, by oral gavage) or three days (L-NMMA, by intraperitoneal route; corresponding to the time previously reported for nitric oxide induction in the animals) after infection. Low doses of oseltamivir were used in order to demonstrate synergy or antagonism. Oseltamivir monotherapy protected 70% of mice from death at 1 mg/kg/day. L-NMMA (40 and 80 mg/kg/day) was ineffective alone in preventing mortality. Compared to oseltamivir treatment alone, L-NMMA combined with oseltamivir was synergistically effective (as evaluated by three-dimensional MacSynergy analysis), resulting in survival increases from 20 to 70% when 40 or 80 mg/kg/day of L-NMMA was combined with 0.3 mg/kg/day of oseltamivir, and from 70 to 100% survival increases when these doses were combined with 1 mg/kg/day of oseltamivir. These data demonstrate that a nitric oxide inhibitor such as L-NMMA has the potential to be beneficial when combined with oseltamivir in treating influenza virus infections.
Collapse
Affiliation(s)
- Donald F Smee
- Department of Animal, Dairy and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| | - Ashley Dagley
- Department of Animal, Dairy and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| | - E B Tarbet
- Department of Animal, Dairy and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| |
Collapse
|
32
|
Leyva-Grado VH, Palese P. Aerosol administration increases the efficacy of oseltamivir for the treatment of mice infected with influenza viruses. Antiviral Res 2017; 142:12-15. [PMID: 28286235 DOI: 10.1016/j.antiviral.2017.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/28/2017] [Accepted: 03/06/2017] [Indexed: 11/24/2022]
Abstract
Oseltamivir is an influenza neuraminidase inhibitor that along with supportive therapy has shown to help critically ill patients infected with H7N9 and H1N1pdm influenza virus strains to recover from disease. The standard of care recommends the administration of oseltamivir via oral route which represents difficulties in patients with gastrointestinal complications. Here we tested the use of aerosol administration of oseltamivir to treat mice infected with influenza A/H7N9 virus or influenza A/H1N1pdm virus and directly compared this approach to the standard of care, oral administration. Using nose only delivery of aerosolized oseltamivir we observed a significant increase in efficacy of the treatment compared to oral administration characterized by reduced body weight loss, increased survival rate and dose sparing. The preclinical data presented here supports the possibility of using this approach in clinical settings.
Collapse
Affiliation(s)
- Victor H Leyva-Grado
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
33
|
Antiviral Resistance in Influenza Viruses: Clinical and Epidemiological Aspects. ANTIMICROBIAL DRUG RESISTANCE 2017. [PMCID: PMC7122614 DOI: 10.1007/978-3-319-47266-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
There are three classes of antiviral drugs approved for the treatment of influenza: the M2 ion channel inhibitors (amantadine, rimantadine), neuraminidase (NA) inhibitors (laninamivir, oseltamivir, peramivir, zanamivir), and the protease inhibitor (favipiravir); some of the agents are only available in selected countries [1, 2]. These agents are effective at treating the signs and symptoms of influenza in patients infected with susceptible viruses. Clinical failure has been demonstrated in patients infected with viruses with primary resistance, i.e., antivirals can be present in the virus initially infecting the patient, or resistance may emerge during the course of therapy [3–5]. NA inhibitors are active against all nine NA subtypes recognized in nature [6], including highly pathogenic avian influenza A/H5N1 and recent low-pathogenic avian influenza A/H7N9 viruses [7]. Since seasonal influenza is usually an acute, self-limited illness in which viral clearance usually occurs rapidly due to innate and adaptive host immune responses, the emergence of drug-resistant variants would be anticipated to have limited effect on clinical recovery in otherwise healthy patients, as has been demonstrated clinically [3, 8, 9]. Unfortunately, immunocompromised or immunologically naïve hosts, such as young children and infants or those exposed to novel strains, are more likely to have mutations that confer resistance emergence during therapy; such resistant variants may also result in clinically significant adverse outcomes [10–13].
Collapse
|
34
|
|
35
|
Hu XP, Shao MM, Song X, Wu XL, Qi L, Zheng K, Fan L, Liao CH, Li CY, He J, Hu YJ, Wu HQ, Li SH, Zhang J, Zhang FX, He ZD. Anti-influenza virus effects of crude phenylethanoid glycosides isolated from ligustrum purpurascens via inducing endogenous interferon-γ. JOURNAL OF ETHNOPHARMACOLOGY 2016; 179:128-136. [PMID: 26190352 DOI: 10.1016/j.jep.2015.07.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 03/18/2015] [Accepted: 07/16/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ligustrum purpurascens Y.C. Yang (Oleaceae) is traditionally recorded as "Ku Ding Cha", a kind of functional tea in southern China for about two thousand years, which has been reported with sore throat alleviating and pathogenic heat expelling effects. However, there are no scientific studies demonstrating its antiviral activity. THE AIM OF THE STUDY This study is aimed at investigating the anti-influenza virus effects of phenylethanoid glycosides isolated from L. purpurascens (LPG) as well as its corresponding mechanisms. MATERIALS AND METHODS In vitro, hemagglutination assay was employed to detect the influenza virus titer; In vivo, C57BL/6J mice were given oral administration of LPG (100mg/kg, 300mg/kg, 900mg/kg) or ribavirin (100mg/kg) once daily for 5 successive days. Meanwhile, on the second day, mice were infected intranasally (i.n.) with A/FM/1/47 H1N1 virus. Mice survival rate and other clinical index were monitored for 15 days. Infected mice were sacrificed to measure the lung lesion and stained with hematoxylin-eosin. Flow cytometry analyses spleen lymphocytes and interferon-γ (IFN-γ) level. The IFN-γ knockout mice (IFN-γ(-/-) mice, C57BL/6J) which had been verified lacking IFN-γ through Western Blot, were applied in the death-protection test to identify the role of IFN-γ played in LPG antiviral effect. RESULTS In vitro, LPG at 0.5mg/ml inhibited Influenza A Virus H1N1 type (H1N1) infection of MDCK cells. In vivo, LPG at 300 and 900mg/kg significantly decreased the mouse lung index (p<0.05), alleviated influenza-induced lethality and clinical symptoms, and therefore enhanced mouse survival (p<0.05). More detailed experiments demonstrated that antiviral cytokine IFN-γ was involved in the antiviral effect of LPG. Flow cytometric analysis revealed that LPG (900mg/kg) significantly induced secretion of IFN-γ by splenic CD4(+) and CD8(+) cells (p<0.05). Moreover, LPG (900mg/kg) protected wild-type C57BL/6J mice from H1N1 injury, whereas LPG-mediated survival protection disappeared in IFN-γ(-/-) mice. CONCLUSION These results suggest that up-regulating endogenous IFN-γ by LPG may represent a novel therapeutic approach for H1N1 infection.
Collapse
Affiliation(s)
- Xiao-peng Hu
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Min-ming Shao
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xun Song
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Xu-li Wu
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Ling Qi
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Kai Zheng
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Long Fan
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Cheng-hui Liao
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Chen-yang Li
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Jiang He
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Ying-jie Hu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hai-qiang Wu
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Shi-he Li
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China
| | - Jian Zhang
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China.
| | - Feng-xue Zhang
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Zhen-dan He
- Department of Pharmacology, School of Medicine, Shenzhen University, Shenzhen 518060, China; Institute of Biotherapy, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
36
|
Influenza Virus Infection Induces Platelet-Endothelial Adhesion Which Contributes to Lung Injury. J Virol 2015; 90:1812-23. [PMID: 26637453 DOI: 10.1128/jvi.02599-15] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/25/2015] [Indexed: 01/13/2023] Open
Abstract
Lung injury after influenza infection is characterized by increased permeability of the lung microvasculature, culminating in acute respiratory failure. Platelets interact with activated endothelial cells and have been implicated in the pathogenesis of some forms of acute lung injury. Autopsy studies have revealed pulmonary microthrombi after influenza infection, and epidemiological studies suggest that influenza vaccination is protective against pulmonary thromboembolism; however, the effect of influenza infection on platelet-endothelial interactions is unclear. We demonstrate that endothelial infection with both laboratory and clinical strains of influenza virus increased the adhesion of human platelets to primary human lung microvascular endothelial cells. Platelets adhered to infected cells as well as to neighboring cells, suggesting a paracrine effect. Influenza infection caused the upregulation of von Willebrand factor and ICAM-1, but blocking these receptors did not prevent platelet-endothelial adhesion. Instead, platelet adhesion was inhibited by both RGDS peptide and a blocking antibody to platelet integrin α5β1, implicating endothelial fibronectin. Concordantly, lung histology from infected mice revealed viral dose-dependent colocalization of viral nucleoprotein and the endothelial marker PECAM-1, while platelet adhesion and fibronectin deposition also were observed in the lungs of influenza-infected mice. Inhibition of platelets using acetylsalicylic acid significantly improved survival, a finding confirmed using a second antiplatelet agent. Thus, influenza infection induces platelet-lung endothelial adhesion via fibronectin, contributing to mortality from acute lung injury. The inhibition of platelets may constitute a practical adjunctive strategy to the treatment of severe infections with influenza.IMPORTANCE There is growing appreciation of the involvement of the lung endothelium in the pathogenesis of severe infections with influenza virus. We have recently shown that the virus can infect human lung endothelial cells, but the functional consequences of this infection are unknown (S. M. Armstrong, C. Wang, J. Tigdi, X. Si, C. Dumpit, S. Charles, A. Gamage, T. J. Moraes, and W. L. Lee, PLoS One 7:e47323, 2012, http://dx.doi.org/10.1371/journal.pone.0047323). Here, we show that this infection causes platelets to adhere to the lung endothelium. Importantly, blocking platelets using two distinct antiplatelet drugs improved survival in a mouse model of severe influenza infection. Thus, platelet inhibition may constitute a novel therapeutic strategy to improve the host response to severe infections with influenza.
Collapse
|
37
|
Treatment of Immunocompromised, Critically Ill Patients with Influenza A H1N1 Infection with a Combination of Oseltamivir, Amantadine, and Zanamivir. Case Rep Infect Dis 2015; 2015:504975. [PMID: 26346659 PMCID: PMC4546743 DOI: 10.1155/2015/504975] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 07/21/2015] [Accepted: 07/28/2015] [Indexed: 01/16/2023] Open
Abstract
Immunocompromised patients are at increased risk of complications of influenza virus infection. We report on two critically ill patients on immunosuppressive medication with influenza pneumonia. In both patients, oseltamivir monotherapy did not result in clearance of the virus after 18 and five days, respectively. After adding zanamivir and amantadine to the treatment, PCRs on pharyngeal and/or plasma specimens turned negative in both patients after four and three days, respectively. We suggest, that in critically ill patients with influenza A H1N1 infection, treatment efficacy should be monitored closely and treatment with a combination of antiviral drugs should be considered.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW This article reviews the clinical and treatment aspects of avian influenza viruses and the Middle East Respiratory Syndrome coronavirus (MERS-CoV). RECENT FINDINGS Avian influenza A(H5N1) and A(H7N9) viruses have continued to circulate widely in some poultry populations and infect humans sporadically. Sporadic human cases of avian A(H5N6), A(H10N8) and A(H6N1) have also emerged. Closure of live poultry markets in China has reduced the risk of A(H7N9) infection. Observational studies have shown that oseltamivir treatment for adults hospitalized with severe influenza is associated with lower mortality and better clinical outcomes, even as late as 4-5 days after symptom onset. Whether higher than standard doses of neuraminidase inhibitor would provide greater antiviral effects in such patients requires further investigation. High-dose systemic corticosteroids were associated with worse outcomes in patients with A(H1N1)pdm09 or A(H5N1). MERS-CoV has continued to spread since its first discovery in 2012. The mortality rates are high in those with comorbid diseases. There is no specific antiviral treatment or vaccine available. The exact mode of transmission from animals to humans remains unknown. SUMMARY There is an urgent need for developing more effective antiviral therapies to reduce morbidity and mortality of these emerging viral respiratory tract infections.
Collapse
|
39
|
Beggs NF, Dobrovolny HM. Determining drug efficacy parameters for mathematical models of influenza. JOURNAL OF BIOLOGICAL DYNAMICS 2015; 9 Suppl 1:332-346. [PMID: 26056712 DOI: 10.1080/17513758.2015.1052764] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Antivirals are the first line of defence against influenza, so drug efficacy should be re-evaluated for each new strain. However, due to the time and expense involved in assessing the efficacy of drug treatments both in vitro and in vivo, treatment regimens are largely not re-evaluated even when strains are found to be resistant to antivirals. Mathematical models of the infection process can help in this assessment, but for accurate model predictions, we need to measure model parameters characterizing the efficacy of antivirals. We use computer simulations to explore whether in vitro experiments can be used to extract drug efficacy parameters for use in viral kinetics models. We find that the efficacy of neuraminidase inhibitors can be determined by measuring viral load during a single cycle assay, while the efficacy of adamantanes can be determined by measuring infected cells during the preparation stage for the single cycle assay.
Collapse
Affiliation(s)
- Noah F Beggs
- a Department of Biology , Hendrix College , Conway , AR , USA
| | | |
Collapse
|
40
|
Sugiyama MG, Armstrong SM, Wang C, Hwang D, Leong-Poi H, Advani A, Advani S, Zhang H, Szaszi K, Tabuchi A, Kuebler WM, Van Slyke P, Dumont DJ, Lee WL. The Tie2-agonist Vasculotide rescues mice from influenza virus infection. Sci Rep 2015; 5:11030. [PMID: 26046800 PMCID: PMC4457136 DOI: 10.1038/srep11030] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/14/2015] [Indexed: 12/13/2022] Open
Abstract
Seasonal influenza virus infections cause hundreds of thousands of deaths annually while viral mutation raises the threat of a novel pandemic strain. Antiviral drugs exhibit limited efficacy unless administered early and may induce viral resistance. Thus, targeting the host response directly has been proposed as a novel therapeutic strategy with the added potential benefit of not eliciting viral resistance. Severe influenza virus infections are complicated by respiratory failure due to the development of lung microvascular leak and acute lung injury. We hypothesized that enhancing lung endothelial barrier integrity could improve the outcome. Here we demonstrate that the Tie2-agonist tetrameric peptide Vasculotide improves survival in murine models of severe influenza, even if administered as late as 72 hours after infection; the benefit was observed using three strains of the virus and two strains of mice. The effect required Tie2, was independent of viral replication and did not impair lung neutrophil recruitment. Administration of the drug decreased lung edema, arterial hypoxemia and lung endothelial apoptosis; importantly, Vasculotide is inexpensive to produce, is chemically stable and is unrelated to any Tie2 ligands. Thus, Vasculotide may represent a novel and practical therapy for severe infections with influenza.
Collapse
Affiliation(s)
- Michael G. Sugiyama
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
- Department of Laboratory Medicine and Pathobiology, University of Toronto
| | - Susan M. Armstrong
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Changsen Wang
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - David Hwang
- Department of Laboratory Medicine and Pathobiology, University of Toronto
- Department of Pathology, Toronto General Hospital, University Health Network, Toronto, ON
| | - Howard Leong-Poi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Suzanne Advani
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Haibo Zhang
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Katalin Szaszi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Arata Tabuchi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Wolfgang M. Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
| | - Paul Van Slyke
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON
| | - Dan J. Dumont
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON
- Department of Laboratory Medicine and Pathobiology, University of Toronto
- Interdepartmental Division of Critical Care and Department of Medicine, University of Toronto.
| |
Collapse
|
41
|
Lu Y, Hardes K, Dahms SO, Böttcher-Friebertshäuser E, Steinmetzer T, Than ME, Klenk HD, Garten W. Peptidomimetic furin inhibitor MI-701 in combination with oseltamivir and ribavirin efficiently blocks propagation of highly pathogenic avian influenza viruses and delays high level oseltamivir resistance in MDCK cells. Antiviral Res 2015; 120:89-100. [PMID: 26022200 DOI: 10.1016/j.antiviral.2015.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/12/2015] [Accepted: 05/20/2015] [Indexed: 12/30/2022]
Abstract
Antiviral medication is used for the treatment of severe influenza infections, of which the neuraminidase inhibitors (NAIs) are the most effective drugs, approved so far. Here, we investigated the antiviral efficacy of the peptidomimetic furin inhibitor MI-701 in combination with oseltamivir carboxylate and ribavirin against the infection of highly pathogenic avian influenza viruses (HPAIV) that are activated by the host protease furin. Cell cultures infected with the strains A/Thailand/1(KAN-1)/2004 (H5N1) and A/FPV/Rostock/1934 (H7N1) were treated with each agent alone, or in double and triple combinations. MI-701 alone achieved a concentration-dependent reduction of virus propagation. Double treatment of MI-701 with oseltamivir carboxylate and triple combination with ribavirin showed synergistic inhibition and a pronounced delay of virus propagation. MI-701 resistant mutants were not observed. Emergence of NA mutation H275Y conferring high oseltamivir resistance was significantly delayed in the presence of MI-701. Our data indicate that combination with a potent furin inhibitor significantly enhances the therapeutic efficacy of conventional antivirals drugs against HPAIV infection.
Collapse
Affiliation(s)
- Yinghui Lu
- Institute of Virology, Philipps University, Hans-Meerwein-Strasse 2, 35043 Marburg, Germany
| | - Kornelia Hardes
- Institute of Pharmaceutical Chemistry, Philipps University, Marbacher Weg 6-10, 35032 Marburg, Germany
| | - Sven O Dahms
- Protein Crystallography Group, Leibniz Institute for Age Research - Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | | | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Philipps University, Marbacher Weg 6-10, 35032 Marburg, Germany
| | - Manuel E Than
- Protein Crystallography Group, Leibniz Institute for Age Research - Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745 Jena, Germany
| | - Hans-Dieter Klenk
- Institute of Virology, Philipps University, Hans-Meerwein-Strasse 2, 35043 Marburg, Germany
| | - Wolfgang Garten
- Institute of Virology, Philipps University, Hans-Meerwein-Strasse 2, 35043 Marburg, Germany.
| |
Collapse
|
42
|
Kelesidis T, Mastoris I, Metsini A, Tsiodras S. How to approach and treat viral infections in ICU patients. BMC Infect Dis 2014; 14:321. [PMID: 25431007 PMCID: PMC4289200 DOI: 10.1186/1471-2334-14-321] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 06/11/2014] [Indexed: 12/21/2022] Open
Abstract
Patients with severe viral infections are often hospitalized in intensive care units (ICUs) and recent studies underline the frequency of viral detection in ICU patients. Viral infections in the ICU often involve the respiratory or the central nervous system and can cause significant morbidity and mortality especially in immunocompromised patients. The mainstay of therapy of viral infections is supportive care and antiviral therapy when available. Increased understanding of the molecular mechanisms of viral infection has provided great potential for the discovery of new antiviral agents that target viral proteins or host proteins that regulate immunity and are involved in the viral life cycle. These novel treatments need to be further validated in animal and human randomized controlled studies.
Collapse
Affiliation(s)
| | | | | | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens School of Medicine, 1 Rimini Street, GR-12462 Haidari, Athens, Greece.
| |
Collapse
|
43
|
Ilyushina NA, Donnelly RP. In vitro anti-influenza A activity of interferon (IFN)-λ1 combined with IFN-β or oseltamivir carboxylate. Antiviral Res 2014; 111:112-20. [PMID: 25245230 DOI: 10.1016/j.antiviral.2014.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/08/2014] [Accepted: 09/13/2014] [Indexed: 11/18/2022]
Abstract
Influenza viruses, which can cross species barriers and adapt to new hosts, pose a constant potential threat to human health. The influenza pandemic of 2009 highlighted the rapidity with which an influenza virus can spread worldwide. Currently available antivirals have a number of limitations against influenza, and novel antiviral strategies, including novel drugs and drug combinations, are urgently needed. Here, we evaluated the in vitro effects of interferon (IFN)-β, IFN-λ1, oseltamivir carboxylate (a neuraminidase (NA) inhibitor), and combinations of these agents against two seasonal (i.e., H1N1 and H3N2) influenza A viruses. We observed that A/California/04/09 (H1N1) and A/Panama/2007/99 (H3N2) isolates were equally sensitive to the antiviral activity of IFN-β and oseltamivir carboxylate in A549 and Calu-3 cells. In contrast, IFN-λ1 exhibited substantially lower protective potential against the H1N1 strain (64-1030-fold ↓, P<0.05), and was ineffective against H3N2 virus in both cell lines. Three dimensional analysis of drug-drug interactions revealed that IFN-λ1 interacted with IFN-β and oseltamivir carboxylate in an additive or synergistic manner, respectively, to inhibit influenza A virus replication in human airway epithelial cells. Overall, the present study demonstrated that anti-influenza agents with different mechanisms of action (e.g., a NA inhibitor combined with IFN-λ1) exerted a significantly greater (P<0.05) synergistic effect compared to co-treatment with drugs that target the same signaling pathway (i.e., IFN-β plus IFN-λ1) in vitro. Our findings provide support for the combined use of interferon plus oseltamivir as a potential means for treating influenza infections.
Collapse
Affiliation(s)
- Natalia A Ilyushina
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Raymond P Donnelly
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
44
|
A Critical Role for Immune System Response in Mediating Anti-influenza Drug Synergies Assessed by Mechanistic Modeling. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e135. [PMID: 25207611 PMCID: PMC4211263 DOI: 10.1038/psp.2014.32] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 06/10/2014] [Indexed: 11/17/2022]
Abstract
Influenza virus infections represent a serious public health problem worldwide, due to the rapid emergence of drug resistance. One strategy to improve treatment efficacy is to combine drugs that act synergistically. Potentially useful drug combinations are typically identified through empirical testing using in vitro and animal models, but the complexity of the clinical situation warrants the use of more careful analysis and sophisticated approaches. To explore new approaches, we constructed a mechanistic model representing the interaction of antiviral drugs with the viral replication pathway and human immune responses. Simulation of combination therapy using oseltamivir and amantadine predicted significant therapeutic synergy only when immune response was included, in agreement with previous in vitro and in vivo studies using amantadine-resistant strains. Our model can be used to predict the optimal doses for combination therapy, and also raises questions about current drug evaluation methods that do not account for immune system interactions.
Collapse
|
45
|
Abstract
Observational data suggest that the treatment of influenza infection with neuraminidase inhibitors decreases progression to more severe illness, especially when treatment is started soon after symptom onset. However, even early treatment might fail to prevent complications in some patients, particularly those infected with novel viruses such as the 2009 pandemic influenza A H1N1, avian influenza A H5N1 virus subtype, or the avian influenza A H7N9 virus subtype. Furthermore, treatment with one antiviral drug might promote the development of antiviral resistance, especially in immunocompromised hosts and critically ill patients. An obvious strategy to optimise antiviral therapy is to combine drugs with different modes of action. Because host immune responses to infection might also contribute to illness pathogenesis, improved outcomes might be gained from the combination of antiviral therapy with drugs that modulate the immune response in an infected individual. We review available data from preclinical and clinical studies of combination antiviral therapy and of combined antiviral-immunomodulator therapy for influenza. Early-stage data draw attention to several promising antiviral combinations with therapeutic potential in severe infections, but there remains a need to substantiate clinical benefit. Combination therapies with favourable experimental data need to be tested in carefully designed aclinical trials to assess their efficacy.
Collapse
|
46
|
Cao B, Hayden FG. Therapy of H7N9 pneumonia: current perspectives. Expert Rev Anti Infect Ther 2014; 11:1123-6. [PMID: 24151830 DOI: 10.1586/14787210.2013.847787] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Bin Cao
- Department of Infectious Diseases and Clinical Microbiology, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, 100020, Beijing, China
| | | |
Collapse
|
47
|
Abstract
Influenza is an acute respiratory disease in mammals and domestic poultry that emerges from zoonotic reservoirs in aquatic birds and bats. Although influenza viruses are among the most intensively studied pathogens, existing control options require further improvement. Influenza vaccines must be regularly updated because of continuous antigenic drift and sporadic antigenic shifts in the viral surface glycoproteins. Currently, influenza therapeutics are limited to neuraminidase inhibitors; novel drugs and vaccine approaches are therefore urgently needed. Advances in vaccinology and structural analysis have revealed common antigenic epitopes on hemagglutinins across all influenza viruses and suggest that a universal influenza vaccine is possible. In addition, various immunomodulatory agents and signaling pathway inhibitors are undergoing preclinical development. Continuing challenges in influenza include the emergence of pandemic H1N1 influenza in 2009, human infections with avian H7N9 influenza in 2013, and sporadic human cases of highly pathogenic avian H5N1 influenza. Here, we review the challenges facing influenza scientists and veterinary and human public health officials; we also discuss the exciting possibility of achieving the ultimate goal of controlling influenza's ability to change its antigenicity.
Collapse
Affiliation(s)
- Robert G Webster
- Division of Virology, Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | |
Collapse
|
48
|
Boivin G. Detection and management of antiviral resistance for influenza viruses. Influenza Other Respir Viruses 2014; 7 Suppl 3:18-23. [PMID: 24215378 DOI: 10.1111/irv.12176] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2013] [Indexed: 12/25/2022] Open
Abstract
Neuraminidase inhibitors (NAIs) are first-line agents for the treatment and prevention of influenza virus infections. As for other antivirals, the development of resistance to NAIs has become an important concern particularly in the case of A(H1N1) viruses and oseltamivir. The most frequently reported change conferring oseltamivir resistance in that viral context is the H275Y neuraminidase mutation (N1 numbering). Recent studies have shown that, in the presence of the appropriate permissive mutations, the H275Y variant can retain virulence and transmissibility in some viral backgrounds. Most oseltamivir-resistant influenza A virus infections can be managed with the use of inhaled or intravenous zanamivir, another NAI. New NAI compounds and non-neuraminidase agents as well as combination therapies are currently in clinical evaluation for the treatment for severe influenza infections.
Collapse
Affiliation(s)
- Guy Boivin
- CHUQ-CHUL and Laval University, Quebec, QC, Canada
| |
Collapse
|
49
|
Jones JC, Baranovich T, Marathe BM, Danner AF, Seiler JP, Franks J, Govorkova EA, Krauss S, Webster RG. Risk assessment of H2N2 influenza viruses from the avian reservoir. J Virol 2014; 88:1175-88. [PMID: 24227848 PMCID: PMC3911670 DOI: 10.1128/jvi.02526-13] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 10/24/2013] [Indexed: 11/20/2022] Open
Abstract
H2N2 influenza A viruses were the cause of the 1957-1958 pandemic. Historical evidence demonstrates they arose from avian virus ancestors, and while the H2N2 subtype has disappeared from humans, it persists in wild and domestic birds. Reemergence of H2N2 in humans is a significant threat due to the absence of humoral immunity in individuals under the age of 50. Thus, examination of these viruses, particularly those from the avian reservoir, must be addressed through surveillance, characterization, and antiviral testing. The data presented here are a risk assessment of 22 avian H2N2 viruses isolated from wild and domestic birds over 6 decades. Our data show that they have a low rate of genetic and antigenic evolution and remained similar to isolates circulating near the time of the pandemic. Most isolates replicated in mice and human bronchial epithelial cells, but replication in swine tissues was low or absent. Multiple isolates replicated in ferrets, and 3 viruses were transmitted to direct-contact cage mates. Markers of mammalian adaptation in hemagglutinin (HA) and PB2 proteins were absent from all isolates, and they retained a preference for avian-like α2,3-linked sialic acid receptors. Most isolates remained antigenically similar to pandemic A/Singapore/1/57 (H2N2) virus, suggesting they could be controlled by the pandemic vaccine candidate. All viruses were susceptible to neuraminidase inhibitors and adamantanes. Nonetheless, the sustained pathogenicity of avian H2N2 viruses in multiple mammalian models elevates their risk potential for human infections and stresses the need for continual surveillance as a component of prepandemic planning.
Collapse
Affiliation(s)
- Jeremy C Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tarbet EB, Vollmer AH, Hurst BL, Barnard DL, Furuta Y, Smee DF. In vitro activity of favipiravir and neuraminidase inhibitor combinations against oseltamivir-sensitive and oseltamivir-resistant pandemic influenza A (H1N1) virus. Arch Virol 2013; 159:1279-91. [PMID: 24311151 DOI: 10.1007/s00705-013-1922-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/06/2013] [Indexed: 12/21/2022]
Abstract
Few anti-influenza drugs are licensed in the United States for the prevention and therapy of influenza A and B virus infections. This shortage, coupled with continuously emerging drug resistance, as detected through a global surveillance network, seriously limits our anti-influenza armamentarium. Combination therapy appears to offer several advantages over traditional monotherapy in not only delaying development of resistance but also potentially enhancing single antiviral activity. In the present study, we evaluated the antiviral drug susceptibilities of fourteen pandemic influenza A (H1N1) virus isolates in MDCK cells. In addition, we evaluated favipiravir (T-705), an investigational drug with a broad antiviral spectrum and a unique mode of action, alone and in dual combination with the neuraminidase inhibitors (NAIs) oseltamivir, peramivir, or zanamivir, against oseltamivir-sensitive pandemic influenza A/California/07/2009 (H1N1) and oseltamivir-resistant A/Hong Kong/2369/2009 (H1N1) virus. Mean inhibitory values showed that the tested virus isolates remained sensitive to commonly used antiviral drugs, with the exception of the Hong Kong virus isolate. Drug dose-response curves confirmed complete drug resistance to oseltamivir, partial sensitivity to peramivir, and retained susceptibility to zanamivir and favipiravir against the A/Hong Kong/2369/2009 virus. Three-dimensional analysis of drug interactions using the MacSynergy(TM) II program indicated an overall synergistic interaction when favipiravir was combined with the NAIs against the oseltamivir-sensitive influenza virus, and an additive effect against the oseltamivir-resistant virus. Although the clinical relevance of these drug combinations remains to be evaluated, results obtained from this study support the use of combination therapy with favipiravir and NAIs for treatment of human influenza virus infections.
Collapse
Affiliation(s)
- E Bart Tarbet
- Institute for Antiviral Research, Utah State University, Logan, UT, 84322, USA,
| | | | | | | | | | | |
Collapse
|