1
|
Dehnavi F, Akhavan M, Bekhradnia A. Advances in quinoxaline derivatives: synthetic routes and antiviral efficacy against respiratory pathogens. RSC Adv 2024; 14:35400-35423. [PMID: 39512644 PMCID: PMC11542553 DOI: 10.1039/d4ra04292a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
The study of quinoxalines as nitrogen-rich heterocyclic compounds has garnered substantial interest within scientific research owing to their multidimensional functionalization capabilities and significant biological activities. The scope of study encompasses their application as potent antiviral agents, particularly within the domain of respiratory pathologies-a topic of pivotal concern in this comprehensive review. They have several prominent pharmacological effects, such as potential influenza inhibitors, potential anti-SARS coronavirus inhibitors, potential anti-SARS-CO-2 coronavirus inhibitors, and miscellaneous respiratory antiviral activities. As a result, some of the literature has described many of these quinoxalines using various synthetic methods for their mentioned biological effects. In the present review, we provided insight into quinoxaline synthesis, structure-activity relationship (SAR), and antiviral activities, along with a compilation of recent studies. The article further encapsulates the gamut of past and ongoing research efforts in the design and synthetic exploration of antiviral scaffolds, with a pronounced emphasis on their strategic deployment against viral pandemics, contextualized against the tapestry of the recent COVID-19 outbreak. This illuminates the quintessential role of quinoxalines in the armamentarium against viral pathogens and provides a platform for the development of next-generation antiviral agents.
Collapse
Affiliation(s)
- Fateme Dehnavi
- Pharmaceutical Sciences Research Center, Department of Medicinal Chemistry, Mazandaran University of Medical Sciences Sari Iran
| | - Malihe Akhavan
- Pharmaceutical Sciences Research Center, Department of Medicinal Chemistry, Mazandaran University of Medical Sciences Sari Iran
| | - Ahmadreza Bekhradnia
- Pharmaceutical Sciences Research Center, Department of Medicinal Chemistry, Mazandaran University of Medical Sciences Sari Iran
| |
Collapse
|
2
|
Park S, Kim JY, Kwon HC, Jang DS, Song YJ. Antiviral Activities of Ethyl Pheophorbides a and b Isolated from Aster pseudoglehnii against Influenza Viruses. Molecules 2022; 28:41. [PMID: 36615236 PMCID: PMC9822050 DOI: 10.3390/molecules28010041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Screening of the antiviral and virucidal activities of ethanol extracts from plants endemic to the Republic of Korea revealed the inhibitory activity of a 70% ethanol extract of the whole plant of A. pseudoglehnii (APE) against influenza virus infection. Two chlorophyll derivatives, ethyl pheophorbides a and b, isolated as active components of APE, exerted virucidal effects with no evident cytotoxicity. These compounds were effective only under conditions of direct incubation with the virus, and exerted no effects on the influenza A virus (IAV) surface glycoproteins hemagglutinin (HA) and neuraminidase (NA). Interestingly, virucidal activities of ethyl pheophorbides a and b were observed against enveloped but not non-enveloped viruses, suggesting that these compounds act by affecting the integrity of the viral membrane and reducing infectivity.
Collapse
Affiliation(s)
- Subin Park
- Department of Life Science, Gachon University, Seongnam 13120, Republic of Korea
| | - Ji-Young Kim
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hak Cheol Kwon
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung Institute, Gangneung 25451, Republic of Korea
| | - Dae Sik Jang
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoon-Jae Song
- Department of Life Science, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
3
|
Chen W, Shao J, Ying Z, Du Y, Yu Y. Approaches for discovery of small-molecular antivirals targeting to influenza A virus PB2 subunit. Drug Discov Today 2022; 27:1545-1553. [PMID: 35247593 DOI: 10.1016/j.drudis.2022.02.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 11/03/2022]
Abstract
Influenza is an acute respiratory infectious disease caused by influenza virus, leading to huge morbidity and mortality in humans worldwide. Despite the availability of antivirals in the clinic, the emergence of resistant strains calls for antivirals with novel mechanisms of action. The PB2 subunit of the influenza A virus polymerase is a promising target because of its vital role in the 'cap-snatching' mechanism. In this review, we summarize the technologies and medicinal chemistry strategies for hit identification, hit-to-lead and lead-to-candidate optimization, and current challenges in PB2 inhibitor development, as well as offering insights for the fight against drug resistance.
Collapse
Affiliation(s)
- Wenteng Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jiaan Shao
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Zhimin Ying
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yushen Du
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China(1)
| | - Yongping Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
4
|
Caffrey M, Lavie A. pH-Dependent Mechanisms of Influenza Infection Mediated by Hemagglutinin. Front Mol Biosci 2022; 8:777095. [PMID: 34977156 PMCID: PMC8718792 DOI: 10.3389/fmolb.2021.777095] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
Influenza hemagglutinin (HA) is a viral membrane bound protein that plays a critical role in the viral life cycle by mediating entry into target cells. HA exploits the lowering of the pH in the endosomal compartment to initiate a series of conformational changes that promote access of the viral genetic material to the cytoplasm, and hence viral replication. In this review we will first discuss what is known about the structural properties of HA as a function of pH. Next, we will discuss the dynamics and intermediate states of HA. We will then discuss the specific residues that are thought to be titrated by the change in pH and possible mechanisms for the pH triggered conformational changes. Finally, we will discuss small molecules that disrupt the pH trigger and thus serve as potential therapeutic strategies to prevent influenza infection.
Collapse
Affiliation(s)
- Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
5
|
Antanasijevic A, Durst MA, Cheng H, Gaisina IN, Perez JT, Manicassamy B, Rong L, Lavie A, Caffrey M. Structure of avian influenza hemagglutinin in complex with a small molecule entry inhibitor. Life Sci Alliance 2020; 3:3/8/e202000724. [PMID: 32611549 PMCID: PMC7335401 DOI: 10.26508/lsa.202000724] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
The binding properties of fusion inhibitor CBS1117 to hemagglutinin by x-ray crystallography, NMR, and mutagenesis give insights into mechanism and guidance for chemical optimization. HA plays a critical role in influenza infection and, thus HA is a potential target for antivirals. Recently, our laboratories have described a novel fusion inhibitor, termed CBS1117, with EC50 ∼3 μM against group 1 HA. In this work, we characterize the binding properties of CBS1117 to avian H5 HA by x-ray crystallography, NMR, and mutagenesis. The x-ray structure of the complex shows that the compound binds near the HA fusion peptide, a region that plays a critical role in HA-mediated fusion. NMR studies demonstrate binding of CBS1117 to H5 HA in solution and show extensive hydrophobic contacts between the compound and HA surface. Mutagenesis studies further support the location of the compound binding site proximal to the HA fusion peptide and identify additional amino acids that are important to compound binding. Together, this work gives new insights into the CBS1117 mechanism of action and can be exploited to further optimize this compound and better understand the group specific activity of small-molecule inhibitors of HA-mediated entry.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthew A Durst
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Han Cheng
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Jasmine T Perez
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
6
|
Palchykov VA, Gaponov AA. 1,3-Amino alcohols and their phenol analogs in heterocyclization reactions. ADVANCES IN HETEROCYCLIC CHEMISTRY 2020. [DOI: 10.1016/bs.aihch.2019.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
7
|
Antanasijevic A, Durst MA, Lavie A, Caffrey M. Identification of a pH sensor in Influenza hemagglutinin using X-ray crystallography. J Struct Biol 2019; 209:107412. [PMID: 31689502 PMCID: PMC7111647 DOI: 10.1016/j.jsb.2019.107412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 11/15/2022]
Abstract
X-ray crystallography identifies a conserved histidine, HA1-H38, that changes side chain conformation at low pH. We attribute the observed conformational change to cation-cation repulsion between protonated HA1-H18 and HA1-H38. We suggest that the HA1-H18 and HA1-H38 pair plays a role in the pathway toward the postfusion conformation of HA.
Hemagglutnin (HA) mediates entry of influenza virus through a series of conformational changes triggered by the low pH of the endosome. The residue or combination of residues acting as pH sensors has not yet been fully elucidated. In this work, we assay pH effects on the structure of H5 HA by soaking HA crystallized at pH 6.5 in a series of buffers with lower pH, mimicking the conditions of the endosome. We find that HA1-H38, which is conserved in Group 1 HA, undergoes a striking change in side chain conformation, which we attribute to its protonation and cation-cation repulsion with conserved HA1-H18. This work suggests that x-ray crystallography can be applied for studying small-scale pH-induced conformational changes providing valuable information on the location of pH sensors in HA. Importantly, the observed change in HA1-H38 conformation is further evidence that the pH-induced conformational changes of HA are the result of a series of protonation events to conserved and non-conserved pH sensors.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, 60607 Chicago, USA
| | - Matthew A Durst
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, 60607 Chicago, USA
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, 60607 Chicago, USA.
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Ave, 60607 Chicago, USA.
| |
Collapse
|
8
|
Hao C, Yu G, He Y, Xu C, Zhang L, Wang W. Marine glycan–based antiviral agents in clinical or preclinical trials. Rev Med Virol 2019; 29:e2043. [PMID: 30942528 DOI: 10.1002/rmv.2043] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Cui Hao
- Systems Biology and Medicine Center for Complex DiseasesAffiliated Hospital of Qingdao University Qingdao PR China
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of EducationOcean University of China Qingdao PR China
| | - Yanli He
- Systems Biology and Medicine Center for Complex DiseasesAffiliated Hospital of Qingdao University Qingdao PR China
| | - Cuijing Xu
- Key Laboratory of Marine Drugs, Ministry of EducationOcean University of China Qingdao PR China
| | - Lijuan Zhang
- Systems Biology and Medicine Center for Complex DiseasesAffiliated Hospital of Qingdao University Qingdao PR China
| | - Wei Wang
- Key Laboratory of Marine Drugs, Ministry of EducationOcean University of China Qingdao PR China
| |
Collapse
|
9
|
Synthesis of cage [4.4.2]propellanes and $${D_{3}}$$ D 3 -trishomocubanes bearing spiro linkage. J CHEM SCI 2018. [DOI: 10.1007/s12039-018-1569-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
10
|
Wang M, Wang S, Wang W, Wang Y, Wang H, Zhu W. Inhibition effects of novel polyketide compound PPQ-B against influenza A virus replication by interfering with the cellular EGFR pathway. Antiviral Res 2017; 143:74-84. [PMID: 28414053 DOI: 10.1016/j.antiviral.2017.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/12/2017] [Indexed: 12/13/2022]
Abstract
Development of anti-influenza A virus (IAV) drugs with novel targets and low toxicity is critical for preparedness against influenza outbreaks. In the current study, our results indicated that the novel polyketide compound purpurquinone B (PPQ-B) derived from acid-tolerant fungus Penicillium purpurogenum strain JS03-21 suppressed the replication of IAV in vitro with low toxicity, and may block some stages after virus adsorption. PPQ-B could inhibit H1N1 (A/Puerto Rico/8/34; PR8), H1N1 (A/California/04/2009; Cal09) and H3N2 (A/swine/Minnesota/02719/2009) virus replication in vitro, suggesting that PPQ-B possesses broad-spectrum anti-IAV activities. PPQ-B's antiviral activity may be largely related to its inhibition of some steps that occur 0-4 h after adsorption. Oral administration of PPQ-B could decrease pulmonary viral titers and improve survival rate in IAV infected mice. PPQ-B also significantly decreased the production of inflammatory factors TNF-α, IL-6, RANTES and KC in IAV infected lungs and A549 cells, suggesting that PPQ-B may also attenuate the inflammatory responses caused by IAV infection. PPQ-B may down-regulate the NF-κB and MAPK pathways to inhibit both virus replication and inflammatory responses. In summary, PPQ-B has the potential to be developed into a novel anti-IAV drug targeting host EGFR pathway in the future.
Collapse
Affiliation(s)
- Miaomiao Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Shuyao Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China; Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, PR China.
| | - Yi Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Hui Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Weiming Zhu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China.
| |
Collapse
|
11
|
Oligothiophene compounds inhibit the membrane fusion between H5N1 avian influenza virus and the endosome of host cell. Eur J Med Chem 2017; 130:185-194. [DOI: 10.1016/j.ejmech.2017.02.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 11/19/2022]
|
12
|
Wang W, Wu J, Zhang X, Hao C, Zhao X, Jiao G, Shan X, Tai W, Yu G. Inhibition of Influenza A Virus Infection by Fucoidan Targeting Viral Neuraminidase and Cellular EGFR Pathway. Sci Rep 2017; 7:40760. [PMID: 28094330 PMCID: PMC5240104 DOI: 10.1038/srep40760] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/09/2016] [Indexed: 11/21/2022] Open
Abstract
Development of novel anti-influenza A virus (IAV) drugs with high efficiency and low toxicity is critical for preparedness against influenza outbreaks. Herein, we investigated the anti-IAV activities and mechanisms of fucoidan in vitro and in vivo. The results showed that a fucoidan KW derived from brown algae Kjellmaniella crassifolia effectively blocked IAV infection in vitro with low toxicity. KW possessed broad anti-IAV spectrum and low tendency of induction of viral resistance, superior to the anti-IAV drug amantadine. KW was capable of inactivating virus particles before infection and blocked some stages after adsorption. KW could bind to viral neuraminidase (NA) and inhibit the activity of NA to block the release of IAV. KW also interfered with the activation of EGFR, PKCα, NF-κB, and Akt, and inhibited both IAV endocytosis and EGFR internalization in IAV-infected cells, suggesting that KW may also inhibit cellular EGFR pathway. Moreover, intranasal administration of KW markedly improved survival and decreased viral titers in IAV-infected mice. Therefore, fucoidan KW has the potential to be developed into a novel nasal drop or spray for prevention and treatment of influenza in the future.
Collapse
Affiliation(s)
- Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Jiandong Wu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
| | - Xiaoshuang Zhang
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Cui Hao
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University Medical College, Qingdao, 266003, P. R. China
| | - Xiaoliang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
| | - Guangling Jiao
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Xindi Shan
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
| | - Wenjing Tai
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| |
Collapse
|
13
|
Wang C, Zhang Y, Han L, Guo L, Zhong H, Wang J. Hemin ameliorates influenza pneumonia by attenuating lung injury and regulating the immune response. Int J Antimicrob Agents 2016; 49:45-52. [PMID: 27884416 DOI: 10.1016/j.ijantimicag.2016.09.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/08/2016] [Accepted: 09/17/2016] [Indexed: 10/20/2022]
Abstract
The anti-influenza activity of hemin, an inducer, activator and the substrate of heme oxygenase-1 (HO-1), was examined both in vitro and in vivo. The human lung carcinoma cell line A549 was used to evaluate the in vitro effect of hemin on influenza A virus (IAV) replication. A mouse model was used to examine the in vivo activity of hemin. Observation indexes included survival rate and body weight of mice, virus load and pathological examination of the lungs, and characterization of the systemic and local immune responses. The results showed that hemin could induce HO-1 expression in A549 cells and inhibit IAV replication in vitro. The in vivo results showed that injection of hemin could protect mice from death and body weight loss caused by IAV infection. Hemin was administered both at initial and progressive stages of influenza pneumonia (1 day and 4 days after virus infection, respectively) and showed significant anti-influenza activity under both conditions. However, the results showed that although hemin could induce HO-1 expression in vivo, it could not inhibit IAV replication in vivo. Pathological examination showed that hemin significantly attenuated lung tissue injury caused by IAV. Further study showed that hemin could regulate the immune response to IAV infection by reducing lymphocytopenia and local inflammatory cytokine increases caused by IAV infection. This study shows that hemin has the potential for the treatment of IAV infection and its effect may be due to attenuation of lung injury and regulation of the immune response.
Collapse
Affiliation(s)
- Conghui Wang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yanjing Zhang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - LianLian Han
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Li Guo
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hui Zhong
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Jianwei Wang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou 310003, China.
| |
Collapse
|
14
|
Antiviral activity of KR-23502 targeting nuclear export of influenza B virus ribonucleoproteins. Antiviral Res 2016; 134:77-88. [DOI: 10.1016/j.antiviral.2016.07.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/24/2016] [Accepted: 07/26/2016] [Indexed: 11/22/2022]
|
15
|
Antanasijevic A, Hafeman NJ, Tundup S, Kingsley C, Mishra RK, Rong L, Manicassamy B, Wardrop D, Caffrey M. Stabilization and Improvement of a Promising Influenza Antiviral: Making a PAIN PAINless. ACS Infect Dis 2016; 2:608-615. [PMID: 27759373 DOI: 10.1021/acsinfecdis.6b00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The viral envelope protein hemagglutinin (HA) plays a critical role in influenza entry and thus is an attractive target for novel therapeutics. The small molecule tert-butylhydroquinone (TBHQ) has previously been shown to bind to HA and inhibit HA-mediated entry with low micromolar potency. However, enthusiasm for the use of TBHQ has diminished due to the compound's antioxidant properties. In this work we show that the antioxidant properties of TBHQ are not responsible for the inhibition of HA-mediated entry. In addition, we have performed a structure-activity relationship (SAR) analysis of TBHQ derivatives. We find that the most promising compound, 3-tert-butyl-4-methoxyphenol, exhibits enhanced potency (IC50 = 0.6 μM), decreased toxicity (CC50 = 340 μM), and increased stability (t1/2 > 48 h). Finally, we have characterized the binding properties of 3-tert-butyl-4-methoxyphenol using NMR and molecular dynamics to guide future efforts for chemical optimization.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| | - Nicholas J Hafeman
- Department of Chemistry, University of Illinois at Chicago , 845 West Taylor Street, Chicago, Illinois 60607, United States
| | - Smanla Tundup
- Department of Microbiology and Immunology, University of Chicago , 920 East 58th Street, Chicago, Illinois 60637, United States
| | - Carolyn Kingsley
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| | - Rama K Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University , 2135 Sheridan Road, Evanston, Illinois 60208, United States
| | - Lijun Rong
- Department of Microbiology & Immunology, University of Illinois at Chicago , 835 South Wolcott, Chicago, Illinois 60612, United States
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Chicago , 920 East 58th Street, Chicago, Illinois 60637, United States
| | - Duncan Wardrop
- Department of Chemistry, University of Illinois at Chicago , 845 West Taylor Street, Chicago, Illinois 60607, United States
| | - Michael Caffrey
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| |
Collapse
|
16
|
Wang J, Li F, Ma C. Recent progress in designing inhibitors that target the drug-resistant M2 proton channels from the influenza A viruses. Biopolymers 2016; 104:291-309. [PMID: 25663018 DOI: 10.1002/bip.22623] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/24/2015] [Indexed: 12/15/2022]
Abstract
Influenza viruses are the causative agents for seasonal influenza, which results in thousands of deaths and millions of hospitalizations each year. Moreover, sporadic transmission of avian or swan influenza viruses to humans often leads to an influenza pandemic, as there is no preimmunity in the human body to fight against such novel strains. The metastable genome of the influenza viruses, coupled with the reassortment of different strains from a wide range of host origins, leads to the continuous evolution of the influenza virus diversity. Such characteristics of influenza viruses present a grand challenge in devising therapeutic strategies to combat influenza virus infection. This review summarizes recent progress in designing small molecule inhibitors that target the drug-resistant influenza A virus M2 proton channels and highlights the contribution of mechanistic studies of proton conductance to drug discovery. The lessons learned throughout the course of M2 drug discovery might provide insights for designing inhibitors that target other therapeutically important ion channels.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721.,BIO5 Institute, University of Arizona, Tucson, AZ, 85721
| | - Fang Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721
| | - Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721
| |
Collapse
|
17
|
Antanasijevic A, Kingsley C, Basu A, Bowlin TL, Rong L, Caffrey M. Application of virus-like particles (VLP) to NMR characterization of viral membrane protein interactions. JOURNAL OF BIOMOLECULAR NMR 2016; 64:255-65. [PMID: 26921030 PMCID: PMC4826305 DOI: 10.1007/s10858-016-0025-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/22/2016] [Indexed: 05/10/2023]
Abstract
The membrane proteins of viruses play critical roles in the virus life cycle and are attractive targets for therapeutic intervention. Virus-like particles (VLP) present the possibility to study the biochemical and biophysical properties of viral membrane proteins in their native environment. Specifically, the VLP constructs contain the entire protein sequence and are comprised of native membrane components including lipids, cholesterol, carbohydrates and cellular proteins. In this study we prepare VLP containing full-length hemagglutinin (HA) or neuraminidase (NA) from influenza and characterize their interactions with small molecule inhibitors. Using HA-VLP, we first show that VLP samples prepared using the standard sucrose gradient purification scheme contain significant amounts of serum proteins, which exhibit high potential for non-specific interactions, thereby complicating NMR studies of ligand-target interactions. We then show that the serum contaminants may be largely removed with the addition of a gel filtration chromatography step. Next, using HA-VLP we demonstrate that WaterLOGSY NMR is significantly more sensitive than Saturation Transfer Difference (STD) NMR for the study of ligand interactions with membrane bound targets. In addition, we compare the ligand orientation to HA embedded in VLP with that of recombinant HA by STD NMR. In a subsequent step, using NA-VLP we characterize the kinetic and binding properties of substrate analogs and inhibitors of NA, including study of the H274Y-NA mutant, which leads to wide spread resistance to current influenza antivirals. In summary, our work suggests that VLP have high potential to become standard tools in biochemical and biophysical studies of viral membrane proteins, particularly when VLP are highly purified and combined with control VLP containing native membrane proteins.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland, Chicago, IL, 60607, USA
| | - Carolyn Kingsley
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland, Chicago, IL, 60607, USA
| | - Arnab Basu
- Microbiotix Inc., Worcester, MA, 01605, USA
| | | | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland, Chicago, IL, 60607, USA.
| |
Collapse
|
18
|
Wang H, Xu R, Shi Y, Si L, Jiao P, Fan Z, Han X, Wu X, Zhou X, Yu F, Zhang Y, Zhang L, Zhang L, Zhou D, Xiao S. Design, synthesis and biological evaluation of novel l-ascorbic acid-conjugated pentacyclic triterpene derivatives as potential influenza virus entry inhibitors. Eur J Med Chem 2016; 110:376-88. [DOI: 10.1016/j.ejmech.2016.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/02/2016] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
|
19
|
Abstract
Since the discovery that certain small viral membrane proteins, collectively termed as viroporins, can permeabilize host cellular membranes and also behave as ion channels, attempts have been made to link this feature to specific biological roles. In parallel, most viroporins identified so far are virulence factors, and interest has focused toward the discovery of channel inhibitors that would have a therapeutic effect, or be used as research tools to understand the biological roles of viroporin ion channel activity. However, this paradigm is being shifted by the difficulties inherent to small viral membrane proteins, and by the realization that protein-protein interactions and other diverse roles in the virus life cycle may represent an equal, if not, more important target. Therefore, although targeting the channel activity of viroporins can probably be therapeutically useful in some cases, the focus may shift to their other functions in following years. Small-molecule inhibitors have been mostly developed against the influenza A M2 (IAV M2 or AM2). This is not surprising since AM2 is the best characterized viroporin to date, with a well-established biological role in viral pathogenesis combined the most extensive structural investigations conducted, and has emerged as a validated drug target. For other viroporins, these studies are still mostly in their infancy, and together with those for AM2, are the subject of the present review.
Collapse
|
20
|
|
21
|
Wang G, Chen L, Xian T, Liang Y, Zhang X, Yang Z, Luo M. Discovery and SAR study of piperidine-based derivatives as novel influenza virus inhibitors. Org Biomol Chem 2015; 12:8048-60. [PMID: 25184371 DOI: 10.1039/c4ob01079e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of piperidine-based derivatives were identified as novel and potent inhibitors of the influenza virus through structural modification of a compound that was selected from a high-throughput screen. Various analogues were synthesized and confirmed as inhibitors. The structure–activity relationship (SAR) studies suggested that the ether linkage between the quinoline and piperidine is critical for the inhibitory activity. The optimized compound tert-butyl 4-(quinolin-4-yloxy)piperidine-1-carboxylate 11e had an excellent inhibitory activity against influenza virus infection from a variety of influenza virus strains, with EC50 values as low as 0.05 μM. The selectivity index value (SI = MLD50/EC50) of 11e is over 160000 based on cytotoxicity, measured by MTT assays of three cell lines. We carried out a time-of-addition experiment to delineate the mechanism of inhibition. The result indicates that 11e interferes with the early to middle stage of influenza virus replication.
Collapse
Affiliation(s)
- Guoxin Wang
- Key Laboratory of Structural Biology and Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | | | | | | | | | | | | |
Collapse
|
22
|
Wu Y, Canturk B, Jo H, Ma C, Gianti E, Klein M, Pinto LH, Lamb RA, Fiorin G, Wang J, DeGrado WF. Flipping in the pore: discovery of dual inhibitors that bind in different orientations to the wild-type versus the amantadine-resistant S31N mutant of the influenza A virus M2 proton channel. J Am Chem Soc 2014; 136:17987-95. [PMID: 25470189 PMCID: PMC4286326 DOI: 10.1021/ja508461m] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Indexed: 12/13/2022]
Abstract
Influenza virus infections lead to numerous deaths and millions of hospitalizations each year. One challenge facing anti-influenza drug development is the heterogeneity of the circulating influenza viruses, which comprise several strains with variable susceptibility to antiviral drugs. For example, the wild-type (WT) influenza A viruses, such as the seasonal H1N1, tend to be sensitive to antiviral drugs, amantadine and rimantadine, while the S31N mutant viruses, such as the pandemic 2009 H1N1 (H1N1pdm09) and seasonal H3N2, are resistant to this class of drugs. Thus, drugs targeting both WT and the S31N mutant are highly desired. We report our design of a novel class of dual inhibitors along with their ion channel blockage and antiviral activities. The potency of the most active compound 11 in inhibiting WT and the S31N mutant influenza viruses is comparable with that of amantadine in inhibiting WT influenza virus. Solution NMR studies and molecular dynamics (MD) simulations of drug-M2 interactions supported our design hypothesis: namely, the dual inhibitor binds in the WT M2 channel with an aromatic group facing down toward the C-terminus, while the same drug binds in the S31N M2 channel with its aromatic group facing up toward the N-terminus. The flip-flop mode of drug binding correlates with the structure-activity relationship (SAR) and has paved the way for the next round of rational design of broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- Yibing Wu
- Department
of Pharmaceutical Chemistry, University
of California, Mission Bay Box 3122, San Francisco, California 94158, United States
| | - Belgin Canturk
- Department
of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Hyunil Jo
- Department
of Pharmaceutical Chemistry, University
of California, Mission Bay Box 3122, San Francisco, California 94158, United States
| | - Chunlong Ma
- Department
of Pharmacology and Toxicology and the BIO5 Institute, The University of Arizona, 1501 N. Campbell Avenue, Tucson, Arizona 85721, United States
| | - Eleonora Gianti
- Institute
for Computational and Molecular Science, Science Education and Research
Center (035-07), Temple University, 1925 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | - Michael
L. Klein
- Institute
for Computational and Molecular Science, Science Education and Research
Center (035-07), Temple University, 1925 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | - Lawrence H. Pinto
- Department
of Neurobiology, Northwestern University, 2205 Tech Drive, Evanston, Illinois 60208, United States
| | - Robert A. Lamb
- Department
of Molecular Biosciences, Northwestern University, 2205 Tech Drive, Evanston, Illinois 60208, United States
- Howard
Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Giacomo Fiorin
- Institute
for Computational and Molecular Science, Science Education and Research
Center (035-07), Temple University, 1925 North 12th Street, Philadelphia, Pennsylvania 19122, United States
| | - Jun Wang
- Department
of Pharmacology and Toxicology and the BIO5 Institute, The University of Arizona, 1501 N. Campbell Avenue, Tucson, Arizona 85721, United States
| | - William F. DeGrado
- Department
of Pharmaceutical Chemistry, University
of California, Mission Bay Box 3122, San Francisco, California 94158, United States
| |
Collapse
|
23
|
Mechanisms of action and efficacy of statins against influenza. BIOMED RESEARCH INTERNATIONAL 2014; 2014:872370. [PMID: 25478576 PMCID: PMC4244940 DOI: 10.1155/2014/872370] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 12/31/2022]
Abstract
The influenza virus (IV) is known to be a resistant virus with frequent mutations, causing severe respiratory diseases in the upper respiratory system. Public health concerns about clinical efficacy of all conventional drugs are ambiguous; therefore, finding additional therapeutic agents is critical to prevent and control influenza outbreaks. Influenza is associated with the induction of proinflammatory cytokines. Scientists have reported that anti-inflammatory drugs, with pleiotropic effects, reduce the burden of severe influenza diseases. Therefore, statins, which are cardioprotective drugs with anti-inflammatory and immunomodulatory effects, may help patients suffering from influenza virus (IV). This review delineates the potential use of statins as an alternative therapy in treating influenza related illness.
Collapse
|
24
|
Smith AAA, Kryger MBL, Wohl BM, Ruiz-Sanchis P, Zuwala K, Tolstrup M, Zelikin AN. Macromolecular (pro)drugs in antiviral research. Polym Chem 2014. [DOI: 10.1039/c4py00624k] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
25
|
Antanasijevic A, Basu A, Bowlin TL, Mishra RK, Rong L, Caffrey M. Mutagenesis studies of the H5 influenza hemagglutinin stem loop region. J Biol Chem 2014; 289:22237-45. [PMID: 24947513 DOI: 10.1074/jbc.m114.572974] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Influenza outbreaks, particularly the pandemic 1918 H1 and avian H5 strains, are of high concern to public health. The hemagglutinin envelope protein of influenza plays a critical role in viral entry and thus is an attractive target for inhibition of virus entry. The highly conserved stem loop region of hemagglutinin has been shown to undergo critically important conformational changes during the entry process and, moreover, to be a site for inhibition of virus entry by antibodies, small proteins, and small drug-like molecules. In this work we probe the structure-function properties of the H5 hemagglutinin stem loop region by site-directed mutagenesis. We find that most mutations do not disrupt expression, proteolytic processing, incorporation into virus, or receptor binding; however, many of the mutations disrupt the entry process. We further assess the effects of mutations on inhibition of entry by a neutralizing monoclonal antibody (C179) and find examples of increased and decreased sensitivity to the antibody, consistent with the antibody binding site observed by x-ray crystallography. In addition, we tested the sensitivity of the mutants to MBX2329, a small molecule inhibitor of influenza entry. Interestingly, the mutants exhibit increased and decreased sensitivities to MBX2329, which gives further insight into the binding site of the compound on HA and potential mechanisms of escape. Finally, we have modeled the binding site of MBX2329 using molecular dynamics and find that the resulting structure is in good agreement with the mutagenesis results. Together these studies underscore the importance of the stem loop region to HA function and suggest potential sites for therapeutic intervention of influenza entry.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- From the Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607
| | - Arnab Basu
- Microbiotix Inc., Worcester, Massachusetts 01605
| | | | - Rama K Mishra
- the Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, and
| | - Lijun Rong
- the Department of Microbiology and Immunology, University of Illinois, Chicago, Illinois 60612
| | - Michael Caffrey
- From the Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607,
| |
Collapse
|
26
|
|
27
|
Pautus S, Sehr P, Lewis J, Fortuné A, Wolkerstorfer A, Szolar O, Guilligay D, Lunardi T, Décout JL, Cusack S. New 7-methylguanine derivatives targeting the influenza polymerase PB2 cap-binding domain. J Med Chem 2013; 56:8915-30. [PMID: 24134208 DOI: 10.1021/jm401369y] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The heterotrimeric influenza virus polymerase performs replication and transcription of viral RNA in the nucleus of infected cells. Transcription by "cap-snatching" requires that host-cell pre-mRNAs are bound via their 5' cap to the PB2 subunit. Thus, the PB2 cap-binding site is potentially a good target for new antiviral drugs that will directly inhibit viral replication. Docking studies using the structure of the PB2 cap-binding domain suggested that 7-alkylguanine derivatives substituted at position N-9 and N-2 could be good candidates. Four series of 7,9-di- and 2,7,9-trialkyl guanine derivatives were synthesized and evaluated by an AlphaScreen assay in competition with a biotinylated cap analogue. Three synthesized compounds display potent in vitro activity with IC50 values lower than 10 μM. High-resolution X-ray structures of three inhibitors in complex with the H5N1 PB2 cap-binding domain confirmed the binding mode and provide detailed information for further compound optimization.
Collapse
Affiliation(s)
- Stéphane Pautus
- Département de Pharmacochimie Moléculaire, Université de Grenoble Alpes/CNRS, UMR 5063 , ICMG FR 2607, 470 rue de la Chimie, BP 53, F-38041 Grenoble, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Antanasijevic A, Cheng H, Wardrop DJ, Rong L, Caffrey M. Inhibition of influenza H7 hemagglutinin-mediated entry. PLoS One 2013; 8:e76363. [PMID: 24194835 PMCID: PMC3806803 DOI: 10.1371/journal.pone.0076363] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/26/2013] [Indexed: 12/20/2022] Open
Abstract
The recent outbreak of H7N9 influenza in China is of high concern to public health. H7 hemagglutinin (HA) plays a critical role in influenza entry and thus HA presents an attractive target for antivirals. Previous studies have suggested that the small molecule tert-butyl hydroquinone (TBHQ) inhibits the entry of influenza H3 HA by binding to the stem loop of HA and stabilizing the neutral pH conformation of HA, thereby disrupting the membrane fusion step. Based on amino acid sequence, structure and immunogenicity, H7 is a related Group 2 HA. In this work we show, using a pseudovirus entry assay, that TBHQ inhibits H7 HA-mediated entry, as well as H3 HA-mediated entry, with an IC50 ~ 6 µM. Using NMR, we show that TBHQ binds to the H7 stem loop region. STD NMR experiments indicate that the aromatic ring of TBHQ makes extensive contact with the H7 HA surface. Limited proteolysis experiments indicate that TBHQ inhibits influenza entry by stabilizing the H7 HA neutral pH conformation. Together, this work suggests that the stem loop region of H7 HA is an attractive target for therapeutic intervention and that TBHQ, which is a widely used food preservative, is a promising lead compound.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Han Cheng
- Department of Microbiology & Immunology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Duncan J. Wardrop
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Lijun Rong
- Department of Microbiology & Immunology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Michael Caffrey
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
29
|
Lugiņina J, Rjabovs V, Belyakov S, Turks M. A concise synthesis of sugar isoxazole conjugates. Tetrahedron Lett 2013. [DOI: 10.1016/j.tetlet.2013.07.103] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Wang J, Ma C, Wang J, Jo H, Canturk B, Fiorin G, Pinto LH, Lamb RA, Klein ML, DeGrado WF. Discovery of novel dual inhibitors of the wild-type and the most prevalent drug-resistant mutant, S31N, of the M2 proton channel from influenza A virus. J Med Chem 2013; 56:2804-12. [PMID: 23437766 DOI: 10.1021/jm301538e] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Anti-influenza drugs, amantadine and rimantadine, targeting the M2 channel from influenza A virus are no longer effective because of widespread drug resistance. S31N is the predominant and amantadine-resistant M2 mutant, present in almost all of the circulating influenza A strains as well as in the pandemic 2009 H1N1 and the highly pathogenic H5N1 flu strains. Thus, there is an urgent need to develop second-generation M2 inhibitors targeting the S31N mutant. However, the S31N mutant presents a huge challenge to drug discovery, and it has been considered undruggable for several decades. Using structural information, classical medicinal chemistry approaches, and M2-specific biological testing, we discovered benzyl-substituted amantadine derivatives with activity against both S31N and WT, among which 4-(adamantan-1-ylaminomethyl)-benzene-1,3-diol (44) is the most potent dual inhibitor. These inhibitors demonstrate that S31N is a druggable target and provide a new starting point to design novel M2 inhibitors that address the problem of drug-resistant influenza A infections.
Collapse
Affiliation(s)
- Jizhou Wang
- Influmedix Inc, 170 North Radnor-Chester Road, Suite 300, Radnor, Pennsylvania 19087, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Structure and inhibition of the drug-resistant S31N mutant of the M2 ion channel of influenza A virus. Proc Natl Acad Sci U S A 2013; 110:1315-20. [PMID: 23302696 DOI: 10.1073/pnas.1216526110] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The influenza A virus M2 proton channel (A/M2) is the target of the antiviral drugs amantadine and rimantadine, whose use has been discontinued due to widespread drug resistance. Among the handful of drug-resistant mutants, S31N is found in more than 95% of the currently circulating viruses and shows greatly decreased inhibition by amantadine. The discovery of inhibitors of S31N has been hampered by the limited size, polarity, and dynamic nature of its amantadine-binding site. Nevertheless, we have discovered small-molecule drugs that inhibit S31N with potencies greater than amantadine's potency against WT M2. Drug binding locks the protein into a well-defined conformation, and the NMR structure of the complex shows the drug bound in the homotetrameric channel, threaded between the side chains of Asn31. Unrestrained molecular dynamics simulations predicted the same binding site. This S31N inhibitor, like other potent M2 inhibitors, contains a charged ammonium group. The ammonium binds as a hydrate to one of three sites aligned along the central cavity that appear to be hotspots for inhibition. These sites might stabilize hydronium-like species formed as protons diffuse through the outer channel to the proton-shuttling residue His37 near the cytoplasmic end of the channel.
Collapse
|
32
|
Gkeka P, Eleftheratos S, Kolocouris A, Cournia Z. Free Energy Calculations Reveal the Origin of Binding Preference for Aminoadamantane Blockers of Influenza A/M2TM Pore. J Chem Theory Comput 2013; 9:1272-81. [DOI: 10.1021/ct300899n] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Paraskevi Gkeka
- Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou,
11527 Athens, Greece
| | - Stelios Eleftheratos
- Faculty
of Pharmacy, Department of Pharmaceutical
Chemistry, University of Athens, Panepistimioupolis-Zografou,
15771 Athens, Greece
| | - Antonios Kolocouris
- Faculty
of Pharmacy, Department of Pharmaceutical
Chemistry, University of Athens, Panepistimioupolis-Zografou,
15771 Athens, Greece
| | - Zoe Cournia
- Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou,
11527 Athens, Greece
| |
Collapse
|
33
|
Hajzer V, Latika A, Durmis J, Šebesta R. EnantioselectiveMichaelAddition of the 2-(1-Ethylpropoxy)acetaldehyde toN-[(1Z)-2-Nitroethenyl]acetamide - Optimization of the Key Step in the Organocatalytic Oseltamivir Synthesis. Helv Chim Acta 2012. [DOI: 10.1002/hlca.201200527] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
34
|
Ye D, Shin WJ, Li N, Tang W, Feng E, Li J, He PL, Zuo JP, Kim H, Nam KY, Zhu W, Seong BL, Tai No K, Jiang H, Liu H. Synthesis of C-4-modified zanamivir analogs as neuraminidase inhibitors and their anti-AIV activities. Eur J Med Chem 2012; 54:764-70. [DOI: 10.1016/j.ejmech.2012.06.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 06/11/2012] [Accepted: 06/15/2012] [Indexed: 01/16/2023]
|
35
|
Feng E, Ye D, Li J, Zhang D, Wang J, Zhao F, Hilgenfeld R, Zheng M, Jiang H, Liu H. Recent advances in neuraminidase inhibitor development as anti-influenza drugs. ChemMedChem 2012; 7:1527-36. [PMID: 22807317 DOI: 10.1002/cmdc.201200155] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/13/2012] [Indexed: 11/06/2022]
Abstract
The recent emergence of the highly pathogenic H5N1 subtype of avian influenza virus (AIV) and of the new type of human influenza A (H1N1) have emphasized the need for the development of effective anti-influenza drugs. Presently, neuraminidase (NA) inhibitors are widely used in the treatment and prophylaxis of human influenza virus infection, and tremendous efforts have been made to develop more potent NA inhibitors to combat resistance and new influenza viruses. In this review, we discuss the structural characteristics of NA catalytic domains and the recent developments of new NA inhibitors using structure-based drug design strategies. These drugs include analogues of zanamivir, analogues of oseltamivir, analogues of peramivir, and analogues of aromatic carboxylic acid and present promising options for therapeutics or leads for further development of NA inhibitors that may be useful in the event of a future influenza pandemic.
Collapse
Affiliation(s)
- Enguang Feng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Graduate School of the Chinese Academy of Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
McCullough C, Wang M, Rong L, Caffrey M. Characterization of influenza hemagglutinin interactions with receptor by NMR. PLoS One 2012; 7:e33958. [PMID: 22815674 PMCID: PMC3397988 DOI: 10.1371/journal.pone.0033958] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 05/18/2012] [Indexed: 02/06/2023] Open
Abstract
In influenza, the envelope protein hemagglutinin (HA) plays a critical role in viral entry by first binding to sialic acid receptors on the cell surface and subsequently mediating fusion of the viral and target membranes. In this work, the receptor binding properties of influenza A HA from different subtypes (H1 A/California/04/09, H5 A/Vietnam/1205/04, H5 A/bar-headed goose/Qinghai/1A/05, and H9 A/Hong Kong/1073/99) have been characterized by NMR spectroscopy. Using saturation transfer difference (STD) NMR, we find that all HAs bind to the receptor analogs 2,3-sialyllactose and 2,6-sialyllactose, with subtle differences in the binding mode. Using competition STD NMR, we determine the receptor preferences for the HA subtypes. We find that H5-Qinghai and H9-Hong Kong HA bind to both receptor analogs with similar affinity. On the other hand, H1 exhibits a clear preference for 2,6-sialyllactose while H5-Vietnam exhibits a clear preference for 2,3-sialyllactose. Together, these results are interpreted within the context of differences in both the amino acid sequence and structures of HA from the different subtypes in determining receptor preference.
Collapse
Affiliation(s)
- Christopher McCullough
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Minxiu Wang
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
37
|
Zheng WH, Lin ZQ, Zhuo M, Du HL, Wang XN. [Research progress on influenza antiviral small RNAs]. YI CHUAN = HEREDITAS 2012; 34:526-532. [PMID: 22659424 DOI: 10.3724/sp.j.1005.2012.00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Worldwide influenza caused by influenza virus is a respiratory disease which threats the public health by seasonal epidemics or global influenza outbreak. Vaccines and drugs are current therapies, but there are many restricted factors such as neurotoxicity, side effects of gastrointestinal, and drug resistance. New technologies, particularly RNAi mediated by small RNAs, has become a potential and robust method in influenza antiviral research for its high efficiency, specific, and speedy. Following the spread and epidemic of the influenza virus, application of small RNAs into influenza antiviral research has been reported increasingly. The small RNAs, PA-2087, NP-1496, and M-950, which targets PA, NP, and M2 genes, respectively, are the most effective anti-influenza siRNAs up to now. siRNA of targeting conservative region of different influenza viral genes has broader effect on virus inhibition. The combination of siRNAs of targeting different genes can achieve better virus inhibition. In this review, we mainly described the progress of siRNAs and miRNAs for anti-influenza virus, and the prospects and hurdles of influenza RNAi therapy as well.
Collapse
Affiliation(s)
- Wei-Hao Zheng
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China.
| | | | | | | | | |
Collapse
|
38
|
Rouchal M, Nečas M, Vícha R. (1-Adamantyl)(3-aminophenyl)methanone. Acta Crystallogr Sect E Struct Rep Online 2011; 67:o3198. [PMID: 22199718 PMCID: PMC3238865 DOI: 10.1107/s1600536811046009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 11/01/2011] [Indexed: 11/17/2022]
Abstract
In the crystal sructure of the title compound, C17H21NO, the molecular packing is stabilized by intermolecular N—H⋯O hydrogen bonds and additional weak N—H⋯π interactions, forming chains that propagate along the b axis. Conjugation of the carbonyl group and the benzene ring is rather attenuated due to a twisting of the carbonyl group from the plane of the benzene ring [torsion angle = 27.1 (2)°].
Collapse
|
39
|
Chou TC, Hsu W, Wang CH, Chen YJ, Fang JM. Rapid and specific influenza virus detection by functionalized magnetic nanoparticles and mass spectrometry. J Nanobiotechnology 2011; 9:52. [PMID: 22088100 PMCID: PMC3248366 DOI: 10.1186/1477-3155-9-52] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 11/16/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The timely and accurate diagnosis of specific influenza virus strains is crucial to effective prophylaxis, vaccine preparation and early antiviral therapy. The detection of influenza A viruses is mainly accomplished using polymerase chain reaction (PCR) techniques or antibody-based assays. In conjugation with the immunoassay utilizing monoclonal antibody, mass spectrometry is an alternative to identify proteins derived from a target influenza virus. Taking advantage of the large surface area-to-volume ratio, antibody-conjugated magnetic nanoparticles can act as an effective probe to extract influenza virus for sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE) and on-bead mass spectrometric analysis. RESULTS Iron oxide magnetic nanoparticles (MNP) were functionalized with H5N2 viral antibodies targeting the hemagglutinin protein and capped with methoxy-terminated ethylene glycol to suppress nonspecific binding. The antibody-conjugated MNPs possessed a high specificity to H5N2 virus without cross-reactivity with recombinant H5N1 viruses. The unambiguous identification of the captured hemagglutinin on magnetic nanoparticles was realized by SDS-PAGE visualization and peptide sequence identification using liquid chromatography-tandem mass spectrometry (LC-MS/MS). CONCLUSIONS The assay combining efficient magnetic separation and MALDI-MS readout offers a rapid and sensitive method for virus screening. Direct on-MNP detection by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) provided high sensitivity (~10(3) EID(50) per mL) and a timely diagnosis within one hour. The magnetic nanoparticles encapsulated with monoclonal antibodies could be used as a specific probe to distinguish different subtypes of influenza.
Collapse
Affiliation(s)
- Tzu-Chi Chou
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan
| | | | | | | | | |
Collapse
|
40
|
Wang W, Zhang P, Hao C, Zhang XE, Cui ZQ, Guan HS. In vitro inhibitory effect of carrageenan oligosaccharide on influenza A H1N1 virus. Antiviral Res 2011; 92:237-46. [DOI: 10.1016/j.antiviral.2011.08.010] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 08/05/2011] [Accepted: 08/10/2011] [Indexed: 01/09/2023]
|
41
|
Wang J, Ma C, Fiorin G, Carnevale V, Wang T, Hu F, Lamb RA, Pinto LH, Hong M, Klein ML, DeGrado WF. Molecular dynamics simulation directed rational design of inhibitors targeting drug-resistant mutants of influenza A virus M2. J Am Chem Soc 2011; 133:12834-41. [PMID: 21744829 DOI: 10.1021/ja204969m] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Influenza A virus M2 (A/M2) forms a homotetrameric proton selective channel in the viral membrane. It has been the drug target of antiviral drugs such as amantadine and rimantadine. However, most of the current virulent influenza A viruses carry drug-resistant mutations alongside the drug binding site, such as S31N, V27A, and L26F, etc., each of which might be dominant in a given flu season. Among these mutations, the V27A mutation was prevalent among transmissible viruses under drug selection pressure. Until now, V27A has not been successfully targeted by small molecule inhibitors, despite years of extensive medicinal chemistry research efforts and high throughput screening. Guided by molecular dynamics (MD) simulation of drug binding and the influence of drug binding on the dynamics of A/M2 from earlier experimental studies, we designed a series of potent spirane amine inhibitors targeting not only WT, but also both A/M2-27A and L26F mutants with IC(50)s similar to that seen for amantadine's inhibition of the WT channel. The potencies of these inhibitors were further demonstrated in experimental binding and plaque reduction assays. These results demonstrate the power of MD simulations to probe the mechanism of drug binding as well as the ability to guide design of inhibitors of targets that had previously appeared to be undruggable.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemistry, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
You L, Cho EJ, Leavitt J, Ma LC, Montelione GT, Anslyn EV, Krug RM, Ellington A, Robertus JD. Synthesis and evaluation of quinoxaline derivatives as potential influenza NS1A protein inhibitors. Bioorg Med Chem Lett 2011; 21:3007-11. [PMID: 21478016 PMCID: PMC3114437 DOI: 10.1016/j.bmcl.2011.03.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 03/10/2011] [Accepted: 03/11/2011] [Indexed: 01/22/2023]
Abstract
A library of quinoxaline derivatives were prepared to target non-structural protein 1 of influenza A (NS1A) as a means to develop anti-influenza drug leads. An in vitro fluorescence polarization assay demonstrated that these compounds disrupted the dsRNA-NS1A interaction to varying extents. Changes of substituent at positions 2, 3 and 6 on the quinoxaline ring led to variance in responses. The most active compounds (35 and 44) had IC(50) values in the range of low micromolar concentration without exhibiting significant dsRNA intercalation. Compound 44 was able to inhibit influenza A/Udorn/72 virus growth.
Collapse
Affiliation(s)
- Lei You
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Eun Jeong Cho
- The Institute for Drug and Diagnostic Development, University of Texas at Austin, Austin, TX, 78712, USA
| | - John Leavitt
- Department of Molecular Genetics and Microbiology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Li-Chung Ma
- Center for Advanced Biotechnology and Medicine, Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Gaetano T. Montelione
- Center for Advanced Biotechnology and Medicine, Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Biochemistry, Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Eric V. Anslyn
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Robert M. Krug
- Department of Molecular Genetics and Microbiology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Ellington
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Jon D. Robertus
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
43
|
Duque MD, Ma C, Torres E, Wang J, Naesens L, Juárez-Jiménez J, Camps P, Luque FJ, DeGrado WF, Lamb RA, Pinto LH, Vázquez S. Exploring the size limit of templates for inhibitors of the M2 ion channel of influenza A virus. J Med Chem 2011; 54:2646-57. [PMID: 21466220 PMCID: PMC3174104 DOI: 10.1021/jm101334y] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Amantadine inhibits the M2 proton channel of influenza A virus, yet its clinical use has been limited by the rapid emergence of amantadine-resistant virus strains. We have synthesized and characterized a series of polycyclic compounds designed as ring-contracted or ring-expanded analogues of amantadine. Inhibition of the wild-type (wt) M2 channel and the A/M2-S31N and A/M2-V27A mutant ion channels were measured in Xenopus oocytes using two-electrode voltage clamp (TEV) assays. Several bisnoradamantane and noradamantane derivatives inhibited the wt ion channel. The compounds bind to a primary site delineated by Val27, Ala30, and Ser31, though ring expansion restricts the positioning in the binding site. Only the smallest analogue 8 was found to inhibit the S31N mutant ion channel. The structure-activity relationship obtained by TEV assay was confirmed by plaque reduction assays with A/H3N2 influenza virus carrying wt M2 protein.
Collapse
Affiliation(s)
- María D. Duque
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Diagonal 643, Barcelona E-08028, Spain
| | - Chunlong Ma
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208-3500
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208-3500
| | - Eva Torres
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Diagonal 643, Barcelona E-08028, Spain
| | - Jun Wang
- Department of Chemistry and Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059
| | - Lieve Naesens
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Jordi Juárez-Jiménez
- Departament de Fisicoquímica, Facultat de Farmàcia, and Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Diagonal 643, E-08028, Barcelona, Spain
| | - Pelayo Camps
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Diagonal 643, Barcelona E-08028, Spain
| | - F. Javier Luque
- Departament de Fisicoquímica, Facultat de Farmàcia, and Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Diagonal 643, E-08028, Barcelona, Spain
| | - William F. DeGrado
- Department of Chemistry and Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059
| | - Robert A. Lamb
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208-3500
- Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208-3500
| | - Lawrence H. Pinto
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208-3500
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Diagonal 643, Barcelona E-08028, Spain
| |
Collapse
|
44
|
De Clercq E. The next ten stories on antiviral drug discovery (part E): advents, advances, and adventures. Med Res Rev 2011; 31:118-60. [PMID: 19844936 PMCID: PMC7168424 DOI: 10.1002/med.20179] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review article presents the fifth part (part E) in the series of stories on antiviral drug discovery. The ten stories belonging to this fifth part are dealing with (i) aurintricarboxylic acid; (ii) alkenyldiarylmethanes; (iii) human immunodeficiency virus (HIV) integrase inhibitors; (iv) lens epithelium‐derived growth factor as a potential target for HIV proviral DNA integration; (v) the status presens of neuraminidase inhibitors NAIs in the control of influenza virus infections; (vi) the status presens on respiratory syncytial virus inhibitors; (vii) tricyclic (1,N‐2‐ethenoguanine)‐based acyclovir and ganciclovir derivatives; (viii) glycopeptide antibiotics as antivirals targeted at viral entry; (ix) the potential (off‐label) use of cidofovir in the treatment of polyoma (JC and BK) virus infections; and (x) finally, thymidine phosphorylase as a target for both antiviral and anticancer agents. © 2009 Wiley Periodicals, Inc. Med Res Rev, 31, No. 1, 118–160, 2010
Collapse
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research, K.U.Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium.
| |
Collapse
|
45
|
Wang J, Ma C, Balannik V, Pinto LH, Lamb RA, DeGrado WF. Exploring the Requirements for the Hydrophobic Scaffold and Polar Amine in inhibitors of M2 from Influenza A Virus. ACS Med Chem Lett 2011; 2:307-312. [PMID: 21691418 DOI: 10.1021/ml100297w] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inhibitors targeting the influenza A virus M2 (A/M2) proton channel, have lost their effectiveness due to widespread resistance. As a first step in the development of new inhibitors that address this problem, we have screened several focused collections of small molecules using two electrode voltage patch clamp assays (TEVC) on Xenopus laevis Oocyte. Diverse head groups and scaffolds of A/M2 inhibitors have been explored. It has been found that not only amine, but also hydroxyl, aminooxyl, guanidine and amidine compounds are active against the A/M2 proton channel. Moreover, the channel is able to accommodate a wide range of structural variation in the apolar scaffold. This study offers information to guide the next generation of A/M2 proton channel inhibitor design.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemistry and ‡Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
- Department of Neurobiology and Physiology, ∥Department of Biochemistry, Molecular Biology and Cell Biology, and ⊥Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208-3500, United States
| | - Chunlong Ma
- Department of Chemistry and ‡Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
- Department of Neurobiology and Physiology, ∥Department of Biochemistry, Molecular Biology and Cell Biology, and ⊥Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208-3500, United States
| | - Victoria Balannik
- Department of Chemistry and ‡Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
- Department of Neurobiology and Physiology, ∥Department of Biochemistry, Molecular Biology and Cell Biology, and ⊥Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208-3500, United States
| | - Lawrence H. Pinto
- Department of Chemistry and ‡Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
- Department of Neurobiology and Physiology, ∥Department of Biochemistry, Molecular Biology and Cell Biology, and ⊥Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208-3500, United States
| | - Robert A. Lamb
- Department of Chemistry and ‡Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
- Department of Neurobiology and Physiology, ∥Department of Biochemistry, Molecular Biology and Cell Biology, and ⊥Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208-3500, United States
| | - William F. DeGrado
- Department of Chemistry and ‡Department of Biochemistry and Biophysics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
- Department of Neurobiology and Physiology, ∥Department of Biochemistry, Molecular Biology and Cell Biology, and ⊥Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois 60208-3500, United States
| |
Collapse
|
46
|
Trajkovic M, Ferjancic Z, Saicic RN. An aldol approach to the enantioselective synthesis of (−)-oseltamivir phosphate. Org Biomol Chem 2011; 9:6927-9. [DOI: 10.1039/c1ob06248d] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
47
|
Kwon HJ, Kim HH, Yoon SY, Ryu YB, Chang JS, Cho KO, Rho MC, Park SJ, Lee WS. In vitro inhibitory activity of Alpinia katsumadai extracts against influenza virus infection and hemagglutination. Virol J 2010; 7:307. [PMID: 21062499 PMCID: PMC2989970 DOI: 10.1186/1743-422x-7-307] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 11/10/2010] [Indexed: 12/04/2022] Open
Abstract
Background Alpinia katsumadai (AK) extracts and fractions were tested for in vitro antiviral activities against influenza virus type A, specially human A/PR/8/34 (H1N1) and avian A/Chicken/Korea/MS96/96 (H9N2), by means of time-of-addition experiments; pre-treatment, simultaneous treatment, and post treatment. Results In pre-treatment assay, the AK extracts and AK fractions did not show significant antiviral activity. During the simultaneous treatment assay, one AK extract and five AK fractions designated as AK-1 to AK-3, AK-5, AK-10, and AK-11 showed complete inhibition of virus infectivity against A/PR/8/34 (H1N1) and A/Chicken/Korea/MS96/96 (H9N2). The 50% effective inhibitory concentrations (EC50) of these one AK extracts and five AK fractions with exception of the AK-9 were from 0.8 ± 1.4 to 16.4 ± 4.5 μg/mL against A/PR/8/34 (H1N1). The two AK extracts and three AK fractions had EC50 values ranging from <0.39 ± 0.4 to 2.3 ± 3.6 μg/mL against A/Chicken/Korea/MS96/96 (H9N2). By the hemagglutination inhibition (HI) assay, the two AK extracts and five AK fractions completely inhibited viral adsorption onto chicken RBCs at less than 100 μg/mL against both A/PR/8/34 (H1N1) and A/Chicken/Korea/MS96/96 (H9N2). Interestingly, only AK-3 was found with inhibition for both viral attachment and viral replication after showing extended antiviral activity during the post treatment assay and quantitative real-time PCR. Conclusions These results suggest that AK extracts and fractions had strong anti-influenza virus activity that can inhibit viral attachment and/or viral replication, and may be used as viral prophylaxis.
Collapse
Affiliation(s)
- Hyung-Jun Kwon
- Eco-Friendly Biomaterial Research Center and AI Control Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 580-185, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ma J, Zhao Y, Ng S, Zhang J, Zeng J, Than A, Chen P, Liu XW. Sugar-Based Synthesis of Tamiflu and Its Inhibitory Effects on Cell Secretion. Chemistry 2010; 16:4533-40. [DOI: 10.1002/chem.200902048] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 11/02/2009] [Indexed: 12/16/2022]
|
49
|
Balannik V, Wang J, Ohigashi Y, Jing X, Magavern E, Lamb RA, Degrado WF, Pinto LH. Design and pharmacological characterization of inhibitors of amantadine-resistant mutants of the M2 ion channel of influenza A virus. Biochemistry 2010; 48:11872-82. [PMID: 19905033 DOI: 10.1021/bi9014488] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The A/M2 proton channel of influenza A virus is a target for the anti-influenza drugs amantadine and rimantadine, whose effectiveness was diminished by the appearance of naturally occurring point mutants in the A/M2 channel pore, among which the most common are S31N, V27A, and L26F. We have synthesized and characterized the properties of a series of compounds, originally derived from the A/M2 inhibitor BL-1743. A lead compound emerging from these investigations, spiro[5.5]undecan-3-amine, is an effective inhibitor of wild-type A/M2 channels and L26F and V27A mutant ion channels in vitro and also inhibits replication of recombinant mutant viruses bearing these mutations in plaque reduction assays. Differences in the inhibition kinetics between BL-1743, known to bind inside the A/M2 channel pore, and amantadine were exploited to demonstrate competition between these compounds, consistent with the conclusion that amantadine binds inside the channel pore. Inhibition by all of these compounds was shown to be voltage-independent, suggesting that their charged groups are within the N-terminal half of the pore, prior to the selectivity filter that defines the region over which the transmembrane potential occurs. These findings not only help to define the location and mechanism of binding of M2 channel-blocking drugs but also demonstrate the feasibility of discovering new inhibitors that target this binding site in a number of amantadine-resistant mutants.
Collapse
Affiliation(s)
- Victoria Balannik
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208-3500, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Synthesis of poly(aspartimide)-based bio-glycoconjugates. Carbohydr Res 2010; 345:33-40. [DOI: 10.1016/j.carres.2009.08.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 08/21/2009] [Accepted: 08/25/2009] [Indexed: 11/20/2022]
|