1
|
Laczi D, Huamán SS, Andrews-Clark T, Laidlaw SM, Salah E, Dumjahn L, Lukacik P, Choudhry H, Walsh MA, Carroll MW, Schofield CJ, Brewitz L. Silaproline-bearing nirmatrelvir derivatives are potent inhibitors of the SARS-CoV-2 main protease highlighting the value of silicon-derivatives in structure-activity-relationship studies. Eur J Med Chem 2025; 291:117603. [PMID: 40220677 DOI: 10.1016/j.ejmech.2025.117603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Nirmatrelvir is a substrate-related inhibitor of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) main protease (Mpro) that is clinically used in combination with ritonavir to treat COVID-19. Derivatives of nirmatrelvir, modified at the substrate P2-equivalent position, have been developed to fine-tune inhibitor properties and are now in clinical use. We report the synthesis of nirmatrelvir derivatives with a (R)-4,4-dimethyl-4-silaproline (silaproline) group at the P2-equivalent position. Mass spectrometry (MS)-based assays demonstrate that silaproline-bearing nirmatrelvir derivatives efficiently inhibit isolated recombinant Mpro, albeit with reduced potency compared to nirmatrelvir. Investigations with SARS-CoV-2 infected VeroE6 cells reveal that the silaproline-bearing inhibitors with a CF3 group at the P4-equivalent position inhibit viral progression, implying that incorporating silicon atoms into Mpro inhibitors can yield in vivo active inhibitors with appropriate optimization. MS and crystallographic studies show that the nucleophilic active site cysteine residue of Mpro (Cys145) reacts with the nitrile group of the silaproline-bearing inhibitors. Substituting the electrophilic nitrile group for a non-activated terminal alkyne shifts the inhibition mode from reversible covalent inhibition to irreversible covalent inhibition. One of the two prochiral silaproline methyl groups occupies space in the S2 pocket that is unoccupied in Mpro:nirmatrelvir complex structures, highlighting the value of sila-derivatives in structure-activity-relationship (SAR) studies. The combined results highlight the potential of silicon-containing molecules for inhibition of Mpro and, by implication, other nucleophilic cysteine enzymes.
Collapse
Affiliation(s)
- Dóra Laczi
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Sofia Schönbauer Huamán
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Taylah Andrews-Clark
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK; Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK; Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Stephen M Laidlaw
- Centre for Human Genetics & Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, OX3 7BN, Oxford, UK
| | - Eidarus Salah
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Leo Dumjahn
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK
| | - Petra Lukacik
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK; Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Hani Choudhry
- Department of Biochemistry, Center for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Martin A Walsh
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK; Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Miles W Carroll
- Centre for Human Genetics & Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, OX3 7BN, Oxford, UK
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK.
| | - Lennart Brewitz
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, OX1 3TA, Oxford, UK.
| |
Collapse
|
2
|
Panayides JL, Riley DL, Hasenmaile F, van Otterlo WAL. The role of silicon in drug discovery: a review. RSC Med Chem 2024; 15:3286-3344. [PMID: 39430101 PMCID: PMC11484438 DOI: 10.1039/d4md00169a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/07/2024] [Indexed: 10/22/2024] Open
Abstract
This review aims to highlight the role of silicon in drug discovery. Silicon and carbon are often regarded as being similar with silicon located directly beneath carbon in the same group in the periodic table. That being noted, in many instances a clear dichotomy also exists between silicon and carbon, and these differences often lead to vastly different physiochemical and biological properties. As a result, the utility of silicon in drug discovery has attracted significant attention and has grown rapidly over the past decade. This review showcases some recent advances in synthetic organosilicon chemistry and examples of the ways in which silicon has been employed in the drug-discovery field.
Collapse
Affiliation(s)
- Jenny-Lee Panayides
- Pharmaceutical Technologies, Future Production: Chemicals, Council for Scientific and Industrial Research (CSIR) Meiring Naude Road, Brummeria Pretoria South Africa
| | - Darren Lyall Riley
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria Lynnwood Road Pretoria South Africa
| | - Felix Hasenmaile
- Department of Chemistry and Polymer Science, Stellenbosch University Matieland Stellenbosch 7600 South Africa
| | - Willem A L van Otterlo
- Department of Chemistry and Polymer Science, Stellenbosch University Matieland Stellenbosch 7600 South Africa
| |
Collapse
|
3
|
Deng C, Wang X, Wang T, Liu W, Yuan X, Huang Y, Cao S. Virtual screening and molecular growth guide the design of inhibitors for the influenza virus drug-resistant mutant M2-V27A/S31N. J Biomol Struct Dyn 2024; 42:5253-5267. [PMID: 37424098 DOI: 10.1080/07391102.2023.2233026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023]
Abstract
The influenza A virus matrix protein 2 (AM2) protein is a proton-gated, proton-selective ion channel essential for influenza replication that has been identified as an antiviral target. The drug-resistance of the M2-V27A/S31N strain, which has been growing more prevalent in recent years and has the potential to spread globally, prevents current amantadine inhibitors from having the desired impact. In this study, we compiled the most common influenza A virus strains from 2001-2020 from the U.S. National Center for Biotechnology Information database and hypothesized that M2-V27A/S31N would become a common strain. The lead compound ZINC299830590 was screened for M2-V27A/S31N in the ZINC15 database using a pharmacophore model and molecular descriptors. This lead compound was then optimized by molecular growth, which allowed us to identify important amino acid residues and create interactions with them to produce compound 4. Molecular dynamics simulation showed that the complex of compound 4 and M2-V27A/S31N had certain degrees of stability and flexibility. The binding free energy of compound 4 was calculated using the MM/PB(GB)SA method and totaled -106.525 kcal/mol. Finally, physicochemical and pharmacokinetic profiles were predicted using the Absorption, Distribution, Metabolism, Excretion, and Toxicity model, which indicated the good bioavailability of compound 4. These results provide the basis for further in vivo and in vitro studies to demonstrate that compound 4 is a promising drug candidate against M2-V27A/S31N.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Changyong Deng
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Xiaobo Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Tangle Wang
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Wei Liu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Xiaolan Yuan
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Yan Huang
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Shuang Cao
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| |
Collapse
|
4
|
Ivleva EA, Zaborskaya MS, Shiryaev VA, Klimochkin YN. One pot synthesis of bridgehead amino alcohols from diamantoid hydrocarbons. SYNTHETIC COMMUN 2023. [DOI: 10.1080/00397911.2023.2177173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- Elena A. Ivleva
- Department of Organic Chemistry, Samara State Technical University, Samara, Russian Federation
| | - Maria S. Zaborskaya
- Department of Organic Chemistry, Samara State Technical University, Samara, Russian Federation
| | - Vadim A. Shiryaev
- Department of Organic Chemistry, Samara State Technical University, Samara, Russian Federation
| | - Yuri N. Klimochkin
- Department of Organic Chemistry, Samara State Technical University, Samara, Russian Federation
| |
Collapse
|
5
|
Hu C, Vo C, Merchant RR, Chen SJ, Hughes JME, Peters BK, Qin T. Uncanonical Semireduction of Quinolines and Isoquinolines via Regioselective HAT-Promoted Hydrosilylation. J Am Chem Soc 2023; 145:25-31. [PMID: 36548026 PMCID: PMC9930105 DOI: 10.1021/jacs.2c11664] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Heterocycles are the backbone of modern medical chemistry and drug development. The derivatization of "an olefin" inside aromatic rings represents an ideal approach to access functionalized saturated heterocycles from abundant aromatic building blocks. Here, we report an operationally simple, efficient, and practical method to selectively access hydrosilylated and reduced N-heterocycles from bicyclic aromatics via a key diradical intermediate. This approach is expected to facilitate complex heterocycle functionalizations that enable access to novel medicinally relevant scaffolds.
Collapse
Affiliation(s)
- Chao Hu
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Cuong Vo
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Rohan R. Merchant
- Department of Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Si-Jie Chen
- Department of Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Jonathan M. E. Hughes
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Byron K. Peters
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Tian Qin
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
6
|
Hu Y, Ma C, Wang J. Cytopathic Effect Assay and Plaque Assay to Evaluate in vitro Activity of Antiviral Compounds Against Human Coronaviruses 229E, OC43, and NL63. Bio Protoc 2022; 12:e4314. [PMID: 35284599 PMCID: PMC8855088 DOI: 10.21769/bioprotoc.4314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/30/2021] [Accepted: 12/10/2021] [Indexed: 10/03/2023] Open
Abstract
Coronaviruses are important human pathogens, among which the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent for the COVID-19 pandemic. To combat the SARS-CoV-2 pandemic, there is a pressing need for antivirals, especially broad-spectrum antivirals that are active against all seven human coronaviruses (HCoVs). For this reason, we are interested in developing antiviral assays to expedite the drug discovery process. Here, we provide the detailed protocol for the cytopathic effect (CPE) assay and the plaque assay for human coronaviruses 229E (HCoV-229E), HCoV-OC43, and HCoV-NL63, to identify novel antivirals against HCoVs. Neutral red was used in the CPE assay, as it is relatively inexpensive and more sensitive than other reagents. Multiple parameters including multiplicity of infection, incubation time and temperature, and staining conditions have been optimized for CPE and plaque assays for HCoV-229E in MRC-5, Huh-7, and RD cell lines; HCoV-OC43 in RD, MRC-5, and BSC-1 cell lines, and HCoV-NL63 in Vero E6, Huh-7, MRC-5, and RD cell lines. Both CPE and plaque assays have been calibrated with the positive control compounds remdesivir and GC-376. Both CPE and plaque assays have high sensitivity, excellent reproducibility, and are cost-effective. The protocols described herein can be used as surrogate assays in the biosafety level 2 facility to identify entry inhibitors and protease inhibitors for SARS-CoV-2, as HCoV-NL63 also uses ACE2 as the receptor for cell entry, and the main proteases of HCoV-OC43 and SARS-CoV-2 are highly conserved. In addition, these assays can also be used as secondary assays to profile the broad-spectrum antiviral activity of existing SARS-CoV-2 drug candidates.
Collapse
Affiliation(s)
- Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
7
|
Hwu JR, Kapoor M, Gupta NK, Tsay SC, Huang WC, Tan KT, Hu YC, Lyssen P, Neyts J. Synthesis and antiviral activities of quinazolinamine–coumarin conjugates toward chikungunya and hepatitis C viruses. Eur J Med Chem 2022; 232:114164. [DOI: 10.1016/j.ejmech.2022.114164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 11/04/2022]
|
8
|
Cáceres CJ, Hu Y, Cárdenas-García S, Wu X, Tan H, Carnaccini S, Gay LC, Geiger G, Ma C, Zhang QY, Rajao D, Perez DR, Wang J. Rational design of a deuterium-containing M2-S31N channel blocker UAWJ280 with in vivo antiviral efficacy against both oseltamivir sensitive and -resistant influenza A viruses. Emerg Microbes Infect 2021; 10:1832-1848. [PMID: 34427541 PMCID: PMC8451667 DOI: 10.1080/22221751.2021.1972769] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 10/28/2022]
Abstract
Seasonal influenza A virus (IAV) infections are among the most important global health problems. FDA-approved antiviral therapies against IAV include neuraminidase inhibitors, M2 inhibitors, and polymerase inhibitor baloxavir. Resistance against adamantanes (amantadine and rimantadine) is widespread as virtually all IAV strains currently circulating in the human population are resistant to adamantanes through the acquisition of the S31N mutation. The neuraminidase inhibitor-resistant strains also contain the M2-S31N mutant, suggesting M2-S31N is a high-profile antiviral drug target. Here we report the development of a novel deuterium-containing M2-S31N inhibitor UAWJ280. UAWJ280 had broad-spectrum antiviral activity against both oseltamivir sensitive and -resistant influenza A strains and had a synergistic antiviral effect in combination with oseltamivir in cell culture. In vivo pharmacokinetic (PK) studies demonstrated that UAWJ280 had favourable PK properties. The in vivo mouse model study showed that UAWJ280 was effective alone or in combination with oseltamivir in improving clinical signs and survival after lethal challenge with an oseltamivir sensitive IAV H1N1 strain. Furthermore, UAWJ280 was also able to ameliorate clinical signs and increase survival when mice were challenged with an oseltamivir-resistant IAV H1N1 strain. In conclusion, we show for the first time that the M2-S31N channel blocker UAWJ280 has in vivo antiviral efficacy in mice that are infected with either oseltamivir sensitive or -resistant IAVs, and it has a synergistic antiviral effect with oseltamivir.
Collapse
Affiliation(s)
- C. Joaquín Cáceres
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Stivalis Cárdenas-García
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Xiangmeng Wu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Haozhou Tan
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Silvia Carnaccini
- Tifton diagnostic laboratory, College of Veterinary Medicine, University of Georgia, Tifton, GA, USA
| | - L. Claire Gay
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Ginger Geiger
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Qing-Yu Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Daniela Rajao
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Daniel R. Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
9
|
Colorimetric detection of Fe3+ ions using Schiff base-chalcone functionalized bis(1,2,3-triazolyl-γ-propyltriethoxysilanes). Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
10
|
Design of pyrene functionalized triazole linked organosilane for specific detection of Ce3+ ions. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130787] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
11
|
Yamagishi H, Saito H, Shimokawa J, Yorimitsu H. Design, Synthesis, and Implementation of Sodium Silylsilanolates as Silyl Transfer Reagents. ACS Catal 2021. [DOI: 10.1021/acscatal.1c02733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Hiroki Yamagishi
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Hayate Saito
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Jun Shimokawa
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Hideki Yorimitsu
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
12
|
Shiryaev VA, Klimochkin YN. Heterocyclic Inhibitors of Viroporins in the Design of Antiviral Compounds. Chem Heterocycl Compd (N Y) 2020; 56:626-635. [PMID: 32836315 PMCID: PMC7366462 DOI: 10.1007/s10593-020-02712-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
Ion channels of viruses (viroporins) represent a common type of protein targets for drugs. The relative simplicity of channel architecture allows convenient computational modeling and enables virtual search for new inhibitors. In this review, we analyze the data published over the last 10 years on known ion channels of viruses that cause socially significant diseases. The effectiveness of inhibition by various types of heterocyclic compounds of the viroporins of influenza virus, hepatitis С virus, human immunodeficiency virus, human papillomaviruses, coronaviruses, and respiratory syncytial virus is discussed. The presented material highlights the promise held by the search for heterocyclic antiviral compounds that act by inhibition of viroporins.
Collapse
Affiliation(s)
- Vadim A. Shiryaev
- Samara State Technical University, 244 Molodogvardeiskaya St, Samara, 443100 Russia
| | - Yuri N. Klimochkin
- Samara State Technical University, 244 Molodogvardeiskaya St, Samara, 443100 Russia
| |
Collapse
|
13
|
Jabłońska J, Kluska M, Erchak N. The challenge of separating and determining biologically active electrostatically stabilized silanates using the high-performance liquid chromatography technique. J Sep Sci 2020; 43:3399-3407. [PMID: 32567759 DOI: 10.1002/jssc.202000453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 11/10/2022]
Abstract
We present the results of research on the optimal conditions for the separation and determination of newly obtained hypercoordinated compounds, which belong to the group of electrostatically stabilized silanates. The research involved five stationary and four mobile phases. The best selectivity was obtained using the graphite phase and the mobile phase consisting of acetonitrile/water (80/20). The maximum selectivity of the determined electrostatically stabilized silanates was 1.13 and 1.06 for (1), (2), (3); 1.10 and 1.15 for (4), (5), (6); and 1.12 and 1.15 for (7), (8), (9). The octadecyl phase (which is recommended as standard) did not yield satisfactory results.
Collapse
Affiliation(s)
- Joanna Jabłońska
- Faculty of Exact and Natural Sciences, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Mariusz Kluska
- Faculty of Exact and Natural Sciences, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | | |
Collapse
|
14
|
Jalily PH, Duncan MC, Fedida D, Wang J, Tietjen I. Put a cork in it: Plugging the M2 viral ion channel to sink influenza. Antiviral Res 2020; 178:104780. [PMID: 32229237 PMCID: PMC7102647 DOI: 10.1016/j.antiviral.2020.104780] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/12/2020] [Accepted: 03/20/2020] [Indexed: 12/17/2022]
Abstract
The ongoing threat of seasonal and pandemic influenza to human health requires antivirals that can effectively supplement existing vaccination strategies. The M2 protein of influenza A virus (IAV) is a proton-gated, proton-selective ion channel that is required for virus replication and is an established antiviral target. While licensed adamantane-based M2 antivirals have been historically used, M2 mutations that confer major adamantane resistance are now so prevalent in circulating virus strains that these drugs are no longer recommended. Here we review the current understanding of IAV M2 structure and function, mechanisms of inhibition, the rise of drug resistance mutations, and ongoing efforts to develop new antivirals that target resistant forms of M2.
Collapse
Affiliation(s)
- Pouria H Jalily
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Maggie C Duncan
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - David Fedida
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tuscon, AZ, USA
| | - Ian Tietjen
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada; The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Musharrafieh R, Ma C, Wang J. Discovery of M2 channel blockers targeting the drug-resistant double mutants M2-S31N/L26I and M2-S31N/V27A from the influenza A viruses. Eur J Pharm Sci 2019; 141:105124. [PMID: 31669761 DOI: 10.1016/j.ejps.2019.105124] [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: 09/26/2019] [Revised: 10/17/2019] [Accepted: 10/25/2019] [Indexed: 12/29/2022]
Abstract
Influenza virus infections are a persistent threat to human health due to seasonal outbreaks and sporadic pandemics. Amantadine and rimantadine are FDA-approved influenza antiviral drugs and work by inhibiting the viral M2 proton channel. However, the therapeutic potential for the antiviral amantadine/rimantadine was curtailed by the emergence of drug-resistant mutations in its target protein M2. In this study, we identified four amantadine-resistant M2 mutants among avian and human influenza A H5N1 strains circulating between 2002 and 2019: the single S31N and V27A mutants, and the S31N/L26I and S31N/V27A double mutants. Herein, utilizing two-electrode voltage clamp (TEVC) assays, we screened a panel of structurally diverse M2 inhibitors against these single and double mutant channels. Three compounds 6, 7, and 15 were found to significantly block all three M2 mutants: M2-S31N, M2-S31N/L26I, and M2-S31N/V27A. Using recombinant viruses generated from reverse genetics, we further showed that these compounds also inhibited the replication of recombinant viruses harboring either the single S31N or double S31N/L26I and S31N/V27A mutants. This work represents the first example in developing antivirals by targeting the drug-resistant double mutants of M2 proton channels.
Collapse
Affiliation(s)
- Rami Musharrafieh
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721 USA
| | - Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721 USA
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721 USA.
| |
Collapse
|
16
|
Yang L, Zhang JH, Zhang XL, Lao GJ, Su GM, Wang L, Li YL, Ye WC, He J. Tandem mass tag-based quantitative proteomic analysis of lycorine treatment in highly pathogenic avian influenza H5N1 virus infection. PeerJ 2019; 7:e7697. [PMID: 31592345 PMCID: PMC6778435 DOI: 10.7717/peerj.7697] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022] Open
Abstract
Highly pathogenic H5N1 influenza viruses (HPAIV) cause rapid systemic illness and death in susceptible animals, leading to a disease with high morbidity and mortality rates. Although vaccines and drugs are the best solution to prevent this threat, a more effective treatment for H5 strains of influenza has yet to be developed. Therefore, the development of therapeutics/drugs that combat H5N1 influenza virus infection is becoming increasingly important. Lycorine, the major component of Amaryllidaceae alkaloids, exhibits better protective effects against A/CK/GD/178/04 (H5N1) (GD178) viruses than the commercial neuraminidase (NA) inhibitor oseltamivir in our prior study. Lycorine demonstrates outstanding antiviral activity because of its inhibitory activity against the export of viral ribonucleoprotein complexes (vRNPs) from the nucleus. However, how lycorine affects the proteome of AIV infected cells is unknown. Therefore, we performed a comparative proteomic analysis to identify changes in protein expression in AIV-infected Madin-Darby Canine Kidney cells treated with lycorine. Three groups were designed: mock infection group (M), virus infection group (V), and virus infection and lycorine-treated after virus infection group (L). The multiplexed tandem mass tag (TMT) approach was employed to analyze protein level in this study. In total, 5,786 proteins were identified from the three groups of cells by using TMT proteomic analysis. In the V/M group, 1,101 proteins were identified, of which 340 differentially expressed proteins (DEPs) were determined during HPAIV infection; among the 1,059 proteins identified from the lycorine-treated group, 258 proteins presented significant change. Here, 71 proteins showed significant upregulation or downregulation of expression in the virus-infected/mock and virus-infected/lycorine-treated comparisons, and the proteins in each fraction were functionally classified further. Interestingly, lycorine treatment decreased the levels of the nuclear pore complex protein 93 (Nup93, E2RSV7), which is associated with nuclear–cytoplasmic transport. In addition, Western blot experiments confirmed that the expression of Nup93 was significantly downregulated in lycorine treatment but induced after viral infection. Our results may provide new insights into how lycorine may trap vRNPs in the nucleus and suggest new potential therapeutic targets for influenza virus.
Collapse
Affiliation(s)
- Li Yang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China.,College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jia Hao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao Li Zhang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Guang Jie Lao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guan Ming Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lei Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Yao Lan Li
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Wen Cai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Jun He
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China.,Institute of Laboratory Animal Science, Jinan University, Guangzhou, China
| |
Collapse
|
17
|
Liu S, Pan P, Fan H, Li H, Wang W, Zhang Y. Photocatalytic C-H silylation of heteroarenes by using trialkylhydrosilanes. Chem Sci 2019; 10:3817-3825. [PMID: 31015923 PMCID: PMC6457191 DOI: 10.1039/c9sc00046a] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
The efficient and selective C-H silylation of heteroarenes, especially the pharmaceutically relevant electron-deficient heteroarenes, represents a great challenge in organic synthesis. Herein we wish to report a distinctive visible light-promoted photocatalytic C-H silylation approach that enables the direct coupling of trialkylhydrosilanes with both electron-deficient and -rich heteroarenes as well as with cyano-substituted arenes in moderate to high yields and with good regioselectivity. The protocol features operational simplicity, mild reaction conditions, and the use of safe and readily available Na2S2O8, bis(trimethylsilyl) peroxide (BTMSPO) or iPr3SiSH as the radical initiators. Notably, the challenging bulky and inert trialkylhydrosilanes, such as (t-butyldimethyl)silane ( t BuMe2SiH) and (triisopropyl)silane (iPr3SiH), work smoothly with the protocol. Moreover, despite the higher stability of t BuMe2Si silylation products, our studies revealed their great reactivity and versatility in diverse C-Si-based chemical transformations, providing an operationally simple, low-cost, and environmentally benign synthetic technology for molecule construction and elaboration.
Collapse
Affiliation(s)
- Shihui Liu
- State Key Laboratory of Bioengineering Reactor , Shanghai Key Laboratory of New Drug Design and School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P. R. China . ;
| | - Peng Pan
- State Key Laboratory of Bioengineering Reactor , Shanghai Key Laboratory of New Drug Design and School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P. R. China . ;
| | - Huaqiang Fan
- State Key Laboratory of Bioengineering Reactor , Shanghai Key Laboratory of New Drug Design and School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P. R. China . ;
| | - Hao Li
- State Key Laboratory of Bioengineering Reactor , Shanghai Key Laboratory of New Drug Design and School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P. R. China . ;
| | - Wei Wang
- State Key Laboratory of Bioengineering Reactor , Shanghai Key Laboratory of New Drug Design and School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P. R. China . ;
- Department of Pharmacology and Toxicology , BIO5 Institute , University of Arizona , Tucson , Arizona 85721-0207 , USA
| | - Yongqiang Zhang
- State Key Laboratory of Bioengineering Reactor , Shanghai Key Laboratory of New Drug Design and School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P. R. China . ;
| |
Collapse
|
18
|
Hu Y, Hau RK, Wang Y, Tuohy P, Zhang Y, Xu S, Ma C, Wang J. Structure-Property Relationship Studies of Influenza A Virus AM2-S31N Proton Channel Blockers. ACS Med Chem Lett 2018; 9:1111-1116. [PMID: 30429954 DOI: 10.1021/acsmedchemlett.8b00336] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
Majority of current circulating influenza A viruses carry the S31N mutation in their M2 genes, rendering AM2-S31N as a high profile antiviral drug target. With our continuous interest in developing AM2-S31N channel blockers as novel antivirals targeting both oseltamivir-sensitive and -resistant influenza A viruses, we report herein the structure-property relationship studies of AM2-S31N inhibitors. The goal was to identify lead compounds with improved microsomal stability and membrane permeability. Two lead compounds, 10d and 10e, were found to have high mouse and human liver microsomal stability (T 1/2 > 145 min) and membrane permeability (>200 nm/s). Both compounds also inhibit both currently circulating oseltamivir-sensitive and -resistant human influenza A viruses (H1N1 and H3N2) with EC50 values ranging from 0.4 to 2.8 μM and a selectivity index of >100. We also showed for the first time that AM2-S31N channel blockers such as 10e inhibited influenza virus replication at both low and high multiply of infection (102-106 pfu/mL) and the inhibition was not cell-type dependent. Overall, these studies have identified two promising lead candidates for further development as antiviral drugs against drug-resistant influenza A viruses.
Collapse
Affiliation(s)
- Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Raymond Kin Hau
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yuanxiang Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Peter Tuohy
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yongtao Zhang
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Shuting Xu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
19
|
Zhao X, Li R, Zhou Y, Xiao M, Ma C, Yang Z, Zeng S, Du Q, Yang C, Jiang H, Hu Y, Wang K, Mok CKP, Sun P, Dong J, Cui W, Wang J, Tu Y, Yang Z, Hu W. Discovery of Highly Potent Pinanamine-Based Inhibitors against Amantadine- and Oseltamivir-Resistant Influenza A Viruses. J Med Chem 2018; 61:5187-5198. [PMID: 29799746 DOI: 10.1021/acs.jmedchem.8b00042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Influenza pandemic is a constant major threat to public health caused by influenza A viruses (IAVs). IAVs are subcategorized by the surface proteins hemagglutinin (HA) and neuraminidase (NA), in which they are both essential targets for drug discovery. While it is of great concern that NA inhibitor oseltamivir resistant strains are frequently identified from human or avian influenza virus, structural and functional characterization of influenza HA has raised hopes for new antiviral therapies. In this study, we explored a structure-activity relationship (SAR) of pinanamine-based antivirals and discovered a potent inhibitor M090 against amantadine-resistant viruses, including the 2009 H1N1 pandemic strains, and oseltamivir-resistant viruses. Mechanism of action studies, particularly hemolysis inhibition, indicated that M090 targets influenza HA and it occupied a highly conserved pocket of the HA2 domain and inhibited virus-mediated membrane fusion by "locking" the bending state of HA2 during the conformational rearrangement process. This work provides new binding sites within the HA protein and indicates that this pocket may be a promising target for broad-spectrum anti-influenza A drug design and development.
Collapse
Affiliation(s)
- Xin Zhao
- State Key Laboratory of Respiratory Disease, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , Guangzhou 510530 , P. R. China.,Department of Pharmacology and Toxicology, College of Pharmacy , The University of Arizona , Tucson , Arizona 85721 , United States
| | - Runfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China
| | - Yang Zhou
- Division of Theoretical Chemistry and Biology, School of Biotechnology , Royal Institute of Technology (KTH), AlbaNova University Center , Stockholm SE-100 44 , Sweden
| | - Mengjie Xiao
- State Key Laboratory of Respiratory Disease, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , Guangzhou 510530 , P. R. China
| | - Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy , The University of Arizona , Tucson , Arizona 85721 , United States.,BIO5 Institute , The University of Arizona , Tucson , Arizona 85721 , United States
| | - Zhongjin Yang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China
| | - Shaogao Zeng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , Guangzhou 510530 , P. R. China
| | - Qiuling Du
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China
| | - Chunguang Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China
| | - Haiming Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy , The University of Arizona , Tucson , Arizona 85721 , United States.,BIO5 Institute , The University of Arizona , Tucson , Arizona 85721 , United States
| | - Kefeng Wang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China
| | - Chris Ka Pun Mok
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China.,HKU-Pasteur Research Pole, School of Public Health, HKU Li Ka Shing Faculty of Medicine , The University of Hong Kong , 5 Sassoon Road , Pokfulam , Hong Kong
| | - Ping Sun
- State Key Laboratory of Respiratory Disease, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China
| | - Jianghong Dong
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , Guangzhou 510530 , P. R. China
| | - Wei Cui
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , Guangzhou 510530 , P. R. China
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy , The University of Arizona , Tucson , Arizona 85721 , United States.,BIO5 Institute , The University of Arizona , Tucson , Arizona 85721 , United States
| | - Yaoquan Tu
- Division of Theoretical Chemistry and Biology, School of Biotechnology , Royal Institute of Technology (KTH), AlbaNova University Center , Stockholm SE-100 44 , Sweden
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital , Guangzhou Medical University , Guangzhou 510120 , P. R. China
| | - Wenhui Hu
- State Key Laboratory of Respiratory Disease, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital , Guangzhou Medical University , Guangzhou 511436 , P. R. China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences , Guangzhou 510530 , P. R. China
| |
Collapse
|
20
|
Dandachi D, Rodriguez-Barradas MC. Viral pneumonia: etiologies and treatment. J Investig Med 2018; 66:957-965. [PMID: 29680828 DOI: 10.1136/jim-2018-000712] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2018] [Indexed: 12/16/2022]
Abstract
Viral pathogens are increasingly recognized as a cause of pneumonia, in immunocompetent patients and more commonly among immunocompromised. Viral pneumonia in adults could present as community-acquired pneumonia (CAP), ranging from mild disease to severe disease requiring hospital admission and mechanical ventilation. Moreover, the role of viruses in hospital-acquired pneumonia and ventilator-associated pneumonia as causative agents or as co-pathogens and the effect of virus detection on clinical outcome are being investigated.More than 20 viruses have been linked to CAP. Clinical presentation, laboratory findings, biomarkers, and radiographic patterns are not characteristic to specific viral etiology. Currently, laboratory confirmation is most commonly done by detection of viral nucleic acid by reverse transcription-PCR of respiratory secretions.Apart from the US Food and Drug Administration-approved medications for treatment of influenza pneumonia, the treatment of non-influenza respiratory viruses is limited. Moreover, the evidence supporting the use of available antivirals to treat immunocompromised patients is modest at best. With the widespread use of molecular diagnostics, an aging population, and advancement in cancer therapy, physicians will face a bigger challenge in managing viral respiratory tract infections. Emphasis on infection control measures to prevent the spread of respiratory viruses especially in healthcare settings is extremely important.
Collapse
Affiliation(s)
- Dima Dandachi
- Infectious Diseases Section, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Maria C Rodriguez-Barradas
- Infectious Diseases Section, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA.,Infectious Diseases Section (MS 111G), Michael E. DeBakey VAMC, Houston, Texas, USA
| |
Collapse
|
21
|
Zhang J, Hu Y, Foley C, Wang Y, Musharrafieh R, Xu S, Zhang Y, Ma C, Hulme C, Wang J. Exploring Ugi-Azide Four-Component Reaction Products for Broad-Spectrum Influenza Antivirals with a High Genetic Barrier to Drug Resistance. Sci Rep 2018; 8:4653. [PMID: 29545578 PMCID: PMC5854701 DOI: 10.1038/s41598-018-22875-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/02/2018] [Indexed: 01/02/2023] Open
Abstract
Influenza viruses are respiratory pathogens that are responsible for seasonal influenza and sporadic influenza pandemic. The therapeutic efficacy of current influenza vaccines and small molecule antiviral drugs is limited due to the emergence of multidrug-resistant influenza viruses. In response to the urgent need for the next generation of influenza antivirals, we utilized a fast-track drug discovery platform by exploring multi-component reaction products for antiviral drug candidates. Specifically, molecular docking was applied to screen a small molecule library derived from the Ugi-azide four-component reaction methodology for inhibitors that target the influenza polymerase PAC-PB1N interactions. One hit compound 5 was confirmed to inhibit PAC-PB1N interactions in an ELISA assay and had potent antiviral activity in an antiviral plaque assay. Subsequent structure-activity relationship studies led to the discovery of compound 12a, which had broad-spectrum antiviral activity and a higher in vitro genetic barrier to drug resistance than oseltamivir. Overall, the discovery of compound 12a as a broad-spectrum influenza antiviral with a high in vitro genetic barrier to drug resistance is significant, as it offers a second line of defense to combat the next influenza epidemics and pandemics if vaccines and oseltamivir fail to confine the disease outbreak.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Christopher Foley
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Yuanxiang Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Rami Musharrafieh
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Shuting Xu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Yongtao Zhang
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Chunlong Ma
- BIO5 Institute, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Christopher Hulme
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States.
- BIO5 Institute, The University of Arizona, Tucson, Arizona, 85721, United States.
| |
Collapse
|
22
|
Wang Y, Hu Y, Xu S, Zhang Y, Musharrafieh R, Hau RK, Ma C, Wang J. In Vitro Pharmacokinetic Optimizations of AM2-S31N Channel Blockers Led to the Discovery of Slow-Binding Inhibitors with Potent Antiviral Activity against Drug-Resistant Influenza A Viruses. J Med Chem 2018; 61:1074-1085. [PMID: 29341607 DOI: 10.1021/acs.jmedchem.7b01536] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Influenza viruses are respiratory pathogens that are responsible for both seasonal influenza epidemics and occasional influenza pandemics. The narrow therapeutic window of oseltamivir, coupled with the emergence of drug resistance, calls for the next-generation of antivirals. With our continuous interest in developing AM2-S31N inhibitors as oral influenza antivirals, we report here the progress of optimizing the in vitro pharmacokinetic (PK) properties of AM2-S31N inhibitors. Several AM2-S31N inhibitors, including compound 10b, were discovered to have potent channel blockage, single to submicromolar antiviral activity, and favorable in vitro PK properties. The antiviral efficacy of compound 10b was also synergistic with oseltamivir carboxylate. Interestingly, binding kinetic studies (Kd, Kon, and Koff) revealed several AM2-S31N inhibitors that have similar Kd values but significantly different Kon and Koff values. Overall, this study identified a potent lead compound (10b) with improved in vitro PK properties that is suitable for the in vivo mouse model studies.
Collapse
Affiliation(s)
- Yuanxiang Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona , Tucson, Arizona 85721, United States
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona , Tucson, Arizona 85721, United States
| | - Shuting Xu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona , Tucson, Arizona 85721, United States
| | - Yongtao Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona , Tucson, Arizona 85721, United States
| | - Rami Musharrafieh
- Department of Chemistry and Biochemistry, The University of Arizona , Tucson, Arizona 85721, United States
| | - Raymond Kin Hau
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona , Tucson, Arizona 85721, United States
| | - Chunlong Ma
- BIO5 Institute, The University of Arizona , Tucson, Arizona 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona , Tucson, Arizona 85721, United States.,BIO5 Institute, The University of Arizona , Tucson, Arizona 85721, United States
| |
Collapse
|
23
|
Ramesh R, Reddy DS. Quest for Novel Chemical Entities through Incorporation of Silicon in Drug Scaffolds. J Med Chem 2017; 61:3779-3798. [DOI: 10.1021/acs.jmedchem.7b00718] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Remya Ramesh
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110 025, India
| | - D. Srinivasa Reddy
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110 025, India
| |
Collapse
|