1
|
Discovery of a Novel Respiratory Syncytial Virus Replication Inhibitor. Antimicrob Agents Chemother 2021; 65:AAC.02576-20. [PMID: 33782012 PMCID: PMC8316115 DOI: 10.1128/aac.02576-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/18/2021] [Indexed: 11/20/2022] Open
Abstract
A high-throughput screen of a Roche internal chemical library based on inhibition of the respiratory syncytial virus (RSV)-induced cytopathic effect (CPE) on HEp-2 cells was performed to identify RSV inhibitors. Over 2,000 hits were identified and confirmed to be efficacious against RSV infection in vitro Here, we report the discovery of a triazole-oxadiazole derivative, designated triazole-1, as an RSV replication inhibitor, and we characterize its mechanism of action. Triazole-1 inhibited the replication of both RSV A and B subtypes with 50% inhibitory concentration (IC50) values of approximately 1 μM, but it was not effective against other viruses, including influenza virus A, human enterovirus 71 (EV71), and vaccinia virus. Triazole-1 was shown to inhibit RSV replication when added at up to 8 h after viral entry, suggesting that it inhibits RSV after viral entry. In a minigenome reporter assay in which RSV transcription regulatory sequences flanking a luciferase gene were cotransfected with RSV N/P/L/M2-1 genes into HEp-2 cells, triazole-1 demonstrated specific and dose-dependent RSV transcription inhibitory effects. Consistent with these findings, deep sequencing of the genomes of triazole-1-resistant mutants revealed a single point mutation (A to G) at nucleotide 13546 of the RSV genome, leading to a T-to-A change at amino acid position 1684 of the L protein, which is the RSV RNA polymerase for both viral transcription and replication. The effect of triazole-1 on minigenome transcription, which was mediated by the L protein containing the T1684A mutation, was significantly reduced, suggesting that the T1684A mutation alone conferred viral resistance to triazole-1.
Collapse
|
2
|
Zheng X, Gao L, Wang L, Liang C, Wang B, Liu Y, Feng S, Zhang B, Zhou M, Yu X, Xiang K, Chen L, Guo T, Shen HC, Zou G, Wu JZ, Yun H. Discovery of Ziresovir as a Potent, Selective, and Orally Bioavailable Respiratory Syncytial Virus Fusion Protein Inhibitor. J Med Chem 2019; 62:6003-6014. [PMID: 31194544 DOI: 10.1021/acs.jmedchem.9b00654] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ziresovir (RO-0529, AK0529) is reported here for the first time as a promising respiratory syncytial virus (RSV) fusion (F) protein inhibitor that currently is in phase 2 clinical trials. This article describes the process of RO-0529 as a potent, selective, and orally bioavailable RSV F protein inhibitor and highlights the in vitro and in vivo anti-RSV activities and pharmacokinetics in animal species. RO-0529 demonstrates single-digit nM EC50 potency against laboratory strains, as well as clinical isolates of RSV in cellular assays, and more than one log viral load reduction in BALB/c mouse model of RSV viral infection. RO-0529 was proven to be a specific RSV F protein inhibitor by identification of drug resistant mutations of D486N, D489V, and D489Y in RSV F protein and the inhibition of RSV F protein-induced cell-cell fusion in cellular assays.
Collapse
Affiliation(s)
- Xiufang Zheng
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Lu Gao
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Lisha Wang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Chungen Liang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Baoxia Wang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Yongfu Liu
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Song Feng
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Bo Zhang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Mingwei Zhou
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Xin Yu
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Kunlun Xiang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Li Chen
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Tao Guo
- International Discovery Service Unit, Research Service Division , WuXi AppTec (Shanghai) Co., Ltd. , Lane 31, Yiwei Road, Waigaoqiao , Shanghai , 200131 , China
| | - Hong C Shen
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Gang Zou
- Ark Biosciences Inc. , 780 Cailun Road, Suite 701, ZhangJiang Hitech Park, Pudong , Shanghai 201203 , China
| | - Jim Zhen Wu
- Ark Biosciences Inc. , 780 Cailun Road, Suite 701, ZhangJiang Hitech Park, Pudong , Shanghai 201203 , China
| | - Hongying Yun
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| |
Collapse
|
3
|
Zheng X, Liang C, Wang L, Miao K, Wang B, Zhang W, Chen D, Wu G, Zhu W, Guo L, Feng S, Gao L, Shen HC, Yun H. Discovery of (aza)indole derivatives as novel respiratory syncytial virus fusion inhibitors. MEDCHEMCOMM 2019; 10:970-973. [PMID: 31303995 DOI: 10.1039/c9md00178f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/30/2019] [Indexed: 12/26/2022]
Abstract
A new class of indole derivatives (3) have been identified as potent RSV fusion inhibitors. SAR exploration revealed that 5-Cl and the sulfonyl side chain of the indole scaffold are crucial for anti-RSV activity. Further optimization led to the discovery of a cyclic sulfone (8i) with 2 nM anti-RSV activity and a much improved PK profile compared to the non-cyclic sulfone counterpart.
Collapse
Affiliation(s)
- Xiufang Zheng
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Chungen Liang
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Lisha Wang
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Kun Miao
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Baoxia Wang
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Weixing Zhang
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Dongdong Chen
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Guolong Wu
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Wei Zhu
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Lei Guo
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Song Feng
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Lu Gao
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Hong C Shen
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| | - Hongying Yun
- Roche Innovation Center Shanghai , Building 5, Lane 720, Cai Lun Road , Shanghai 201203 , China .
| |
Collapse
|
4
|
Zheng X, Liang C, Wang L, Wang B, Liu Y, Feng S, Wu JZ, Gao L, Feng L, Chen L, Guo T, Shen HC, Yun H. Discovery of Benzoazepinequinoline (BAQ) Derivatives as Novel, Potent, Orally Bioavailable Respiratory Syncytial Virus Fusion Inhibitors. J Med Chem 2018; 61:10228-10241. [PMID: 30339388 DOI: 10.1021/acs.jmedchem.8b01394] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A novel benzoazepinequnoline (BAQ) series was discovered as RSV fusion inhibitors. BAQ series originated from compound 2, a hit from similarity-based virtual screening. In SAR exploration, benzoazepine allowed modifications in the head moiety. Benzylic sulfonyl on benzoazepine and 6-Me on quinoline were crucial for good anti-RSV activity. Although the basic amine in the head portion was crucial for anti-RSV activity, the attenuated basicity was required to reduce Vss. Introducing oxetane to the head portion led to discovery of compound 1, which demonstrated single-digit nM anti-RSV activity against different RSV strains, reasonable oral exposure in plasma, and 78-fold higher exposure in lung. Compound 1 also displayed 1 log viral reduction in a female BALB/c mice RSV model by b.i.d. oral dosing at 12.5 mg/kg. A single resistant mutant at L138F in fusion protein proved compound 1 to be a RSV fusion inhibitor.
Collapse
Affiliation(s)
- Xiufang Zheng
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Chungen Liang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Lisha Wang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Baoxia Wang
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Yongfu Liu
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Song Feng
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Jim Zhen Wu
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Lu Gao
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Lichun Feng
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Li Chen
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Tao Guo
- International Discovery Service Unit, Research Service Division , WuXi AppTec (Shanghai) Co., Ltd. , Lane 31, Yiwei Road , Waigaoqiao, Shanghai 200131 China
| | - Hong C Shen
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| | - Hongying Yun
- Roche Pharma Research and Early Development , Roche Innovation Center Shanghai , Building 5, 720 Cailun Road , Shanghai 201203 , China
| |
Collapse
|
5
|
Salem MA, Helel MH, Ammar YA, El-Gaby MSA, Thabet HK, Gouda MA. Diphenic acid derivatives: Synthesis, reactions, and applications. SYNTHETIC COMMUN 2017. [DOI: 10.1080/00397911.2017.1298805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- M. A. Salem
- Department of Chemistry, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
- Department of Chemistry, Faculty of Arts and Science, King Khalid University, Mohail Assir, KSA
| | - M. H. Helel
- Department of Chemistry, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
- Department of Chemistry, Faculty of Arts and Science, Northern Border University, Rafha, KSA
| | - Y. A. Ammar
- Department of Chemistry, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| | - M. S. A. El-Gaby
- Department of Chemistry, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - H. Kh. Thabet
- Department of Chemistry, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
- Department of Chemistry, Faculty of Arts and Science, Northern Border University, Rafha, KSA
| | - M. A. Gouda
- Department of Chemistry, Faculty of Arts and Science, Taibah University, Ulla, KSA
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
6
|
Zheng X, Wang L, Wang B, Miao K, Xiang K, Feng S, Gao L, Shen HC, Yun H. Discovery of Piperazinylquinoline Derivatives as Novel Respiratory Syncytial Virus Fusion Inhibitors. ACS Med Chem Lett 2016; 7:558-62. [PMID: 27326326 PMCID: PMC4904258 DOI: 10.1021/acsmedchemlett.5b00234] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 04/20/2016] [Indexed: 12/26/2022] Open
Abstract
A novel series of piperazinylquinoline derivatives were discovered as respiratory syncytial virus (RSV) fusion inhibitors by the ligand-based screening approach. Among 3,000 hits, 1-amino-3-[[2-(4-phenyl-1-piperidyl)-4-quinolyl]amino]propan-2-ol (7) was proven to be active against the RSV long (A) strain. The anti-RSV activity was improved by converting piperidine to benzylcarbonyl substituted piperazine. The basic side chain was also found to be crucial for anti-RSV activity. The selected analogues, 45 and 50, demonstrated anti-RSV activities up to EC50 = 0.028 μM and 0.033 μM, respectively. A direct anti-RSV effect was confirmed by a plaque reduction assay and a fusion inhibition assay. Both 45 and 50 showed promising DMPK properties with good oral bioavailability, and could potentially lead to novel therapeutic agents targeting the RSV fusion process.
Collapse
Affiliation(s)
- Xiufang Zheng
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Lisha Wang
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Baoxia Wang
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Kun Miao
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Kunlun Xiang
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Song Feng
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Lu Gao
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Hong C. Shen
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| | - Hongying Yun
- Roche Innovation Center Shanghai, Building 5, Lane 720, Cai Lun Road, Shanghai 201203, China
| |
Collapse
|
7
|
Wang G, Deval J, Hong J, Dyatkina N, Prhavc M, Taylor J, Fung A, Jin Z, Stevens SK, Serebryany V, Liu J, Zhang Q, Tam Y, Chanda SM, Smith DB, Symons JA, Blatt LM, Beigelman L. Discovery of 4'-chloromethyl-2'-deoxy-3',5'-di-O-isobutyryl-2'-fluorocytidine (ALS-8176), a first-in-class RSV polymerase inhibitor for treatment of human respiratory syncytial virus infection. J Med Chem 2015; 58:1862-78. [PMID: 25667954 DOI: 10.1021/jm5017279] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading pathogen of childhood and is associated with significant morbidity and mortality. To date, ribavirin is the only approved small molecule drug, which has limited use. The only other RSV drug is palivizumab, a monoclonal antibody, which is used for RSV prophylaxis. Clearly, there is an urgent need for small molecule RSV drugs. This article reports the design, synthesis, anti-RSV activity, metabolism, and pharmacokinetics of a series of 4'-substituted cytidine nucleosides. Among tested compounds 4'-chloromethyl-2'-deoxy-2'-fluorocytidine (2c) exhibited the most promising activity in the RSV replicon assay with an EC50 of 0.15 μM. The 5'-triphosphate of 2c (2c-TP) inhibited RSV polymerase with an IC50 of 0.02 μM without appreciable inhibition of human DNA and RNA polymerases at 100 μM. ALS-8176 (71), the 3',5'-di-O-isobutyryl prodrug of 2c, demonstrated good oral bioavailability and a high level of 2c-TP in vivo. Compound 71 is a first-in-class nucleoside RSV polymerase inhibitor that demonstrated excellent anti-RSV efficacy and safety in a phase 2 clinical RSV challenge study.
Collapse
Affiliation(s)
- Guangyi Wang
- Alios BioPharma, Inc. , 260 East Grand Avenue, South San Francisco, California 94080, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Douglas JL. In search of a small-molecule inhibitor for respiratory syncytial virus. Expert Rev Anti Infect Ther 2014; 2:625-39. [PMID: 15482225 DOI: 10.1586/14787210.2.4.625] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Respiratory syncytial virus has been an ongoing health problem for 50 years. Hospitalization rates due to virus-induced respiratory illness continue to be substantial for infants, small children, the elderly and the immunocompromised. The only currently available treatments are a broad-spectrum antiviral and two immunoprophylactic antibodies, all of which are reserved for high-risk patients. The combination of this limited therapeutic repertoire and the lack of a vaccine clearly demonstrates the need to continue the search for more efficacious and safe agents against respiratory syncytial virus. The following is a review on the current progress of that search.
Collapse
|
9
|
Sun Z, Pan Y, Jiang S, Lu L. Respiratory syncytial virus entry inhibitors targeting the F protein. Viruses 2013; 5:211-25. [PMID: 23325327 PMCID: PMC3564118 DOI: 10.3390/v5010211] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 12/17/2012] [Accepted: 01/11/2013] [Indexed: 11/16/2022] Open
Abstract
Human respiratory syncytial virus (RSV) is the main viral cause of respiratory tract infection in infants as well as some elderly and high-risk adults with chronic pulmonary disease and the severely immunocompromised. So far, no specific anti-RSV therapeutics or effective anti-RSV vaccines have been reported. Only one humanized monoclonal antibody, Palivizumab, has been approved for use in high-risk infants to prevent RSV infection. Ribavirin is the only drug licensed for therapy of RSV infection, but its clinical use is limited by its nonspecific anti-RSV activity, toxic effect, and relatively high cost. Therefore, development of novel effective anti-RSV therapeutics is urgently needed. The RSV envelope glycoprotein F plays an important role in RSV fusion with, and entry into, the host cell and, consequently, serves as an attractive target for developing RSV entry inhibitors. This article reviews advances made in studies of the structure and function of the F protein and the development of RSV entry inhibitors targeting it.
Collapse
Affiliation(s)
- Zhiwu Sun
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China; E-Mails: (Z.S.); (S.J.)
| | - Yanbin Pan
- Aris (Nantong) Pharmaceuticals Co. Ltd., Nantong Economic and Technological Area, Jiangsu Province 226006, China; E-Mail:
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China; E-Mails: (Z.S.); (S.J.)
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China; E-Mails: (Z.S.); (S.J.)
| |
Collapse
|
10
|
Costello HM, Ray WC, Chaiwatpongsakorn S, Peeples ME. Targeting RSV with vaccines and small molecule drugs. Infect Disord Drug Targets 2012; 12:110-28. [PMID: 22335496 DOI: 10.2174/187152612800100143] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 01/01/2012] [Indexed: 12/21/2022]
Abstract
Respiratory syncytial virus (RSV) is the most significant cause of pediatric respiratory infections. Palivizumab (Synagis®), a humanized monoclonal antibody, has been used successfully for a number of years to prevent severe RSV disease in at-risk infants. However, despite intense efforts, there is no approved vaccine or small molecule drug for RSV. As an enveloped virus, RSV must fuse its envelope with the host cell membrane, which is accomplished through the actions of the fusion (F) glycoprotein, with attachment help from the G glycoprotein. Because of their integral role in initiation of infection and their accessibility outside the lipid bilayer, these proteins have been popular targets in the discovery and development of antiviral compounds and vaccines against RSV. This review examines advances in the development of antiviral compounds and vaccine candidates.
Collapse
Affiliation(s)
- Heather M Costello
- Center for Vaccines & Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | | | | | | |
Collapse
|
11
|
Moore BP, Chung DH, Matharu DS, Golden JE, Maddox C, Rasmussen L, Noah JW, Sosa MI, Ananthan S, Tower NA, White EL, Jia F, Prisinzano TE, Aubé J, Jonsson CB, Severson WE. (S)-N-(2,5-Dimethylphenyl)-1-(quinoline-8-ylsulfonyl)pyrrolidine-2-carboxamide as a small molecule inhibitor probe for the study of respiratory syncytial virus infection. J Med Chem 2012; 55:8582-7. [PMID: 23043370 DOI: 10.1021/jm300612z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A high-throughput, cell-based screen was used to identify chemotypes as inhibitors for human respiratory syncytial virus (hRSV). Optimization of a sulfonylpyrrolidine scaffold resulted in compound 5o that inhibited a virus-induced cytopathic effect in the entry stage of infection (EC₅₀ = 2.3 ± 0.8 μM) with marginal cytotoxicity (CC₅₀ = 30.9 ± 1.1 μM) and reduced viral titer by 100-fold. Compared to ribavirin, sulfonylpyrrolidine 5o demonstrated an improved in vitro potency and selectivity index.
Collapse
Affiliation(s)
- Blake P Moore
- Southern Research Specialized Biocontainment Screening Center, Southern Research Institute, Birmingham, Alabama 35205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hao M, Li Y, Wang Y, Zhang S. A classification study of respiratory Syncytial Virus (RSV) inhibitors by variable selection with random forest. Int J Mol Sci 2011; 12:1259-80. [PMID: 21541057 PMCID: PMC3083704 DOI: 10.3390/ijms12021259] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 02/10/2011] [Accepted: 02/11/2011] [Indexed: 12/29/2022] Open
Abstract
Experimental pEC50s for 216 selective respiratory syncytial virus (RSV) inhibitors are used to develop classification models as a potential screening tool for a large library of target compounds. Variable selection algorithm coupled with random forests (VS-RF) is used to extract the physicochemical features most relevant to the RSV inhibition. Based on the selected small set of descriptors, four other widely used approaches, i.e., support vector machine (SVM), Gaussian process (GP), linear discriminant analysis (LDA) and k nearest neighbors (kNN) routines are also employed and compared with the VS-RF method in terms of several of rigorous evaluation criteria. The obtained results indicate that the VS-RF model is a powerful tool for classification of RSV inhibitors, producing the highest overall accuracy of 94.34% for the external prediction set, which significantly outperforms the other four methods with the average accuracy of 80.66%. The proposed model with excellent prediction capacity from internal to external quality should be important for screening and optimization of potential RSV inhibitors prior to chemical synthesis in drug development.
Collapse
Affiliation(s)
- Ming Hao
- School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116012, China; E-Mails: (M.H.); (S.Z.)
| | - Yan Li
- School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116012, China; E-Mails: (M.H.); (S.Z.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-411-84986062; Fax: +86-411-84986063
| | - Yonghua Wang
- Center of Bioinformatics, Northwest A&F University, Yangling, Shaanxi 712100, China; E-Mail:
| | - Shuwei Zhang
- School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116012, China; E-Mails: (M.H.); (S.Z.)
| |
Collapse
|
13
|
Gordo S, Giralt E. Knitting and untying the protein network: modulation of protein ensembles as a therapeutic strategy. Protein Sci 2009; 18:481-93. [PMID: 19241367 DOI: 10.1002/pro.43] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proteins constitute the working machinery and structural support of all organisms. In performing a given function, they must adopt highly specific structures that can change with their level of activity, often through the direct or indirect action of other proteins. Indeed, proteins typically function within an ensemble, rather than individually. Hence, they must be sufficiently flexible to interact with each other and execute diverse tasks. The discovery that errors within these groups can ultimately cause disease has led to a paradigm shift in drug discovery, from an emphasis on single protein targets to a holistic approach whereby entire ensembles are targeted.
Collapse
Affiliation(s)
- Susana Gordo
- Institute for Research in Biomedicine, Parc Científic de Barcelona, Barcelona, Spain
| | | |
Collapse
|
14
|
Huber M, Olson WC, Trkola A. Antibodies for HIV treatment and prevention: window of opportunity? Curr Top Microbiol Immunol 2007; 317:39-66. [PMID: 17990789 DOI: 10.1007/978-3-540-72146-8_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Monoclonal antibodies are routinely used as therapeutics in a number of disease settings and have thus also been explored as potential treatment for human immunodeficiency virus (HIV)-1 infection. Antibodies targeting viral antigens, and those directed to the cellular receptors, have been considered for use in prevention and therapy. For virus-targeted antibodies, attention has focused primarily on their neutralizing activity, but such antibodies also have the potential to exert antiviral effects via effector functions, such as antibody-dependent cellular cytotoxicity (ADCC), opsonization, or complement activation. Anti-cell antibodies act through occlusion or down-modulation of the viral receptors with notable impact in vivo, as recent trials have shown. This review summarizes the diverse specificities and modes of action of therapeutic antibodies against HIV-1 infection. Successes, challenges, and future opportunities of harnessing antibodies for therapy of HIV-1 infection are discussed.
Collapse
Affiliation(s)
- M Huber
- Division of Infectious Diseases, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | | | | |
Collapse
|
15
|
Yin H, Hamilton AD. Strategies for targeting protein-protein interactions with synthetic agents. Angew Chem Int Ed Engl 2006; 44:4130-63. [PMID: 15954154 DOI: 10.1002/anie.200461786] [Citation(s) in RCA: 380] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The development of small-molecule modulators of protein-protein interactions is a formidable goal, albeit one that possesses significant potential for the discovery of novel therapeutics. Despite the daunting challenges, a variety of examples exists for the inhibition of two large protein partners with low-molecular-weight ligands. This review discusses the strategies for targeting protein-protein interactions and the state of the art in the rational design of molecules that mimic the structures and functions of their natural targets.
Collapse
Affiliation(s)
- Hang Yin
- Yale University, New Haven, CT, USA
| | | |
Collapse
|
16
|
|
17
|
Steffensen MB, Hollink E, Kuschel F, Bauer M, Simanek EE. Dendrimers Based on [1,3,5]-Triazines. ACTA ACUST UNITED AC 2006; 44:3411-3433. [PMID: 19953202 DOI: 10.1002/pola.21333] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A comprehensive and chronological account of dendrimers based on [1,3,5]-triazines is provided. Synthetic strategies to install the triazine through cycloaddition, cyclotrimerization, and nucleophilic aromatic substitution of cyanuric chloride are discussed. Motivations and applications of these architectures are surveyed, including the preparation of supra-molecular assemblies in the solution and solid states and their use in medicines, advanced materials, and separations when anchored to solid supports.
Collapse
|
18
|
Yu KL, Wang XA, Civiello RL, Trehan AK, Pearce BC, Yin Z, Combrink KD, Gulgeze HB, Zhang Y, Kadow KF, Cianci CW, Clarke J, Genovesi EV, Medina I, Lamb L, Wyde PR, Krystal M, Meanwell NA. Respiratory syncytial virus fusion inhibitors. Part 3: Water-soluble benzimidazol-2-one derivatives with antiviral activity in vivo. Bioorg Med Chem Lett 2005; 16:1115-22. [PMID: 16368233 DOI: 10.1016/j.bmcl.2005.11.109] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Revised: 11/24/2005] [Accepted: 11/28/2005] [Indexed: 10/25/2022]
Abstract
The introduction of acidic and basic functionality into the side chains of respiratory syncytial virus (RSV) fusion inhibitors was examined in an effort to identify compounds suitable for evaluation in vivo in the cotton rat model of RSV infection following administration as a small particle aerosol. The acidic compounds 2r, 2u, 2v, 2w, 2z, and 2aj demonstrated potent antiviral activity in cell culture and exhibited efficacy in the cotton rat comparable to ribavirin. In a BALB/c mouse model, the oxadiazolone 2aj reduced virus titers following subcutaneous dosing, whilst the ester 2az and amide 2aab exhibited efficacy following oral administration. These results established the potential of this class of RSV fusion inhibitors to interfere with infection in vivo following topical or systemic administration.
Collapse
Affiliation(s)
- Kuo-Long Yu
- Department of Chemistry, The Bristol-Myers Squibb Pharmaceutical Research Institute, 5, Research Parkway, Wallingford, CT 06492, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yin H, Hamilton AD. Strategien zur Modulation von Protein-Protein-Wechselwirkungen mit synthetischen Substanzen. Angew Chem Int Ed Engl 2005. [DOI: 10.1002/ange.200461786] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
20
|
Nikitenko A, Raifeld Y, Mitsner B, Newman H. Pyrimidine containing RSV fusion inhibitors. Bioorg Med Chem Lett 2005; 15:427-30. [PMID: 15603966 DOI: 10.1016/j.bmcl.2004.10.056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Revised: 10/15/2004] [Accepted: 10/21/2004] [Indexed: 11/24/2022]
Abstract
The knowledge of SAR in a series of biphenyl anionic RSV inhibitors has been broadened by synthesis and testing of analogs with pyrimidine linkers. Generally, pyrimidine compounds were much harder to synthesize, and their anti-RSV activity was lower in comparison with triazine analogs.
Collapse
Affiliation(s)
- Antonia Nikitenko
- Chemical and Screening Sciences, Wyeth Research, 401 N. Middletown Rd, Pearl River, NY 10965, USA.
| | | | | | | |
Collapse
|
21
|
|
22
|
Preparation and surfactancy of branched copolymers from poly(oxyalkylene)-amine and trichlorotriazine coupling. J Appl Polym Sci 2005. [DOI: 10.1002/app.21273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Cianci C, Langley DR, Dischino DD, Sun Y, Yu KL, Stanley A, Roach J, Li Z, Dalterio R, Colonno R, Meanwell NA, Krystal M. Targeting a binding pocket within the trimer-of-hairpins: small-molecule inhibition of viral fusion. Proc Natl Acad Sci U S A 2004; 101:15046-51. [PMID: 15469910 PMCID: PMC523459 DOI: 10.1073/pnas.0406696101] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Indexed: 11/18/2022] Open
Abstract
Trimeric class I virus fusion proteins undergo a series of conformational rearrangements that leads to the association of C- and N-terminal heptad repeat domains in a "trimer-of-hairpins" structure, facilitating the apposition of viral and cellular membranes during fusion. This final fusion hairpin structure is sustained by protein-protein interactions, associations thought initially to be refractory to small-molecule inhibition because of the large surface area involved. By using a photoaffinity analog of a potent respiratory syncytial virus fusion inhibitor, we directly probed the interaction of the inhibitor with its fusion protein target. Studies have shown that these inhibitors bind within a hydrophobic cavity formed on the surface of the N-terminal heptad-repeat trimer. In the fusogenic state, this pocket is occupied by key amino acid residues from the C-terminal heptad repeat that stabilize the trimer-of-hairpins structure. The results indicate that a low-molecular-weight fusion inhibitor can interfere with the formation or consolidation of key structures within the hairpin moiety that are essential for membrane fusion. Because analogous cavities are present in many class I viruses, including HIV, these results demonstrate the feasibility of this approach as a strategy for drug discovery.
Collapse
Affiliation(s)
- Christopher Cianci
- Bristol-Myers Squibb Pharmaceutical Research Institute, Wallingford, CT 06492, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Yu KL, Zhang Y, Civiello RL, Trehan AK, Pearce BC, Yin Z, Combrink KD, Gulgeze HB, Wang XA, Kadow KF, Cianci CW, Krystal M, Meanwell NA. Respiratory syncytial virus inhibitors. Part 2: Benzimidazol-2-one derivatives. Bioorg Med Chem Lett 2004; 14:1133-7. [PMID: 14980651 DOI: 10.1016/j.bmcl.2003.12.072] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2003] [Revised: 12/14/2003] [Accepted: 12/18/2003] [Indexed: 11/30/2022]
Abstract
Structure-activity relationships for a series of benzimidazol-2-one-based inhibitors of respiratory syncytial virus are described. These studies focused on structural variation of the benzimidazol-2-one substituent, a vector inaccessible in a series of benzotriazole derivatives on which 2 is based, and revealed a broad tolerance for substituent size and functionality.
Collapse
Affiliation(s)
- Kuo-Long Yu
- Department of Chemistry, The Bristol-Myers Squibb Pharmaceutical Research Institute, 5 Research Parkway, Wallingford, CT 06492, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cianci C, Genovesi EV, Lamb L, Medina I, Yang Z, Zadjura L, Yang H, D'Arienzo C, Sin N, Yu KL, Combrink K, Li Z, Colonno R, Meanwell N, Clark J, Krystal M. Oral efficacy of a respiratory syncytial virus inhibitor in rodent models of infection. Antimicrob Agents Chemother 2004; 48:2448-54. [PMID: 15215093 PMCID: PMC434195 DOI: 10.1128/aac.48.7.2448-2454.2004] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BMS-433771 is a potent inhibitor of respiratory syncytial virus (RSV) replication in vitro. Mechanism of action studies have demonstrated that BMS-433771 halts virus entry through inhibition of F protein-mediated membrane fusion. BMS-433771 also exhibited in vivo efficacy following oral administration in a mouse model of RSV infection (C. Cianci, K. Y. Yu, K. Combrink, N. Sin, B. Pearce, A. Wang, R. Civiello, S. Voss, G. Luo, K. Kadow, E. Genovesi, B. Venables, H. Gulgeze, A. Trehan, J. James, L. Lamb, I. Medina, J. Roach, Z. Yang, L. Zadjura, R. Colonno, J. Clark, N. Meanwell, and M. Krystal, Antimicrob. Agents Chemother. 48:413-422, 2004). In this report, the in vivo efficacy of BMS-433771 against RSV was further examined in the BALB/c mouse and cotton rat host models of infection. By using the Long strain of RSV, prophylactic efficacy via oral dosing was observed in both animal models. A single oral dose, administered 1 h prior to intranasal RSV inoculation, was as effective against infection as a 4-day b.i.d. dosing regimen in which the first oral dose was given 1 h prior to virus inoculation. Results of dose titration experiments suggested that RSV infection was more sensitive to inhibition by BMS-433771 treatment in the BALB/c mouse host than in the cotton rat. This was reflected by the pharmacokinetic and pharmacodynamic analysis of the efficacy data, where the area under the concentration-time curve required to achieve 50% of the maximum response was approximately 7.5-fold less for mice than for cotton rats. Inhibition of RSV by BMS-433771 in the mouse is the result of F1-mediated inhibition, as shown by the fact that a virus selected for resistance to BMS-433771 in vitro and containing a single amino acid change in the F1 region was also refractory to treatment in the mouse host. BMS-433771 efficacy against RSV infection was also demonstrated for mice that were chemically immunosuppressed by cyclophosphamide treatment, indicating that compound inhibition of the virus did not require an active host immune response.
Collapse
Affiliation(s)
- Christopher Cianci
- The Bristol-Myers Squibb Pharmaceutical Research Institute, 5 Research Parkway, Wallingford, CT 06492, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yu KL, Zhang Y, Civiello RL, Kadow KF, Cianci C, Krystal M, Meanwell NA. Fundamental structure-activity relationships associated with a new structural class of respiratory syncytial virus inhibitor. Bioorg Med Chem Lett 2003; 13:2141-4. [PMID: 12798322 DOI: 10.1016/s0960-894x(03)00383-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Structure-activity relationships surrounding the dialkylamino side chain of a series of benzotriazole-derived inhibitors of respiratory syncytial virus fusion based on the screening lead 1a were examined. The results indicate that the topology of the side chain is important but the terminus element offers considerable latitude to modulate physical properties.
Collapse
Affiliation(s)
- Kuo Long Yu
- Department of Chemistry, The Bristol-Myers Squibb Pharmaceutical Research Institute, 5, Research Parkway, 06492, Wallingford, CT, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Weiss WJ, Murphy T, Lynch ME, Frye J, Buklan A, Gray B, Lenoy E, Mitelman S, O'Connell J, Quartuccio S, Huntley C. Inhalation efficacy of RFI-641 in an African green monkey model of RSV infection. J Med Primatol 2003; 32:82-8. [PMID: 12823630 DOI: 10.1034/j.1600-0684.2003.00014.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human respiratory syncytial virus (RSV) is a major cause of acute upper and lower respiratory tract infections. RFI-641 is a novel RSV fusion inhibitor with potent in vitro activity. In vivo efficacy of RFI was determined in an African green monkey model of RSV infection involving prophylactic and therapeutic administration by inhalation exposure. Inhalation was with an RFI-641 nebulizer reservoir concentration of 15 mg/ml for 15 minutes (short exposure) or 2 hours (long exposure). Efficacy and RFI-641 exposure was determined by collection of throat swabs, nasal washes and bronchial alveolar lavage (BAL) on selected days. The short-exposure group (15 minutes) exhibited no effect on the nasal, throat or BAL samples. The throat and nasal samples for the long-exposure group failed to show a consistent reduction in viral titers. RFI-641 2 hours exposure-treated monkeys showed a statistically significantly log reduction for BAL samples of 0.73-1.34 PFU/ml (P-value 0.003) over all the sampling days. Analysis indicates that the long-exposure group titer was lower than the control titer on day 7 and when averaged across days. The results of this study demonstrate the ability of RFI-641 to reduce the viral load of RSV after inhalation exposure in the primate model of respiratory infection.
Collapse
Affiliation(s)
- W J Weiss
- Wyeth Research, 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Razinkov V, Huntley C, Ellestad G, Krishnamurthy G. RSV entry inhibitors block F-protein mediated fusion with model membranes. Antiviral Res 2002; 55:189-200. [PMID: 12076763 DOI: 10.1016/s0166-3542(02)00050-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
RSV fusion is mediated by F-protein, a major viral surface glycoprotein. CL-309623, a specific inhibitor of RSV, interacts tightly with F-protein, which results in a hydrophobic environment at the binding site. The binding is selective for F-protein and does not occur with G-protein, a surface glycoprotein that facilitates the binding of RSV to target cells, or with lipid membranes at concentrations in the sub-millimolar range. Using an assay based on the relief of self-quenching of octadecyl rhodamine (R18) incorporated in the RSV envelope, we show that the virus fuses efficiently with large unilamellar vesicles containing cholesterol, in the absence of specific receptor analogs. Fusion of cp-52, a mutant virus lacking the G and SH surface glycoproteins, with vesicles is inhibited by CL-309623 and RFI-641 due to specific interactions of the inhibitor(s) with the fusion protein. Both virus-vesicle and virus-cell fusion are inhibited with equal potency. The formation of the binary complex of CL-309623 with F-protein in its native state, resulting in the inhibition of fusion and entry of virus, is a prerequisite for the observed anti-RSV activity in cell cultures.
Collapse
Affiliation(s)
- Vladimir Razinkov
- Department of Biological Chemistry, Wyeth Research, Pearl River, NY 10965, USA
| | | | | | | |
Collapse
|
29
|
Huntley CC, Weiss WJ, Gazumyan A, Buklan A, Feld B, Hu W, Jones TR, Murphy T, Nikitenko AA, O'Hara B, Prince G, Quartuccio S, Raifeld YE, Wyde P, O'Connell JF. RFI-641, a potent respiratory syncytial virus inhibitor. Antimicrob Agents Chemother 2002; 46:841-7. [PMID: 11850270 PMCID: PMC127488 DOI: 10.1128/aac.46.3.841-847.2002] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human respiratory syncytial virus (RSV), a paramyxovirus, is a major cause of acute upper and lower respiratory tract infections in infants, young children, and adults. RFI-641 is a novel anti-RSV agent with potent in vitro and in vivo activity. RFI-641 is active against both RSV type A and B strains. The viral specificity and the large therapeutic window of RFI-641 (>100-fold) indicate that the antiviral activity of the compound is not due to adverse effects on normal cells. The potent in vitro activity of RFI-641 can be translated to efficacy in vivo: RFI-641 is efficacious when administered prophylactically by the intranasal route in mice, cotton rats, and African green monkeys. RFI-641 is also efficacious when administered therapeutically (24 h postinfection) in the monkey model. Mechanism of action studies indicate that RFI-641 blocks viral F protein-mediated fusion and cell syncytium formation.
Collapse
Affiliation(s)
- Clayton C Huntley
- Department of Infectious Disease and Chemical Sciences, Wyeth-Ayerst Research, Pearl River, New York 10965, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Razinkov V, Gazumyan A, Nikitenko A, Ellestad G, Krishnamurthy G. RFI-641 inhibits entry of respiratory syncytial virus via interactions with fusion protein. CHEMISTRY & BIOLOGY 2001; 8:645-59. [PMID: 11451666 DOI: 10.1016/s1074-5521(01)00042-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND RFI-641, a small dendrimer-like compound, is a potent and selective inhibitor of respiratory syncytial virus (RSV), which is currently a clinical candidate for the treatment of upper and lower respiratory tract infections caused by RSV. RFI-641 inhibits RSV growth with an IC(50) value of 50 nM and prevents syncytia formation in tissue culture. RSV contains of three surface glycoproteins, a small hydrophobic (SH) protein of unknown function, and attachment (G) and fusion (F) proteins that enable binding and fusion of virus, respectively, with target cells. Because of their role in attachment and fusion, the G and F surface proteins are prominent targets for therapeutic intervention. RFI-641 was previously shown to bind purified preparations of RSV fusion protein. Based on this observation, in conjunction with the biological results, it was speculated that the fusion event might be the target of these inhibitors. RESULTS A fusion assay based upon the relief of self-quenching of octadecyl rhodamine R18 was used to determine effects of the inhibitors on binding and fusion of RSV. The results show that RFI-641 inhibits both RSV-cell binding and fusion events. The inhibition of RSV is mediated via binding to the fusion protein on the viral surface. A closely related analog, WAY-158830, which is much less active in the virus-infectivity assay does not inhibit binding and fusion of RSV with Vero cells. CONCLUSIONS RFI-641, an in vivo active RSV inhibitor, is shown to inhibit both binding and fusion of RSV with cells, events that are early committed steps in RSV entry and pathogenicity. The results described here demonstrate that a non-peptidic, small molecule can inhibit binding and fusion of enveloped virus specifically via interaction with the viral fusion protein.
Collapse
Affiliation(s)
- V Razinkov
- Department of Biological Chemistry, Wyeth-Ayerst Research, Pearl River, NY 10965, USA
| | | | | | | | | |
Collapse
|
31
|
Nikitenko AA, Raifeld YE, Wang TZ. The discovery of RFI-641 as a potent and selective inhibitor of the respiratory syncytial virus. Bioorg Med Chem Lett 2001; 11:1041-4. [PMID: 11327584 DOI: 10.1016/s0960-894x(01)00150-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The design and synthesis of a new potent and selective inhibitor of the respiratory syncytial virus are described. This compound, RFI-641, emerged from analysis of the structure-activity relationship in a series of biphenyl triazine anionic compounds possessing specific anti-RSV activity. The key synthetic step involves coupling of diaminobiphenyl 11 with two equivalents of chlorotriazine 10 under microwave conditions. RFI-641 inhibited RSV in vitro and in vivo models.
Collapse
Affiliation(s)
- A A Nikitenko
- Chemical Sciences, Wyeth-Ayerst Research, Pearl River, NY 10965, USA.
| | | | | |
Collapse
|
32
|
Abstract
Respiratory syncytial virus (RSV), the most common cause of lower respiratory tract disease in infants and young children, is a ubiquitous respiratory pathogen, infecting or reinfecting much of the population every year and causing severe, sometimes fatal disease in high-risk populations of infants and adults, particularly in developing countries. Spurred by the medical and economic burdens of RSV disease and enticed by the economic potential of therapeutic drugs, particularly in the absence to date of a safe and effective RSV vaccine, scientists in many industrial, academic and government laboratories have developed a wide variety of candidate RSV antiviral agents. Most of these have been screened thus far only in cell culture, a few in animal models. Aside from ribavirin, however, none has proven effective in therapeutic clinical trials and even ribavirin usage has declined precipitously in recent years due to concerns over efficacy, safety, ease of use and cost. All of the antiviral compounds discussed in this review were evaluated primarily for their ability to reduce viral load, with little or no attention paid to the role of host inflammation in the pathogenesis of RSV disease. Recent research has highlighted the prominent role of inflammatory mediators and an increasing number of reports suggest that a therapeutic strategy that combines antiviral and anti-inflammatory components will be the most effective way of treating RSV disease.
Collapse
Affiliation(s)
- G A Prince
- Virion Systems, Inc., 9610 Medical Center Drive, Suite 100, Rockville, Maryland 20850-3347, USA.
| |
Collapse
|
33
|
Respiratory syncytial virus: recent progress towards the discovery of effective prophylactic and therapeutic agents. Drug Discov Today 2000; 5:241-252. [PMID: 10825730 DOI: 10.1016/s1359-6446(00)01500-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Although respiratory syncytial virus (RSV) was discovered in 1955, the burden associated with this infectious agent on all population groups is only now beginning to be fully appreciated. The successful launch of the humanized monoclonal antibody Synagis (developed by MedImmune, Gaithersburg, MD, USA), as a prophylactic in September 1998 has helped to heighten awareness of the extent of mortality and morbidity associated with annual RSV epidemics. Small, drug-like molecules that would provide the clinician with effective and conveniently administered prophylactic and therapeutic agents for the prevention and treatment of RSV have not yet advanced into clinical studies. This review will summarize recent developments in the area of RSV drug discovery and development.
Collapse
|