1
|
Antisense Oligonucleotide-Based Therapy of Viral Infections. Pharmaceutics 2021; 13:pharmaceutics13122015. [PMID: 34959297 PMCID: PMC8707165 DOI: 10.3390/pharmaceutics13122015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid-based therapeutics have demonstrated their efficacy in the treatment of various diseases and vaccine development. Antisense oligonucleotide (ASO) technology exploits a single-strand short oligonucleotide to either cause target RNA degradation or sterically block the binding of cellular factors or machineries to the target RNA. Chemical modification or bioconjugation of ASOs can enhance both its pharmacokinetic and pharmacodynamic performance, and it enables customization for a specific clinical purpose. ASO-based therapies have been used for treatment of genetic disorders, cancer and viral infections. In particular, ASOs can be rapidly developed for newly emerging virus and their reemerging variants. This review discusses ASO modifications and delivery options as well as the design of antiviral ASOs. A better understanding of the viral life cycle and virus-host interactions as well as advances in oligonucleotide technology will benefit the development of ASO-based antiviral therapies.
Collapse
|
2
|
Balakrishnan A, Mun AB. Ribavirin inhibits Chandipura virus replication in Vero cells. J Med Virol 2020; 92:2969-2975. [PMID: 32543712 DOI: 10.1002/jmv.26184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/10/2020] [Indexed: 01/04/2023]
Abstract
Chandipura virus (CHPV) is an emerging tropical pathogen in India. The virus has been reported to be associated with an acute encephalitis syndrome in young children with a case fatality rate of 55% to 75%. Clinical management with symptomatic treatment is the only option available to treat infected patients. No vaccines are available for prophylaxis. In light of the prophylactic limitations, antiviral therapy would play an important role in control of CHPV infection. In the present study, ribavirin (RBV), an antiviral drug widely accepted for human use and having an antiviral effect on many RNA and DNA viruses, was tested against the CHPV. A screening assay that scores for the virus-mediated plaque formation in the cultured Vero cells was used. RBV exhibited 50% inhibitory concentration (IC50 ) at 89.84 ± 1.8 µM. The drug was very effective when the cells were treated either within an hour postinfection or 4 to 6 hours before infection. The plaque morphology was different in RBV treated cells; the plaques were smaller in size as compared with the plaques in the virus infected cells. The study reports the antiviral activity of RBV against CHPV, and hence, suggests the possible utility of RBV in CHPV infected patients to mitigate the disease. A further clinical trial is needed before introducing the drug for human use against CHPV infection.
Collapse
Affiliation(s)
| | - Amol B Mun
- ICMR-National Institute of Research in Tribal Health, Jabalpur, Madhya Pradesh, India
| |
Collapse
|
3
|
Piyush R, Rajarshi K, Chatterjee A, Khan R, Ray S. Nucleic acid-based therapy for coronavirus disease 2019. Heliyon 2020; 6:e05007. [PMID: 32984620 PMCID: PMC7501848 DOI: 10.1016/j.heliyon.2020.e05007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/02/2020] [Accepted: 09/17/2020] [Indexed: 12/14/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19), the pandemic that originated in China has already spread into more than 190 countries, resulting in huge loss of human life and many more are at the stake of losing it; if not intervened with the best therapeutics to contain the disease. For that aspect, various scientific groups are continuously involved in the development of an effective line of treatment to control the novel coronavirus from spreading rapidly. Worldwide scientists are evaluating various biomolecules and synthetic inhibitors against COVID-19; where the nucleic acid-based molecules may be considered as potential drug candidates. These molecules have been proved potentially effective against SARS-CoV, which shares high sequence similarity with SARS-CoV-2. Recent advancements in nucleic acid-based therapeutics are helpful in targeted drug delivery, safely and effectively. The use of nucleic acid-based molecules also known to regulate the level of gene expression inside the target cells. This review mainly focuses on various nucleic acid-based biologically active molecules and their therapeutic potentials in developing vaccines for SARS-CoV-2.
Collapse
Affiliation(s)
- Ravikant Piyush
- School of Biotechnology, Madurai Kamaraj University, Madurai, Tamil Nadu 625021, India
| | - Keshav Rajarshi
- School of Community Science and Technology (SOCSAT) Indian Institute of Engineering Science and Technology (IIEST), Shibpur, Howrah, West Bengal 711103, India
| | - Aroni Chatterjee
- Indian Council of Medical Research (ICMR)-Virus Research Laboratory, NICED, Kolkata, India
| | - Rajni Khan
- Motihari College of Engineering, Bariyarpur, Motihari, NH 28A, Furshatpur, Motihari, Bihar 845401, India
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University Motihari, 845401, India
| |
Collapse
|
4
|
Asha K, Kumar P, Sanicas M, Meseko CA, Khanna M, Kumar B. Advancements in Nucleic Acid Based Therapeutics against Respiratory Viral Infections. J Clin Med 2018; 8:jcm8010006. [PMID: 30577479 PMCID: PMC6351902 DOI: 10.3390/jcm8010006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
Abstract
Several viruses cause pulmonary infections due to their shared tropism with cells of the respiratory tract. These respiratory problems due to viral infection become a public health concern due to rapid transmission through air/aerosols or via direct-indirect contact with infected persons. In addition, the cross-species transmission causes alterations to viral genetic makeup thereby increasing the risk of emergence of pathogens with new and more potent infectivity. With the introduction of effective nucleic acid-based technologies, post translational gene silencing (PTGS) is being increasingly used to silence viral gene targets and has shown promising approach towards management of many viral infections. Since several host factors are also utilized by these viruses during various stages of infection, silencing these host factors can also serve as promising therapeutic tool. Several nucleic acid-based technologies such as short interfering RNAs (siRNA), antisense oligonucleotides, aptamers, deoxyribozymes (DNAzymes), and ribozymes have been studied and used against management of respiratory viruses. These therapeutic nucleic acids can be efficiently delivered through the airways. Studies have also shown efficacy of gene therapy in clinical trials against respiratory syncytial virus (RSV) as well as models of respiratory diseases including severe acute respiratory syndrome (SARS), measles and influenza. In this review, we have summarized some of the recent advancements made in the area of nucleic acid based therapeutics and highlighted the emerging roles of nucleic acids in the management of some of the severe respiratory viral infections. We have also focused on the methods of their delivery and associated challenges.
Collapse
Affiliation(s)
- Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| | - Prashant Kumar
- Amity Institute of Virology and Immunology, Amity University, Noida 201303, India.
| | - Melvin Sanicas
- Sanofi Pasteur, Asia and JPAC Region, Singapore 257856, Singapore.
| | - Clement A Meseko
- Regional Centre for Animal Influenza, National Veterinary Research Institute, Vom 930010, Nigeria.
| | - Madhu Khanna
- Department of Respiratory Virology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110007, India.
| | - Binod Kumar
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| |
Collapse
|
5
|
Espinoza JA, Bueno SM, Riedel CA, Kalergis AM. Induction of protective effector immunity to prevent pathogenesis caused by the respiratory syncytial virus. Implications on therapy and vaccine design. Immunology 2014; 143:1-12. [PMID: 24801878 DOI: 10.1111/imm.12313] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 01/22/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) is the leading cause of respiratory illness in infants and young children around the globe. This pathogen, which was discovered in 1956, continues to cause a huge number of hospitalizations due to respiratory disease and it is considered a health and economic burden worldwide, especially in developing countries. The immune response elicited by hRSV infection leads to lung and systemic inflammation, which results in lung damage but is not efficient at preventing viral replication. Indeed, natural hRSV infection induces a poor immune memory that allows recurrent infections. Here, we review the most recent knowledge about the lifecycle of hRSV, the immune response elicited by this virus and the subsequent pathology induced in response to infection in the airways. Novel findings about the alterations that this virus causes in the central nervous system and potential therapies and vaccines designed to treat or prevent hRSV infection are discussed.
Collapse
Affiliation(s)
- Janyra A Espinoza
- Millennium Institute on Immunology, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | |
Collapse
|
6
|
Recent advances in diagnosis, prevention, and treatment of human respiratory syncytial virus. Adv Virol 2013; 2013:595768. [PMID: 24382964 PMCID: PMC3872095 DOI: 10.1155/2013/595768] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/30/2013] [Indexed: 12/25/2022] Open
Abstract
Human respiratory syncytial virus (RSV) is a common cause of respiratory infection in infants and the elderly, leading to significant morbidity and mortality. The interdisciplinary fields, especially biotechnology and nanotechnology, have facilitated the development of modern detection systems for RSV. Many anti-RSV compounds like fusion inhibitors and RNAi molecules have been successful in laboratory and clinical trials. But, currently, there are no effective drugs for RSV infection even after decades of research. Effective diagnosis can result in effective treatment, but the progress in both of these facets must be concurrent. The development in prevention and treatment measures for RSV is at appreciable pace, but the implementation into clinical practice still seems a challenge. This review attempts to present the promising diverse research approaches and advancements in the area of diagnosis, prevention, and treatment that contribute to RSV management.
Collapse
|
7
|
Abstract
Early detection of pediatric viruses is critical to effective intervention. A successful clinical tool must have a low detection limit, be simple to use and report results quickly. No current method meets all three of these criteria. In this report, we describe an approach that combines simple, rapid processing and label free detection. The method detects viral RNA using DNA hairpin structures covalently attached to a gold filament. In this design, the gold filament serves both to simplify processing and enable fluorescence detection. The approach was evaluated by assaying for the presence of respiratory syncytial virus (RSV) using the DNA hairpin probe 5' [C6Thiol]TTTTTTTTTTCGACGAAAAATGGGGCAAATACGTCG[CAL] 3' covalently attached to a 5 cm length of a 100 microm diameter gold-clad filament. This sequence was designed to target a portion of the gene end-intergenic gene start signals which is repeated multiple times within the negative-sense genome giving multiple targets for each strand of genomic viral RNA present. The filament functionalized with probes was immersed in a 200 microm capillary tube containing viral RNA, moved to subsequent capillary tubes for rinsing and then scanned for fluorescence. The response curve had a typical sigmoidal shape and plateaued at about 300 plaque forming units (PFU) of viral RNA in 20 microL. The lower limit of detection was determined to be 11.9 PFU. This lower limit of detection was approximately 200 times better than a standard comparison ELISA. The simplicity of the core assay makes this approach attractive for further development as a viral detection platform in a clinical setting.
Collapse
Affiliation(s)
- Jonas W. Perez
- Vanderbilt University, Department of Chemistry, Station B 351822, Nashville, TN, 37235, USA. ; Fax: +1 615 343 1234; Tel: +1 615 322 2636
| | - Frederick R. Haselton
- Vanderbilt University, Department of Biomedical Engineering, Station B 351631, Nashville, TN, 37235, USA
| | - David W. Wright
- Vanderbilt University, Department of Chemistry, Station B 351822, Nashville, TN, 37235, USA. ; Fax: +1 615 343 1234; Tel: +1 615 322 2636
| |
Collapse
|
8
|
Li ZH, Li CM, Ling P, Shen FH, Chen SH, Liu CC, Yu CK, Chen SH. Ribavirin reduces mortality in enterovirus 71-infected mice by decreasing viral replication. J Infect Dis 2008; 197:854-7. [PMID: 18279075 PMCID: PMC7109938 DOI: 10.1086/527326] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Enterovirus 71 (EV71) causes fatal encephalitis in young children. However, there is no effective antiviral drug available for infected patients. Ribavirin is currently used for the treatment of several RNA virus infections clinically, so its anti-EV71 efficacy was evaluated. In vitro results showed that ribavirin effectively reduced the viral yields (with an IC50 of 65 μg/mL) and virus-induced cytopathic effect in human and mouse cell lines. In vivo results showed that ribavirin reduced the mortality, morbidity, and subsequent paralysis sequelae in infected mice by decreasing viral loads in tissues. Thus, ribavirin could be a potential anti-EV71 drug
Collapse
Affiliation(s)
- Zhao-Hong Li
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University,Taiwan, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Lai SH, Stein DA, Guerrero-Plata A, Liao SL, Ivanciuc T, Hong C, Iversen PL, Casola A, Garofalo RP. Inhibition of respiratory syncytial virus infections with morpholino oligomers in cell cultures and in mice. Mol Ther 2008; 16:1120-8. [PMID: 18443602 PMCID: PMC2782410 DOI: 10.1038/mt.2008.81] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infection in infants, young children, and high-risk adults. Currently, there is no vaccine to prevent RSV infection, and the available therapeutic agents are of limited utility. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) are a class of antisense agents that can enter cells readily and interfere with viral protein expression through steric blocking of complementary RNA. Two antisense PPMOs, designed to target sequence that includes the 5'-terminal region and translation start-site region of RSV L mRNA, were tested for anti-RSV activity in cultures of two human-airway cell lines. Both PPMOs showed minimal cytotoxicity and one of them, (AUG-2), reduced viral titers by >2.0 log(10). Intranasal (i.n.) treatment of BALB/c mice with AUG-2 PPMO before the RSV inoculation produced a reduction in viral titer of 1.2 log(10) in lung tissue at day 5 postinfection (p.i.), and attenuated pulmonary inflammation at day 7 postinfection. These data show that the AUG-2 PPMO possesses potent anti-RSV activity and is worthy of further investigation as a candidate for potential therapeutic application.
Collapse
Affiliation(s)
- Shen-Hao Lai
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Chang Gung Children’s Hospital, 5-7, Fu-Hsing Street, Kuei Shan Hsiang, Taoyuan Hsien, Taiwan, R.O.C., and Chang Gung University, 259, Wen Hwa 1 Rd., Taoyuan, Taiwan 33333
| | - David A. Stein
- AVI BioPharma Inc., 4575 SW Research Way, Corvallis OR, 97333, USA
| | - Antonieta Guerrero-Plata
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | - Sui-Ling Liao
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Chang Gung Children’s Hospital, 5-7, Fu-Hsing Street, Kuei Shan Hsiang, Taoyuan Hsien, Taiwan, R.O.C., and Chang Gung University, 259, Wen Hwa 1 Rd., Taoyuan, Taiwan 33333
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | - Chao Hong
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | | | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| | - Roberto P. Garofalo
- Department of Pediatrics, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA
| |
Collapse
|
10
|
Abstract
The final step in the maturation of paramyxoviruses, orthomyxoviruses and viruses of several other families, entails the budding of the viral nucleocapsid through the plasma membrane of the host cell. Many medically important viruses, such as influenza, parainfluenza, respiratory syncytial virus (RSV) and Ebola, can form filamentous particles when budding. Although filamentous virions have been previously studied, details of how viral filaments bud from the plasma membrane remain largely unknown. Using molecular beacon (MB)-fluorescent probes to image the viral genomic RNA (vRNA) of human RSV (hRSV) in live Vero cells, the dynamics of assembled viral filaments was observed to consist of three primary types of motion prior to egress from the plasma membrane: (i) filament projection and rotation, (ii) migration and (iii) non-directed motion. In addition, from information gained by imaging the 3D distribution of cellular vRNA, observing and characterizing vRNA dynamics, imaging vRNA/Myosin Va colocalization, and studying the effects of cytochalasin D (actin depolymerizing agent) exposure, a model for filamentous virion egress is presented.
Collapse
Affiliation(s)
| | - Gang Bao
- *To whom correspondence should be addressed. ;
| |
Collapse
|
11
|
Sidwell RW, Barnard DL. Respiratory syncytial virus infections: Recent prospects for control. Antiviral Res 2006; 71:379-90. [PMID: 16806515 DOI: 10.1016/j.antiviral.2006.05.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 05/18/2006] [Accepted: 05/22/2006] [Indexed: 10/24/2022]
Abstract
Respiratory syncytial virus (RSV) infections remain a significant public health problem throughout the world, although recently developed and clinically approved anti-RSV antibodies administered prophylactically to at-risk populations appear to have significantly affected the disease development. Much effort has been expended to develop effective anti-RSV therapies, using both in vitro assay systems and mouse, cotton rat, and primate models, with several products now in various stages of clinical study. Several products are also being considered for the treatment of clinical symptoms of RSV. In this review, updates on the status of the approved anti-RSV antibodies, ribavirin, and recent results of studies with potential new anti-RSV compounds are summarized and discussed.
Collapse
Affiliation(s)
- Robert W Sidwell
- Institute for Antiviral Research, Utah State University, 5600 Old Main Hill, Logan, UT 84322-5600, United States.
| | | |
Collapse
|
12
|
Abed Y, Boivin G. Treatment of respiratory virus infections. Antiviral Res 2006; 70:1-16. [PMID: 16513187 PMCID: PMC7114251 DOI: 10.1016/j.antiviral.2006.01.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Revised: 12/18/2005] [Accepted: 01/08/2006] [Indexed: 02/06/2023]
Abstract
Respiratory viral infections (RVIs) can be associated with a wide range of clinical manifestations ranging from self-limited upper respiratory tract infections to more devastating conditions, such as pneumonia. RVIs constitute the most frequent reason for medical consultations in the world and they have a considerable impact on quality of life and productivity. Therefore, the prevention and control of RVIs remain major clinical goals. Currently, there are approximately 200 known respiratory viruses that can be grouped into one family of DNA viruses (Adenoviridae) and four families of RNA viruses (Orthomyxoviridae, Paramyxoviridae, Picornaviridae and Coronaviridae). In this paper, we review the major respiratory viruses that cause disesases in humans, with an emphasis on current treatment options.
Collapse
Affiliation(s)
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHUQ-CHUL and Laval University, Québec City, Que., Canada
| |
Collapse
|
13
|
Abstract
Respiratory syncytial virus (RSV) continues as an emerging infectious disease not only among infants and children, but also for the immune-suppressed, hospitalised and the elderly. To date, ribavirin (Virazole, ICN Pharmaceuticals, Inc.) remains the only therapeutic agent approved for the treatment of RSV. However, its clinical benefits are small and occur only in a fraction of RSV-infected patients. The prophylactic administration of palivizumab (Synagis, MedImmune, Inc.) is problematic and costly and, therefore, only recommended for use in high-risk infants. Clearly, the need for an effective and safe drug remains high. This review discusses several different antisense approaches and compares them with traditional strategies, such as RSV-targeting antibodies and antivirals, as well as developments in vaccine research.
Collapse
Affiliation(s)
- Hagen Cramer
- Ridgeway Biosystems, Inc., 9500 Euclid Avenue, ND-50, Cleveland, OH 44195, USA.
| |
Collapse
|
14
|
Santangelo P, Nitin N, LaConte L, Woolums A, Bao G. Live-cell characterization and analysis of a clinical isolate of bovine respiratory syncytial virus, using molecular beacons. J Virol 2006; 80:682-8. [PMID: 16378971 PMCID: PMC1346841 DOI: 10.1128/jvi.80.2.682-688.2006] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Understanding viral pathogenesis is critical for prevention of outbreaks, development of antiviral drugs, and biodefense. Here, we utilize molecular beacons to directly detect the viral genome and characterize a clinical isolate of bovine respiratory syncytial virus (bRSV) in living cells. Molecular beacons are dual-labeled, hairpin oligonucleotide probes with a reporter fluorophore at one end and a quencher at the other; they are designed to fluoresce only when hybridizing to a complementary target. By imaging the fluorescence signal of molecular beacons, the spread of bRSV was monitored for 7 days with a signal-to-noise ratio of 50 to 200, and the measured time course of infection was quantified with a mathematical model for viral growth. We found that molecular beacon signal could be detected in single living cells infected with a viral titer of 2 x 10(3.6) 50% tissue culture infective doses/ml diluted 1,000 fold, demonstrating high detection sensitivity. Low background in uninfected cells and simultaneous staining of fixed cells with molecular beacons and antibodies showed high detection specificity. Furthermore, using confocal microscopy to image the viral genome in live, infected cells, we observed a connected, highly three-dimensional, amorphous inclusion body structure not seen in fixed cells. Taken together, the use of molecular beacons for active virus imaging provides a powerful tool for rapid viral infection detection, the characterization of RNA viruses, and the design of new antiviral drugs.
Collapse
Affiliation(s)
- Philip Santangelo
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr., Atlanta, GA 30332, USA
| | | | | | | | | |
Collapse
|
15
|
Abstract
Human respiratory syncytial virus (RSV) is the most common worldwide cause of lower respiratory tract infections (LRI) in infants less than 6 months of age. The prophylaxis against RSV infection by vaccination has been unsuccessful because of its adverse effects. As antiviral drug, ribavirin spray (aerosol) had been used clinically and reduces the amount of virus load, without reducing the necessity of symptomatic therapy and the duration of hospitalization. Therefore RSV LRI has been treated mainly symptomatically. Recently humanized anti-RSV F protein monoclonal antibody was developed and prescribed for prevention in high-risk infants such as premature ones and those with chronic lung and congenital heart diseases. It reduced the incidence of hospitalization significantly. It has been introduced in clinical use in Japan following to Western countries. On the other hand, a number of anti-RSV drugs have now been investigation; however, no valuable drugs for clinical use have been yet developed.
Collapse
Affiliation(s)
- Hiroyuki Tsutsumi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Japan.
| |
Collapse
|
16
|
|
17
|
Xu Z, Kuang M, Okicki JR, Cramer H, Chaudhary N. Potent inhibition of respiratory syncytial virus by combination treatment with 2-5A antisense and ribavirin. Antiviral Res 2004; 61:195-206. [PMID: 15168801 DOI: 10.1016/j.antiviral.2003.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2003] [Accepted: 10/24/2003] [Indexed: 11/26/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory diseases in infants, young children, and the elderly. Ribavirin, the only currently approved drug for the treatment of RSV infections in the U.S., requires high doses to be effective. Therefore, it has only a limited clinical efficacy in the treatment of RSV infections. It has been shown that a cellular ribonuclease, RNase L, can be recruited by 2'-5' linked tetra-adenylates (2-5A) attached to an antisense sequence complementary to the RSV genome to specifically cleave RSV genomic RNA. Here we confirm the antiviral activity of the lead 2-5A antisense compound, RBI034, by using several different viral assays. We demonstrate that RBI034 is more efficient than antisense lacking 2-5A or small interfering dsRNA (siRNA) in inhibiting RSV replication. Although the best antiviral activity of RBI034 was observed with co-treatment of RSV infection, it remained effective even when administered 24 h after the initiation of infection. Interestingly, the activity of RBI034 can be further enhanced by a combination treatment with ribavirin. At suboptimal concentrations, neither ribavirin nor RBI034 was effective in suppressing RSV replication. However, a combination of these two drugs at the same suboptimal concentrations showed a potent inhibitory activity. The potent reduction of RSV replication by combination treatment was also confirmed in primary human airway epithelial cells. Therefore, a combination therapy of the 2-5A antisense compound RBI034 and ribavirin might be a more effective therapeutic approach for treating RSV infections than ribavirin alone.
Collapse
Affiliation(s)
- Zan Xu
- Ridgeway Biosystems, Inc., 9500 Euclid Avenue, ND50, Cleveland, OH 44195, USA.
| | | | | | | | | |
Collapse
|
18
|
Easton AJ, Domachowske JB, Rosenberg HF. Animal pneumoviruses: molecular genetics and pathogenesis. Clin Microbiol Rev 2004; 17:390-412. [PMID: 15084507 PMCID: PMC387412 DOI: 10.1128/cmr.17.2.390-412.2004] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pneumoviruses are single-stranded, negative-sense, nonsegmented RNA viruses of the family Paramyxoviridae, subfamily Pneumovirinae, and include pathogens that infect humans (respiratory syncytial virus and human metapneumovirus), domestic mammals (bovine, ovine, and caprine respiratory syncytial viruses), rodents (pneumonia virus of mice), and birds (avian metapneumovirus). Among the topics considered in this review are recent studies focused on the roles of the individual virus-encoded components in promoting virus replication as well as in altering and evading innate antiviral host defenses. Advances in the molecular technology of pneumoviruses and the emergence of recombinant pneumoviruses that are leading to improved virus-based vaccine formulations are also discussed. Since pneumovirus infection in natural hosts is associated with a profound inflammatory response that persists despite adequate antiviral therapy, we also review the recent experimental treatment strategies that have focused on combined antiviral, anti-inflammatory, and immunomodulatory approaches.
Collapse
|
19
|
Leaman DW, Longano FJ, Okicki JR, Soike KF, Torrence PF, Silverman RH, Cramer H. Targeted therapy of respiratory syncytial virus in African green monkeys by intranasally administered 2-5A antisense. Virology 2002; 292:70-7. [PMID: 11878909 DOI: 10.1006/viro.2001.1213] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of respiratory disease in infants, young children, immunocompromised patients, and the institutionalized elderly. Previous work had shown that RNase L, an antiviral enzyme of the interferon system, could be recruited to cleave RSV genomic RNA by attaching tetrameric 2prime prime or minute-5prime prime or minute-linked oligoadenylates (2-5A) to an oligonucleotide complementary to repetitive gene-start sequences within the RSV genome (2-5A antisense). A 2prime prime or minute-O-methyl RNA-modified analog of the lead 2-5A anti-RSV chimera is shown here to have enhanced antiviral activity in cell culture studies while also cleaving RSV genomic RNA in an RNase L- and sequence-specific manner. When administered intranasally to RSV-infected African green monkeys, this chimera reduced nasal RSV replication by up to four log(10) units in a dose- and time-dependent manner.
Collapse
Affiliation(s)
- Douglas W Leaman
- Ridgeway Biosystems Inc., 9500 Euclid Avenue, NE50, Cleveland, Ohio 44195, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
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
|
21
|
Matsuse H, Behera AK, Kumar M, Lockey RF, Mohapatra SS. Differential cytokine mRNA expression in Dermatophagoides farinae allergen-sensitized and respiratory syncytial virus-infected mice. Microbes Infect 2000; 2:753-9. [PMID: 10955955 DOI: 10.1016/s1286-4579(00)00500-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The interaction between mite allergen sensitization and respiratory syncytial virus (RSV) infection at the level of cytokine mRNA expression was examined in a murine model in the present study. Primary RSV infection enhances expression of inflammatory cytokines such as IL-6, IFN-gamma, and eotaxin in the lung and upregulates the expression of Th2-like cytokines IL-10 and IL-13 in the spleen in BALB/c mice. Mite antigen-sensitized and RSV-infected (ASRSV) mice show enhanced (P < 0.05) total serum IgE compared to antigen-sensitized mice. However, the levels of viral mRNA in the lung tissues are comparable between RSV-infected and ASRSV mice. It is concluded that compartmentalization of cytokine expression following RSV infection plays a role in the augmentation of Th2-like and IgE antibody response to RSV.
Collapse
Affiliation(s)
- H Matsuse
- The Joy McCann Culverhouse Airway Disease Center, Department of Internal Medicine, University of South Florida and James A. Haley VA Hospital, Tampa 33612, USA
| | | | | | | | | |
Collapse
|
22
|
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
|
23
|
Shigeta S. Recent progress in antiviral chemotherapy for respiratory syncytial virus infections. Expert Opin Investig Drugs 2000; 9:221-35. [PMID: 11060673 DOI: 10.1517/13543784.9.2.221] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The recent progress in antiviral chemotherapy against respiratory syncytial virus (RSV) infections was reviewed. RSV infections among high risk individuals, such as premature babies, infants with congenital disease of cardiopulmonary system or immune system and the aged, hospitalised patients with immunosuppressed status are threatened, with high mortality rates and thus need anti-viral chemotherapy. Clinical efficacy of ribavirin and humanized monoclonal antibody (mAb) against RSV infections as well as experimental reports of novel anti-RSV compounds under investigation such as membrane fusion inhibitors were introduced.
Collapse
Affiliation(s)
- S Shigeta
- Department of Microbiology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| |
Collapse
|
24
|
Mills J. Prevention and treatment of respiratory syncytial virus infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 458:39-53. [PMID: 10549378 DOI: 10.1007/978-1-4615-4743-3_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Affiliation(s)
- J Mills
- Macfarlane Burnet Centre for Medical Research, Fairfield, Victoria, Australia
| |
Collapse
|
25
|
Abstract
Respiratory syncytial virus (RSV), long recognised as the major viral pathogen of the lower respiratory tract of infants, has also been implicated in severe lung disease in adults, especially the elderly. This fact, and the demonstration that passive prophylaxis with either polyclonal or monoclonal antibody to RSV prevents severe lung disease in high-risk infants and children, has led to renewed interest in the immune mechanisms surrounding protection, and the development of vaccines
Collapse
Affiliation(s)
- E A Simoes
- Department of Pediatrics, University of Colorado School of Medicine, and Children's Hospital, Denver 80218, USA
| |
Collapse
|
26
|
Abstract
Billions of people are infected with respiratory viruses annually. Infants and young children, the elderly, immunocompromised individuals and those debilitated by other diseases or nutritional deficiencies are most at risk for serious disease. There are few vaccines available for use against these viruses, and even where there are (influenza, measles and adenovirus), infections remain common. The continued prevalence of respiratory virus infections has lead to renewed efforts to find safe agents effective against the most medically important respiratory viruses: influenza, respiratory syncytial, parainfluenza, measles, rhino- and adenovirus. Copyright 1999 Harcourt Publishers Ltd.
Collapse
Affiliation(s)
- Philip R. Wyde
- Department of Microbiology, Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
27
|
Domachowske JB, Rosenberg HF. Respiratory syncytial virus infection: immune response, immunopathogenesis, and treatment. Clin Microbiol Rev 1999; 12:298-309. [PMID: 10194461 PMCID: PMC88919 DOI: 10.1128/cmr.12.2.298] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the single most important cause of lower respiratory tract infection during infancy and early childhood. Once RSV infection is established, the host immune response includes the production of virus-neutralizing antibodies and T-cell-specific immunity. The humoral immune response normally results in the development of anti-RSV neutralizing-antibody titers, but these are often suboptimal during an infant's initial infection. Even when the production of RSV neutralizing antibody following RSV infection is robust, humoral immunity wanes over time. Reinfection during subsequent seasons is common. The cellular immune response to RSV infection is also important for the clearance of virus. This immune response, vital for host defense against RSV, is also implicated in the immunopathogenesis of severe lower respiratory tract RSV bronchiolitis. Many details of the immunology and immunopathologic mechanisms of RSV disease known at present have been learned from rodent models of RSV disease and are discussed in some detail. In addition, the roles of immunoglobulin E, histamine, and eosinophils in the immunopathogenesis of RSV disease are considered. Although the treatment of RSV bronchiolitis is primarily supportive, the role of ribavirin is briefly discussed. Novel approaches to the development of new antiviral drugs with promising anti-RSV activity in vitro are also described.
Collapse
Affiliation(s)
- J B Domachowske
- State University of New York Health Science Center at Syracuse, Syracuse, New York 13210,
| | | |
Collapse
|
28
|
Barnard DL, Sidwell RW, Xiao W, Player MR, Adah SA, Torrence PF. 2-5A-DNA conjugate inhibition of respiratory syncytial virus replication: effects of oligonucleotide structure modifications and RNA target site selection. Antiviral Res 1999; 41:119-34. [PMID: 10320045 DOI: 10.1016/s0166-3542(99)00005-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
To define more fully the conditions for 2-5A-antisense inhibition of respiratory syncytial virus (RSV), relationships between 2-5A antisense oligonucleotide structure and the choice of RNA target sites to inhibition of RSV replication have been explored. The lead 2-5A-antisense chimera for this study was the previously reported NIH8281 that targets the RSV M2 RNA. We have confirmed and extended the earlier study by showing that NIH8281 inhibited RSV strain A2 replication in a variety of antiviral assays, including virus yield reduction assays performed in monkey (EC90 = 0.02 microM) and human cells (EC90 = microM). This 2-5A-antisense chimera also inhibited other A strains, B strains and bovine RSV in cytopathic effect inhibition and Neutral Red Assays (EC50 values = 0.1-1.6 microM). The 2'-O-methylation modification of NIH8281 to increase affinity for the complementary RNA and provide nuclease resistance, the introduction of phosphothioate groups in the antisense backbone to enhance resistance to exo- and endonucleases, and the addition of cholesterol to the 3'-terminus of the antisense oligonucleotide to increase cellular uptake, all resulted in loss of activity. Of the antisense chimeras targeting other RSV mRNAs (NS1, NS2, P, M. G, F, and L), only those complementary to L mRNA were inhibitory. These results suggest that lower abundance mRNAs may be the best targets for 2-5A-antisense; moreover, the active 2-5A antisense chimeras in this study may serve as useful guides for the development of compounds with improved stability, uptake and anti-RSV activity.
Collapse
Affiliation(s)
- D L Barnard
- Institute For Antiviral Research, Utah State University, Logan 84322-5600, USA
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Respiratory syncytial virus (RSV) is a major virus pathogen of infants and young children, an important cause of disease in adults and is responsible for a significant amount of excess morbidity and mortality in the elderly. It also can be devastating in immunosuppressed populations. Vaccines are being developed, but none are currently licensed. Moreover, even if one or more are approved, they may not be suitable for some populations vulnerable to RSV (e.g. very young infants and the immunosuppressed). Ribavirin and immunoglobulin preparations with high titers of RSV-specific neutralizing antibodies are currently approved for use to treat and prevent RSV infection. However, neither of these is cost-effective or simple to administer. New agents are needed to reduce the impact of RSV. This review is concerned with the means currently available for controlling RSV, the search for new agents effective against this virus, and future prospects for preventing and treating RSV infections.
Collapse
Affiliation(s)
- P R Wyde
- Department of Microbiology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Player MR, Barnard DL, Torrence PF. Potent inhibition of respiratory syncytial virus replication using a 2-5A-antisense chimera targeted to signals within the virus genomic RNA. Proc Natl Acad Sci U S A 1998; 95:8874-9. [PMID: 9671772 PMCID: PMC21170 DOI: 10.1073/pnas.95.15.8874] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The 2-5A system is a recognized mechanistic component of the antiviral action of interferon. Interferon-induced 2-5A synthetase generates 2-5A, which, in turn, activates the latent constitutive RNase L that degrades viral RNA. Chemical conjugation of 2-5A to an antisense oligonucleotide can target the 2-5A-dependent RNase L to the antisense-specified RNA and effect its selective destruction. Such a 2-5A-antisense chimera (NIH351) has been developed that targets a consensus sequence within the respiratory syncytial virus (RSV) genomic RNA. NIH351 was 50- to 90-fold more potent against RSV strain A2 than was ribavirin, the presently approved drug for clinical management of RSV infection. It was similarly active against a variety of RSV strains of both A and B subgroups and possessed a cell culture selectivity index comparable to ribavirin. In addition, the anti-RSV activity of NIH351 was shown to be virus-specific and a result of a true antisense effect, because a scrambled nucleotide sequence in the antisense domain of NIH351 caused a significant decrease in antiviral activity. The 2-5A system's RNase L was implicated in the mechanism of action of NIH351 because a congener with a disabled 2-5A moiety was of greatly reduced anti-RSV effectiveness. These findings represent an innovative approach to the control of RSV replication.
Collapse
Affiliation(s)
- M R Player
- Section on Biomedical Chemistry, Laboratory of Medicinal Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0805, USA
| | | | | |
Collapse
|
31
|
Abstract
RNA coliphage SP was propagated for several generations on a host expressing an inhibitory antisense RNA complementary to bases 31-270 of the positive-stranded genome. Phages evolved that escaped inhibition. Typically, these escape mutants contained 3-4 base substitutions, but different sequences were observed among different isolates. The mutations were located within three different types of structural features within the predicted secondary structure of SP genomic RNA: (i) hairpin loops; (ii) hairpin stems; and (iii) the 5' region of the phage genome complementary to the antisense molecule. Computer modelling of the mutant genomic RNAs showed that all of the substitutions within hairpin stems improved the Watson-Crick pairing of the stem. No major structural rearrangements were predicted for any of the mutant genomes, and most substitutions in coding regions did not alter the amino acid sequence. Although the evolved phage populations were polymorphic for substitutions, many substitutions appeared independently in two selected lines. The creation of a new, perfect, antisense RNA against an escape mutant resulted in the inhibition of that mutant but not of other escape mutants nor of the ancestral, unevolved phage. Thus, at least in this system, a population of viruses that evolved to escape from a single antisense RNA would require a cocktail of several antisense RNAs for inhibition.
Collapse
Affiliation(s)
- J J Bull
- Department of Zoology and Institute of Cellular and Molecular Biology, University of Texas, Austin 78712-1064, USA.
| | | | | | | |
Collapse
|
32
|
Abstract
The causative agents of acute respiratory infections (ARI) in infants and children are mostly thought to be viruses. Some ARI in adult patients may be caused by bacteria but most often the causes are virus infections. When ARI affect immunocompromised patients or the elderly the mortality rates are significantly higher than in immunocompetent individuals. Many types of viruses cause ARI. Among them, influenza viruses A and B and respiratory syncytial virus (RSV) are thought to be the most important because of the severity of illness after infection and their high communicability in the human population. Recently, several novel antiviral drugs against ARI have been developed and some are proceeding in clinical trials. This review covers current investigations into antiviral compounds targeted at several points in the virus life-cycle. This includes PM-523, which broadly inhibits ortho- and paramyxo-viruses, two neuraminidase inhibitors for influenza virus, neutralizing antibody to RSV and chimeric soluble ICAM-1-IgA molecules targeted against rhinoviruses.
Collapse
Affiliation(s)
- S Shigeta
- Department of Microbiology, Fukushima Medical College, Japan
| |
Collapse
|
33
|
Affiliation(s)
- A K Field
- Hybridon, Inc., Cambridge, MA 02139, USA
| |
Collapse
|
34
|
Kilkuskie RE, Field AK. Antisense inhibition of virus infections. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1997; 40:437-83. [PMID: 9217933 PMCID: PMC7129323 DOI: 10.1016/s1054-3589(08)60147-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This chapter summarizes the new approaches to identify novel antiviral drug targets and to develop novel antiviral strategies. The chapter also reviews genetic pharmacology as it relates to antiviral antisense research and drug development. Antisense oligonucleotides are selective compounds by virtue of their interaction with specific segments of RNA. For potential antivirals, identification of appropriate target RNA sequences for antisense oligonucleotides is performed at two levels: the optimal gene within the virus, and the optimal sequence within the RNA. The importance of these oligonucleotide modifications in designing effective drugs is just now being evaluated, both in animal model systems and in the clinic. The first generation of widely used antisense oligonucleotides has been the phosphorothioate (PS) compounds and a body of data on biodistribution, pharmacokinetics, and metabolism in animals and in humans is now available. Since the identification and sequencing of human immunodeficiency virus (HIV), there has been a strong interest in identifying a potent oligonucleotide inhibitor that would have the potential for development as a therapy for acquired immunodeficiency syndrome (AIDS). Numerous phosphorothioate oligonucleotides, with no apparent antisense sequence specificity, can have an anti-herpes simplex virus (HSV) effect. Oligonucleotides can be effective anti-influenza agents in cell culture assays. Hepatitis B virus (HBV) X protein that is a transactivator has been also reported to be targeted successfully by antisense oligonucleotides in vivo. Several of picornaviruses have been targets for antisense oligonucleotide inhibition, and the studies demonstrate the versatility of the antisense approach. However, the fact that oligonucleotides may contribute numerous mechanisms toward the antiviral activity, in addition to the antisense mechanism, may in some cases be an asset in the pursuit of clinically useful antiviral drugs.
Collapse
|