Abstract
There are currently only two drugs approved for respiratory syncytial virus (RSV). Palivizumab is a monoclonal antibody for the prevention of RSV in high-risk children and ribavirin is approved for treatment of severe RSV disease, however its effectiveness in improving outcomes is questionable. Over the past 40 years, many obstacles have delayed the development of safe and effective vaccines and treatment regimens. This article reviews these obstacles and presents the novel development strategies used to overcome many of them. Also discussed are promising new antiviral treatment candidates and their associated mechanism of action, the significant advances made in vaccine development, and exciting, new studies directed at improving outcomes through pharmacologic manipulation of the host response to RSV disease.
Keywords: RSV, Vaccine, Treatment, Prevention, Pharmacology
Introduction
RSV is the leading cause of pediatric viral respiratory tract infections. The World Health Organization estimates an annual mortality rate of ∼160,000 worldwide[1]; more inclusive all-cause mortality rates related to RSV approach 600,000[2]. RSV is also the second leading cause of viral death in the elderly[3]. By 18 months of age, 87% of children have developed RSV-specific antibodies[4]; by 3 years of age, virtually all children have been infected. In the United States alone, RSV infection results in over 120,000 childhood hospitalizations and up to 500 deaths[5]. Compared to influenza, RSV accounts for over nine times more deaths in children under the age of 1 year[6-11].
Only two FDA approved drugs are currently available for RSV disease. Palivizumab is indicated for prevention in high risk infants, including those with chronic lung disease, congenital heart disease (CHD) and those born prematurely[12]. This indication is based on hospitalization rates that are ∼5 times greater in high versus non-high risk infants. However, among all infants hospitalized with severe RSV disease, about 70% are term infants with no underlying risk factors compared to 10-20% of high risk infants[13]. Thus, previously healthy infants make up the majority of RSV hospital admissions[14, 15] but are not considered candidates for palivizumab therapy. Ribavirin has been used for treatment of severe infections despite limited evidence of benefit[16, 17], potential for toxicity to health care workers[18], and high cost[19]. The American Academy of Pediatrics does not generally recommend ribavirin treatment for RSV infections[20]. There is an urgent need for safe and effective drugs to treat and prevent RSV disease.
This review begins with a description of the RSV structure and mechanism of cellular invasion. It then summarizes the pharmacology and innovative strategies used to develop RSV treatment and prevention drugs. Concluding remarks emphasize promising anti-RSV drug candidates and their current development status.
Viral Drug Targets
RSV is a negative sense, single-stranded, enveloped RNA paramyxovirus. The RSV genome encodes a total of 11 proteins, many which are under investigation as possible drug targets (Figure 1; Table 1)[21-23]. There are two major antigenic subgroups, A and B, which are defined by different envelope proteins and co-circulate each year. Infectivity of the virus is determined by the surface glycoproteins, F and G, which also serve as targets for neutralizing antibodies[24]. The F glcyoprotein exists only on the surface membrane and is highly conserved across both major antigenic subgroups of RSV. The G glycoprotein exists in both a membrane-bound and secreted form, whereby the secreted form serves as a decoy to neutralizing antibody. Much of the antigenic diversity between and within RSV subgroups is due to variations in the G glycoprotein. Antibody responses to the G glycoprotein are subgroup specific; as low as 35% homology between subgroups A and B has been reported[25]. Antibody responses to the F glycoprotein are generally cross-reactive between RSV A and B subgroups (∼85% homology)[26]. Viral proteins are produced by RSV-specific RNA-dependent RNA polymerase (RdRp)[27]. The RdRp is comprised of at least five viral components, including genomic RNA, and the L, N, P, and M2-1 proteins[28]. The genomic RNA is encapsidated by the nucleocapsid (N) protein[29], which, together with the P and L proteins, forms the necessary unit for RNA replication (Figure 1)[22, 30-33]. These components form a ribonucleoprotein (RNP) complex whose activity results in the production of progeny virus particles.
Figure 1. A cartoon showing the RSV structure and pharmacologic targets.
Five of the eleven RSV proteins have been evaluated in preclinical or clinical trials as potential drug targets for anti-RSV therapy. These include the G and F glycoproteins responsible for viral attachment and fusion to host cells and the genome associated N, P, and L proteins required for RSV replication; together they are termed, the RNP complex.
Table 1. RSV proteins and targeted drug candidates.
Protein/Function | Drug Candidates | Mechanism | Development Status | |
---|---|---|---|---|
Envelope glycoproteins | ||||
F | Mediates fusion and entry of the virion into the host cell and promotes fusion of infected host cells to facilitate cell-to-cell transmission (syncytial formation) [24] | Motavizumab, MEDI-524 [107] | mAba | Phase 1-III, complete; BLAb submitted February, 2008; pending approval |
TMC-353121 [56, 108, 109] | Fusion inhibitor | Preclinical, ongoing | ||
BMS-433771 [110] | Fusion inhibitor | Phase I/II, discontinued | ||
RFI-641 [62] | Fusion inhibitor | Phase I/II, discontinued | ||
VP-14637 [111] | Fusion inhibitor | Phase I, discontinued | ||
BTA9881 [36] | Fusion inhibitor | Phase I, ongoing | ||
G | Mediates viral attachment [24] | MBX-300 [60, 61] | Attachment inhibitor | Preclinical, ongoing |
mAb 131-2G [46, 112] | mAb | Preclinical, ongoing | ||
SH | Structural component; believed to inhibit c TNFα signaling [ 113 ] | |||
Nucleocapsid proteins [28] | ||||
N | Major nucleocapsid protein [29] | ALN-RSV01 [114] | d siRNA | Phase I, complete; Phase II, ongoing |
RSV-604 [65] | N-protein inhibitor | Phase I, complete; Phase II, ongoing | ||
P | Phosphoprotein | |||
L | Large polymerase subunit | YM-53403 [64] | Benzazepine derivative | Preclinical, ongoing |
AUG-2 [115] | Peptide-conjugated e PMO | Preclinical, ongoing | ||
Nucleocapsid-associated proteins [116] | ||||
M2-1 | Transcription elongation factor | |||
M2-2 | Regulator of transcription | |||
Matrix Protein | ||||
M1 | Mediates virus assembly [117, 118] | |||
Nonstructural proteins | ||||
NS1 | Antagonize interferon-induced antiviral responses [119] | |||
NS2 |
mAb = monoclonal antibody;
BLA = Biologic License Agreement
TNFα = tumor necrosis factor alpha;
siRNA = small interfering ribonucleic acid;
PMO = phosphorodiamidate morpholino oligomers.
Drug Candidates Targeting RSV
Polyclonal antibodies
In 1996 MedImmune's RSV Immune Globulin Intravenous (RSV-IGIV), consisting of a high concentration of polyclonal, anti-RSV IgG antibodies purified from the plasma of healthy individuals[34], became the first FDA approved agent for RSV disease prevention. The subsequent approval of palivuzmab, a monoclonal antibody (mAb) directed at the RSV F glycoprotein (discussed below), led many to question the superiority of one product over the other; see reference[35] for an excellent comparative meta-analysis. Despite qualitatively similar reductions in RSV hospitalizations and ICU admissions[35], RSV-IGIV had major disadvantages. These included significant adverse events in children with CHD, the need for IV access, and the risk of infectious disease transmission associated with human plasma-derived products. The availability of palivizumab, a safe and effective alternative, in 1998 led to the removal of RSV-IGIV from the US market that same year[35]. Despite this, a new candidate hyperimmune IVIG product, RI-001, (ADMA Biologicals, Inc.) (personal communication, Dr. Mark Sorrentino; Oct 20, 2009) is now being evaluated in Phase II clinical trials in immunosuppressed, RSV infected patients at risk for lower respiratory tract (LRT) illness[36]. Although human plasma products harbor certain risks, stringent purification requirements have been implemented for all human plasma-derived products to significantly minimize the risk of infection transmission[37]. Moreover, polyclonal antibodies contain a mixed population of antibodies targeting multiple viral epitopes, thus overcoming the mutagenic potential intrinsic among viruses. Results of these studies are ongoing and have not yet been published.
Monoclonal antibodies
Monoclonal anti-RSV antibodies target a single viral epitope. Palivizumab, the only FDA approved mAb for RSV, targets the highly conserved RSV F glycoprotein, inhibiting viral entry into host cells[38]. It has demonstrated no clinical benefit for the treatment of RSV disease, thus is indicated only for RSV prevention. Motavizumab (MEDI-524, MedImmune, Gaithersburgh, MD) is a second-generation humanized IgG1 monoclonal antibody, developed from palivizumab[39], with approximately 70-fold higher affinity for the RSV F glycoprotein and 20-fold greater neutralizing capacity[40]. In a rat model, motavizumab had 50 to 100 times greater anti-RSV activity in the lower respiratory tract compared to palivizumab[41] and reduced RSV viral load in the upper airways where palivizumab has minimal effect[40]. In a large phase III non-inferiority study comparing motavizumab to palivizumab for RSV prevention in high-risk children, motavizumab demonstrated 26% fewer RSV hospitalizations (p<0.01) and a 50% reduction in the incidence of RSV-specific outpatient LRIs (p=0.005) [42]. Moreover, motavizumab significantly reduced viral load by day 1 post-treatment in children hospitalized with RSV, suggesting it may be beneficial for RSV treatment as well as prevention[43]. Motavizumab is currently pending FDA approval.
The majority of RSV mAb candidates target the more conserved F glycoprotein; however, recent evidence suggests that mAbs targeting the G glycoprotein may impart dual anti-RSV activity. The RSV G glycoprotein has been shown to induce lung inflammation by binding to the chemokine receptor CXC3R1, and initiating a cascade of inflammatory mediators[44, 45]. Although still in early preclinical studies, a mAb targeting the CXC3 motif on the RSV G glycoprotein (mAb 131-2G) was shown to reduce both lung inflammation and RSV titers in BALB/c mice[46].
Antisense anti-RSV Drugs
Antisense technology, introduced in the 1980s, involves the targeting of mRNA and viral RNA by oligonucleotides. First generation oligodeoxyribonucleotides (ODNs) were comprised of synthetic DNA molecules 15-20 nucleotides in length that were complementary to small segments of target mRNA. To improve stability against cellular nucleases, phosphodiester bonds were later substituted with phosphorothioate, however, this occurred at the expense of lower sequence specificity and higher toxicity[47]. More recently, a variety of other mechanisms of oligonucleotide-induced antiviral activity have emerged. Those showing the most promise include 2-5A antisense compounds, phosphorodiamidate morpholino oligomers (PMOs), and small interfering RNAs (siRNAs). Of these, only siRNAs have advanced to clinical trials and will be discussed herein. For a thorough review of antisense approaches targeting RSV, see reference[48].
RNA interference (RNAi) is a posttranscriptional mechanism of gene silencing that occurs in plants, animals, and humans[49, 50]. It is important for the regulation of gene expression and participates in host defense against viral infections. The discovery that synthetic double-stranded, small interfering RNAs (siRNAs) could be used to inhibit protein synthesis by targeting mRNA transcripts in mammalian cells led to the emergence of a new field of drug discovery[51] spanning a variety of human diseases including cancer, metabolic diseases, and viral infections[52]. For RSV, siRNAs targeting the P protein[53], NS1 protein[54], and N protein genes[29] have been evaluated. Of these, a siRNA targeting the N protein (ALN-RSV01; Alnylam Pharmaceuticals, Cambridge, MA) is currently being studied in humans. In a phase II clinical trial, ALN-RSV01 or placebo was administered intranasally to 85 healthy adult volunteers two days prior to and three days following viral inoculation. Subjects receiving ALN-RSV01 experienced a 38.1% reduction in RSV infection (p < 0.01) and a 95% increase in the number of subjects who remained free from infection compared to placebo-treated subjects. In a second, recently completed phase II study, safety and tolerability of ALN-RSV01 among adult lung transplant patients naturally infected with RSV was demonstrated. Although not powered to study efficacy, results showed improvement in lung function at the 90 day endpoint[55]. Larger clinical trials in infants are needed to evaluate safety and efficacy; however ALN-RSV01 offers a promising targeted approach to treating infant RSV disease.
Fusion inhibitors
Fusion is a critical step in the life cycle of RSV. Inhibition of this step leads to reduction in viral load and syncytia formation[56, 57]. Upon viral coalescence with target cell membranes, the F glycoprotein undergoes a conformational change exposing hydrophobic pockets or epitopes (Figure 2)[58]. Binding of these exposed targets by RSV fusion inhibitors prevents viral entry in the host cell[59]. Several small-molecule fusion inhibitors have been screened, each targeting a slightly different epitope within the F glycoprotein (Table 2). Despite this, only two remain under investigation (Table 1). BTA9881 is currently in phase I clinical trials and preclinical studies in rats suggest that TMC-353121 is a highly potent (up to 90% inhibition of virus replication) anti-RSV drug candidate. A comprehensive review of RSV fusion inhibitors was recently published by Bonfanti and Roymans[56].
Figure 2.
The RSV F protein is a trimeric class I fusion protein in which each F1 monomer contains two heptad repeats, HR1 and HR2. The fusion peptides are adjacent to the HR1 segments at the amino terminus end of the F1 subunit. These peptides are responsible for fusion to target cell membranes. The HR2 segments located at the F1 carboxy terminus anchor the F glycoprotein to the viral membrane. Of the fusion inhibitors with known binding sites, all bind to the HR1 portion of the fusion glycoprotein (Table 2). Following fusion initiation, conformational changes occur in which the trimeric coiled-coil structure of three HR1 repeats is created. Three HR2 repeats collapse upon the coiled-coil structure to form a “six-helix bundle” (6HB)-complex (also referred to as a “hairpin structure” or “coiled coil heptad repeat”).[58] Modified from Drugs of the Future 2000; 25(3): 287-294. Copyright 2000 Prous Science, S.A. All rights reserved.
Table 2. RSV fusion inhibitors.
Compound | Company | a F1 Interaction | Structure | Route of administration | Sub-group | Model | Reference |
---|---|---|---|---|---|---|---|
TMC-353121 | Johnson & Johnson/Tibotec | Hydrophobic cavity on bHR1 surface of c6HB | Benzimidazole derivatives | Inhaled/oral | A/B | Cotton rat | [56, 108, 109] |
VP-14637 | ViroPharma Inc/RSVCO | Hydrophobic cavity on HR1 surface of 6HB; similar mechanism to TMC-353121 | Triphenolic compound | Inhaled | A/B | Cotton rat/humans | [56, 110, 111, 120] |
BMS-433771 | Bristol-Myers Squibb | Hydrophobic cavity on surface of HR1 trimeric coiled-coil | Benzimidazole derivatives | Oral | A/B | Cotton rat/mice | [110, 121] |
RFI-641 | Wyeth Research | Interacts with F-protein in its native state | Disulfonated stilbene | Inhaled | A/B | Mice/African green monkeys/humans | [122] |
BTA9881 | Biota Holdings Limited/AstraZeneca | Inhibition of F protein assumed based on inhibition of syncytium formation | Imidazoiso-indolone derivative | Oral | A/B | Rodents/humans | [123, 124] |
F1 - Fusion 1 protein; HR – Heptad repeat; 6HB – Six-helix bundle.
Other small-molecule compounds
Other small-molecule compounds have been engineered to inhibit RSV by binding RSV-specific epitopes, including G, L, and N proteins (Figure 1; Table 1). MBX-300 (Microbiotix, Worcester, MA, US) targets the RSV G glycoprotein, resulting in inhibition of viral attachment to host cells [60]. As previously discussed, binding of the G glycoprotein also alters RSV-induced inflammatory responses[44]. In preclinical studies, MBX-300 was found to be safe in both rats and monkeys and demonstrated specific and potent anti-RSV activity[60-62].
The majority of anti-RSV compounds being actively pursued disrupt viral entry into the cell either through the F or G glycoproteins. YM-53403 (Yamanouchi Pharmaceutical Co., Deerfield, Illinois) is a novel compound targeting the RSV L protein, which together with the P and N proteins make up viral RNA polymerase (Figure 1)[63]. Sudo and colleagues demonstrated potent anti-RSV activity against both subgroups A and B, presumably by interfering with RNA synthesis[64]. YM-53403 EC50 values against all RSV strains studied were 76-105-fold more potent than ribavirin. Preclinical studies are ongoing for YM-53403.
RSV604 is an oral benzodiazepine targeting the RSV N protein (Arrow Therapeutics, London, England and Novartis, East Hanover, NJ). Like YM-53403, its putative mechanism of anti-RSV activity is inhibition of viral RNA polymerase. It displays submicromolar activity against many clinical isolates of A and B RSV antigenic subgroups. In contrast to most fusion inhibitors, RSV604 was shown to be active when administered post-infection. Moreover, it significantly reduced viral spread in vitro when given up to 24h post-infection[65]. RSV604 is in ongoing phase II clinical trials.
Vaccines
Numerous obstacles have prevented the development of an effective RSV vaccine. The population most vulnerable to severe RSV disease, children less than 6 months of age[66, 67], may respond inadequately to vaccination due to immunologic immaturity or immune suppression caused by maternal antibodies[5, 68, 69]. In the 1960's, a formalin inactivated (FI)RSV vaccine was studied in human infants for the first time. Not only did it fail to protect against subsequent wild-type (wt) RSV disease, it induced an exaggerated clinical response causing as many as 80% of vaccinated children to be hospitalized and two infant deaths[70-73]. Infiltration of excess eosinophils into the peribronchial spaces[70, 73] and deposits of nonprotective antibodies complexed with virus in affected tissue were blamed for the vaccine-enhanced disease[73-75].
Live vaccines
RSV vaccine development is currently focused on live attenuated strains for intranasal administration. This strategy accomplishes several goals: it induces local mucosal and systemic immunity; the intranasal route partially escapes the suppressive effects of maternal serum antibodies[76]; compared to inactivated vaccines, live intranasal vaccines are more immunogenic and provide broader protection[77]; and live, attenuated vaccines are not associated with enhanced disease[78]. It is unlikely that a single vaccination will impart complete protection against RSV disease as evidenced by natural reinfection occurring throughout life[5, 79, 80]. Thus, the goal for a successful vaccine should be to prevent serious RSV-associated lower respiratory tract infections in those at risk.
The balance between attenuation and immunogenicity is critical in vaccine development. Live vaccine candidates have been developed using serial passages at decreasing temperatures (cold passage[cp]) and chemical mutagenesis to produce temperature-sensitive (ts) mutants. These cpts viruses will replicate at the low temperatures of the upper respiratory tract, but not at the high temperatures of the lower respiratory tract[78]. Initial vaccine candidates developed using these attenuation methods were found to be either too reactive or over-attenuated and mutations were often unstable[81, 82]. The latest strategy to safely and effectively attenuate RSV is through reverse genetics[78], which involves producing infectious virus in cell culture completely from cloned cDNAs[83, 84]. This method introduces targeted mutations to achieve more precise levels of attenuation while maintaining sufficient immunogenicity. rRSVA2cpts248/404/1030/ΔSH (MEDI-559; MedImmune/National Institute of Allergy and Infectious Diseases, Bethesda, MD) is a recombinant, tsRSV with a deletion of the SH gene[85, 86]. The SH protein has been shown to decrease Th1 responses, thereby inhibiting the host anti-viral response. A virus lacking the SH protein would thus impart greater immunogenicity[87]. It is the first vaccine candidate to be sufficiently attenuated for young infants (1-2 months of age). A phase 1/2a study is currently recruiting healthy children between the age of 1 and 24 months to evaluate immunogenicity, viral shedding, safety, and tolerability[36]. Other vaccine candidates under development using these attenuation strategies include rRSVA2cpts248/404/ΔNS2 and rRSVA2cpts530/1009ΔNS2, which include a deletion in the NS genes. The NS protein decreases type I IFN signaling, thus inhibiting host response[88]. Similar to SH deletions, virus lacking the NS proteins will be more immunogenic. Despite often having up to 5 mutations to protect against reversion to wtRSV, there is still concern regarding genetic stability with these vaccine candidates. To address this concern, highly attenuating gene deletion vaccines were developed, including ΔNS1, ΔM2-2, and ΔM2-2NS2[89, 90]. These vaccine candidates maintained a high level of immunogenicity when evaluated in chimpanzees and induced protection following wtRSV challenge; further evaluation in humans is needed[89-91].
Vector vaccines
An alternative method for overcoming genetic instability, while maintain immunogenicity is through the delivery of RSV proteins using viruses with substantially greater growth and stability[78]. The vector vaccine candidate rb/h PIV3/RSV F2 (MEDI-534) delivers RSV F using a bovine/human chimeric parainfluenza type 3 genome. rb/h PIV3/RSVF2 protected monkeys against challenge with wtRSV and generated high titers of RSV and hPIV3 neutralizing antibodies[92]. Safety was demonstrated in a Phase I study of RSV seropositive adults; further studies are needed to determine safety and immunogenicity in children[93]. Other viruses engineered to express RSV F and/or G glycoproteins include Newcastle disease and Sendai viruses, both of which demonstrated immune protection in rodent models[94, 95].
Subunit vaccines
Purified RSV F, G, and M proteins have been evaluated for their potential to induce neutralizing and protective antibodies. The following subunit vaccines have advanced to clinical trials: a) three RSV F subunit vaccines (purified F protein 1 [PFP-1], PFP-2, and PFP-3)[96, 97] b) a combined subunit vaccine containing F, G, and M proteins(Sanofi Pasteur, Swiftwater, PA)[98] and c) BBG2Na, a G peptide conjugated to streptococcal protein G[99]. Only modest rises in antibody titers have been observed in seropositive populations. Safety and efficacy in RSV naïve infants and young children have not been determined. Drawbacks to this vaccine approach include poor immunogenicity, immunosuppressive effects of maternally acquired antibodies, and potential for vaccine-enhanced disease.
Drugs targeting RSV disease pathophysiology
Despite over 50 years of RSV research, the immunopathology and incomplete immunity associated with infant RSV disease remain problematic in the development of effective vaccines and treatments. Novel approaches for altering the host response to RSV, rather than directly targeting the virus, are in the early stages of investigation. Some of these include MBX-300, fosfomycin, and the active metabolite of leflunomide (A77-1726). MBX-300, as previously discussed, targets the RSV G glycoprotein directly, but also competes with the potent chemokine, fractalkine, for binding to CX3CR1 in host cells resulting in reduction of the RSV-induced inflammatory response[44].
Fosfomycin is a structurally unique antibiotic shown to possess in vitro and in vivo immunomodulatory activity[100-102]. Initial studies performed in airway epithelial cells demonstrated that fosfomycin suppressed the RSV-induced transcription of RANTES[103], a chemokine shown to play an important role in RSV lung inflammation[104].
Davis and colleagues demonstrated that RSV is associated with reduced alveolar fluid clearance (AFC), a process that is crucial for efficient gas exchange in the lungs[105]. They went on to show that intranasal administration of A77-1726 to RSV-infected BALB/c mice prevents the RSV-induced decrease of AFC and the onset of arterial hypoxemia[106].
Concluding remarks
Palivizumab remains, perhaps the greatest advancement in RSV pharmacotherapy. Motavizumab, its more potent successor, demonstrates activity in both the upper and lower airways. Despite its pending FDA approval for RSV prevention in high-risk children, evidence suggests it may also play a role in RSV treatment[43]. Of the many RSV treatment candidates evaluated, three have advanced to clinical trials and remain ongoing (Table 1). The siRNA, ALN-RSV01, has received a great deal of attention for its innovative mechanism of action and promising clinical data, however, safety and efficacy in infants remains to be determined.
Vaccine development has made considerable progress over the past 50 years. rRSVA2cpts248/404/1030/ΔSH (MEDI-559) remains the first and only vaccine candidate to be tested in the target infant population since the 1960s FI-RSV trials. Its immunogenicity among infants is currently being evaluated in ongoing clinical trials. The vectored vaccine candidate, rb/hPIV3/RSVF2, takes advantage of the substantially greater growth and stability of the HPIVs compared to RSV. Clinical studies will be needed to determine safety and immunogenicity in infants.
Lastly, it is likely that immunomodulating agents will have a significant impact on RSV disease. Although still in early, preclinical stages, immunomodulatory agents will likely play an important role in combination with direct anti-viral agents.
Acknowledgments
This publication was made possible by Grant Number KL2 RR024154 from the National Center for Research Resources (NCRR), a component of the National Institutes of Health (NIH), and NIH Roadmap for Medical Research. Its contents are solely the responsibility of the authors and do not necessarily represent the official view of NCRR or NIH. Information on NCRR is available at http://www.ncrr.nih.gov/. Information on Re-engineering the Clinical Research Enterprise can be obtained from http://nihroadmap.nih.gov/clinicalresearch/overview-translational.asp.
Footnotes
All authors certify that they have no potential conflicts of interest to disclose.
References
- 1.Acute Respiratory Infections (Update 2009) Initiative for Vaccine Research. 2009 cited 2009; Available from: http//www.who.int/vaccine_research/diseases/ari/en/
- 2.Howard TS, Hoffman LH, Stang PE, Simoes EA. Respiratory syncytial virus pneumonia in the hospital setting: length of stay, charges, and mortality. The Journal of pediatrics. 2000 Aug;137(2):227–32. doi: 10.1067/mpd.2000.107525. [DOI] [PubMed] [Google Scholar]
- 3.Falsey AR, Hennessey PA, Formica MA, Cox C, Walsh EE. Respiratory syncytial virus infection in elderly and high-risk adults. The New England journal of medicine. 2005 Apr 28;352(17):1749–59. doi: 10.1056/NEJMoa043951. [DOI] [PubMed] [Google Scholar]
- 4.Simoes EA. Respiratory syncytial virus infection. Lancet. 1999 Sep 4;354(9181):847–52. doi: 10.1016/S0140-6736(99)80040-3. [DOI] [PubMed] [Google Scholar]
- 5.Dudas RA, Karron RA. Respiratory syncytial virus vaccines. Clin Microbiol Rev. 1998 Jul;11(3):430–9. doi: 10.1128/cmr.11.3.430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Greenough A. Respiratory syncytial virus infection: clinical features, management, and prophylaxis. Curr Opin Pulm Med. 2002 May;8(3):214–7. doi: 10.1097/00063198-200205000-00011. [DOI] [PubMed] [Google Scholar]
- 7.Leader S, Kohlhase K. Recent trends in severe respiratory syncytial virus (RSV) among US infants, 1997 to 2000. The Journal of pediatrics. 2003 Nov;143(5 Suppl):S127–32. doi: 10.1067/s0022-3476(03)00510-9. [DOI] [PubMed] [Google Scholar]
- 8.Shay DK, Holman RC, Newman RD, Liu LL, Stout JW, Anderson LJ. Bronchiolitis-associated hospitalizations among US children, 1980-1996. JAMA. 1999 Oct 20;282(15):1440–6. doi: 10.1001/jama.282.15.1440. [DOI] [PubMed] [Google Scholar]
- 9.Simoes EA, Carbonell-Estrany X. Impact of severe disease caused by respiratory syncytial virus in children living in developed countries. The Pediatric infectious disease journal. 2003 Feb;22(2 Suppl):S13–8. S8–20. doi: 10.1097/01.inf.0000053881.47279.d9. discussion. [DOI] [PubMed] [Google Scholar]
- 10.Thompson WW, Shay DK, Weintraub E, et al. Mortality associated with influenza and respiratory syncytial virus in the United States. JAMA. 2003 Jan 8;289(2):179–86. doi: 10.1001/jama.289.2.179. [DOI] [PubMed] [Google Scholar]
- 11.Welliver RC. Respiratory syncytial virus and other respiratory viruses. The Pediatric infectious disease journal. 2003 Feb;22(2 Suppl):S6–10. S–2. doi: 10.1097/01.inf.0000053880.92496.db. discussion. [DOI] [PubMed] [Google Scholar]
- 12.Meissner HC, Long SS. Revised indications for the use of palivizumab and respiratory syncytial virus immune globulin intravenous for the prevention of respiratory syncytial virus infections. Pediatrics. 2003 Dec;112(6 Pt 1):1447–52. doi: 10.1542/peds.112.6.1447. [DOI] [PubMed] [Google Scholar]
- 13.Meissner HC, Rennels MB, Pickering LK, Hall CB. Risk of severe respiratory syncytial virus disease, identification of high risk infants and recommendations for prophylaxis with palivizumab. The Pediatric infectious disease journal. 2004 Mar;23(3):284–5. doi: 10.1097/01.inf.0000121203.33560.f9. [DOI] [PubMed] [Google Scholar]
- 14.Hall CB, Weinberg GA, Iwane MK, et al. The burden of respiratory syncytial virus infection in young children. The New England journal of medicine. 2009 Feb 5;360(6):588–98. doi: 10.1056/NEJMoa0804877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Stevens WW, Falsey AR, Braciale TJ. RSV 2007: recent advances in respiratory syncytial virus research. Viral Immunol. 2008 Jun;21(2):133–40. doi: 10.1089/vim.2008.0012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hall CB, McBride JT, Walsh EE, et al. Aerosolized ribavirin treatment of infants with respiratory syncytial viral infection. A randomized double-blind study. The New England journal of medicine. 1983 Jun 16;308(24):1443–7. doi: 10.1056/NEJM198306163082403. [DOI] [PubMed] [Google Scholar]
- 17.Ventre K, Randolph AG. Ribavirin for respiratory syncytial virus infection of the lower respiratory tract in infants and young children. Cochrane Database Syst Rev. 2007;(1) doi: 10.1002/14651858.CD000181.pub3. CD000181. [DOI] [PubMed] [Google Scholar]
- 18.Prod Info Rebetol®. 2004 [Google Scholar]
- 19.Glanville AR, Scott AI, Morton JM, et al. Intravenous ribavirin is a safe and cost-effective treatment for respiratory syncytial virus infection after lung transplantation. J Heart Lung Transplant. 2005 Dec;24(12):2114–9. doi: 10.1016/j.healun.2005.06.027. [DOI] [PubMed] [Google Scholar]
- 20.Respiratory Syncytial Virus. Red Book. 2006 January 1;2006(1):560–6. 2006. [Google Scholar]
- 21.Tripp RA. Pneumovirus and metapneumovirus: respiratory syncytial virus and human metapneumovirus. In: Mahy BWJ, M V, editors. Topley and Wilson's Microbiology and Microbial Infections. 10. Vol. 2. London: Hoffer Arnold; 2005. [Google Scholar]
- 22.Liuzzi M, Mason SW, Cartier M, et al. Inhibitors of respiratory syncytial virus replication target cotranscriptional mRNA guanylylation by viral RNA-dependent RNA polymerase. J Virol. 2005 Oct;79(20):13105–15. doi: 10.1128/JVI.79.20.13105-13115.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kingsbury DW. The Paramyxoviruses. New York: Plenum Publishing Corp; 1991. [Google Scholar]
- 24.Bukreyev A, Yang L, Fricke J, et al. The secreted form of respiratory syncytial virus G glycoprotein helps the virus evade antibody-mediated restriction of replication by acting as an antigen decoy and through effects on Fc receptor-bearing leukocytes. J Virol. 2008 Dec;82(24):12191–204. doi: 10.1128/JVI.01604-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Johnson TR, Varga SM, Braciale TJ, Graham BS. Vbeta14(+) T cells mediate the vaccine-enhanced disease induced by immunization with respiratory syncytial virus (RSV) G glycoprotein but not with formalin-inactivated RSV. J Virol. 2004 Aug;78(16):8753–60. doi: 10.1128/JVI.78.16.8753-8760.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Johnson PR, Spriggs MK, Olmsted RA, Collins PL. The G glycoprotein of human respiratory syncytial viruses of subgroups A and B: extensive sequence divergence between antigenically related proteins. Proceedings of the National Academy of Sciences of the United States of America. 1987 Aug;84(16):5625–9. doi: 10.1073/pnas.84.16.5625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mason SW, Lawetz C, Gaudette Y, et al. Polyadenylation-dependent screening assay for respiratory syncytial virus RNA transcriptase activity and identification of an inhibitor. Nucleic Acids Res. 2004;32(16):4758–67. doi: 10.1093/nar/gkh809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Collins PL, C R, Murphy B. Respiratory Syncytial Virus. In: Knipe DM, H P, editors. Fields Virology. Philadelphia: Lippincott Williams & Wilkins; 2001. pp. 1443–85. [Google Scholar]
- 29.Alvarez R, Elbashir S, Borland T, et al. RNAi-Mediated Silencing of the Respiratory Syncytial Virus Nucleocapsid Defines a Potent Anti-Viral Strategy. Antimicrobial agents and chemotherapy. 2009 Jun 8; doi: 10.1128/AAC.00014-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.McNamara PS, Smyth RL. The pathogenesis of respiratory syncytial virus disease in childhood. Br Med Bull. 2002;61:13–28. doi: 10.1093/bmb/61.1.13. [DOI] [PubMed] [Google Scholar]
- 31.Domachowske JB, Rosenberg HF. Respiratory syncytial virus infection: immune response, immunopathogenesis, and treatment. Clin Microbiol Rev. 1999 Apr;12(2):298–309. doi: 10.1128/cmr.12.2.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Collins PL, Hill MG, Cristina J, Grosfeld H. Transcription elongation factor of respiratory syncytial virus, a nonsegmented negative-strand RNA virus. Proceedings of the National Academy of Sciences of the United States of America. 1996 Jan 9;93(1):81–5. doi: 10.1073/pnas.93.1.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hardy RW, Wertz GW. The product of the respiratory syncytial virus M2 gene ORF1 enhances readthrough of intergenic junctions during viral transcription. J Virol. 1998 Jan;72(1):520–6. doi: 10.1128/jvi.72.1.520-526.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Siber GR, Leombruno D, Leszczynski J, et al. Comparison of antibody concentrations and protective activity of respiratory syncytial virus immune globulin and conventional immune globulin. The Journal of infectious diseases. 1994 Jun;169(6):1368–73. doi: 10.1093/infdis/169.6.1368. [DOI] [PubMed] [Google Scholar]
- 35.Morris SK, Dzolganovski B, Beyene J, Sung L. A meta-analysis of the effect of antibody therapy for the prevention of severe respiratory syncytial virus infection. BMC Infect Dis. 2009;9:106. doi: 10.1186/1471-2334-9-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Clinicaltrials.gov. 2009 cited; Available from: www.clinicaltrials.gov.
- 37.Buchacher A, Iberer G. Purification of intravenous immunoglobulin G from human plasma--aspects of yield and virus safety. Biotechnol J. 2006 Feb;1(2):148–63. doi: 10.1002/biot.200500037. [DOI] [PubMed] [Google Scholar]
- 38.Weisman LE. Motavizumab, a second-generation humanized mAb for the prevention of respiratory syncytial virus infection in high-risk populations. Curr Opin Mol Ther. 2009 Apr;11(2):208–18. [PubMed] [Google Scholar]
- 39.Abarca K, Jung E, Fernandez P, et al. Safety, tolerability, pharmacokinetics, and immunogenicity of motavizumab, a humanized, enhanced-potency monoclonal antibody for the prevention of respiratory syncytial virus infection in at-risk children. The Pediatric infectious disease journal. 2009 Apr;28(4):267–72. doi: 10.1097/INF.0b013e31818ffd03. [DOI] [PubMed] [Google Scholar]
- 40.Wu H, Pfarr DS, Johnson S, et al. Development of motavizumab, an ultra-potent antibody for the prevention of respiratory syncytial virus infection in the upper and lower respiratory tract. J Mol Biol. 2007 May 4;368(3):652–65. doi: 10.1016/j.jmb.2007.02.024. [DOI] [PubMed] [Google Scholar]
- 41.Wu H, Pfarr DS, Tang Y, et al. Ultra-potent antibodies against respiratory syncytial virus: effects of binding kinetics and binding valence on viral neutralization. J Mol Biol. 2005 Jul 1;350(1):126–44. doi: 10.1016/j.jmb.2005.04.049. [DOI] [PubMed] [Google Scholar]
- 42.Carbonell-Estrany X, Losonsky GA, Micki H, Edward C. Phase 3 trial of motavizumab (MEDI-524), an enhanced potency respiratory syncytial virus (RSV) specific monoclonal antibody (Mab) for the prevention of serious RSV disease in high risk infants. Pediatric American Society; Toronto, Canada: 2007. [Google Scholar]
- 43.Lagos R, Devincenzo JP, Munoz A, et al. Safety and antiviral activity of motavizumab, a respiratory syncytial virus (rsv)-specific humanized monoclonal antibody, when administered to rsv-infected children. The Pediatric infectious disease journal. 2009 Sep;28(9):835–7. doi: 10.1097/INF.0b013e3181a165e4. [DOI] [PubMed] [Google Scholar]
- 44.Tripp RA, Jones LP, Haynes LM, Zheng H, Murphy PM, Anderson LJ. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nat Immunol. 2001 Aug;2(8):732–8. doi: 10.1038/90675. [DOI] [PubMed] [Google Scholar]
- 45.Haynes LM, Jones LP, Barskey A, Anderson LJ, Tripp RA. Enhanced disease and pulmonary eosinophilia associated with formalin-inactivated respiratory syncytial virus vaccination are linked to G glycoprotein CX3C-CX3CR1 interaction and expression of substance P. J Virol. 2003 Sep;77(18):9831–44. doi: 10.1128/JVI.77.18.9831-9844.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Haynes LM, Caidi H, Radu GU, et al. Therapeutic monoclonal antibody treatment targeting respiratory syncytial virus (RSV) G protein mediates viral clearance and reduces the pathogenesis of RSV infection in BALB/c mice. The Journal of infectious diseases. 2009 Aug 1;200(3):439–47. doi: 10.1086/600108. [DOI] [PubMed] [Google Scholar]
- 47.Maggon K, Barik S. New drugs and treatment for respiratory syncytial virus. Rev Med Virol. 2004 May-Jun;14(3):149–68. doi: 10.1002/rmv.423. [DOI] [PubMed] [Google Scholar]
- 48.Cramer H. Antisense approaches for inhibiting respiratory syncytial virus. Expert Opin Biol Ther. 2005 Feb;5(2):207–20. doi: 10.1517/14712598.5.2.207. [DOI] [PubMed] [Google Scholar]
- 49.Bosher JM, Labouesse M. RNA interference: genetic wand and genetic watchdog. Nat Cell Biol. 2000 Feb;2(2):E31–6. doi: 10.1038/35000102. [DOI] [PubMed] [Google Scholar]
- 50.Hannon GJ. RNA interference. Nature. 2002 Jul 11;418(6894):244–51. doi: 10.1038/418244a. [DOI] [PubMed] [Google Scholar]
- 51.Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001 May 24;411(6836):494–8. doi: 10.1038/35078107. [DOI] [PubMed] [Google Scholar]
- 52.de Fougerolles A, Vornlocher HP, Maraganore J, Lieberman J. Interfering with disease: a progress report on siRNA-based therapeutics. Nat Rev Drug Discov. 2007 Jun;6(6):443–53. doi: 10.1038/nrd2310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bitko V, Musiyenko A, Shulyayeva O, Barik S. Inhibition of respiratory viruses by nasally administered siRNA. Nat Med. 2005 Jan;11(1):50–5. doi: 10.1038/nm1164. [DOI] [PubMed] [Google Scholar]
- 54.Zhang W, Yang H, Kong X, et al. Inhibition of respiratory syncytial virus infection with intranasal siRNA nanoparticles targeting the viral NS1 gene. Nat Med. 2005 Jan;11(1):56–62. doi: 10.1038/nm1174. [DOI] [PubMed] [Google Scholar]
- 55.Alnylam. ALN-RSV01: Respiratory Syncytial Virus. 2009. cited 2009 2009; Web page. Available from: [Google Scholar]
- 56.Bonfanti JF, Roymans D. Prospects for the development of fusion inhibitors to treat human respiratory syncytial virus infection. Curr Opin Drug Discov Devel. 2009 Jul;12(4):479–87. [PubMed] [Google Scholar]
- 57.Colman PM, Lawrence MC. The structural biology of type I viral membrane fusion. Nat Rev Mol Cell Biol. 2003 Apr;4(4):309–19. doi: 10.1038/nrm1076. [DOI] [PubMed] [Google Scholar]
- 58.Lawless-Delmedico MK, Sista P, Sen R, et al. Heptad-repeat regions of respiratory syncytial virus F1 protein form a six-membered coiled-coil complex. Biochemistry. 2000 Sep 26;39(38):11684–95. doi: 10.1021/bi000471y. [DOI] [PubMed] [Google Scholar]
- 59.Zhao X, Singh M, Malashkevich VN, Kim PS. Structural characterization of the human respiratory syncytial virus fusion protein core. Proceedings of the National Academy of Sciences of the United States of America. 2000 Dec 19;97(26):14172–7. doi: 10.1073/pnas.260499197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kimura K, Mori S, Tomita K, et al. Antiviral activity of NMSO3 against respiratory syncytial virus infection in vitro and in vivo. Antiviral Res. 2000 Jul;47(1):41–51. doi: 10.1016/s0166-3542(00)00091-7. [DOI] [PubMed] [Google Scholar]
- 61.Douglas JL. In search of a small-molecule inhibitor for respiratory syncytial virus. Expert Rev Anti Infect Ther. 2004 Aug;2(4):625–39. doi: 10.1586/14787210.2.4.625. [DOI] [PubMed] [Google Scholar]
- 62.Sidwell RW, Barnard DL. Respiratory syncytial virus infections: recent prospects for control. Antiviral Res. 2006 Sep;71(23):379–90. doi: 10.1016/j.antiviral.2006.05.014. [DOI] [PubMed] [Google Scholar]
- 63.Garcia-Barreno B, Delgado T, Melero JA. Identification of protein regions involved in the interaction of human respiratory syncytial virus phosphoprotein and nucleoprotein: significance for nucleocapsid assembly and formation of cytoplasmic inclusions. J Virol. 1996 Feb;70(2):801–8. doi: 10.1128/jvi.70.2.801-808.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sudo K, Miyazaki Y, Kojima N, et al. YM-53403, a unique anti-respiratory syncytial virus agent with a novel mechanism of action. Antiviral Res. 2005 Feb;65(2):125–31. doi: 10.1016/j.antiviral.2004.12.002. [DOI] [PubMed] [Google Scholar]
- 65.Chapman J, Abbott E, Alber DG, et al. RSV604, a novel inhibitor of respiratory syncytial virus replication. Antimicrobial agents and chemotherapy. 2007 Sep;51(9):3346–53. doi: 10.1128/AAC.00211-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lee MS, Walker RE, Mendelman PM. Medical burden of respiratory syncytial virus and parainfluenza virus type 3 infection among US children. Implications for design of vaccine trials. Hum Vaccin. 2005 Jan-Feb;1(1):6–11. doi: 10.4161/hv.1.1.1424. [DOI] [PubMed] [Google Scholar]
- 67.Yamazaki H, Tsutsumi H, Matsuda K, Nagai K, Ogra PL, Chiba S. Effect of maternal antibody on IgA antibody response in nasopharyngeal secretion in infants and children during primary respiratory syncytial virus infection. J Gen Virol. 1994 Aug;75(Pt 8):2115–9. doi: 10.1099/0022-1317-75-8-2115. [DOI] [PubMed] [Google Scholar]
- 68.Osborn JJ, Dancis J, Julia JF. Studies of the immunology of the newborn infant. II. Interference with active immunization by passive transplacental circulating antibody. Pediatrics. 1952 Sep;10(3):328–34. [PubMed] [Google Scholar]
- 69.Karron RA, Steinhoff MC, Subbarao EK, et al. Safety and immunogenicity of a cold-adapted influenza A (H1N1) reassortant virus vaccine administered to infants less than six months of age. The Pediatric infectious disease journal. 1995 Jan;14(1):10–6. doi: 10.1097/00006454-199501000-00002. [DOI] [PubMed] [Google Scholar]
- 70.Chin J, Magoffin RL, Shearer LA, Schieble JH, Lennette EH. Field evaluation of a respiratory syncytial virus vaccine and a trivalent parainfluenza virus vaccine in a pediatric population. Am J Epidemiol. 1969 Apr;89(4):449–63. doi: 10.1093/oxfordjournals.aje.a120957. [DOI] [PubMed] [Google Scholar]
- 71.Fulginiti VA, Eller JJ, Sieber OF, Joyner JW, Minamitani M, Meiklejohn G. Respiratory virus immunization. I. A field trial of two inactivated respiratory virus vaccines; an aqueous trivalent parainfluenza virus vaccine and an alum-precipitated respiratory syncytial virus vaccine. Am J Epidemiol. 1969 Apr;89(4):435–48. doi: 10.1093/oxfordjournals.aje.a120956. [DOI] [PubMed] [Google Scholar]
- 72.Kapikian AZ, Mitchell RH, Chanock RM, Shvedoff RA, Stewart CE. An epidemiologic study of altered clinical reactivity to respiratory syncytial (RS) virus infection in children previously vaccinated with an inactivated RS virus vaccine. Am J Epidemiol. 1969 Apr;89(4):405–21. doi: 10.1093/oxfordjournals.aje.a120954. [DOI] [PubMed] [Google Scholar]
- 73.Kim HW, Canchola JG, Brandt CD, et al. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol. 1969 Apr;89(4):422–34. doi: 10.1093/oxfordjournals.aje.a120955. [DOI] [PubMed] [Google Scholar]
- 74.Polack FP, Teng MN, Collins PL, et al. A role for immune complexes in enhanced respiratory syncytial virus disease. The Journal of experimental medicine. 2002 Sep 16;196(6):859–65. doi: 10.1084/jem.20020781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Murphy BR, Alling DW, Snyder MH, et al. Effect of age and preexisting antibody on serum antibody response of infants and children to the F and G glycoproteins during respiratory syncytial virus infection. J Clin Microbiol. 1986 Nov;24(5):894–8. doi: 10.1128/jcm.24.5.894-898.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Crowe JE, Jr, Bui PT, Siber GR, Elkins WR, Chanock RM, Murphy BR. Cold-passaged, temperature-sensitive mutants of human respiratory syncytial virus (RSV) are highly attenuated, immunogenic, and protective in seronegative chimpanzees, even when RSV antibodies are infused shortly before immunization. Vaccine. 1995 Jun;13(9):847–55. doi: 10.1016/0264-410x(94)00074-w. [DOI] [PubMed] [Google Scholar]
- 77.Johnson PR, Feldman S, Thompson JM, Mahoney JD, Wright PF. Immunity to influenza A virus infection in young children: a comparison of natural infection, live cold-adapted vaccine, and inactivated vaccine. The Journal of infectious diseases. 1986 Jul;154(1):121–7. doi: 10.1093/infdis/154.1.121. [DOI] [PubMed] [Google Scholar]
- 78.Collins PL, Murphy BR. New generation live vaccines against human respiratory syncytial virus designed by reverse genetics. Proc Am Thorac Soc. 2005;2(2):166–73. doi: 10.1513/pats.200501-011AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Schickli JH, Dubovsky F, Tang RS. Challenges in developing a pediatric RSV vaccine. Hum Vaccin. 2009 Sep;5(9):582–91. doi: 10.4161/hv.9131. [DOI] [PubMed] [Google Scholar]
- 80.Murphy BR, Hall SL, Kulkarni AB, et al. An update on approaches to the development of respiratory syncytial virus (RSV) and parainfluenza virus type 3 (PIV3) vaccines. Virus Res. 1994 Apr;32(1):13–36. doi: 10.1016/0168-1702(94)90059-0. [DOI] [PubMed] [Google Scholar]
- 81.Karron RA, Wright PF, Crowe JE, Jr, et al. Evaluation of two live, cold-passaged, temperature-sensitive respiratory syncytial virus vaccines in chimpanzees and in human adults, infants, and children. The Journal of infectious diseases. 1997 Dec;176(6):1428–36. doi: 10.1086/514138. [DOI] [PubMed] [Google Scholar]
- 82.Wright PF, Karron RA, Belshe RB, et al. Evaluation of a live, cold-passaged, temperature-sensitive, respiratory syncytial virus vaccine candidate in infancy. The Journal of infectious diseases. 2000 Nov;182(5):1331–42. doi: 10.1086/315859. [DOI] [PubMed] [Google Scholar]
- 83.Collins PL, Hill MG, Camargo E, Grosfeld H, Chanock RM, Murphy BR. Production of infectious human respiratory syncytial virus from cloned cDNA confirms an essential role for the transcription elongation factor from the 5′ proximal open reading frame of the M2 mRNA in gene expression and provides a capability for vaccine development. Proceedings of the National Academy of Sciences of the United States of America. 1995 Dec 5;92(25):11563–7. doi: 10.1073/pnas.92.25.11563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Firestone CY, Whitehead SS, Collins PL, Murphy BR, Crowe JE., Jr Nucleotide sequence analysis of the respiratory syncytial virus subgroup A cold-passaged (cp) temperature sensitive (ts) cpts-248/404 live attenuated virus vaccine candidate. Virology. 1996 Nov 15;225(2):419–22. doi: 10.1006/viro.1996.0618. [DOI] [PubMed] [Google Scholar]
- 85.Karron RA, Wright PF, Belshe RB, et al. Identification of a recombinant live attenuated respiratory syncytial virus vaccine candidate that is highly attenuated in infants. The Journal of infectious diseases. 2005 Apr 1;191(7):1093–104. doi: 10.1086/427813. [DOI] [PubMed] [Google Scholar]
- 86.Medimmune. 2009 cited 2009 November 5. Available from: www.medimmune.com/research_pipeline_candidates.aspx.
- 87.Tripp RA, Moore D, Jones L, Sullender W, Winter J, Anderson LJ. Respiratory syncytial virus G and/or SH protein alters Th1 cytokines, natural killer cells, and neutrophils responding to pulmonary infection in BALB/c mice. J Virol. 1999 Sep;73(9):7099–107. doi: 10.1128/jvi.73.9.7099-7107.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ramaswamy M, Shi L, Varga SM, Barik S, Behlke MA, Look DC. Respiratory syncytial virus nonstructural protein 2 specifically inhibits type I interferon signal transduction. Virology. 2006 Jan 20;344(2):328–39. doi: 10.1016/j.virol.2005.09.009. [DOI] [PubMed] [Google Scholar]
- 89.Jin H, Cheng X, Traina-Dorge VL, et al. Evaluation of recombinant respiratory syncytial virus gene deletion mutants in African green monkeys for their potential as live attenuated vaccine candidates. Vaccine. 2003 Sep 8;21(2526):3647–52. doi: 10.1016/s0264-410x(03)00426-2. [DOI] [PubMed] [Google Scholar]
- 90.Teng MN, Whitehead SS, Bermingham A, et al. Recombinant respiratory syncytial virus that does not express the NS1 or M2-2 protein is highly attenuated and immunogenic in chimpanzees. J Virol. 2000 Oct;74(19):9317–21. doi: 10.1128/jvi.74.19.9317-9321.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Cheng X, Zhou H, Tang RS, Munoz MG, Jin H. Chimeric subgroup A respiratory syncytial virus with the glycoproteins substituted by those of subgroup B and RSV without the M2-2 gene are attenuated in African green monkeys. Virology. 2001 Apr 25;283(1):59–68. doi: 10.1006/viro.2001.0894. [DOI] [PubMed] [Google Scholar]
- 92.Tang RS, MacPhail M, Schickli JH, et al. Parainfluenza virus type 3 expressing the native or soluble fusion (F) Protein of Respiratory Syncytial Virus (RSV) confers protection from RSV infection in African green monkeys. J Virol. 2004 Oct;78(20):11198–207. doi: 10.1128/JVI.78.20.11198-11207.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Tang RS, Spaete RR, Thompson MW, et al. Development of a PIV-vectored RSV vaccine: preclinical evaluation of safety, toxicity, and enhanced disease and initial clinical testing in healthy adults. Vaccine. 2008 Nov 25;26(50):6373–82. doi: 10.1016/j.vaccine.2008.09.018. [DOI] [PubMed] [Google Scholar]
- 94.Martinez-Sobrido L, Gitiban N, Fernandez-Sesma A, et al. Protection against respiratory syncytial virus by a recombinant Newcastle disease virus vector. J Virol. 2006 Feb;80(3):1130–9. doi: 10.1128/JVI.80.3.1130-1139.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Takimoto T, Hurwitz JL, Coleclough C, et al. Recombinant Sendai virus expressing the G glycoprotein of respiratory syncytial virus (RSV) elicits immune protection against RSV. J Virol. 2004 Jun;78(11):6043–7. doi: 10.1128/JVI.78.11.6043-6047.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Simoes EA, Tan DH, Ohlsson A, Sales V, Wang EE. Respiratory syncytial virus vaccine: a systematic overview with emphasis on respiratory syncytial virus subunit vaccines. Vaccine. 2001 Dec 12;20(56):954–60. doi: 10.1016/s0264-410x(01)00388-7. [DOI] [PubMed] [Google Scholar]
- 97.Wathen MW, Kakuk TJ, Brideau RJ, Hausknecht EC, Cole SL, Zaya RM. Vaccination of cotton rats with a chimeric FG glycoprotein of human respiratory syncytial virus induces minimal pulmonary pathology on challenge. The Journal of infectious diseases. 1991 Mar;163(3):477–82. doi: 10.1093/infdis/163.3.477. [DOI] [PubMed] [Google Scholar]
- 98.Falsey AR, Walsh EE, Capellan J, et al. Comparison of the safety and immunogenicity of 2 respiratory syncytial virus (rsv) vaccines--nonadjuvanted vaccine or vaccine adjuvanted with alum--given concomitantly with influenza vaccine to high-risk elderly individuals. The Journal of infectious diseases. 2008 Nov 1;198(9):1317–26. doi: 10.1086/592168. [DOI] [PubMed] [Google Scholar]
- 99.Power UF, Nguyen TN, Rietveld E, et al. Safety and immunogenicity of a novel recombinant subunit respiratory syncytial virus vaccine (BBG2Na) in healthy young adults. The Journal of infectious diseases. 2001 Dec 1;184(11):1456–60. doi: 10.1086/324426. [DOI] [PubMed] [Google Scholar]
- 100.Honda J, Okubo Y, Kusaba M, Kumagai M, Saruwatari N, Oizumi K. Fosfomycin (FOM: 1 R-2S-epoxypropylphosphonic acid) suppress the production of IL-8 from monocytes via the suppression of neutrophil function. Immunopharmacology. 1998 May;39(2):149–55. doi: 10.1016/s0162-3109(98)00003-4. [DOI] [PubMed] [Google Scholar]
- 101.Morikawa K, Nonaka M, Torii I, Morikawa S. Modulatory effect of fosfomycin on acute inflammation in the rat air pouch model. Int J Antimicrob Agents. 2003 Apr;21(4):334–9. doi: 10.1016/s0924-8579(02)00358-8. [DOI] [PubMed] [Google Scholar]
- 102.Morikawa K, Watabe H, Araake M, Morikawa S. Modulatory effect of antibiotics on cytokine production by human monocytes in vitro. Antimicrobial agents and chemotherapy. 1996 Jun;40(6):1366–70. doi: 10.1128/aac.40.6.1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Okabayashi T, Yokota S, Yoto Y, Tsutsumi H, Fujii N. Fosfomycin suppresses chemokine induction in airway epithelial cells infected with respiratory syncytial virus. Clin Vaccine Immunol. 2009 Jun;16(6):859–65. doi: 10.1128/CVI.00033-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Casola A, Garofalo RP, Haeberle H, et al. Multiple cis regulatory elements control RANTES promoter activity in alveolar epithelial cells infected with respiratory syncytial virus. J Virol. 2001 Jul;75(14):6428–39. doi: 10.1128/JVI.75.14.6428-6439.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Davis IC, Sullender WM, Hickman-Davis JM, Lindsey JR, Matalon S. Nucleotide-mediated inhibition of alveolar fluid clearance in BALB/c mice after respiratory syncytial virus infection. Am J Physiol Lung Cell Mol Physiol. 2004 Jan;286(1):L112–20. doi: 10.1152/ajplung.00218.2003. [DOI] [PubMed] [Google Scholar]
- 106.Davis IC, Lazarowski ER, Chen FP, Hickman-Davis JM, Sullender WM, Matalon S. Post-infection A77-1726 blocks pathophysiologic sequelae of respiratory syncytial virus infection. American journal of respiratory cell and molecular biology. 2007 Oct;37(4):379–86. doi: 10.1165/rcmb.2007-0142OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Madaan A. Motavizumab: Humanized anti-RSV monoclonal antibody, prevention of RSV infection. Drugs of the Future. 2008;33(3):203–5. [Google Scholar]
- 108.Andries K, Moeremans M, Gevers T, et al. Substituted benzimidazoles with nanomolar activity against respiratory syncytial virus. Antiviral Res. 2003 Nov;60(3):209–19. doi: 10.1016/j.antiviral.2003.07.004. [DOI] [PubMed] [Google Scholar]
- 109.Bonfanti JF, Meyer C, Doublet F, et al. Selection of a respiratory syncytial virus fusion inhibitor clinical candidate. 2. Discovery of a morpholinopropylaminobenzimidazole derivative (TMC353121) J Med Chem. 2008 Feb 28;51(4):875–96. doi: 10.1021/jm701284j. [DOI] [PubMed] [Google Scholar]
- 110.Cianci C, Langley DR, Dischino DD, et al. Targeting a binding pocket within the trimer-of-hairpins: small-molecule inhibition of viral fusion. Proceedings of the National Academy of Sciences of the United States of America. 2004 Oct 19;101(42):15046–51. doi: 10.1073/pnas.0406696101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Douglas JL, Panis ML, Ho E, et al. Small molecules VP-14637 and JNJ-2408068 inhibit respiratory syncytial virus fusion by similar mechanisms. Antimicrobial agents and chemotherapy. 2005 Jun;49(6):2460–6. doi: 10.1128/AAC.49.6.2460-2466.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Miao C, Radu GU, Caidi H, Tripp RA, Anderson LJ, Haynes LM. Treatment with respiratory syncytial virus G glycoprotein monoclonal antibody or F(ab′)2 components mediates reduced pulmonary inflammation in mice. J Gen Virol. 2009 May;90(Pt 5):1119–23. doi: 10.1099/vir.0.009308-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Fuentes S, Tran KC, Luthra P, Teng MN, He B. Function of the respiratory syncytial virus small hydrophobic protein. J Virol. 2007 Aug;81(15):8361–6. doi: 10.1128/JVI.02717-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.DeVincenzo J, Cehelsky JE, Alvarez R, et al. Evaluation of the safety, tolerability and pharmacokinetics of ALN-RSV01, a novel RNAi antiviral therapeutic directed against respiratory syncytial virus (RSV) Antiviral Res. 2008 Mar;77(3):225–31. doi: 10.1016/j.antiviral.2007.11.009. [DOI] [PubMed] [Google Scholar]
- 115.Lai SH, Stein DA, Guerrero-Plata A, et al. Inhibition of respiratory syncytial virus infections with morpholino oligomers in cell cultures and in mice. Mol Ther. 2008 Jun;16(6):1120–8. doi: 10.1038/mt.2008.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Fearns R, Collins PL. Role of the M2-1 transcription antitermination protein of respiratory syncytial virus in sequential transcription. J Virol. 1999 Jul;73(7):5852–64. doi: 10.1128/jvi.73.7.5852-5864.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ghildyal R, Ho A, Dias M, et al. The respiratory syncytial virus matrix protein possesses a Crm1-mediated nuclear export mechanism. J Virol. 2009 Jun;83(11):5353–62. doi: 10.1128/JVI.02374-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Garcia J, Garcia-Barreno B, Vivo A, Melero JA. Cytoplasmic inclusions of respiratory syncytial virus-infected cells: formation of inclusion bodies in transfected cells that coexpress the nucleoprotein, the phosphoprotein, and the 22K protein. Virology. 1993 Jul;195(1):243–7. doi: 10.1006/viro.1993.1366. [DOI] [PubMed] [Google Scholar]
- 119.Schlender J, Bossert B, Buchholz U, Conzelmann KK. Bovine respiratory syncytial virus nonstructural proteins NS1 and NS2 cooperatively antagonize alpha/beta interferon-induced antiviral response. J Virol. 2000 Sep;74(18):8234–42. doi: 10.1128/jvi.74.18.8234-8242.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wyde PR, Laquerre S, Chetty SN, Gilbert BE, Nitz TJ, Pevear DC. Antiviral efficacy of VP14637 against respiratory syncytial virus in vitro and in cotton rats following delivery by small droplet aerosol. Antiviral Res. 2005 Oct;68(1):18–26. doi: 10.1016/j.antiviral.2005.06.009. [DOI] [PubMed] [Google Scholar]
- 121.Cianci C, Meanwell N, Krystal M. Antiviral activity and molecular mechanism of an orally active respiratory syncytial virus fusion inhibitor. J Antimicrob Chemother. 2005 Mar;55(3):289–92. doi: 10.1093/jac/dkh558. [DOI] [PubMed] [Google Scholar]
- 122.Razinkov V, Huntley C, Ellestad G, Krishnamurthy G. RSV entry inhibitors block F-protein mediated fusion with model membranes. Antiviral Res. 2002 Jul;55(1):189–200. doi: 10.1016/s0166-3542(02)00050-5. [DOI] [PubMed] [Google Scholar]
- 123.Bond S, Draffan A, Lambert J, et al. Discovery of a new class of polycyclic RSV inhibitors. Antiviral Research. 2007 Jun;74(3):A30–A. [Google Scholar]
- 124.Biota. 2009 2009 [cited 2009; Available from: www.biota.com.au.