Skip to main content
Elsevier Sponsored Documents logoLink to Elsevier Sponsored Documents
. 2017 Jan 11;35(3):469–480. doi: 10.1016/j.vaccine.2016.11.054

Animal models of respiratory syncytial virus infection

Geraldine Taylor 1
PMCID: PMC5244256  PMID: 27908639

Highlights

  • Respiratory syncytial virus (RSV) is a major cause respiratory disease, worldwide.

  • Paediatric and elderly populations are most vulnerable to severe disease.

  • Vaccine development has been hampered by the experience of vaccine-enhanced disease.

  • Animal models do not necessarily predict vaccine efficacy and safety.

  • The strengths and limitations of animal models of RSV infection are summarised.

Keywords: Respiratory syncytial virus, Animal models

Abstract

Human respiratory syncytial virus (hRSV) is a major cause of respiratory disease and hospitalisation of infants, worldwide, and is also responsible for significant morbidity in adults and excess deaths in the elderly. There is no licensed hRSV vaccine or effective therapeutic agent. However, there are a growing number of hRSV vaccine candidates that have been developed targeting different populations at risk of hRSV infection. Animal models of hRSV play an important role in the preclinical testing of hRSV vaccine candidates and although many have shown efficacy in preclinical studies, few have progressed to clinical trials or they have had only limited success. This is, at least in part, due to the lack of animal models that fully recapitulate the pathogenesis of hRSV infection in humans. This review summarises the strengths and limitations of animal models of hRSV, which include those in which hRSV is used to infect non-human mammalian hosts, and those in which non-human pneumoviruses, such as bovine (b)RSV and pneumonia virus of mice (PVM) are studied in their natural host.

Apart from chimpanzees, other non-human primates (NHP) are only semi-permissive for hRSV replication and experimental infection with large doses of virus result in little or no clinical signs of disease, and generally only mild pulmonary pathology. Other animal models such as cotton rats, mice, ferrets, guinea pigs, hamsters, chinchillas, and neonatal lambs are also only semi-permissive for hRSV. Nevertheless, mice and cotton rats have been of value in the development of monoclonal antibody prophylaxis for infants at high risk of severe hRSV infection and have provided insights into mechanisms of immunity to and pathogenesis of hRSV. However, the extent to which they predict hRSV vaccine efficacy and safety is unclear and several hRSV vaccine candidates that are completely protective in rodent models are poorly effective in chimpanzees and other NHP, such as African Green monkeys. Furthermore, interpretation of findings from many rodent and NHP models of vaccine-enhanced hRSV disease has been confounded by sensitisation to non-viral antigens present in the vaccine and challenge virus.

Studies of non-human pneumoviruses in their native hosts are more likely to reflect the pathogenesis of natural hRSV infection, and experimental infection of calves with bRSV and of mice with PVM result in clinical disease and extensive pulmonary pathology. These animal models have not only been of value in studies on mechanisms of immunity to and the pathogenesis of pneumovirus infections but have also been used to evaluate hRSV vaccine concepts. Furthermore, the similarities between the epidemiology of bRSV in calves and hRSV in infants and the high level of genetic and antigenic similarity between bRSV and hRSV, make the calf model of bRSV infection a relevant model for preclinical evaluation of hRSV vaccine candidates which contain proteins that are conserved between hRSV and bRSV.

1. Introduction

Human respiratory syncytial virus (hRSV) is the commonest cause of lower respiratory tract infection (LRTI) in children, worldwide, causing disease in an estimated 34 million children, >3 million hospitalizations, and 66,000–199,000 deaths in children under 5 years, each year [1]. Most children are infected during the first year of life, and all have been infected by their second year. Although there is limited viral antigenic variation, the duration of immunity induced by hRSV is short-lived and recurrent infections occur throughout life [2]. The peak incidence of severe disease is in infants 2–7 months of age. However, hRSV also contributes to excess mortality in the elderly [3], [4] and severe hRSV disease occurs in immunosuppressed individuals of any age [5]. The economic impact of hRSV disease in adults is estimated to be greater than that of influenza in relation to numbers of days lost from work [6], [7]. There is no effective anti-viral therapy or licensed hRSV vaccine. However, monoclonal antibody (mAb) prophylaxis is effective in reducing hRSV hospitalisations by 55% in infants at increased risk of severe disease [8].

Hurdles to vaccine development include the need to vaccinate at an early age when maternal antibodies are present, the failure of natural infection to prevent reinfection, a history of vaccine-enhanced disease in young children given a formalin-inactivated (FI) hRSV vaccine [9], and the lack of animal models that fully reproduce the pathogenesis of hRSV infection in man. Since hRSV infection does not prime for enhanced disease, live attenuated vaccine candidates are considered safe, although their protective efficacy may not be very durable. Development of other vaccines for paediatric use is hampered by the difficulty of reliably demonstrating their safety and efficacy in preclinical studies.

2. HRSV infection in man

HRSV is mainly transmitted by large particle aerosol or direct contact. Initial viral replication occurs in the nasopharynx with an incubation period of 4–5 days, and can be followed by spread to the LRT. Disease severity ranges from that of a mild common cold to bronchiolitis, with airway obstruction and hypoxia in infants, wheezing, and pneumonia [4]. Apnea is seen in 1–24% of hRSV-infected infants [10], and severe hRSV bronchiolitis is associated with subsequent episodes of wheezing which can persist until ∼11 years of age [11].

HRSV replicates primarily in ciliated airway epithelial cells and type I and type II alveolar pneumocytes (Fig. 1A) [12]. Histopathological changes of fatal cases of hRSV infection are characterised by peribronchiolar and perivascular mononuclear cell accumulations, interstitial pneumonia, bronchiolar epithelial cell necrosis, and occlusion of the bronchiolar lumen by desquamated epithelial cells, macrophages, neutrophils, fibrin, and mucin, resulting in airway obstruction (Fig. 1B) [12]. Cells in the peribronchial areas of the lung of a child with hRSV who died from an unrelated cause, consisted of monocytes, CD3+CD4CD8 T cells and CD3+CD8+ [12]. However, CD4+ and CD8+ lymphocytes were rare in lungs of fatal hRSV LRTI [13]. Neutrophils predominate in bronchoalveolar lavage (BAL) from infants with hRSV bronchiolitis [14], [15], and respiratory secretions contain high levels of proinflammatory cytokines (e.g. tumour necrosis factor-α (TNFα), interleukin-6 (IL-6), IL-1α), CXC/CC chemokines (e.g. IL-8, MIP-1α, MCP-1 and RANTES) [16], and interferon (IFN)γ, IL-4, IL-5, IL-10, IL-9 and IL-17 [17], [18]. There is an association between severity of hRSV LRTI and polymorphisms in a number of host response genes (e.g. IL-8 promoter, and CC chemokine receptor 5 (CCR5), which binds Mip-1α and RANTES) [19], [20], suggesting that a robust inflammatory response contributes to hRSV pathogenesis. However, intrinsic properties of different virus strains may also contribute to variations in disease severity [21].

Fig. 1.

Fig. 1

(A) Localisation of human (h)RSV antigen in bronchiolar epithelial cells of a child with an acute hRSV infection. HRSV antigen-positive cells are brown; (B) section of lung from a child with an acute hRSV infection showing acute bronchiolitis in a medium-sized airway with intraluminal cellular debris and inflammatory cells (A and B are adapted from [12], reprinted by permission from Macmillan Publishers Ltd: Modern Pathology, 20:108–119, Johnson et al., copyright 2007); (C) localisation of bovine (b)RSV antigen in bronchial, bronchiolar, and alveolar cells of a calf experimentally infected with bRSV, 6 days previously. BRSV antigen-positive cells are red (adapted from [114], reprinted from The American Journal of Pathology, 161:2195–2207, copyright 2002, with permission from Elsevier); (D) section of lung from a calf, inoculated intranasally and intracheally 6 days previously with bRSV, showing bronchiolitis with intraluminal cellular debris and inflammatory cells, and interstitial pneumonia; (E) section of chimpanzee lung, naturally infected with hRSV, showing alveolar and interstitial pneumonitis and bronchitis (reprinted from [25], J Com Pathol, 110:207–212, copyright 1994, with permission from Elsevier; (F) section of lung from an infant rhesus monkey infected by aerosol with hRSV, 7 days previously, showing mild lymphocytic and histiocytic inflammatory cell infiltrates in the walls and lumens of the terminal conducting airways, and in the septa and lumens of adjacent alveoli (adapted from [45], reprinted from Vaccine, 23:2928–2942, copyright 2005, with permission from Elsevier); (G) section of lung from a lamb infected by aerosol with hRSV, 8 days previously, showing viral antigen within epithelial cells lining the bronchioles (brown cells); (H) section of lung from a lamb infected by aerosol with hRSV, 8 days previously, showing bronchiolitis with degenerate/necrotic individual epithelial cells (thin arrow), occasional syncytial cells (long arrow), accumulation of degenerate neutrophils (short arrow), and occasional macrophages (G and H adapted from [59]; (I) section of lung from a cotton rat infected 5 days previously with hRSV, showing mild peribronchiolitis (adapted from [70], reprinted from Vaccine, 31:306–312, copyright 2013, with permission from Elsevier).

3. Animal models of RSV

Animal models of hRSV include those in which hRSV is used to infect non-human mammalian hosts, and those in which non-human pneumoviruses are studied in their natural host. The main non-human mammalian hosts that have been used are non-human primates, cotton rats, mice, and lambs. Apart from chimpanzees, these animals are semi-permissive for hRSV replication and experimental infection with large doses of virus result in little or no clinical signs of disease. In contrast, natural hosts of non-human pneumoviruses, such as bovine (b)RSV and pneumonia virus of mice (PVM), are fully permissive for virus replication and experimental infection results in disease.

3.1. Non-human primates

3.1.1. Chimpanzees

HRSV was first isolated from a chimpanzee in a captive colony of 20 animals, 14 of which had a respiratory illness characterised by coughing, sneezing, and a mucopurulent nasal discharge [22]. The virus, which was originally termed chimpanzee coryza agent (CCA), caused a similar upper respiratory tract (URT) disease in 2 out of 3 chimpanzees after intranasal (IN) inoculation with 104 TCID50 of tissue culture-passaged material. The animal that did not develop disease was the only one that had pre-existing hRSV-specific serum antibodies. The virus was transmitted not only between chimpanzees but also from chimpanzees to a laboratory worker.

Chimpanzees experimentally infected with ∼104 pfu hRSV shed large quantities of virus (105 and 106 pfu/ml of nasopharyngeal swab sample and tracheal lavage, respectively), for 6–10 days, which is comparable to that seen in children (Table 1) [23], [24]. Although LRTI has not been seen in experimentally infected chimpanzees, fatal cases of hRSV-associated bronchopneumonia, some of which also involved Streptococcus pneumoniae, have been reported in captive chimpanzees in zoos and in habituated chimpanzees in a research station in Côte d’Ivoire [25], [26], [27], [28]. HRSV antigen can be detected in the lungs and histopathological changes of extensive purulent bronchopneumonia and interstitial pneumonia with occasional syncytial cells resemble those in human infants (Fig. 1E) [25]. S. pneumoniae has been shown to enhance hRSV infection in differentiated human airway epithelial cells, in vitro, and in cotton rats [29].

Table 1.

Non-human primate models of hRSV infection.

Species Age Virus straina Inoculum Routeb Viral replication Clinical signs of diseasec Pathology Reference
Chimpanzees (Pan troglodytes) 20–24 mths hRSV 104 TCID50 IN Virus isolated URT illness Not done [22]
15–18 mths hRSV A2 103.5 pfu IN High URT illness Not done [23]



Owl monkeys (Aotus trivirgatus) Adult hRSV A2 103.7–106 pfu IN or IT Moderate Serous rhinorrea Minor histological changes [32], [54], [55]



Baboons (Papio cynocephalus anubis) 4 wks hRSV A2 107.9 pfu IT Virus tires declined from day 1 Tachypnoea & dyspnoea Gross changes of vascular congestion & oedema; Interstitial pneumonia, sloughing of bronchiolar epithelium, obstruction of the bronchiolar lumen [58]



Cebus monkeys (Cebus apella, Cebus albifrons) 6–20 mths hRSV A2 108 pfu IT Moderate Rhinorrhea & conjunctivitis Extensive interstitial pneumonia, alveolitis, syncytial cells [57]



African green monkeys (Cercopithecus aethiops) Adolescent & adult hRSV A2 103 pfu IN & IT Moderate Rhinorrhea, sneezing, & wheezing Patchy inflammation in terminal bronchioles, interstitium & alveoli, syncytial cells; Slight increase in neutrophils in BAL [42]
Not reported hRSV M37 105.8 pfu IN Moderate None Not done [40]



Rhesus macaques (Macaca mulatta) 1 wk hRSV A2 103.2 pfu IN Low None Not done [23]
Young adults Rhesus-adapted hRSV Long 105.7 TCID50 IN Moderate Not reported Not done [50]
1.5–5.5 mths Clinical isolate hRSV 105.7–107 TCID50 Aerosol Low Slight fever & increased RR Foci of broncho-interstitial pneumonia [44], [45]
Young adults Macaque-adapted clinical isolate 105 pfu IN Moderate None Not done [51]



Bonnet monkeys (Macacca radiata) Juveniles hRSV Long 106–106.7 pfu IT or intra-bronchial Moderate Tachypnoea & chest retractions Foci of broncho-interstitial pneumonia & alveolitis [46], [47], [48]



Cynomolgus monkeys (Macacca fascicularis) 8–15 mths Macaque-adapted hRSV 106 TCID50 IT Low None Foci of broncho-interstitial pneumonia, alveolitis, syncytial cells [49]
4 mths, 1 yr, adults hRSV 105 TCID50 IN Low None Not done [52]
a

Animals were infected with different isolates of human respiratory syncytial virus (hRSV).

b

Animals were inoculated by the intranasal (IN), intra-tracheal (IT) or a combination of both IN and IT routes.

c

URT = upper respiratory tract.

Chimpanzees have been used to evaluate the virulence and protective efficacy of live, attenuated hRSV vaccine candidates [24], [30], [31]. The replication and genetic stability of the mutant viruses in chimpanzees parallels that in sero-negative children, and the mutants induced protection against subsequent wild-type hRSV challenge in chimpanzees [24], [32], [33], [34]. However, chimpanzees vaccinated with recombinant vaccinia viruses (rVV) expressing the hRSV surface glycoproteins F and G developed only low levels of neutralising antibodies, and were poorly protected against hRSV challenge [24], [35]. These findings contrast with the almost complete protection induced in mice, cotton rats and owl monkeys by the same rVV [24], [35].

3.1.2. African green monkeys

African green monkeys (AGMs), which are semi-permissive for hRSV replication (Table 1), have been used in a number of studies to evaluate hRSV vaccine candidates [36], [37], [38], [39], [40], [41]. Clinical signs of disease are uncommon following hRSV infection, and AGMs develop only mild histopathological changes in the lung [38], [42], [43], The proportion of neutrophils in BAL of monkeys infected with the Memphis M37 (M37) strain of hRSV increased only to 9%, [40], whereas neutrophils are the predominant cell in BAL from hRSV-infected children [14].

Vaccine-enhanced pulmonary pathology has been demonstrated following hRSV challenge of AGMs vaccinated with FI-hRSV [42]. Although FI-hRSV induced partial protection against hRSV replication, animals developed severe peribronchiolar and parenchymal inflammation. Enhanced histopathological changes were also induced in hRSV-infected AGMs that had been vaccinated with FI-herpes simplex virus (FI-HSV), although they were less severe than those in FI-hRSV animals. These findings suggest that an immune response to cell culture components present in the vaccine and the hRSV challenge may have contributed to the enhanced pathology seen in FI-hRSV vaccinated animals.

A comparison of the protective efficacy of a number of different non-replicating hRSV vaccines, all of which had been shown to induce complete protection against hRSV in mice and/or cotton rats, demonstrated varying efficacy in AGMs [40]. The vaccines included purified hRSV F and G proteins in different adjuvants, replication incompetent recombinant vesicular stomatitis virus expressing F or G, replication incompetent recombinant adenovirus (rAdV) expressing F or G, and DNA plasmids encoding F or G, administered on 3 occasions in various combinations. Only IN priming with rAdV-F/G followed by F/G protein boosting induced complete protection in both the URT and LRT. However, F/G proteins formulated in Alhydrogel and CpG induced a significant reduction in virus replication. There was no evidence of enhanced pulmonary inflammation in any of the vaccinated monkeys. These observations highlight the failure of small animal models to predict hRSV vaccine efficacy in AGMs.

3.1.3. Macaques

Three species of macaques (rhesus, cynomolgus and bonnet monkeys) have been experimentally infected with hRSV (Table 1). Although infection does not usually produce clinical signs of disease, mild interstitial pneumonia has been seen following IT or aerosol inoculation of high titre virus (>106 pfu) (Fig. 1F) [44], [45], [46], [47], [48], [49], [50], [51]. Macaques are not very permissive for hRSV replication, and susceptibility to infection does not appear to be influenced by age [52]. Viral loads are usually determined by RT-PCR. However, virus has been isolated from the respiratory tract in some studies, and viral mRNA was detected in alveolar and perivascular sites, but not in ciliated epithelial cells [46], [47], [48].

Vaccine-enhanced pulmonary pathology has been described in macaques [48], [49]. Pulmonary pathology in hRSV infected, FI-hRSV-vaccinated bonnet monkeys was associated with enhanced lung virus load, and characterised by peribronchiolar and perivascular accumulations of macrophages, lymphocytes, neutrophils and eosinophils, alveolitis and interstitial pneumonia [48]. However, the extent of pulmonary pathology in FI-hRSV vaccinated animals was not significantly different from that in monkeys vaccinated with FI-Vero cells, suggesting that sensitisation to non-viral antigens contributed to the vaccine-enhanced pathology. Antibody-mediated enhanced uptake and replication of hRSV in macrophages was proposed as the mechanism for increased lung viral load [53]. However, enhanced pulmonary pathology was not associated with increased virus replication in the lungs of FI-hRSV-immunised AGMs [42] or cynomolgus monkeys [49]. Vaccine-enhanced pulmonary pathology in FI-hRSV vaccinated cynomolgus monkeys was associated with pulmonary eosinophilia, and priming of T cells for production of IL-13, IL-5 and IL-2 [49]. Two vaccinated monkeys developed pulmonary hyperinflation and were moribund, 12 days after hRSV challenge. However, the lungs of these animals did not show overt inflammatory lesions or pulmonary eosinophilia. Following challenge with hRSV, lymphocytes from FI-hRSV-immunised monkeys produced IL-13 in response to stimulation with Vero cell lysate suggesting that sensitisation to non-viral antigens may have contributed to the vaccine-enhanced pulmonary pathology.

3.1.4. Other non-human primates

A number of other non-human primates (NHP) have been used as models of hRSV infection, although all are only semi-permissive for virus replication (Table 1).

HRSV-infected owl monkeys develop serous rhinorrhea, which is less severe than that seen in chimpanzees [32], [54] and virus replicates efficiently in the nasopharynx for 8–17 days [54]. However, IT inoculation with higher doses of hRSV (106 pfu) induced only minor histological lung changes [55]. Owl monkeys resembled chimpanzees in their response to infection with live, attenuated hRSV vaccine candidates [32], and therapeutic administration of human IgG containing virus neutralising (VN) antibodies significantly reduced lung viral load, but did not affect pulmonary pathology [56].

HRSV replicates in the URT of cebus monkeys following IN inoculation, but they do not develop clinical signs of disease [23]. However, following IT inoculation with 108 pfu, monkeys developed mild rhinorrhea and conjunctivitis, one developed coughing, dyspnea, anorexia and lethargy, there was a high virus lung load, and nearly all had evidence of pneumonic lesions by radiography [57]. The lungs of all animals had areas of gross pathological changes, histopathological changes were characterised by interstitial pneumonia, and an abundance of viral antigen was detected in ciliated epithelial cells of the nasal turbinate and bronchioles, and in alveolar cells. However, cebus monkeys do not appear to have been exploited for vaccine evaluation.

Infant baboons inoculated IT with a large dose of virus (107.9 pfu) developed clinical signs of LRTI, with tachypnoea, dyspnoea, and a decrease in blood oxygen saturation (<97%) in a proportion of animals [58]. However, virus titres in BAL were maximal 24 h after inoculation, and declined thereafter. Lungs showed vascular congestion and oedema, and microscopic lesions were characterised by interstitial pneumonia, sloughing of the bronchiolar epithelium, and obstruction of the bronchiolar lumen with inflammatory cells and sloughed epithelial cells. Viral antigen was mainly detected in lung parenchyma, and a few scattered epithelial cells. There was a rapid increase in BAL neutrophils 24 h after infection, which declined by day 3, and was followed by an increase in macrophage numbers in BAL. Further studies are needed to determine the extent to which the pulmonary inflammatory response is the result of virus replication or is a non-specific inflammatory response to the high dose inoculum.

3.2. Sheep

Neonatal lambs can be infected with high doses of hRSV (Table 2). Infection with hRSV A2 caused a slight fever and cough, macroscopic lung lesions characterised by multifocal areas of consolidation, and microscopic lesions similar to those of hRSV-infected children [59]. Viral antigen was detected in ciliated bronchial epithelial cells and syncytial cells (Fig. 1G and H). Viral mRNA in BAL increased from low levels at day 3 pi to peak at day 6 pi and was cleared by day 14. Lambs infected with high doses of hRSV M37 developed increased respiratory effort and wheezing [60], [61]. Virus titres in BAL increased 100-fold from day 1 to day 6, and infection resulted in increased numbers of macrophages, CD4+ and CD8+ cells, and levels of IFNγ, IL-8, MCP-1, MIP-1α, RANTES, and surfactant protein A (SP-A) mRNA increased in the lungs [60], [62].

Table 2.

Lamb and rodent models of hRSV infection.

Animal Age Virus straina Inoculum Routeb Viral replicationc Clinical signs of disease Pathology Reference
Neonatal lambs 2–3 days hRSV A2 108 pfu Intra-bronchial or aerosol RSV mRNA detected by RT-PCR Increased temperature, coughing Multifocal areas of pulmonary consolidation. Mild to moderate bronchiolitis, interstitial pneumonia [59], [62]
2–3 days hRSV M37 108 pfu Aerosol Moderate in LRT Increased respiratory effort wheezing Multifocal areas of pulmonary consolidation. Bronchitis/bronchiolitis, interstitial pneumonia, alveolitis [61]



BALB/c mice 4–32 wks hRSV A2 104.2–107 pfu IN Moderate Weight loss at high doses Mild to moderate bronchiolitis [80], [81]



Cotton rats Neonatal to adults hRSV Long 104 pfu IN Moderate None Mild, proliferative bronchiolitis [68]



Ferrets Neonatal to adults hRSV Long 103.5 pfu IN High in nose None Mild, focal desquamative rhinitis [104]
9–12 mths hRSV-A clinical isolate 105 TCID50 IT Moderate in LRT None Sporadic neutrophils in tracheal epithelium, bronchiolar & alveolar lumina [69]



Guinea pigs Adult hRSV Long 103.6 pfu IN Moderate None Bronchiolar epithelial necrosis, peribronchial mononuclear & neutrophil infiltrates [105]



Chinchillas Juvenile hRSV A2 107 pfu IN Moderate Ruffled fur & lethargy Mild inflammation of nasopharynx and eustachian tube [109], [110]



Syrian hamsters 3 wks hRSV A2 106 pfu IN Moderate None None [106]
a

Animals were infected with different isolates of human respiratory syncytial virus (hRSV).

b

Animals were inoculated by the intranasal (IN) route.

c

LRT = lower respiratory tract.

FI-hRSV vaccinated lambs were protected against virus infection and the development of macroscopic lesions [63]. Although there was a more extensive peribronchiolar accumulation of cells in vaccinated lambs, other features of pulmonary pathology were significantly reduced, and there was no evidence of exacerbation of disease. The effect of maternal immunisation on the levels of hRSV-specific antibodies and induction of resistance to hRSV infection in their offspring has been evaluated [64]. Maternal antibodies are transferred to lambs via colostrum and immunisation of pregnant ewes with hRSV F protein in adjuvant induced high levels of virus neutralising (VN) antibodies in serum and colostrum. Lambs receiving colostrum from vaccinated ewes had VN titres (VNT) about 50-fold higher than those in lambs born to non-vaccinated ewes [64]. Following hRSV challenge of lambs at 2–3 days of age, lung viral loads were reduced by ∼70% and there was a significant reduction in pulmonary pathology in lambs born to vaccinated ewes compared with controls. These studies suggest that maternal immunisation is a safe and effective approach to increase protection against hRSV LRTI in young infants.

3.3. Rodents

Cotton rats and mice have been used extensively as models of hRSV infection and have provided insights into mechanisms of immunity to and the pathogenesis of hRSV infections [65], [66].

3.3.1. Cotton rats

Although cotton rats are semi-permissive for virus replication, they are about 100-fold more permissive than BALB/c mice per inoculum dose of virus, but they do not develop clinical signs of disease (Table 2) [67]. Following IN inoculation, hRSV replicates to high titres in the nose and lungs, but to lower titres in the trachea. Cotton rats are susceptible to infection throughout life, but virus replication is greater and persists for longer in the nasal passages of 3 day-old rats than in older animals. Viral antigen can be detected in ciliated epithelial cells of the nose and lungs, but not trachea [68], [69], [70], and induces an increase in IFNγ, IL-10, IL-6, MCP-1 and growth-regulated oncogene (GRO) (IL-8 homologue) mRNA in the lungs [71], and histopathological changes characterised by a desquamative, exudative rhinitis, and a mild proliferative bronchiolitis (Fig. 1I) [68], [72]. Virus loads are greater and virus persists for longer in cotton rats immunosuppressed by cyclophosphamide, and animals develop a more severe pulmonary inflammatory response characterised by foamy macrophages, and bronchial and tracheal intraluminal debris [73], [74].

There is a direct correlation between levels of VN serum antibodies associated with protection in cotton rats and infants. Serum VNT of >1:380 are protective in passively immunised cotton rats and similar levels of maternally derived serum antibodies (MDA) in human infants <2 months old correlate with increased resistance to hRSV LRTI [75]. The cotton rat has played an important role in the development of mAb prophylaxis for the prevention of LRTI in infants at high risk of severe hRSV disease [75], [76], and is a well-established model of FI-hRSV vaccine-enhanced disease. Although FI-hRSV vaccination reduced hRSV replication, cotton rats developed an enhanced peribronchiolitis, alveolitis and interstitial pneumonitis [77]. Whereas peribronchiolitis is thought to be a normal component of the immune-mediated resolution of hRSV infection, alveolitis is considered to be the primary marker of vaccine-enhanced disease in the cotton rat model [65]. However, the severity of alveolitis in FI-hRSV-vaccinated cotton rats is associated with priming of T cells specific for non-viral antigens, and this response is potentiated by hRSV antigens [72]. This is reminiscent of the response of T cells from FI-hRSV vaccinated macaques, but not of those from monkeys vaccinated with FI-measles virus, which responded to vero cell antigen following hRSV challenge [49]. These findings indicate that caution should be used when extrapolating results from this model to assess the safety of human vaccine candidates.

3.3.2. Mice

Although inbred mouse strains vary in their susceptibility to experimental hRSV infection [78], the BALB/c mouse, which shows intermediate susceptibility, has been widely used to study the pathogenesis of and mechanisms of immunity to hRSV [66]. Following IN inoculation, hRSV replicates in the nasal passages and lungs (Table 2) [78], [79]. Although mice are susceptible to hRSV infection throughout life, older mice are more susceptible than younger animals [80], [81]. High (>106 pfu) doses of hRSV are needed to induce clinical signs of disease, which is characterised by weight loss, ruffled fur and a hunched posture. Microscopic lesions consist of perivascular and peribronchiolar accumulations of mononuclear cells [80] and increased mucus production. Levels of pro-inflammatory cytokines and chemokines are increased during the acute phase of infection [82], [83], [84]. Both CD4+ and CD8+ T cells play a role in virus clearance and contribute to disease. Depletion of either subset, or both, prolongs virus infection and reduces weight loss, illness, and pulmonary pathology [85]. These observations contrast with the prolonged viral shedding, severe respiratory disease, and giant cell pneumonia seen in immunosuppressed humans [4].

Laboratory strains of hRSV replicate primarily in type I alveolar pneumocytes. However, there is extensive replication of hRSV in bronchiolar epithelial cells in mice deficient in type I IFN pathways [82] suggesting that the mechanisms that hRSV employs to evade type I IFN responses are poorly effective in the mouse. Some clinical isolates also infect bronchial epithelial cells in mice, and clinical isolates vary in their virulence in mice [86]. HRSV infection of mice induces airway obstruction and airway hyper-responsiveness (AHR) in response to methacholine [83], and the mouse model has been used to determine the immunological mechanisms that result in virally-induced AHR (reviewed [11], [87]).

FI-hRSV vaccine-enhanced disease in mice is associated with priming of a Th2 response and is characterised by pulmonary eosinophilia, increased pulmonary pathology, mucus hypersecretion, weight loss, increased airway obstruction and AHR, and a reduction in lung viral load [88], [89]. Depletion of CD4+ cells or IL-4 and IL-10 abrogates vaccine-enhanced pathology [90], [91]. Low avidity, non-neutralising antibodies induced by FI-hRSV vaccination of mice have also been shown to contribute to enhanced AHR and pulmonary pathology [92], [93]. As seen in other animal models, immune responses to non-viral antigens contribute to vaccine-enhanced disease in the mouse [72], [94], [95]. However, a study in which vaccine virus was produced in serum-free medium in different cells from that used for challenge virus, showed that FI-hRSV primed both Th1 and Th2 responses. Th2 responses mediated enhanced pulmonary pathology, pulmonary eosinophilia, mucus hypersecretion and AHR, whereas TNF-α contributed to airway obstruction and weight loss; eosinophils did not contribute to any disease parameters associated with FI-hRSV vaccine-enhanced disease in mice [89].

The protective effects of mAbs, in vivo, was first demonstrated in the mouse model of hRSV [96], and led to the development of a protective, humanised mAb specific for the RSV F protein [97] and ultimately to the development of Palivizumab prophylaxis. The mouse model of hRSV is useful for the negative selection of vaccine candidates. However, since there are many differences in innate and adaptive immune responses between mice and humans [98], the responses of mice to vaccination may differ substantially from those which occur in higher species. For example, recombinant vaccinia Ankara (MVA) expressing the hRSV F or G proteins completely protects mice from hRSV infection [99], but did not induce protection in macaques [100].

Infection of a humanised mouse model, which lacks functional CD8+ T cells, with 106 pfu of hRSV line 19 induced weight loss and pulmonary pathology characterised by peribronchiolar inflammation, a predominance of neutrophils in BAL, and enhanced mucus production [101]. The predominance of neutrophils in BAL is similar to that in hRSV-infected children, and contrasts with hRSV-infection of BALB/c mice, where the proportion of neutrophils in BAL is usually ∼10% [102]. A humanised mouse model may be a more relevant model for evaluating hRSV vaccine candidates than conventional mice.

3.3.3. Ferrets

Following IN infection, HRSV replicates in the nasal passages of ferrets of all ages, but only in the lungs of infant animals (Table 2) [103], [104]. However, replication of hRSV in the LRT of adult ferrets, with viral antigen seen in ciliated epithelial cells of the trachea and bronchi has been demonstrated following IT inoculation with a low passage, clinical isolate of hRSV [69]. Viral antigen could be detected in bronchiolar epithelial cells only in immunosuppressed ferrets, in which virus replicated to higher titres and clearance was delayed [69]. Although ferrets did not develop clinical signs of disease, sporadic neutrophils were seen in the tracheal epithelium, and the bronchiolar and alveolar lumina. The possible transmission of infection from infant ferrets to their mothers [104], suggests that ferrets could be used to determine the ability of vaccines to block virus transmission.

3.3.4. Other rodents

Although guinea pigs can be experimentally infected with hRSV, and develop histopathological changes characterised by bronchiolar epithelial necrosis, peribronchial mononuclear and neutrophil infiltrates, and necrotic cells and debris in some bronchioles, they have not been used to evaluate hRSV vaccine efficacy (Table 2) [105].

Syrian hamsters are semi-permissive for hRSV and although virus replicates in the URT and LRT, they are ∼100-fold less permissive than cotton rats (Table 2) [106], and infection does not produce clinical signs of disease or microscopic evidence of pulmonary pathology [106]. Hamsters have been used to evaluate the protective efficacy of recombinant parainfluenza viruses expressing hRSV surface glycoproteins [107], [108].

Chinchillas can also be infected with high doses of hRSV, and a small inoculum volume restricts infection to the URT (Table 2) [109]. Virus was isolated from the nasopharynx for at least 8 days, and infection resulted in mild multifocal neutrophilic and lymphocytic infiltrates of the nasal, goblet cell hyperplasia, hypersecretion of mucus in the Eustachian tube lumen, a mild retraction of the tympanic membrane and a decrease in middle ear pressure. Virus was detected in ciliated epithelial cells of the nasopharynx, nasoturbinates, and Eustachian tube [110]. The chinchilla may be a useful model for evaluation of vaccines to control hRSV infection of the URT, and to investigate the mechanisms by which hRSV infection predisposes to middle ear bacterial infections in children.

4. Non-human pneumovirus animal models

An advantage of studying non-human pneumoviruses in their native host is that the host is fully permissive. Studies of bovine (b)RSV [111], and pneumonia virus of mice (PVM) [112] in their natural host have increased our understanding of pneumovirus pathogenesis and support vaccine development concepts.

4.1. BRSV in calves

BRSV is a leading cause of respiratory disease in young calves, worldwide [113]. The epidemiology and pathogenesis of naturally occurring bRSV infections are similar to those of hRSV in children. The peak incidence of severe disease is in calves <6 months of age and, as seen in children, MDA do not protect against bRSV infection, but the incidence and severity of respiratory disease is inversely related to the level of maternally-derived VN antibodies [114]. The severity of infection can range from subclinical, to severe with LRTI, and death. BRSV-infected calves are also susceptible to secondary bacterial infections, although some outbreaks can be attributed to bRSV alone. In the UK, 70% of calves have been infected by 9 months of age, and although cattle can be reinfected with bRSV, disease following reinfection is usually subclinical or mild. However, primary bRSV infections causing severe disease have been reported in older cattle [115]. The incubation period is estimated to be between 2 and 5 days, and clinical signs include mild depression, nasal seromucoid discharge, cough, tachypnea, dyspnoea, and fever, with abnormal breath on ausculation. Macroscopic lung lesions have a mainly cranioventral distribution and are characterised by areas of consolidation. BRSV replicates primarily in ciliated airway epithelial cells and type II pneumocytes [116] (Fig. 1C), and induces microscopic lesions of bronchointerstitial pneumonia, epithelial necrosis, an exudative or proliferative alveolitis, and occasional syncytial cells (Fig. 1D). The lumen of the bronchi and bronchioles are obstructed by cellular debris consisting of neutrophils, desquamated epithelial cells, macrophages, and mucin [117].

Although bRSV is genetically and antigenically closely related to hRSV (Table 3), bRSV is highly restricted in non-human primates [118]. The similarities in the epidemiology and pathogenesis of hRSV and bRSV infections, and the close antigenic relationship between these viruses, make bRSV infection in calves a relevant animal model of hRSV. Initial attempts to induce clinical signs of respiratory disease in calves experimentally infected with bRSV yielded inconsistent results [111]. However, reproducible clinical signs of disease have been produced in calves following inoculation with ∼104 pfu of low- or non-cell-culture-passaged bRSV (Table 4) [119], [120], [121], [122]. The calf allows daily sampling of the URT, and virus replication in the LRT and the inflammatory response can be analysed in BAL or in the lung tissue at post-mortem examination. Virus titres in the URT peak at ∼104 pfu/ml and at ∼105 pfu/ml in BAL, and virus is normally cleared by day 8–9. Clinical signs of disease usually appear between day 5 and 6 and are maximal at day 7–8. Macroscopic and microscopic lesions of experimentally infected calves are similar to those observed in naturally occurring bRSV infections (Fig. 1), and can be induced in the absence of other respiratory pathogens. Experimentally infected calves have a significant decrease in arterial oxygen tension and a diffuse pattern of patchy consolidation can be detected by thoracic radiography [120], [121]. BRSV infection induces increased levels of pro-inflammatory cytokines and chemokines, and a marked increase in neutrophils in BAL similar to that in children with hRSV bronchiolitis [117], [123], [124], [125].

Table 3.

Amino acid sequence identity between the protein of hRSV subgroup A, hRSV subgroup B (hRSV-B), bovine (b)RSV and pneumonia virus of mice (PVM).

Viral protein % Amino acid sequence identity
hRSV-B bRSV PVM
NS1 Non-structural protein 1 87 69 16
NS2 Non-structural protein 2 92 84 20
N Nucleoprotein 96 93 60
P Phosphoprotein 91 81 33
M Matrix protein 91 89 42
SH Small hydrophobic protein 76 38 23
G Attachment glycoprotein 53 30 12
F Fusion protein 89 81 43
M2-1 Matrix protein 2–1 92 80 43
M2-2 Matrix protein 2–2 72 42 10
L RNA polymerase 93 84 53

Table 4.

Models utilising non-human pneumoviruses.

Animal Age Virus straina Inoculum Routeb Viral replicationc Clinical signs of disease Pathology Reference
Calves 3–6 wks bRSV Snook 103.7–104 pfu IN & IT or aerosol Moderate Nasal discharge, raised respiratory rate, cough, fever, dyspnoea Macroscopic lung lesions. Broncho-interstitial pneumonia, epithelial necrosis, exudative or proliferative alveolitis, occlusion of bronchiolar lumina [122], [135]



BALB/c mice Adult PVM J3666 120 pfu IN High in LRT Weight loss, hunched posture, ruffled fur, laboured breathing, cyanosis, death Multifocal acute alveolitis, intra-alveolar oedema, scattered haemorrhage, moderate granulocytic infiltrates [152]
Adult PVM 15 300–600 pfu IN High in LRT Weight loss, hunched posture, ruffled fur Broadly dispersed lesions with cellular infiltrates in the alveolar space and surrounding tissues & oedema [151], [153]
a

Animals were infected with bovine respiratory syncytial virus (bRSV) or pneumonia virus of mice (PVM).

b

Animals were inoculated by the intranasal (IN), or a combination of both IN and intratracheal (IT) routes.

c

LRT = lower respiratory tract.

CD8+ T cells are important in virus clearance of bRSV from both the URT and LRT and, in contrast to T-cell depleted hRSV-infected mice, prolonged bRSV infection in CD8+-depleted calves is associated with more severe pulmonary pathology [126]. Neutralising mAbs to antigenic sites corresponding to amino acids 255–275 (antigenic site II) and 417–438 (antigenic site IV) of the F protein neutralise both hRSV and bRSV, and passive immunisation with bovine mAbs specific for these antigenic sites protect calves from bRSV LRTI [127], [128].

There is evidence of vaccine-enhanced disease in calves following a natural bRSV infection. Severe disease with a high mortality was seen in 34% of 7 month-old calves vaccinated with a commercial β-propiolactone-inactivated bRSV vaccine, whereas disease in non-vaccinated, 2–3 month-old calves was less severe and no animals died [129]. BRSV antigen was detected in the lungs of the vaccinated calves and lung lesions were characterised by interstitial pneumonia, with bronchitis and bronchiolitis obliterans, pneumocyte type II hyperplasia, syncytial cell, and an infiltration of neutrophils and eosinophils. These features of altered age distribution for severe disease and the pattern of lung pathology were similar to those seen in FI-hRSV- vaccinated infants [9]. Exacerbated disease has also been seen in calves vaccinated IM with a live, attenuated bRSV vaccine administered during a concurrent bRSV infection [130]. However, attempts to experimentally reproduce vaccine-enhanced disease with FI-bRSV have yielded conflicting results. Some studies have demonstrated enhanced disease with pulmonary eosinophilia in vaccinated calves [131], whereas others have not reported an increase in eosinophils [132]. FI-bRSV vaccination has induced varying levels of protection against infection, and either enhanced pulmonary pathology, or an earlier onset of disease followed by a faster resolution and less severe pathology [120], [131], [132], [133]. These differences may be related to the vaccine antigen dose, the level of MDA at vaccination, and/or the interval between vaccination and challenge. An advantage of using the calf as a model of vaccine-enhanced disease is that virus for vaccine production and challenge are grown in bovine cells maintained in calf serum, so there is less likelihood that calves will be sensitised to foreign cell culture antigens.

The calf model of bRSV infection has been used to evaluate the virulence, immunogenicity and protective efficacy of a variety of recombinant bRSV in which various genes have been deleted [134], [135], [136]. The findings from these studies are similar to those seen in chimpanzees infected with analogous mutants of hRSV [31]. Calves have been used to evaluate hRSV vaccine concepts such as DNA vaccines [137], subunit vaccines [138], immunostimulating complexes (ISCOMs) [139], live attenuated viruses [140], recombinant virus vectors [141], [142], and novel adjuvants with inactivated bRSV [143], and to determine the effects of maternal antibodies on vaccination [144]. Calves can also be part of the preclinical assessment of hRSV vaccine candidates which contain proteins that are conserved between hRSV and bRSV. For example, calves vaccinated IN with rAdV expressing a fusion protein consisting of the hRSV F, N and M2-1 proteins and boosted IM with MVA expressing the same antigens were completely protected against subsequent bRSV challenge [145]. These same vaccine vectors can effectively boost immune responses in healthy adult volunteers [146]. Furthermore, the ability to repeatedly sample different regions of the respiratory tract of the same animal during the course of bRSV vaccination and challenge studies could contribute to a greater understanding of mucosal immunity and identification of correlates of protection against bRSV infection.

4.2. PVM in mice

PVM was originally isolated form mouse lungs and is a common pathogen of laboratory mouse colonies [147]. Experimental infection of mice with PVM results in disease that varies in severity depending on the virus dose, mouse strain, and age [148], [149], [150]. BALB/c mice are susceptible and C57BL/6 mice are relatively resistant. Following IN inoculation of BALB/c mice with low doses (60–600 pfu) of virulent strains of PVM, virus replicates to high titres in the lung, causing severe morbidity with weight loss, ruffled fur, hunched posture, laboured breathing, and cyanosis, and 100% mortality (Table 4) [151], [152]. Virus replication occurs initially in alveolar cells and subsequently in bronchial epithelial cells [152], [153], and induces an early increase in eosinophils in BAL followed by a predominantly neutrophil response [154]. Lung histopathological changes consist of alveolar epithelial cell apoptosis, bronchial epithelial necrosis, multifocal acute alveolitis, intra-alveolar oedema, haemorrhage, and granulocytic infiltration. The pro-inflammatory chemokines and cytokine responses in the lungs of PVM-infected mice are similar to those of hRSV-infected mice [155]. However MIP-1α, which controls the inflammatory response induced by PVM [156], does not control the inflammatory response induced by hRSV in mice [157]. These findings highlight differences in the pathogenesis of pneumoviruses in a natural host and in an unnatural, semi-permissive animal model. Both CD4+ and CD8+ T cells contribute to virus clearance, clinical disease and pulmonary pathology in mice infected with sub-lethal doses of PVM. However, CD8+ T cells are largely responsible for the cytokine storm [158].

Enhanced disease in FI-PVM-vaccinated mice is associated with a Th2-biased response, production of IL-4, IL-5, IL-13, and pulmonary eosinophilia [159]. In contrast, neutrophils predominate in BAL from mice vaccinated with FI-uninfected cell antigen, and Th2 cytokine levels are low. However, mice vaccinated with FI-PVM had a reduced lung virus load and were partially protected against weight loss, despite the absence of VN antibodies. Sensitisation to non-virus antigens are unlikely to have contributed to enhanced pathology in FI-PVM-vaccinated mice as the challenge virus had been passaged in vivo, and was therefore devoid of foreign non-viral antigens that could cross-react with those in the PVM vaccine. Whilst hRSV vaccine approaches can be studied in the mouse model of PVM, and vaccine candidates can be negatively selected, their ability to predict efficacy in higher species is not known. IN vaccination of mice with rAdV expressing either the M or N protein of PVM protected mice against a lethal challenge dose of PVM, although mice developed a transient weight loss [160].

Whilst the advantage of the mouse model of PVM is the ability to study a pneumovirus infection in a natural host, the many differences in the innate and adaptive immune response between mice and humans, the differences in lung anatomy, and the antigenic differences in the viral proteins, limits its relevance to hRSV in humans. Nevertheless, an understanding of the pathogenesis and mechanisms of immunity to PVM can inform studies in higher animal species and man.

5. Concluding remarks

The natural progression of hRSV infection in humans, which is probably initiated by exposure to small doses of virus in the URT, is bypassed in the majority of animal models of hRSV infection, where large doses of virus are introduced directly into the lungs. As a result, the innate immune defences in the LRT are rapidly mobilised to deal with a large load of foreign antigen, which will impact on the timing and magnitude of the adaptive immune response. This has consequences for understanding the pathogenesis of hRSV infections and for evaluating the protective efficacy and safety of vaccines. Immunopathology is likely to result from a very high viral load encountering large numbers of virus-specific effector cells and/or poorly neutralising antibodies in the lungs. In the normal course of events, the initial period of virus replication in the URT provides an opportunity for the adaptive immune response to be activated prior to the accumulation of large virus loads in the lungs. Even in non-human pneumovirus animal models, virus is delivered directly to the lungs in order to ensure reproducible LRTI.

Interpretation of findings from animal models of vaccine-enhanced disease has been confounded by sensitisation to non-viral antigens in the vaccine and challenge virus. In order to minimise priming of lymphocytes to non-viral antigens in vaccine formulations that might confound results obtained from challenge of vaccinated animals, the cells and medium components used to produce vaccine preparations and virus challenge should be considered carefully.

The chimpanzee is the only animal host that is fully permissive to hRSV replication, allowing animal-to-animal transmission, and in which infection reliably produces URT disease. However, the close genetic and antigenic relatedness of hRSV and bRSV, and the similarities between hRSV in children and bRSV infection in calves suggest that the calf model has many advantages. However, virulent bRSV is difficult to grow to high titres in cell culture, calves require specialised housing, and the availability of reagents to dissect the immune response is not as extensive as that for mice and humans. Mice and cotton rats can be used for the negative selection of vaccine candidates. However, the extent to which they predict vaccine efficacy in man is not clear; therefore higher animal species should also be used for preclinical evaluation of new vaccine candidates.

Conflict of interest

The author has no conflict of interest to declare.

Acknowledgements

GT is a Jenner Investigator and is supported by the BBSRC Institute Strategic Programme on Livestock Viral Diseases at The Pirbright Institute.

References

  • 1.Nair H. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet. 2010;375(9725):1545–1555. doi: 10.1016/S0140-6736(10)60206-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hall C.B. Immunity to and frequency of reinfection with respiratory syncytial virus. J Infect Dis. 1991;163(4):693–698. doi: 10.1093/infdis/163.4.693. [DOI] [PubMed] [Google Scholar]
  • 3.Falsey A.R., Walsh E.E. Viral pneumonia in older adults. Clin Infect Dis. 2006;42(4):518–524. doi: 10.1086/499955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hall C.B., Simoes E.A., Anderson L.J. Clinical and epidemiologic features of respiratory syncytial virus. Curr Top Microbiol Immunol. 2013;372:39–57. doi: 10.1007/978-3-642-38919-1_2. [DOI] [PubMed] [Google Scholar]
  • 5.Chemaly R.F., Shah D.P., Boeckh M.J. Management of respiratory viral infections in hematopoietic cell transplant recipients and patients with hematologic malignancies. Clin Infect Dis. 2014;59(Suppl. 5):S344–S351. doi: 10.1093/cid/ciu623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Falsey A.R. Respiratory syncytial virus infection in elderly and high-risk adults. N Engl J Med. 2005;352(17):1749–1759. doi: 10.1056/NEJMoa043951. [DOI] [PubMed] [Google Scholar]
  • 7.Fleming D.M., Elliot A.J. Respiratory syncytial virus: a sleeping giant? Eur Respir J. 2007;30(6):1029–1031. doi: 10.1183/09031936.00120707. [DOI] [PubMed] [Google Scholar]
  • 8.Cardenas S., Auais A., Piedimonte G. Palivizumab in the prophylaxis of respiratory syncytial virus infection. Expert Rev Anti Infect Ther. 2005;3(5):719–726. doi: 10.1586/14787210.3.5.719. [DOI] [PubMed] [Google Scholar]
  • 9.Kim H.W. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol. 1969;89(4):422–434. doi: 10.1093/oxfordjournals.aje.a120955. [DOI] [PubMed] [Google Scholar]
  • 10.Ralston S., Hill V. Incidence of apnea in infants hospitalized with respiratory syncytial virus bronchiolitis: a systematic review. J Pediatr. 2009;155(5):728–733. doi: 10.1016/j.jpeds.2009.04.063. [DOI] [PubMed] [Google Scholar]
  • 11.Lotz M.T., Moore M.L., Peebles R.S., Jr Respiratory syncytial virus and reactive airway disease. Curr Top Microbiol Immunol. 2013;372:105–118. doi: 10.1007/978-3-642-38919-1_5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Johnson J.E. The histopathology of fatal untreated human respiratory syncytial virus infection. Mod Pathol. 2007;20(1):108–119. doi: 10.1038/modpathol.3800725. [DOI] [PubMed] [Google Scholar]
  • 13.Welliver T.P. Severe human lower respiratory tract illness caused by respiratory syncytial virus and influenza virus is characterized by the absence of pulmonary cytotoxic lymphocyte responses. J Infect Dis. 2007;195(8):1126–1136. doi: 10.1086/512615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Everard M.L. Analysis of cells obtained by bronchial lavage of infants with respiratory syncytial virus infection. Arch Dis Child. 1994;71(5):428–432. doi: 10.1136/adc.71.5.428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Smith P.K. Leucocyte populations in respiratory syncytial virus-induced bronchiolitis. J Paediatr Child Health. 2001;37(2):146–151. doi: 10.1046/j.1440-1754.2001.00618.x. [DOI] [PubMed] [Google Scholar]
  • 16.Rosenberg H.F., Domachowske J.B. Inflammatory responses to respiratory syncytial virus (RSV) infection and the development of immunomodulatory pharmacotherapeutics. Curr Med Chem. 2012;19(10):1424–1431. doi: 10.2174/092986712799828346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Brearey S.P., Smyth R.L. Pathogenesis of RSV in children. In: Cane P., editor. Respiratory syncytial virus. Elsevier; 2007. pp. 141–162. [Google Scholar]
  • 18.Stoppelenburg A.J. Elevated Th17 response in infants undergoing respiratory viral infection. Am J Pathol. 2014;184(5):1274–1279. doi: 10.1016/j.ajpath.2014.01.033. [DOI] [PubMed] [Google Scholar]
  • 19.Choi E.H., Lee H.J., Chanock S.J. Human genetics and respiratory syncytial virus disease: current findings and future approaches. Curr Top Microbiol Immunol. 2013;372:121–137. doi: 10.1007/978-3-642-38919-1_6. [DOI] [PubMed] [Google Scholar]
  • 20.Hull J. Genetic susceptibility to RSV disease. In: Cane P., editor. Respiratory syncytial virus. Elsevier; 2007. pp. 115–139. [Google Scholar]
  • 21.Thompson T.M. Viral specific factors contribute to clinical respiratory syncytial virus disease severity differences in infants. Clin Microbiol. 2015;4(3) doi: 10.4172/2327-5073.1000206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Morris J.A., Blount R.E., Jr, Savage R.E. Recovery of cytopathogenic agent from chimpanzees with coryza. Proc Soc Exp Biol Med. 1956;92(3):544–549. doi: 10.3181/00379727-92-22538. [DOI] [PubMed] [Google Scholar]
  • 23.Belshe R.B. Experimental respiratory syncytial virus infection of four species of primates. J Med Virol. 1977;1(3):157–162. doi: 10.1002/jmv.1890010302. [DOI] [PubMed] [Google Scholar]
  • 24.Crowe J.E., Jr A comparison in chimpanzees of the immunogenicity and efficacy of live attenuated respiratory syncytial virus (RSV) temperature-sensitive mutant vaccines and vaccinia virus recombinants that express the surface glycoproteins of RSV. Vaccine. 1993;11(14):1395–1404. doi: 10.1016/0264-410x(93)90168-w. [DOI] [PubMed] [Google Scholar]
  • 25.Clarke C.J. Respiratory syncytial virus-associated bronchopneumonia in a young chimpanzee. J Comp Pathol. 1994;110(2):207–212. doi: 10.1016/s0021-9975(08)80191-0. [DOI] [PubMed] [Google Scholar]
  • 26.Szentiks C.A. Lethal pneumonia in a captive juvenile chimpanzee (Pan troglodytes) due to human-transmitted human respiratory syncytial virus (HRSV) and infection with Streptococcus pneumoniae. J Med Primatol. 2009;38(4):236–240. doi: 10.1111/j.1600-0684.2009.00346.x. [DOI] [PubMed] [Google Scholar]
  • 27.Unwin S., Chatterton J., Chantrey J. Management of severe respiratory tract disease caused by human respiratory syncytial virus and Streptococcus pneumoniae in captive chimpanzees (Pan troglodytes) J Zoo Wildl Med. 2013;44(1):105–115. doi: 10.1638/1042-7260-44.1.105. [DOI] [PubMed] [Google Scholar]
  • 28.Kondgen S. Pandemic human viruses cause decline of endangered great apes. Curr Biol. 2008;18(4):260–264. doi: 10.1016/j.cub.2008.01.012. [DOI] [PubMed] [Google Scholar]
  • 29.Nguyen D.T. Streptococcus pneumoniae enhances human respiratory syncytial virus infection in vitro and in vivo. PLoS ONE. 2015;10(5):e0127098. doi: 10.1371/journal.pone.0127098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Crowe J.E., Jr A further attenuated derivative of a cold-passaged temperature-sensitive mutant of human respiratory syncytial virus retains immunogenicity and protective efficacy against wild-type challenge in seronegative chimpanzees. Vaccine. 1994;12(9):783–790. doi: 10.1016/0264-410x(94)90286-0. [DOI] [PubMed] [Google Scholar]
  • 31.Collins P.L., Murphy B.R. New generation live vaccines against human respiratory syncytial virus designed by reverse genetics. Proc Am Thorac Soc. 2005;2(2):166–173. doi: 10.1513/pats.200501-011AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Richardson L.S. Evaluation of five temperature-sensitive mutants of respiratory syncytial virus in primates: I. Viral shedding, immunologic response, and associated illness. J Med Virol. 1978;3(2):91–100. doi: 10.1002/jmv.1890030202. [DOI] [PubMed] [Google Scholar]
  • 33.Crowe J.E., Jr 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;13(9):847–855. doi: 10.1016/0264-410x(94)00074-w. [DOI] [PubMed] [Google Scholar]
  • 34.Karron R.A., Buchholz U.J., Collins P.L. Live-attenuated respiratory syncytial virus vaccines. Curr Top Microbiol Immunol. 2013;372:259–284. doi: 10.1007/978-3-642-38919-1_13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Collins P.L. Evaluation in chimpanzees of vaccinia virus recombinants that express the surface glycoproteins of human respiratory syncytial virus. Vaccine. 1990;8(2):164–168. doi: 10.1016/0264-410x(90)90141-8. [DOI] [PubMed] [Google Scholar]
  • 36.Jin H. Evaluation of recombinant respiratory syncytial virus gene deletion mutants in African green monkeys for their potential as live attenuated vaccine candidates. Vaccine. 2003;21(25–26):3647–3652. doi: 10.1016/s0264-410x(03)00426-2. [DOI] [PubMed] [Google Scholar]
  • 37.Le Nouen C. Attenuation of human respiratory syncytial virus by genome-scale codon-pair deoptimization. Proc Natl Acad Sci USA. 2014;111(36):13169–13174. doi: 10.1073/pnas.1411290111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jones B.G. Sendai virus-based RSV vaccine protects African green monkeys from RSV infection. Vaccine. 2012;30(5):959–968. doi: 10.1016/j.vaccine.2011.11.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tang R.S. 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;78(20):11198–11207. doi: 10.1128/JVI.78.20.11198-11207.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Eyles J.E. Nonreplicating vaccines can protect African green monkeys from the Memphis 37 strain of respiratory syncytial virus. J Infect Dis. 2013;208(2):319–329. doi: 10.1093/infdis/jit169. [DOI] [PubMed] [Google Scholar]
  • 41.Bates J.T. Immunogenicity and efficacy of alphavirus-derived replicon vaccines for respiratory syncytial virus and human metapneumovirus in nonhuman primates. Vaccine. 2016;34(7):950–956. doi: 10.1016/j.vaccine.2015.12.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kakuk T.J. A human respiratory syncytial virus (RSV) primate model of enhanced pulmonary pathology induced with a formalin-inactivated RSV vaccine but not a recombinant FG subunit vaccine. J Infect Dis. 1993;167(3):553–561. doi: 10.1093/infdis/167.3.553. [DOI] [PubMed] [Google Scholar]
  • 43.Ispas G. Antiviral activity of TMC353121, a Respiratory Syncytial Virus (RSV) fusion inhibitor, in a non-human primate model. PLoS ONE. 2015;10(5):e0126959. doi: 10.1371/journal.pone.0126959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.McArthur-Vaughan K., Gershwin L.J. A rhesus monkey model of respiratory syncytial virus infection. J Med Primatol. 2002;31(2):61–73. doi: 10.1034/j.1600-0684.2002.01006.x. [DOI] [PubMed] [Google Scholar]
  • 45.Vaughan K., Rhodes G.H., Gershwin L.J. DNA immunization against respiratory syncytial virus (RSV) in infant rhesus monkeys. Vaccine. 2005;23(22):2928–2942. doi: 10.1016/j.vaccine.2004.10.046. [DOI] [PubMed] [Google Scholar]
  • 46.Babu P.G. A primate model of respiratory syncytial virus infection. Indian J Exp Biol. 1998;36(8):758–762. [PubMed] [Google Scholar]
  • 47.Simoes E.A. Respiratory syncytial virus infects the Bonnet monkey, Macaca radiata. Pediatr Dev Pathol. 1999;2(4):316–326. doi: 10.1007/s100249900129. [DOI] [PubMed] [Google Scholar]
  • 48.Ponnuraj E.M. Increased replication of respiratory syncytial virus (RSV) in pulmonary infiltrates is associated with enhanced histopathological disease in bonnet monkeys (Macaca radiata) pre-immunized with a formalin-inactivated RSV vaccine. J Gen Virol. 2001;82(Pt 11):2663–2674. doi: 10.1099/0022-1317-82-11-2663. [DOI] [PubMed] [Google Scholar]
  • 49.De Swart R.L. Immunization of macaques with formalin-inactivated respiratory syncytial virus (RSV) induces interleukin-13-associated hypersensitivity to subsequent RSV infection. J Virol. 2002;76(22):11561–11569. doi: 10.1128/JVI.76.22.11561-11569.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Weltzin R. Intranasal monoclonal IgA antibody to respiratory syncytial virus protects rhesus monkeys against upper and lower respiratory tract infection. J Infect Dis. 1996;174(2):256–261. doi: 10.1093/infdis/174.2.256. [DOI] [PubMed] [Google Scholar]
  • 51.Grunwald T. Novel vaccine regimen elicits strong airway immune responses and control of respiratory syncytial virus in nonhuman primates. J Virol. 2014;88(8):3997–4007. doi: 10.1128/JVI.02736-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Grandin C. Evidence for an intranasal immune response to human respiratory syncytial virus infection in cynomolgus macaques. J Gen Virol. 2015;96(Pt 4):782–792. doi: 10.1099/vir.0.000039. [DOI] [PubMed] [Google Scholar]
  • 53.Ponnuraj E.M. Antibody-dependent enhancement, a possible mechanism in augmented pulmonary disease of respiratory syncytial virus in the Bonnet monkey model. J Infect Dis. 2003;187(8):1257–1263. doi: 10.1086/374604. [DOI] [PubMed] [Google Scholar]
  • 54.Prince G.A. Respiratory syncytial virus infection in owl monkeys: viral shedding, immunological response, and associated illness caused by wild-type virus and two temperature-sensitive mutants. Infect Immun. 1979;26(3):1009–1013. doi: 10.1128/iai.26.3.1009-1013.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hemming V.G. Studies of passive immunotherapy for infections of respiratory syncytial virus in the respiratory tract of a primate model. J Infect Dis. 1985;152(5):1083–1087. doi: 10.1093/infdis/152.5.1083. [DOI] [PubMed] [Google Scholar]
  • 56.Hemming V.G. Hyperimmune globulins in prevention and treatment of respiratory syncytial virus infections. Clin Microbiol Rev. 1995;8(1):22–33. doi: 10.1128/cmr.8.1.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Richardson L.S. Experimental respiratory syncytial virus pneumonia in cebus monkeys. J Med Virol. 1978;2(1):45–59. doi: 10.1002/jmv.1890020108. [DOI] [PubMed] [Google Scholar]
  • 58.Papin J.F. Infant baboons infected with respiratory syncytial virus develop clinical and pathological changes that parallel those of human infants. Am J Physiol Lung Cell Mol Physiol. 2013;304(8):L530–L539. doi: 10.1152/ajplung.00173.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Olivier A. Human respiratory syncytial virus A2 strain replicates and induces innate immune responses by respiratory epithelia of neonatal lambs. Int J Exp Pathol. 2009;90(4):431–438. doi: 10.1111/j.1365-2613.2009.00643.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Derscheid R.J. Human respiratory syncytial virus Memphis 37 causes acute respiratory disease in perinatal lamb lung. Biores Open Access. 2014;3(2):60–69. doi: 10.1089/biores.2013.0044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Larios Mora A. Kinetics of Respiratory Syncytial Virus (RSV) Memphis Strain 37 (M37) infection in the respiratory tract of newborn lambs as an RSV infection model for human infants. PLoS ONE. 2015;10(12):e0143580. doi: 10.1371/journal.pone.0143580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sow F.B. Respiratory syncytial virus is associated with an inflammatory response in lungs and architectural remodeling of lung-draining lymph nodes of newborn lambs. Am J Physiol Lung Cell Mol Physiol. 2011;300(1):L12–L24. doi: 10.1152/ajplung.00169.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Derscheid R.J. Effects of formalin-inactivated respiratory syncytial virus (FI-RSV) in the perinatal lamb model of RSV. PLoS ONE. 2013;8(12):e81472. doi: 10.1371/journal.pone.0081472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Garg R. Maternal immunization with respiratory syncytial virus fusion protein formulated with a novel combination adjuvant provides protection from RSV in newborn lambs. Vaccine. 2016;34(2):261–269. doi: 10.1016/j.vaccine.2015.11.029. [DOI] [PubMed] [Google Scholar]
  • 65.Boukhvalova M.S., Blanco J.C. The cotton rat Sigmodon hispidus model of respiratory syncytial virus infection. Curr Top Microbiol Immunol. 2013;372:347–358. doi: 10.1007/978-3-642-38919-1_17. [DOI] [PubMed] [Google Scholar]
  • 66.Openshaw P.J. The mouse model of respiratory syncytial virus disease. Curr Top Microbiol Immunol. 2013;372:359–369. doi: 10.1007/978-3-642-38919-1_18. [DOI] [PubMed] [Google Scholar]
  • 67.Byrd L.G., Prince G.A. Animal models of respiratory syncytial virus infection. Clin Infect Dis. 1997;25(6):1363–1368. doi: 10.1086/516152. [DOI] [PubMed] [Google Scholar]
  • 68.Prince G.A. The pathogenesis of respiratory syncytial virus infection in cotton rats. Am J Pathol. 1978;93(3):771–791. [PMC free article] [PubMed] [Google Scholar]
  • 69.Stittelaar K.J. Ferrets as a novel animal model for studying human respiratory syncytial virus infections in immunocompetent and immunocompromised hosts. Viruses. 2016;8(6) doi: 10.3390/v8060168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lemon K. Recombinant subgroup B human respiratory syncytial virus expressing enhanced green fluorescent protein efficiently replicates in primary human cells and is virulent in cotton rats. J Virol. 2015;89(5):2849–2856. doi: 10.1128/JVI.03587-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Boukhvalova M.S. Age-related differences in pulmonary cytokine response to respiratory syncytial virus infection: modulation by anti-inflammatory and antiviral treatment. J Infect Dis. 2007;195(4):511–518. doi: 10.1086/510628. [DOI] [PubMed] [Google Scholar]
  • 72.Shaw C.A. The role of non-viral antigens in the cotton rat model of respiratory syncytial virus vaccine-enhanced disease. Vaccine. 2013;31(2):306–312. doi: 10.1016/j.vaccine.2012.11.006. [DOI] [PubMed] [Google Scholar]
  • 73.Boukhvalova M. Treatment with novel RSV Ig RI-002 controls viral replication and reduces pulmonary damage in immunocompromised Sigmodon hispidus. Bone Marrow Transplant. 2016;51(1):119–126. doi: 10.1038/bmt.2015.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wong D.T. Respiratory syncytial virus infection in immunosuppressed animals: implications in human infection. J Med Virol. 1985;17(4):359–370. doi: 10.1002/jmv.1890170408. [DOI] [PubMed] [Google Scholar]
  • 75.Prince G.A., Horswood R.L., Chanock R.M. Quantitative aspects of passive immunity to respiratory syncytial virus infection in infant cotton rats. J Virol. 1985;55(3):517–520. doi: 10.1128/jvi.55.3.517-520.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.The IMpact-RSV Study Group Palivizumab, a humanized respiratory syncytial virus monoclonal antibody, reduces hospitalization from respiratory syncytial virus infection in high-risk infants. Pediatrics 1998;102(3 Pt 1):531–7. [PubMed]
  • 77.Prince G.A. Vaccine-enhanced respiratory syncytial virus disease in cotton rats following immunization with Lot 100 or a newly prepared reference vaccine. J Gen Virol. 2001;82(Pt 12):2881–2888. doi: 10.1099/0022-1317-82-12-2881. [DOI] [PubMed] [Google Scholar]
  • 78.Prince G.A. Respiratory syncytial virus infection in inbred mice. Infect Immun. 1979;26(2):764–766. doi: 10.1128/iai.26.2.764-766.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Rameix-Welti M.A. Visualizing the replication of respiratory syncytial virus in cells and in living mice. Nat Commun. 2014;5:5104. doi: 10.1038/ncomms6104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Taylor G. Respiratory syncytial virus infection in mice. Infect Immun. 1984;43(2):649–655. doi: 10.1128/iai.43.2.649-655.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Graham B.S. Primary respiratory syncytial virus infection in mice. J Med Virol. 1988;26(2):153–162. doi: 10.1002/jmv.1890260207. [DOI] [PubMed] [Google Scholar]
  • 82.Bhoj V.G. MAVS and MyD88 are essential for innate immunity but not cytotoxic T lymphocyte response against respiratory syncytial virus. Proc Natl Acad Sci USA. 2008;105(37):14046–14051. doi: 10.1073/pnas.0804717105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Jafri H.S. Respiratory syncytial virus induces pneumonia, cytokine response, airway obstruction, and chronic inflammatory infiltrates associated with long-term airway hyperresponsiveness in mice. J Infect Dis. 2004;189(10):1856–1865. doi: 10.1086/386372. [DOI] [PubMed] [Google Scholar]
  • 84.Mukherjee S. IL-17-induced pulmonary pathogenesis during respiratory viral infection and exacerbation of allergic disease. Am J Pathol. 2011;179(1):248–258. doi: 10.1016/j.ajpath.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Graham B.S. Role of T lymphocyte subsets in the pathogenesis of primary infection and rechallenge with respiratory syncytial virus in mice. J Clin Invest. 1991;88(3):1026–1033. doi: 10.1172/JCI115362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Stokes K.L. Differential pathogenesis of respiratory syncytial virus clinical isolates in BALB/c mice. J Virol. 2011;85(12):5782–5793. doi: 10.1128/JVI.01693-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Peebles R.S., Jr, Graham B.S. Pathogenesis of respiratory syncytial virus infection in the murine model. Proc Am Thorac Soc. 2005;2(2):110–115. doi: 10.1513/pats.200501-002AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Graham B.S. Priming immunization determines T helper cytokine mRNA expression patterns in lungs of mice challenged with respiratory syncytial virus. J Immunol. 1993;151(4):2032–2040. [PubMed] [Google Scholar]
  • 89.Knudson C.J. RSV vaccine-enhanced disease is orchestrated by the combined actions of distinct CD4 T cell subsets. PLoS Pathog. 2015;11(3):e1004757. doi: 10.1371/journal.ppat.1004757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Connors M. Enhanced pulmonary histopathology induced by respiratory syncytial virus (RSV) challenge of formalin-inactivated RSV-immunized BALB/c mice is abrogated by depletion of interleukin-4 (IL-4) and IL-10. J Virol. 1994;68(8):5321–5325. doi: 10.1128/jvi.68.8.5321-5325.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Connors M. Pulmonary histopathology induced by respiratory syncytial virus (RSV) challenge of formalin-inactivated RSV-immunized BALB/c mice is abrogated by depletion of CD4+ T cells. J Virol. 1992;66(12):7444–7451. doi: 10.1128/jvi.66.12.7444-7451.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Polack F.P. A role for immune complexes in enhanced respiratory syncytial virus disease. J Exp Med. 2002;196(6):859–865. doi: 10.1084/jem.20020781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Delgado M.F. Lack of antibody affinity maturation due to poor Toll-like receptor stimulation leads to enhanced respiratory syncytial virus disease. Nat Med. 2009;15(1):34–41. doi: 10.1038/nm.1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Boelen A. Both immunisation with a formalin-inactivated respiratory syncytial virus (RSV) vaccine and a mock antigen vaccine induce severe lung pathology and a Th2 cytokine profile in RSV-challenged mice. Vaccine. 2001;19:989–991. doi: 10.1016/s0264-410x(00)00213-9. [DOI] [PubMed] [Google Scholar]
  • 95.Kruijsen D. Local innate and adaptive immune responses regulate inflammatory cell influx into the lungs after vaccination with formalin inactivated RSV. Vaccine. 2011;29(15):2730–2741. doi: 10.1016/j.vaccine.2011.01.087. [DOI] [PubMed] [Google Scholar]
  • 96.Taylor G. Monoclonal antibodies protect against respiratory syncytial virus infection in mice. Immunology. 1984;52(1):137–142. [PMC free article] [PubMed] [Google Scholar]
  • 97.Tempest P.R. Reshaping a human monoclonal antibody to inhibit human respiratory syncytial virus infection in vivo. Biotechnology (N Y) 1991;9(3):266–271. doi: 10.1038/nbt0391-266. [DOI] [PubMed] [Google Scholar]
  • 98.Mestas J., Hughes C. Of mice and men: differences between mouse and human immunology. J Immunol. 2004;172:2731–2738. doi: 10.4049/jimmunol.172.5.2731. [DOI] [PubMed] [Google Scholar]
  • 99.Wyatt L. Priming and boosting immunity to respiratory syncytial virus by recombinant replication-defective vaccinia virus MVA. Vaccine. 2000;18:392–397. doi: 10.1016/s0264-410x(99)00257-1. [DOI] [PubMed] [Google Scholar]
  • 100.de Waal L. Vaccination of infant macaques with a recombinant modified vaccinia virus Ankara expressing the respiratory syncytial virus F and G genes does not predispose for immunopathology. Vaccine. 2004;22(8):923–926. doi: 10.1016/j.vaccine.2003.10.010. [DOI] [PubMed] [Google Scholar]
  • 101.Sharma A. Respiratory Syncytial Virus (RSV) pulmonary infection in humanized mice induces human anti-RSV immune responses and pathology. J Virol. 2016;90(10):5068–5074. doi: 10.1128/JVI.00259-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Harker J.A. Delayed sequelae of neonatal respiratory syncytial virus infection are dependent on cells of the innate immune system. J Virol. 2014;88(1):604–611. doi: 10.1128/JVI.02620-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Coates H., Chanock R.M. Experimental infection with respiratory syncytial virus in several species of animals. Am J Epidemiol. 1962;76:302–312. doi: 10.1093/oxfordjournals.aje.a120285. [DOI] [PubMed] [Google Scholar]
  • 104.Prince G.A., Porter D.D. The pathogenesis of respiratory syncytial virus infection in infant ferrets. Am J Pathol. 1976;82(2):339–352. [PMC free article] [PubMed] [Google Scholar]
  • 105.Hegele R.G. Production of acute bronchiolitis in guinea-pigs by human respiratory syncytial virus. Eur Respir J. 1993;6(9):1324–1331. [PubMed] [Google Scholar]
  • 106.Wright P.F., Woodend W.G., Chanock R.M. Temperature-sensitive mutants of respiratory syncytial virus: in-vivo studies in hamsters. J Infect Dis. 1970;122(6):501–512. doi: 10.1093/infdis/122.6.501. [DOI] [PubMed] [Google Scholar]
  • 107.Schmidt A.C. Recombinant bovine/human parainfluenza virus type 3 (B/HPIV3) expressing the respiratory syncytial virus (RSV) G and F proteins can be used to achieve simultaneous mucosal immunization against RSV and HPIV3. J Virol. 2001;75(10):4594–4603. doi: 10.1128/JVI.75.10.4594-4603.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mackow N. Attenuated Human Parainfluenza Virus Type 1 (HPIV1) expressing the fusion glycoprotein of human Respiratory Syncytial Virus (RSV) as a bivalent HPIV1/RSV vaccine. J Virol. 2015;89(20):10319–10332. doi: 10.1128/JVI.01380-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gitiban N. Chinchilla and murine models of upper respiratory tract infections with respiratory syncytial virus. J Virol. 2005;79(10):6035–6042. doi: 10.1128/JVI.79.10.6035-6042.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Grieves J.L. Mapping the anatomy of respiratory syncytial virus infection of the upper airways in chinchillas (Chinchilla lanigera) Comp Med. 2010;60(3):225–232. [PMC free article] [PubMed] [Google Scholar]
  • 111.Taylor G. Bovine model of respiratory syncytial virus infection. Curr Top Microbiol Immunol. 2013;372:327–345. doi: 10.1007/978-3-642-38919-1_16. [DOI] [PubMed] [Google Scholar]
  • 112.Dyer K.D. The Pneumonia Virus of Mice (PVM) model of acute respiratory infection. Viruses. 2012;4(12):3494–3510. doi: 10.3390/v4123494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sacco R.E. Respiratory syncytial virus infection in cattle. Vet Pathol. 2014;51(2):427–436. doi: 10.1177/0300985813501341. [DOI] [PubMed] [Google Scholar]
  • 114.Kimman T.G., Westenbrink F. Immunity to human and bovine respiratory syncytial virus. Arch Virol. 1990;112(1–2):1–25. doi: 10.1007/BF01348982. [DOI] [PubMed] [Google Scholar]
  • 115.Elvander M. Severe respiratory disease in dairy cows caused by infection with bovine respiratory syncytial virus. Vet Rec. 1996;138(5):101–105. doi: 10.1136/vr.138.5.101. [DOI] [PubMed] [Google Scholar]
  • 116.Viuff B. Replication and clearance of respiratory syncytial virus: apoptosis is an important pathway of virus clearance after experimental infection with bovine respiratory syncytial virus. Am J Pathol. 2002;161(6):2195–2207. doi: 10.1016/S0002-9440(10)64496-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Valarcher J.F., Taylor G. Bovine respiratory syncytial virus infection. Vet Res. 2007;38(2):153–180. doi: 10.1051/vetres:2006053. [DOI] [PubMed] [Google Scholar]
  • 118.Buchholz U.J. Chimeric bovine respiratory syncytial virus with glycoprotein gene substitutions from human respiratory syncytial virus (HRSV): effects on host range and evaluation as a live-attenuated HRSV vaccine. J Virol. 2000;74(3):1187–1199. doi: 10.1128/jvi.74.3.1187-1199.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.van der Poel W.H. Experimental reproduction of respiratory disease in calves with non-cell-culture-passaged bovine respiratory syncytial virus. Vet Q. 1996;18(3):81–86. doi: 10.1080/01652176.1996.9694622. [DOI] [PubMed] [Google Scholar]
  • 120.West K. The effect of formalin-inactivated vaccine on respiratory disease associated with bovine respiratory syncytial virus infection in calves. Vaccine. 1999;17(7–8):809–820. doi: 10.1016/s0264-410x(98)00265-5. [DOI] [PubMed] [Google Scholar]
  • 121.Woolums A.R. Evaluation of severe disease induced by aerosol inoculation of calves with bovine respiratory syncytial virus. Am J Vet Res. 1999;60(4):473–480. [PubMed] [Google Scholar]
  • 122.Blodorn K. A bovine respiratory syncytial virus model with high clinical expression in calves with specific passive immunity. BMC Vet Res. 2015;11:76. doi: 10.1186/s12917-015-0389-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Rontved C.M. Increased pulmonary secretion of tumor necrosis factor-alpha in calves experimentally infected with bovine respiratory syncytial virus. Vet Immunol Immunopathol. 2000;76(3–4):199–214. doi: 10.1016/S0165-2427(00)00214-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Antonis A.F. Age-dependent differences in the pathogenesis of bovine respiratory syncytial virus infections related to the development of natural immunocompetence. J Gen Virol. 2010;91(Pt 10):2497–2506. doi: 10.1099/vir.0.020842-0. [DOI] [PubMed] [Google Scholar]
  • 125.Taylor G., Thomas L.H., Stott E.J. Effect of vaccination on cell populations in lung washes from calves after infection with respiratory syncytial virus. Res Vet Sci. 1989;47(2):231–235. [PubMed] [Google Scholar]
  • 126.Taylor G. Role of T-lymphocyte subsets in recovery from respiratory syncytial virus infection in calves. J Virol. 1995;69(11):6658–6664. doi: 10.1128/jvi.69.11.6658-6664.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Thomas L.H. Passive protection of gnotobiotic calves using monoclonal antibodies directed at different epitopes on the fusion protein of bovine respiratory syncytial virus. J Infect Dis. 1998;177(4):874–880. doi: 10.1086/515234. [DOI] [PubMed] [Google Scholar]
  • 128.Taylor G. Protective epitopes on the fusion protein of respiratory syncytial virus recognized by murine and bovine monoclonal antibodies. J Gen Virol. 1992;73(Pt 9):2217–2223. doi: 10.1099/0022-1317-73-9-2217. [DOI] [PubMed] [Google Scholar]
  • 129.Schreiber P. High mortality rate associated with bovine respiratory syncytial virus (BRSV) infection in Belgian white blue calves previously vaccinated with an inactivated BRSV vaccine. J Vet Med B Infect Dis Vet Public Health. 2000;47(7):535–550. doi: 10.1046/j.1439-0450.2000.00380.x. [DOI] [PubMed] [Google Scholar]
  • 130.Kimman T.G. A severe outbreak of respiratory tract disease associated with bovine respiratory syncytial virus probably enhanced by vaccination with modified live vaccine. Vet Q. 1989;11(4):250–253. doi: 10.1080/01652176.1989.9694231. [DOI] [PubMed] [Google Scholar]
  • 131.Antonis A.F. Vaccine-induced immunopathology during bovine respiratory syncytial virus infection: exploring the parameters of pathogenesis. J Virol. 2003;77(22):12067–12073. doi: 10.1128/JVI.77.22.12067-12073.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Gershwin L.J. A bovine model of vaccine enhanced respiratory syncytial virus pathophysiology. Vaccine. 1998;16(11–12):1225–1236. doi: 10.1016/s0264-410x(98)80123-0. [DOI] [PubMed] [Google Scholar]
  • 133.Kalina W.V. Formalin-inactivated bovine RSV vaccine enhances a Th2 mediated immune response in infected cattle. Vaccine. 2004;22(11–12):1465–1474. doi: 10.1016/j.vaccine.2003.10.024. [DOI] [PubMed] [Google Scholar]
  • 134.Schmidt U. Mucosal immunization with live recombinant bovine respiratory syncytial virus (BRSV) and recombinant BRSV lacking the envelope glycoprotein G protects against challenge with wild-type BRSV. J Virol. 2002;76(23):12355–12359. doi: 10.1128/JVI.76.23.12355-12359.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Valarcher J.F. Role of alpha/beta interferons in the attenuation and immunogenicity of recombinant bovine respiratory syncytial viruses lacking NS proteins. J Virol. 2003;77(15):8426–8439. doi: 10.1128/JVI.77.15.8426-8439.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Taylor G. Recombinant bovine respiratory syncytial virus with deletion of the SH gene induces increased apoptosis and pro-inflammatory cytokines in vitro, and is attenuated and induces protective immunity in calves. J Gen Virol. 2014;95(Pt 6):1244–1254. doi: 10.1099/vir.0.064931-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Taylor G. DNA vaccination against respiratory syncytial virus in young calves. Vaccine. 2005;23(10):1242–1250. doi: 10.1016/j.vaccine.2004.09.005. [DOI] [PubMed] [Google Scholar]
  • 138.Riffault S. A new subunit vaccine based on nucleoprotein nanoparticles confers partial clinical and virological protection in calves against bovine respiratory syncytial virus. Vaccine. 2010;28(21):3722–3734. doi: 10.1016/j.vaccine.2010.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hagglund S. Bovine respiratory syncytial virus ISCOMs-immunity, protection and safety in young conventional calves. Vaccine. 2011;29(47):8719–8730. doi: 10.1016/j.vaccine.2011.07.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Vangeel I. Efficacy of a modified live intranasal bovine respiratory syncytial virus vaccine in 3-week-old calves experimentally challenged with BRSV. Vet J. 2007;174(3):627–635. doi: 10.1016/j.tvjl.2006.10.013. [DOI] [PubMed] [Google Scholar]
  • 141.Antonis A.F. Vaccination with recombinant modified vaccinia virus Ankara expressing bovine respiratory syncytial virus (bRSV) proteins protects calves against RSV challenge. Vaccine. 2007;25(25):4818–4827. doi: 10.1016/j.vaccine.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 142.Taylor G. Recombinant vaccinia viruses expressing the F, G or N, but not the M2, protein of bovine respiratory syncytial virus (BRSV) induce resistance to BRSV challenge in the calf and protect against the development of pneumonic lesions. J Gen Virol. 1997;78(Pt 12):3195–3206. doi: 10.1099/0022-1317-78-12-3195. [DOI] [PubMed] [Google Scholar]
  • 143.Mapletoft J.W. Formulation with CpG oligodeoxynucleotides increases cellular immunity and protection induced by vaccination of calves with formalin-inactivated bovine respiratory syncytial virus. Virology. 2006;353(2):316–323. doi: 10.1016/j.virol.2006.06.001. [DOI] [PubMed] [Google Scholar]
  • 144.Ellis J.A., Gow S.P., Goji N. Response to experimentally induced infection with bovine respiratory syncytial virus following intranasal vaccination of seropositive and seronegative calves. J Am Vet Med Assoc. 2010;236(9):991–999. doi: 10.2460/javma.236.9.991. [DOI] [PubMed] [Google Scholar]
  • 145.Taylor G. Efficacy of a virus-vectored vaccine against human and bovine respiratory syncytial virus infections. Sci Transl Med. 2015;7(300):300ra127. doi: 10.1126/scitranslmed.aac5757. [DOI] [PubMed] [Google Scholar]
  • 146.Green C.A. Chimpanzee adenovirus- and MVA-vectored respiratory syncytial virus vaccine is safe and immunogenic in adults. Sci Transl Med. 2015;7(300):300ra126. doi: 10.1126/scitranslmed.aac5745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Horsfall F.L., Hahn R.G. A latent virus in normal mice capable of producing pneumonia in its natural host. J Exp Med. 1940;71(3):391–408. doi: 10.1084/jem.71.3.391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Anh D.B., Faisca P., Desmecht D.J. Differential resistance/susceptibility patterns to pneumovirus infection among inbred mouse strains. Am J Physiol Lung Cell Mol Physiol. 2006;291(3):L426–L435. doi: 10.1152/ajplung.00483.2005. [DOI] [PubMed] [Google Scholar]
  • 149.Bonville C.A. Diminished inflammatory responses to natural pneumovirus infection among older mice. Virology. 2007;368(1):182–190. doi: 10.1016/j.virol.2007.06.020. [DOI] [PubMed] [Google Scholar]
  • 150.Bonville C.A. Inflammatory responses to acute pneumovirus infection in neonatal mice. Virol J. 2010;7:320. doi: 10.1186/1743-422X-7-320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Krempl C.D., Collins P.L. Reevaluation of the virulence of prototypic strain 15 of pneumonia virus of mice. J Virol. 2004;78(23):13362–13365. doi: 10.1128/JVI.78.23.13362-13365.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Cook P.M., Eglin R.P., Easton A.J. Pathogenesis of pneumovirus infections in mice: detection of pneumonia virus of mice and human respiratory syncytial virus mRNA in lungs of infected mice by in situ hybridization. J Gen Virol. 1998;79(Pt 10):2411–2417. doi: 10.1099/0022-1317-79-10-2411. [DOI] [PubMed] [Google Scholar]
  • 153.Watkiss E.R. Innate and adaptive immune response to pneumonia virus of mice in a resistant and a susceptible mouse strain. Viruses. 2013;5(1):295–320. doi: 10.3390/v5010295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Domachowske J.B. Pulmonary eosinophilia and production of MIP-1alpha are prominent responses to infection with pneumonia virus of mice. Cell Immunol. 2000;200(2):98–104. doi: 10.1006/cimm.2000.1620. [DOI] [PubMed] [Google Scholar]
  • 155.Bonville C.A. Respiratory dysfunction and proinflammatory chemokines in the pneumonia virus of mice (PVM) model of viral bronchiolitis. Virology. 2006;349(1):87–95. doi: 10.1016/j.virol.2006.02.017. [DOI] [PubMed] [Google Scholar]
  • 156.Domachowske J.B. The chemokine macrophage-inflammatory protein-1 alpha and its receptor CCR1 control pulmonary inflammation and antiviral host defense in paramyxovirus infection. J Immunol. 2000;165(5):2677–2682. doi: 10.4049/jimmunol.165.5.2677. [DOI] [PubMed] [Google Scholar]
  • 157.Domachowske J.B. MIP-1alpha is produced but it does not control pulmonary inflammation in response to respiratory syncytial virus infection in mice. Cell Immunol. 2000;206(1):1–6. doi: 10.1006/cimm.2000.1730. [DOI] [PubMed] [Google Scholar]
  • 158.Walsh K.B. Animal model of respiratory syncytial virus: CD8+ T cells cause a cytokine storm that is chemically tractable by sphingosine-1-phosphate 1 receptor agonist therapy. J Virol. 2014;88(11):6281–6293. doi: 10.1128/JVI.00464-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Percopo C.M. Pulmonary eosinophils and their role in immunopathologic responses to formalin-inactivated pneumonia virus of mice. J Immunol. 2009;183(1):604–612. doi: 10.4049/jimmunol.0802270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Maunder H.E. Intranasal immunisation with recombinant adenovirus vaccines protects against a lethal challenge with pneumonia virus of mice. Vaccine. 2015;33(48):6641–6649. doi: 10.1016/j.vaccine.2015.10.105. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES