Skip to main content
American Journal of Respiratory and Critical Care Medicine logoLink to American Journal of Respiratory and Critical Care Medicine
. 2015 Jan 1;191(1):34–44. doi: 10.1164/rccm.201405-0901PP

Toward Primary Prevention of Asthma. Reviewing the Evidence for Early-Life Respiratory Viral Infections as Modifiable Risk Factors to Prevent Childhood Asthma

Amy S Feldman 1, Yuan He 2, Martin L Moore 1,2, Marc B Hershenson 3, Tina V Hartert 1,
PMCID: PMC4299628  PMID: 25369458

Abstract

A first step in primary disease prevention is identifying common, modifiable risk factors that contribute to a significant proportion of disease development. Infant respiratory viral infection and childhood asthma are the most common acute and chronic diseases of childhood, respectively. Common clinical features and links between these diseases have long been recognized, with early-life respiratory syncytial virus (RSV) and rhinovirus (RV) lower respiratory tract infections (LRTIs) being strongly associated with increased asthma risk. However, there has long been debate over the role of these respiratory viruses in asthma inception. In this article, we systematically review the evidence linking early-life RSV and RV LRTIs with asthma inception and whether they could therefore be targets for primary prevention efforts.

Keywords: respiratory syncytial virus, RSV, rhinovirus, RV, asthma


At a Glance Commentary

Scientific Knowledge on the Subject

Early-life infections represent ubiquitous and potentially modifiable exposures and hold the potential to be important targets for primary and/or secondary asthma prevention. Evidence from many studies that has never previously been compiled provides a body of evidence that links these risk factors with asthma genesis.

What This Study Adds to the Field

This is the first objective and systematic overview that compiles all available data on the role of respiratory syncytial virus and rhinovirus in asthma inception, identifying the remaining knowledge gaps and research opportunities.

An important first step in primary prevention is identification of risk factors for disease and establishment of a causal relationship. This review tackles a long-standing debate on the role of these viruses in asthma inception and presents the currently available evidence to support or refute the role of infant respiratory syncytial virus (RSV) and rhinovirus (RV) infections as potentially causal and modifiable risk factors for asthma development. For RSV and RV we review and discuss the following evidence: (1) the link between host determinants of infant RSV and RV infection severity and asthma risk; (2) the role of viral determinants of infant infection severity and asthma risk; (3) the influence of other environmental factors on respiratory viral infection and asthma risk; (4) the data supporting a causal relationship between infant viral respiratory infections and asthma risk, including available mechanistic, observational, and intervention studies; and (5) identification of knowledge gaps and recommendations for future directions.

Host Genetic and Familial Determinants Linking Infant RSV and RV Infection Severity and Asthma Risk

Whether infant viral lower respiratory infections are merely the first manifestation of asthma, whether there is a shared genetic predisposition to asthma and severe sequelae of RSV and RV, or whether these viruses are causal in asthma development has been long debated. Patients with asthma have been shown to have an increased susceptibility to certain viral and bacterial infections (17). Patients with asthma have an increased risk for colonization with certain bacteria, increased risk for latent infections, increased risk for community-acquired pneumonia (7, 8), increased morbidity with influenza infection (5), increased likelihood of persistent rhinovirus in the airway epithelium (9, 10), and increased risk of invasive infections, such as rhinoviremia and invasive pneumococcal disease (1113). This increased risk of colonization, latent infection, infection morbidity, and infection severity may result from underlying immune differences that increase overall susceptibility to infections and asthma in the infant, thus making infants more susceptible to acute infections and to the chronic sequelae of early-life infection.

Certain heritable factors have been identified that support a link between these early-life infections and asthma risk. Among these are genetic polymorphisms that are associated with RSV infections and asthma. We and others have previously compiled the genes that have been demonstrated to be associated with RSV and asthma. Among these are polymorphisms in a number of immune response genes, suggesting immune perturbations common to both diseases: CX3CR1, TLR-4, SP-A, SP-D, IL-10, CCR5, TLR-10, IL-4, IL-13, IL-10, IL-8, IL-18, tumor necrosis factor, TLR-4, MS4A2, VDR, IL-4Rα, RANTES, TGF-β1, and ADAM33 (14, 15). In addition, a recent clinical multicenter cohort study found that human IL1RL1 gene variants and nasopharyngeal IL1RL1-α levels were associated with severe RSV bronchiolitis. The potential biological role of IL1RL1 in the pathogenesis of severe RSV bronchiolitis was supported by high local concentrations of IL1RL1 in children with the most severe disease (16). An important genetic association linking RV with asthma has been identified between host 17q21 locus variants and RV wheezing illness (17). The association between the 17q21 locus, in particular ORMDL3 and childhood-onset asthma, has been replicated in several different cohorts (18).

Studies using familial asthma, atopy, and allergic sensitization to assess the hereditable link between infant viral infection and asthma have also demonstrated an increase in the relative odds of RV acute respiratory infection (ARI) and more severe infant RV ARI among infants born to mothers with atopic asthma. This suggests that a familial predisposition to asthma increases the risk of severe RV ARIs before the onset of asthma (19). Previous studies have also shown that children with atopic asthma have more frequent and more severe RV illnesses compared with patients with nonatopic asthma (4, 6, 20).

Host Immune Response to Infant RSV and RV Infection

The airway epithelium is an important component of host defense because airway epithelial cells (ECs) are the interface between the environment and the host. The airway epithelium is a major target of respiratory viral infections. ECs have surface receptors (Toll-like receptors and other pattern recognition receptors) that can recognize specific patterns on pathogen molecules (pathogen-associated molecular patterns). Once these ECs recognize pathogen-associated molecular patterns, they become activated, release cytokines and antimicrobial peptides, increase expression of chemokines, and activate the adaptive immune system. In the short- and long term, this leads to increased inflammation, Th2 cell activation, and alternative macrophage activation. These Th2 and alternative macrophages can then regulate EC production of growth factors, such as TGF-β and VEGF, that lead to airway remodeling (21). ECs also help to regulate the acute response to viral infections through production of cytokines such as type I, II, and III IFNs. However, in the setting of high virus replication or other determinants of severe infection, IFN produced by ECs may not be sufficient to control the viral infection, and severe sequelae of infection may result. Infants, who have an immature immune system, and individuals who are genetically predisposed to asthma might also manifest immune responses to RSV and RV, predisposing them to more severe infant infection and possibly the chronic consequences of these infections. Increasing evidence suggests that infant and adult responses to infection are not identical, with infants demonstrating a bias away from type 1 and toward type 2 immunity (22). This bias could have ramifications for the development of allergic disease after early-life viral infection.

Patients with established asthma have increased morbidity from certain infections compared with persons without asthma (12). There are known immune and organ-specific differences characteristic of asthma that predispose patients to more severe infection. These include altered EC response to viral infection, increased mucus production, impaired mucociliary clearance, impaired alveolar macrophage function, altered IFN response, and increased Th2 activation (2329). A deficiency of IFN production in patients with asthma may also contribute to impaired immunity (23, 24, 29) by causing a shift from Th1 responses to Th2 responses (27). Infants who develop asthma are born with lower lung function, perhaps also predisposing them to more severe infections (3034).

Viral Strain Determinants of Infant Infection Severity and Asthma Risk

RSV is an enveloped, nonsegmented, negative-sense, single-stranded RNA virus of the Paramyxoviridae family. RSV is the leading cause of severe lower respiratory tract infection (LRTI) in the pediatric population. Almost all children are infected by their second or third year of life. RSV infection is estimated to cause 33 million LRTIs in children under 5 years of age worldwide, among whom 3.4 million are hospitalized and 66,000 to 199,000 die annually (35). RSV has one serotype and two antigenic subgroups, A and B (36). Within the antigenic subgroups, RSV can be further classified into clades according to the nucleotide sequence of the variable attachment glycoprotein (G) genes. Subgroup A strains can be divided into at least seven clades (GA1–GA7), and subgroup B strains can be divided into at least four clades (GB1–GB4) (37). The emergence of a new RSV-B genotype with a 60-nucleotide duplication in the G-protein gene (G gene) has also been reported (38). Strains of A and B subgroups cocirculate, but one strain or a low number of strains usually predominate within a single outbreak, with replacement of dominant genotypes in subsequent years (39).

Clinical studies have demonstrated an association of RSV genotype with severity of illness. Group A RSV infection results in greater disease severity than group B infection among hospitalized infants (40). The GA3 clade has been associated with greater severity of illness compared with clades GA2 and GA4 (41). Differential pathogenesis of RSV A subgroup strains has been reported in an animal model of infection of BALB/cJ mice with RSV A2001/2–20 (2–20). A subgroup strain resulted in greater disease severity, higher lung IL-13 levels, and higher lung gob-5 levels and induced airway mucin expression, supporting differential pathogenicity dependent on strain in these genetically identical mice (42).

RVs are positive-sense, single-stranded RNA viruses belonging to the family Picornaviridae and the genus Enterovirus. RVs are classified into RV-A, -B, and -C based on phylogenetic sequence criteria (43). Previously, 99 serotypes were known, and these were divided into two species: RV-A (containing 74 serotypes) and RV-B (containing 25 serotypes) (44). RV-C was identified in 2009 (45). Currently, at least 50 different types of RV-C have been identified (46). RV-C and RV-A were shown to cause moderate to severe illness in young children compared with milder infection with RV-B (47, 48).

Influence of Other Environmental Exposures on Infant Viral Infection Severity and Asthma Risk

Infant viral infections do not act in isolation in asthma development. There are likely multiple known and unknown risk factors that, acting independently or in conjunction, contribute to the overall inception of asthma. Currently recognized environmental factors associated with severity of infant infection and asthma risk include second-hand smoke exposure, diet, and exposures that alter the infant microbiome. These risk factors for asthma may act through influencing the infant’s developing immune system and/or altering infant responses to viral infections. Second-hand smoke exposure has been well established to be associated with increased risk of more severe respiratory morbidity and with asthma and atopy among children (4952). Maternal smoking has also been shown to alter the neonate’s innate immune response with higher neonatal Th2 responses, higher cord blood IgE levels, and decreased innate TLR responses compared with infants of nonsmokers (5355). These alterations in the infant’s immune response may predispose these infants to more severe infection.

The maternal and infant diet also plays an important role in the response to viral infection and asthma. In the prenatal stage, diet likely modifies asthma risk in part through epigenetic modifications. Excessive folate supplementation, for example, has been demonstrated to increase DNA CG methylation of the Runx3 gene, decreasing its expression and increasing the risk of an asthma-like phenotype in an animal model of asthma (56, 57). In humans, infants have been shown to have greater risk of bronchiolitis if they were born to mothers who received folic acid supplementation in the first trimester compared with those who did not receive folic acid supplementation (58).

Vitamin D is also involved in epigenetic modifications and in the regulation of several genes involved in inflammation and immunity. Vitamin D deficiency may lead to increased inflammation, increased risk of viral infections, and the development of asthma (5963).

Selenium deficiency has been shown to modify respiratory epithelium and to alter the immune response to viral infection in mice. Selenium-deficient mice have decreased GPX1, decreased catalase activity, increased mucus production, and increased Muc5AC mRNA levels. These mice had more severe influenza infection and increased IL-6 production, decreased IP-10 production, and increased influenza-induced apoptosis (64).

Acetaminophen and ibuprofen use during infancy have also been implicated in asthma inception (65). These drugs are likely used during infant respiratory viral illnesses. The current evidence suggests that acetaminophen use in combination with a genetic polymorphism in TLR4 may be associated with asthma (66).

Not much is known about the infant microbiome in relation to respiratory infections. However, we do know that the type or pattern of bacterial colonization of the airways of infants is associated with asthma risk, and it seems likely that bacteria and viruses interact in maintaining health and in influencing disease. Infants colonized with Streptococcus pneumoniae, Haemophilus influenzae, or Moraxella catarrhalis in their airway are at an increased risk for asthma (67). In another recent study, M. catarrhalis and S. pneumoniae detected during rhinovirus infection were associated with increased moderate asthma exacerbations and asthma symptoms (68). A recent randomized controlled trial of pre- and probiotic supplements showed prevention of RV infection in preterm infants (69). Thus, modification of the infant microbiome could be a mechanism through which RV wheezing illnesses might be prevented, in turn preventing later asthma. These studies suggest that either the infant immune system among children who will develop asthma results in differential colonization and/or that the airway microbiome influences the immune system and subsequently the type and severity of viral infection an infant develops. These environmental factors may therefore act through independent mechanisms but likely also interact with early-life RSV and RV infections by altering the microbiome and developing immune system and thus may increase risk and severity of these infant infections and later asthma.

In summary, a combination of environmental factors acting at critical time periods during gestation and early life likely interact with early-life viral infections in the development of childhood asthma.

Causal Evidence for Early-Life RSV, RV Infection, and Asthma Risk

Although early-life RSV and RV are associated with asthma development, this does not establish causality. We will review the evidence that supports a causal relationship between infection with infant RSV and RV and asthma, asking the following questions (Table 1): (1) Does the factor precede development of disease? (2) Is there a dose-dependent relationship? (3) Is there a biological mechanism(s) through which the causal factor contributes to disease development? and (4) Is there an intervention or proof of concept that demonstrates that eliminating the risk factor prevents disease? It is important to recognize that establishing Koch’s third postulate (exposure of the host to virus causes asthma), albeit intended for establishing causality of infectious diseases, can never experimentally be done in humans. In the case of risk factors that result in significant harm or chronic disease, randomized controlled trials of early-life infection in humans are not ethical to conduct, and we must rely on studies of prevention of the risk factor to demonstrate whether these early-life infections are causal and whether prevention is an effective primary asthma prevention strategy. Human, in vitro, and animal models provide the closest models we have for demonstrating an understanding Koch’s third postulate of how viral infection may lead to asthma development (23, 7072).

Table 1.

Summary of the Available Evidence in Support of, against, or Lacking a Link between Respiratory Syncytial Virus and Rhinovirus Lower Respiratory Tract Infection with Asthma Inception

  RSV LRTI
RV LRTI
Evidence* Data Summary Evidence* Data Summary
Association with asthma + Among infants with RSV LRTI, the estimated risk of later developing asthma ranges from OR 2.07 to 12.7 (95% CI, 1.2–47.1) (13, 3032, 48, 76, 7982). + Among infants with RV LRTI, the estimated risk of later developing asthma ranges from OR 1.99 to 10 (95% CI, 1.04–23) (13, 3032, 48, 76, 81).
RV is a frequent cause of asthma exacerbations (119)
Precedes asthma onset + Longitudinal studies demonstrate that RSV LRTI precedes atopic sensitization and asthma onset (76, 77). +/− Longitudinal studies demonstrate that RV LRTI precedes asthma onset (76, 77).
Allergic sensitization precedes RV wheezing in some infants (12, 78).
Dose–response relationship demonstrated + RSV LRTI severity is associated in a dose-dependent fashion with both increasing asthma risk and increasing asthma severity (34, 76). 0/+ A dose–response relationship has not yet been demonstrated with RV LRTI.
A dose–response relationship with no infection, mild infection, and infection with wheezing has been demonstrated for RSV (73). A dose–response relationship with no infection, mild infection, and infection with wheezing has been demonstrated for RV (73).
Contributes to a substantial proportion of asthma + A majority of infant LRTIs are attributable to RSV infection (111, 112). + Although the risk of asthma associated with RV LRTI is higher in most studies compared with RSV LRTI, RV LRTI may contribute to a smaller proportion of asthma because infant RV LRTI is less common (32).
Infant RSV LRTIs therefore contribute to a higher proportion of asthma in the population.
Defined risk groups + Family history of asthma (12, 77) + Family history of asthma/atopy (77)
Premature birth (77, 116, 118) Precedent allergen sensitization (77)
Male sex (77) Genetic polymorphisms (17)
White race (77)
Seasonality of birth (118, 120)
Genetic polymorphisms commonly in immune response genes (1, 9)
Host genetic and viral genetic determinants of disease risk and severity + Host: Several genes are associated with both RSV infection and asthma, suggesting a genetic susceptibility to both (1, 9, 10). + Host: 17q21 variants are associated with asthma in children with RV wheezing illnesses in early life (17).
Virus: RSV strain differences have been shown in mouse and human studies to affect the pathogenicity, which await demonstration as to whether they are associated with asthma risk after infant infection (41, 43). Virus: RV strain differences may have an impact on pathogenicity (47, 48).
Biologic mechanisms through which these viruses may cause asthma + Pathology: RSV in animal models causes acute and chronic lung changes similar to asthma (80, 90, 98). + Pathology: RV in animal models causes acute and chronic lung changes similar to asthma (92, 100).
Physiology: RSV infection is associated with prolonged airway hyperresponsiveness (90, 99, 101). Physiology: RV infection is associated with prolonged airway hyperresponsiveness (92, 100).
Immune development: In animal models RSV infection results in long-term immunomodulatory changes and impairs regulatory T cells (88, 103, 107110). Immune development: In animal models RV infection results in long-term immunomodulatory changes (92, 93).
Epithelial barrier function: In a cell culture model, RSV degrades epithelial barrier function, which could increase allergen sensitization through the airways (96). Epithelial barrier function: RV degrades epithelial barrier function in cell culture and infected mice, which could lead to increase allergen sensitization through the airways (97).
Currently available interventions + Avoidance 0 Avoidance
Birth timing (118, 120) Different classes of RV inhibitors have been evaluated in clinical trials but are no longer being developed (117, 121).
RSV immunoprophylaxis (110113) Pre- and probiotics may prevent rhinovirus infection in premature infants (69).
Ribavirin (114)
Acceptable interventions in pregnant women and children + Most would consider both birth timing and the currently available RSV immunoprophylaxis as acceptable interventions (110113). 0 There is currently no available vaccine or preventive treatment other than avoidance.
Proof of concept studies available by challenging, preventing or removing + Randomized controlled trial of RSV immunoprophylaxis among premature infants demonstrated reduced risk of wheezing at 1 yr (110). 0 No evidence, and there is currently no available vaccine or preventive treatment to test.
Observational studies of infants treated with ribavirin or RSV immunoprophylaxis demonstrated significantly lower incidence of asthma or recurrent wheezing (111114).

Definition of abbreviations: CI = confidence interval; LRTI = lower respiratory tract infection; OR = odds ratio; RSV = respiratory syncytial virus; RV = rhinovirus.

*

+ = evidence in support of a causal relationship; − = evidence against a causal relationship; 0 = no available evidence or none available.

Numerous longitudinal studies have demonstrated this first important line of evidence that RSV and RV LRTIs precede the development of asthma (33, 73). RSV LRTI also precedes allergic sensitization; however, allergic sensitization has been shown to precede wheezing with RV infection, suggesting that wheezing with RV may follow or require atopic sensitization (33, 74). A number of studies next demonstrate the strength of the association of early-life RSV and RV LRTI with later asthma development; estimates of asthma after RSV LRTI range from 2- to 12-fold increased risk (odds ratio, 2.07–12.7; 95% confidence interval, 1.2–47.1), and estimates after RV LRTI range from 2- to 10-fold increased risk (odds ratio, 1.99–10; 95% confidence interval, 1.04–23) (20, 47, 73, 7581). Several birth cohort studies also demonstrate a dose–response relationship between infant respiratory viral infection severity and asthma risk, with increasing infant infection severity associated with greater childhood asthma risk and asthma severity (73, 82). It is important to consider, however, that prevention of early-life wheezing will likely never negate the possibility of another later environmental causal factor resulting in increased asthma risk given the strong genetic susceptibility to asthma. The findings of observational longitudinal studies demonstrate decreased associations with early-life risk factors as subjects age (81, 83, 84). A randomized controlled trial in which there would in theory be no differences between groups in these subsequent causal exposures could answer the question about the duration of effect of preventing early-life exposures on asthma risk.

Biologic Mechanisms through Which Respiratory Viral Infections Cause Asthma

Children hospitalized with RSV have mild airway obstruction and airway hyperreactivity (27). Is this a sequelae of the viral infection, an underlying host characteristic of children destined to develop asthma, or both? Although children who develop asthma are born with lower lung function (3034), severe RSV infection has been shown to result in long-term impairments in lung function (31). Acute infection with RSV causes expression of proinflammatory cytokines and chemokines such as IL-1α, IL-1β, IL-6, IL-8, IFN-γ, tumor necrosis factor α, monocyte chemotactic protein 1, macrophage inflammatory protein (MIP)-1α, and MIP-1β, promoting a prolonged inflammatory environment within the lung that could contribute to the development of asthma (85). Studies of natural colds, predominantly with RV, have also shown the release of inflammatory cytokines (86). In addition, levels of IL-17, secreted by type 17 helper T cells (Th17), are consistently elevated in human infection with RSV and in asthmatic sputum and bronchoalveolar lavage fluid (87). Thus, it is possible that elevated Th17 responses after RSV infection may contribute to asthma development or indicate similar host risks for RSV and asthma (88).

Viruses can also program the immune response toward a type 2 helper T cell (Th2) proallergic phenotype. Infection with RSV has been shown to induce a Th2 response characterized by production of IL-4, IL-5, and IL-13 in some but not all studies. A Th2-biased immune response is known to contribute to disease in murine models of allergic airway inflammation and is characteristic of human asthma (89). Studies using a paramyxovirus in an experimental mouse model resembling asthma and chronic obstructive pulmonary disease demonstrated that an innate NKT cell–macrophage–IL-13 immune axis may be activated in human disease conditions, similar to the virus-induced mouse model of chronic airway disease, providing another possible connection between infection and chronic inflammatory diseases (90). More recently, epithelial cells have been shown to produce novel “innate cytokines,” TSLP, IL-25, and IL-33, which create a permissive environment for type 2 differentiation of dendritic cells, T cells, and innate lymphoid cells, leading to production of the proasthmatic cytokines IL-4, IL-5, and IL-13. Studies in humans and mice demonstrate that viruses, specifically RV and influenza, can elicit this epithelial cell response (9194). RSV infection may also contribute to the development of allergy by breaking immune tolerance to allergens early in life. RSV infection induces GATA-3 expression and Th2 cytokine production in forkhead box P3+ Treg cells and compromises the suppressive function of pulmonary Treg cells, dependent on IL-4 receptor α (IL-4Rα) expression in the host. Thus, RSV induces a TH2-like effector phenotype in Treg cells, which attenuates tolerance to an unrelated antigen (allergen) (95). RSV and RV may also increase airway sensitization by altering the epithelial barrier, another mechanism through which viruses may lead to the airway hyperreactivity that characterizes asthma (96, 97). Finally, rodent models suggest that respiratory viral infections of immature and mature animals (90) result in chronic pathologic changes in the lung, airway hyperreactivity, and immune system changes similar to those seen in human asthma (98101). In addition, these early-life infections may alter subsequent responses to viral infection in adult animals (102, 103).

Rhinovirus has also been demonstrated to result in immunologic changes and in the induction of factors and airway changes that could result in chronically altered lung and immune function. Certain strains of RV have been demonstrated to have the unique ability to bypass antigen presentation and directly infect and activate CD4+ and CD8+ T cells. This could explain the strong association of rhinovirus with exacerbation of airway diseases and may have relevance to early-life altered immune response to RV that could be strain specific rather than illness severity specific (104). RV infection has also been shown to increase deposition of the extracellular matrix proteins collagen and endothelial growth factor in cultured human bronchial ECs possibly mediated through TLRs. Furthermore, gene expression was increased in lung homogenates of mice infected with RV-1b (105). In RV neonatal mouse models, infection has been demonstrated to result in prolonged asthma-like responses (airway responsiveness and mucous metaplasia) that were dependent on IL-13, IL-25, and type 2 innate lymphoid cells (92, 100). These could be mechanisms through which early-life RSV and RV infections of the lower airways might permanently alter lung development, airway physiology, and immune development.

Although animal models are important, in addition to findings in mice not always translating to humans, there are several limitations of the mouse models of RSV and RV infection. First, RSV infection does not result in extensive epithelial damage and desquamation in the mouse as it does in humans. As a result, the epithelial barrier remains more intact in the mouse and prevents greater subepithelial exposure to antigens than likely occurs in humans after infection. A second shortcoming is that the airway epithelium of the mouse is not as permissive for RSV or RV infection, and the epithelial replication in the mouse is not as robust as it is in humans. A third shortcoming is the difficulty in quantifying airway physiologic changes to early-life infection in mice because the measurement tools are made for older and larger animals. Fourth, only the transgenic, human ICAM-1 receptor mouse used to study RV infection is susceptible to major group RV infection, although similar results were obtained in wild-type mice infected with minor group virus (106). Finally, interpretation of animal models should consider whether neonatal or adult mice were studied. Given the differences in the developing lung and immune system of neonatal and adult mice and of humans, inferences as to the long-lasting impact of early-life viral infection are best extrapolated from neonatal mouse models.

Despite the limitations of the types of studies that can be conducted in young infants and the available animal models, taken together the available studies provide biological mechanisms through which RSV and RV could lead to physiologic, pathologic, and immunologic changes that characterize asthma.

The Contribution of Early-Life Infection to Asthma

The next important consideration is the population-level contribution of RSV and RV LRTIs to the burden of asthma. First, as risk factors, RSV and RV are common, nearly universal early-life infections. An important distinction is that current studies of RSV LRTI illness have been predominantly in hospitalized children because RSV LRTI is a more common cause of severe LRTI during early infancy, whereas RV illnesses tend to be less severe and are more commonly outpatient illnesses. With advancing age, there is reversal of the predominant viral etiology of LRTIs, with RV becoming more common and RSV becoming less common. Second, RSV and RV are strongly associated with childhood asthma. However, association does not establish causality or establish whether these viruses contribute to a significant proportion of resultant asthma. This is best demonstrated by the population-attributable risk for asthma after these early-life LRTIs. Among infants, the prevalence of LRTI is approximately 18 to 32% in the first year of life and approximately 9 to 17% in the second year of life (33). Among infants with LRTI, the prevalence of RSV can be as high as 80%, especially in the first 3 months of life (107, 108). The prevalence of RV in infants with LRTI is approximately 20 to 30% (77). Although the odds of developing asthma after RV LRTI are higher in most studies than for RSV LRTI, RSV LRTI in the first year of life is significantly more common and therefore, if causal, may be responsible for a greater proportion of asthma that develops in children with RSV compared with RV (73, 82, 109). Thus, the phenotype of asthma after infant RSV LRTI accounts for up to 31% of early childhood asthma, with a population-attributable risk estimated at about 13% in several diverse populations (80, 109). No available estimates exist for RV, but one would estimate that RV LRTI, if causal, becomes more important as a risk factor with advancing age, where RV LRTI becomes more common and RSV LRTI becomes less common.

The strongest data supporting a causal relationship of RSV LRTI with recurrent wheezing comes from a recent randomized, controlled trial of a highly specific monoclonal IgG antibody directed against the RSV fusion (F) glycoprotein (palivizumab) (110). In this trial of late preterm infants (33–35 wk), RSV immunoprophylaxis resulted in a nearly 50% reduction of recurrent wheezing (11 vs. 21%) in the first year of life. This study was not powered to determine whether RSV immunoprophylaxis results in a reduction of asthma at 6 years of age. However, observational studies of premature infants eligible to receive palivizumab immunoprophylaxis have reported comparable risk reduction for recurrent wheezing during the first 3.5 years of life (110113). A study of children who received antiviral treatment (ribavirin) for RSV during infancy also demonstrated significantly lower incidence of asthma or recurrent wheezing (114). Maternally derived RSV antibodies measured in cord blood, presumably representing passive immunization, have also been associated with a decrease in infant RSV hospitalization. However, very high RSV cord blood antibody titers were associated with an increased risk of recurrent wheeze in children with and without RSV LRTI hospitalization (115). This could represent a more severe infection, including in utero RSV infection, that may alter infant airway structure and immune function, predisposing the infant to an increased risk of asthma, as Piedimonte and colleagues showed in their murine model of vertical RSV transmission (116). High RSV titers may also indicate a genetic predisposition to severe respiratory infections and asthma. This evidence supports the hypothesis that preventing at least early-life RSV LRTI may help to prevent wheezing or asthma. Although there is a strong evidence base to support the role of RSV infection in asthma development, there are insufficient data at this time to support a causal role of infant RV infection with asthma inception. The later age of first wheezing with RV and precedent allergic sensitization in some children before wheezing with RV supports the well-known association of RV with exacerbation of prevalent asthma but could certainly be consistent with the contribution of RV as a causal factor during a later susceptibility period during childhood or one that alters the natural history of asthma, analogous to infectious exacerbations in COPD.

Future Directions and Recommendations

Infant viral lower respiratory tract infections with RSV and RV have been strongly associated with childhood asthma. Whether this is from a shared inherited risk for asthma and enhanced susceptibility to these viruses, a result of these viruses’ capacity to cause asthma through alteration of the host’s immune response and lung function, or both is not known. What is clear is that these viruses represent ubiquitous, potentially modifiable early-life exposures that are well established to be associated with disease and hold promise for primary or secondary prevention strategies for asthma. The strongest body of current evidence supports testing prevention of RSV LRTI in primary prevention trials (birth timing and RSV immunoprophylaxis have been demonstrated to decrease risk in observational studies) and shorter-term intervention studies on the outcome of recurrent wheezing. Important in these considerations is the selection of interventions that would be of acceptable risk in vulnerable populations of infants and pregnant women. For RV, the lack of a preventive intervention is an obstacle to advancing the field and should be a research priority for this ubiquitous early-life risk factor (117). Because not all infants who develop RSV or RV LRTI develop asthma, future studies will also need to focus on the genetics of both the host and the virus to better understand the host response to infection and if “asthmagenic” strains of RSV and RV exist that might be targets for vaccine or targeted small molecule development. In addition, further understanding the human immune response differences to early-life infection that predispose infants to developing asthma or aid in resolving early-life infection will be important. Altering the host immune response could be another potential early-life intervention preventing morbidity from early-life infection as well as recurrent wheezing and virally induced asthma exacerbations. Finally, because RV is most strongly associated with asthma exacerbations in children, continued efforts to advance our understanding of the altered immune response to viruses across the entire age continuum in patients with asthma and in atopic patients will aide in secondary prevention strategies and will likely provide insights into infant host susceptibility (118).

Footnotes

This work was supported by National Institutes of Health grants HL81420 (M.B.H.) and K24 AI 077930, UL1 RR024975, and U19AI 095227 (T.V.H.).

Author Contributions: A.S.F. contributed to assembling the evidence and drafting the manuscript. Y.H. contributed to the assembling of evidence on the immunobiology and animal models of disease and drafting these portions of the manuscript. M.L.M. contributed to reviewing the evidence on the immunobiology and animal models and editing these portions of the manuscript. M.B.H. contributed to assembling evidence on the immunobiology and animal models of disease and editing these portions of the manuscript. T.V.H. conceived of the idea for this perspective, oversaw the literature review, and edited the manuscript.

Originally Published in Press as DOI: 10.1164/rccm.201405-0901PP on November 4, 2014

Author disclosures are available with the text of this article at www.atsjournals.org.

References

  • 1.Almirall J, Bolíbar I, Serra-Prat M, Roig J, Hospital I, Carandell E, Agustí M, Ayuso P, Estela A, Torres A Community-Acquired Pneumonia in Catalan Countries (PACAP) Study Group. New evidence of risk factors for community-acquired pneumonia: a population-based study. Eur Respir J. 2008;31:1274–1284. doi: 10.1183/09031936.00095807. [DOI] [PubMed] [Google Scholar]
  • 2.Corne JM, Marshall C, Smith S, Schreiber J, Sanderson G, Holgate ST, Johnston SL. Frequency, severity, and duration of rhinovirus infections in asthmatic and non-asthmatic individuals: a longitudinal cohort study. Lancet. 2002;359:831–834. doi: 10.1016/S0140-6736(02)07953-9. [DOI] [PubMed] [Google Scholar]
  • 3.Eldeirawi K, McConnell R, Furner S, Freels S, Stayner L, Hernandez E, Amoruso L, Torres S, Persky V. Frequent ear infections in infancy and the risk of asthma in Mexican American children. J Asthma. 2010;47:473–477. doi: 10.3109/02770901003759428. [DOI] [PubMed] [Google Scholar]
  • 4.Green RM, Custovic A, Sanderson G, Hunter J, Johnston SL, Woodcock A. Synergism between allergens and viruses and risk of hospital admission with asthma: case-control study. BMJ. 2002;324:763. doi: 10.1136/bmj.324.7340.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Miller EK, Griffin MR, Edwards KM, Weinberg GA, Szilagyi PG, Staat MA, Iwane MK, Zhu Y, Hall CB, Fairbrother G, et al. New Vaccine Surveillance Network. Influenza burden for children with asthma. Pediatrics. 2008;121:1–8. doi: 10.1542/peds.2007-1053. [DOI] [PubMed] [Google Scholar]
  • 6.Olenec JP, Kim WK, Lee WM, Vang F, Pappas TE, Salazar LE, Evans MD, Bork J, Roberg K, Lemanske RF, Jr., et al. Weekly monitoring of children with asthma for infections and illness during common cold seasons. J Allergy Clin Immunol. 2010;125:1001–1006.e1. doi: 10.1016/j.jaci.2010.01.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Teepe J, Grigoryan L, Verheij TJ. Determinants of community-acquired pneumonia in children and young adults in primary care. Eur Respir J. 2010;35:1113–1117. doi: 10.1183/09031936.00101509. [DOI] [PubMed] [Google Scholar]
  • 8.Vinogradova Y, Hippisley-Cox J, Coupland C. Identification of new risk factors for pneumonia: population-based case-control study. Br J Gen Pract. 2009;59:e329–e338. doi: 10.3399/bjgp09X472629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Malmström K, Pitkäranta A, Carpen O, Pelkonen A, Malmberg LP, Turpeinen M, Kajosaari M, Sarna S, Lindahl H, Haahtela T, et al. Human rhinovirus in bronchial epithelium of infants with recurrent respiratory symptoms. J Allergy Clin Immunol. 2006;118:591–596. doi: 10.1016/j.jaci.2006.04.032. [DOI] [PubMed] [Google Scholar]
  • 10.Wos M, Sanak M, Soja J, Olechnowicz H, Busse WW, Szczeklik A. The presence of rhinovirus in lower airways of patients with bronchial asthma. Am J Respir Crit Care Med. 2008;177:1082–1089. doi: 10.1164/rccm.200607-973OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Juhn YJ, Kita H, Yawn BP, Boyce TG, Yoo KH, McGree ME, Weaver AL, Wollan P, Jacobson RM. Increased risk of serious pneumococcal disease in patients with asthma. J Allergy Clin Immunol. 2008;122:719–723. doi: 10.1016/j.jaci.2008.07.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Talbot TR, Hartert TV, Mitchel E, Halasa NB, Arbogast PG, Poehling KA, Schaffner W, Craig AS, Griffin MR. Asthma as a risk factor for invasive pneumococcal disease. N Engl J Med. 2005;352:2082–2090. doi: 10.1056/NEJMoa044113. [DOI] [PubMed] [Google Scholar]
  • 13.Xatzipsalti M, Kyrana S, Tsolia M, Psarras S, Bossios A, Laza-Stanca V, Johnston SL, Papadopoulos NG. Rhinovirus viremia in children with respiratory infections. Am J Respir Crit Care Med. 2005;172:1037–1040. doi: 10.1164/rccm.200502-315OC. [DOI] [PubMed] [Google Scholar]
  • 14.Singh AM, Moore PE, Gern JE, Lemanske RF, Jr, Hartert TV. Bronchiolitis to asthma: a review and call for studies of gene-virus interactions in asthma causation. Am J Respir Crit Care Med. 2007;175:108–119. doi: 10.1164/rccm.200603-435PP. [DOI] [PubMed] [Google Scholar]
  • 15.Wu P, Hartert TV. Evidence for a causal relationship between respiratory syncytial virus infection and asthma. Expert Rev Anti Infect Ther. 2011;9:731–745. doi: 10.1586/eri.11.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Faber TE, Schuurhof A, Vonk A, Koppelman GH, Hennus MP, Kimpen JL, Janssen R, Bont LJ. IL1RL1 gene variants and nasopharyngeal IL1RL-a levels are associated with severe RSV bronchiolitis: a multicenter cohort study. PLoS ONE. 2012;7:e34364. doi: 10.1371/journal.pone.0034364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Calışkan M, Bochkov YA, Kreiner-Møller E, Bønnelykke K, Stein MM, Du G, Bisgaard H, Jackson DJ, Gern JE, Lemanske RF, Jr, et al. Rhinovirus wheezing illness and genetic risk of childhood-onset asthma. N Engl J Med. 2013;368:1398–1407. doi: 10.1056/NEJMoa1211592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, Depner M, von Berg A, Bufe A, Rietschel E, et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature. 2007;448:470–473. doi: 10.1038/nature06014. [DOI] [PubMed] [Google Scholar]
  • 19.Carroll KN, Gebretsadik T, Minton P, Woodward K, Liu Z, Miller EK, Williams JV, Dupont WD, Hartert TV. Influence of maternal asthma on the cause and severity of infant acute respiratory tract infections. J Allergy Clin Immunol. 2012;129:1236–1242. doi: 10.1016/j.jaci.2012.01.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jackson DJ, Gangnon RE, Evans MD, Roberg KA, Anderson EL, Pappas TE, Printz MC, Lee WM, Shult PA, Reisdorf E, et al. Wheezing rhinovirus illnesses in early life predict asthma development in high-risk children. Am J Respir Crit Care Med. 2008;178:667–672. doi: 10.1164/rccm.200802-309OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sidhu SS, Yuan S, Innes AL, Kerr S, Woodruff PG, Hou L, Muller SJ, Fahy JV. Roles of epithelial cell-derived periostin in TGF-beta activation, collagen production, and collagen gel elasticity in asthma. Proc Natl Acad Sci USA. 2010;107:14170–14175. doi: 10.1073/pnas.1009426107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lee HH, Hoeman CM, Hardaway JC, Guloglu FB, Ellis JS, Jain R, Divekar R, Tartar DM, Haymaker CL, Zaghouani H. Delayed maturation of an IL-12-producing dendritic cell subset explains the early Th2 bias in neonatal immunity. J Exp Med. 2008;205:2269–2280. doi: 10.1084/jem.20071371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Contoli M, Message SD, Laza-Stanca V, Edwards MR, Wark PA, Bartlett NW, Kebadze T, Mallia P, Stanciu LA, Parker HL, et al. Role of deficient type III interferon-lambda production in asthma exacerbations. Nat Med. 2006;12:1023–1026. doi: 10.1038/nm1462. [DOI] [PubMed] [Google Scholar]
  • 24.Copenhaver CC, Gern JE, Li Z, Shult PA, Rosenthal LA, Mikus LD, Kirk CJ, Roberg KA, Anderson EL, Tisler CJ, et al. Cytokine response patterns, exposure to viruses, and respiratory infections in the first year of life. Am J Respir Crit Care Med. 2004;170:175–180. doi: 10.1164/rccm.200312-1647OC. [DOI] [PubMed] [Google Scholar]
  • 25.Fitzpatrick AM, Holguin F, Teague WG, Brown LA. Alveolar macrophage phagocytosis is impaired in children with poorly controlled asthma. J Allergy Clin Immunol. 2008;121:1372–1378, 1378.e1–3. doi: 10.1016/j.jaci.2008.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hammad H, Lambrecht BN. Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma. Nat Rev Immunol. 2008;8:193–204. doi: 10.1038/nri2275. [DOI] [PubMed] [Google Scholar]
  • 27.Meyer EH, DeKruyff RH, Umetsu DT. T cells and NKT cells in the pathogenesis of asthma. Annu Rev Med. 2008;59:281–292. doi: 10.1146/annurev.med.59.061506.154139. [DOI] [PubMed] [Google Scholar]
  • 28.Ordoñez CL, Khashayar R, Wong HH, Ferrando R, Wu R, Hyde DM, Hotchkiss JA, Zhang Y, Novikov A, Dolganov G, et al. Mild and moderate asthma is associated with airway goblet cell hyperplasia and abnormalities in mucin gene expression. Am J Respir Crit Care Med. 2001;163:517–523. doi: 10.1164/ajrccm.163.2.2004039. [DOI] [PubMed] [Google Scholar]
  • 29.Wark PA, Johnston SL, Bucchieri F, Powell R, Puddicombe S, Laza-Stanca V, Holgate ST, Davies DE. Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus. J Exp Med. 2005;201:937–947. doi: 10.1084/jem.20041901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bisgaard H, Jensen SM, Bønnelykke K. Interaction between asthma and lung function growth in early life. Am J Respir Crit Care Med. 2012;185:1183–1189. doi: 10.1164/rccm.201110-1922OC. [DOI] [PubMed] [Google Scholar]
  • 31.Korppi M, Piippo-Savolainen E, Korhonen K, Remes S. Respiratory morbidity 20 years after RSV infection in infancy. Pediatr Pulmonol. 2004;38:155–160. doi: 10.1002/ppul.20058. [DOI] [PubMed] [Google Scholar]
  • 32.Martinez FD. What have we learned from the Tucson Children’s Respiratory Study? Paediatr Respir Rev. 2002;3:193–197. doi: 10.1016/s1526-0542(02)00188-4. [DOI] [PubMed] [Google Scholar]
  • 33.Taussig LM, Wright AL, Holberg CJ, Halonen M, Morgan WJ, Martinez FD. Tucson Children’s Respiratory Study: 1980 to present. J Allergy Clin Immunol. 2003;111:661–675, quiz 676. doi: 10.1067/mai.2003.162. [DOI] [PubMed] [Google Scholar]
  • 34.Turner SW, Young S, Landau LI, Le Souëf PN. Reduced lung function both before bronchiolitis and at 11 years. Arch Dis Child. 2002;87:417–420. doi: 10.1136/adc.87.5.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nair H, Nokes DJ, Gessner BD, Dherani M, Madhi SA, Singleton RJ, O’Brien KL, Roca A, Wright PF, Bruce N, et al. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet. 2010;375:1545–1555. doi: 10.1016/S0140-6736(10)60206-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Anderson LJ, Hierholzer JC, Tsou C, Hendry RM, Fernie BF, Stone Y, McIntosh K. Antigenic characterization of respiratory syncytial virus strains with monoclonal antibodies. J Infect Dis. 1985;151:626–633. doi: 10.1093/infdis/151.4.626. [DOI] [PubMed] [Google Scholar]
  • 37.Botosso VF, Zanotto PM, Ueda M, Arruda E, Gilio AE, Vieira SE, Stewien KE, Peret TC, Jamal LF, Pardini MI, et al. VGDN Consortium. Positive selection results in frequent reversible amino acid replacements in the G protein gene of human respiratory syncytial virus. PLoS Pathog. 2009;5:e1000254. doi: 10.1371/journal.ppat.1000254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Trento A, Casas I, Calderón A, Garcia-Garcia ML, Calvo C, Perez-Breña P, Melero JA. Ten years of global evolution of the human respiratory syncytial virus BA genotype with a 60-nucleotide duplication in the G protein gene. J Virol. 2010;84:7500–7512. doi: 10.1128/JVI.00345-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Peret TC, Hall CB, Hammond GW, Piedra PA, Storch GA, Sullender WM, Tsou C, Anderson LJ. Circulation patterns of group A and B human respiratory syncytial virus genotypes in 5 communities in North America. J Infect Dis. 2000;181:1891–1896. doi: 10.1086/315508. [DOI] [PubMed] [Google Scholar]
  • 40.Walsh EE, McConnochie KM, Long CE, Hall CB. Severity of respiratory syncytial virus infection is related to virus strain. J Infect Dis. 1997;175:814–820. doi: 10.1086/513976. [DOI] [PubMed] [Google Scholar]
  • 41.Simões EA, Carbonell-Estrany X, Rieger CH, Mitchell I, Fredrick L, Groothuis JR Palivizumab Long-Term Respiratory Outcomes Study Group. The effect of respiratory syncytial virus on subsequent recurrent wheezing in atopic and nonatopic children. J Allergy Clin Immunol. 2010;126:256–262. doi: 10.1016/j.jaci.2010.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Stokes KL, Chi MH, Sakamoto K, Newcomb DC, Currier MG, Huckabee MM, Lee S, Goleniewska K, Pretto C, Williams JV, et al. Differential pathogenesis of respiratory syncytial virus clinical isolates in BALB/c mice. J Virol. 2011;85:5782–5793. doi: 10.1128/JVI.01693-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Palmenberg AC, Rathe JA, Liggett SB. Analysis of the complete genome sequences of human rhinovirus. J Allergy Clin Immunol. 2010;125:1190–1199; quiz 1200–1201. doi: 10.1016/j.jaci.2010.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Palmenberg AC, Spiro D, Kuzmickas R, Wang S, Djikeng A, Rathe JA, Fraser-Liggett CM, Liggett SB. Sequencing and analyses of all known human rhinovirus genomes reveal structure and evolution. Science. 2009;324:55–59. doi: 10.1126/science.1165557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lamson D, Renwick N, Kapoor V, Liu Z, Palacios G, Ju J, Dean A, St George K, Briese T, Lipkin WI. MassTag polymerase-chain-reaction detection of respiratory pathogens, including a new rhinovirus genotype, that caused influenza-like illness in New York State during 2004-2005. J Infect Dis. 2006;194:1398–1402. doi: 10.1086/508551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Bochkov YA, Gern JE. Clinical and molecular features of human rhinovirus C. Microbes Infect. 2012;14:485–494. doi: 10.1016/j.micinf.2011.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lee WM, Lemanske RF, Jr, Evans MD, Vang F, Pappas T, Gangnon R, Jackson DJ, Gern JE. Human rhinovirus species and season of infection determine illness severity. Am J Respir Crit Care Med. 2012;186:886–891. doi: 10.1164/rccm.201202-0330OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Miller EK, Mackay IM. From sneeze to wheeze: what we know about rhinovirus Cs. J Clin Virol. 2013;57:291–299. doi: 10.1016/j.jcv.2013.04.015. [DOI] [PubMed] [Google Scholar]
  • 49.DiFranza JR, Aligne CA, Weitzman M. Prenatal and postnatal environmental tobacco smoke exposure and children’s health. Pediatrics. 2004;113(Suppl):1007–1015. [PubMed] [Google Scholar]
  • 50.Hu FB, Persky V, Flay BR, Richardson J. An epidemiological study of asthma prevalence and related factors among young adults. J Asthma. 1997;34:67–76. doi: 10.3109/02770909709071205. [DOI] [PubMed] [Google Scholar]
  • 51.Jedrychowski W, Flak E. Maternal smoking during pregnancy and postnatal exposure to environmental tobacco smoke as predisposition factors to acute respiratory infections. Environ Health Perspect. 1997;105:302–306. doi: 10.1289/ehp.97105302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Martinez FD, Cline M, Burrows B. Increased incidence of asthma in children of smoking mothers. Pediatrics. 1992;89:21–26. [PubMed] [Google Scholar]
  • 53.Devereux G, Barker RN, Seaton A. Antenatal determinants of neonatal immune responses to allergens. Clin Exp Allergy. 2002;32:43–50. doi: 10.1046/j.0022-0477.2001.01267.x. [DOI] [PubMed] [Google Scholar]
  • 54.Noakes PS, Hale J, Thomas R, Lane C, Devadason SG, Prescott SL. Maternal smoking is associated with impaired neonatal toll-like-receptor-mediated immune responses. Eur Respir J. 2006;28:721–729. doi: 10.1183/09031936.06.00050206. [DOI] [PubMed] [Google Scholar]
  • 55.Noakes PS, Holt PG, Prescott SL. Maternal smoking in pregnancy alters neonatal cytokine responses. Allergy. 2003;58:1053–1058. doi: 10.1034/j.1398-9995.2003.00290.x. [DOI] [PubMed] [Google Scholar]
  • 56.Fainaru O, Shseyov D, Hantisteanu S, Groner Y. Accelerated chemokine receptor 7-mediated dendritic cell migration in Runx3 knockout mice and the spontaneous development of asthma-like disease. Proc Natl Acad Sci USA. 2005;102:10598–10603. doi: 10.1073/pnas.0504787102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hollingsworth JW, Maruoka S, Boon K, Garantziotis S, Li Z, Tomfohr J, Bailey N, Potts EN, Whitehead G, Brass DM, et al. In utero supplementation with methyl donors enhances allergic airway disease in mice. J Clin Invest. 2008;118:3462–3469. doi: 10.1172/JCI34378. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 58.Veeranki SP, Gebretsadik T, Dorris SL, Mitchel EF, Hartert TV, Cooper WO, Tylavsky FA, Dupont W, Hartman TJ, Carroll KN. Association of folic acid supplementation during pregnancy and infant bronchiolitis. Am J Epidemiol. 2014;179:938–946. doi: 10.1093/aje/kwu019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Belderbos ME, Houben ML, Wilbrink B, Lentjes E, Bloemen EM, Kimpen JL, Rovers M, Bont L. Cord blood vitamin D deficiency is associated with respiratory syncytial virus bronchiolitis. Pediatrics. 2011;127:e1513–e1520. doi: 10.1542/peds.2010-3054. [DOI] [PubMed] [Google Scholar]
  • 60.Camargo CA, Jr, Ingham T, Wickens K, Thadhani R, Silvers KM, Epton MJ, Town GI, Pattemore PK, Espinola JA, Crane J New Zealand Asthma and Allergy Cohort Study Group. Cord-blood 25-hydroxyvitamin D levels and risk of respiratory infection, wheezing, and asthma. Pediatrics. 2011;127:e180–e187. doi: 10.1542/peds.2010-0442. [DOI] [PubMed] [Google Scholar]
  • 61.Ginde AA, Mansbach JM, Camargo CA., Jr Association between serum 25-hydroxyvitamin D level and upper respiratory tract infection in the Third National Health and Nutrition Examination Survey. Arch Intern Med. 2009;169:384–390. doi: 10.1001/archinternmed.2008.560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sundar IK, Rahman I. Vitamin d and susceptibility of chronic lung diseases: role of epigenetics. Front Pharmacol. 2011;2:50. doi: 10.3389/fphar.2011.00050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wayse V, Yousafzai A, Mogale K, Filteau S. Association of subclinical vitamin D deficiency with severe acute lower respiratory infection in Indian children under 5 y. Eur J Clin Nutr. 2004;58:563–567. doi: 10.1038/sj.ejcn.1601845. [DOI] [PubMed] [Google Scholar]
  • 64.Jaspers I, Zhang W, Brighton LE, Carson JL, Styblo M, Beck MA. Selenium deficiency alters epithelial cell morphology and responses to influenza. Free Radic Biol Med. 2007;42:1826–1837. doi: 10.1016/j.freeradbiomed.2007.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Valkhoff VE, Schade R.’t Jong GW, Romio S, Schuemie MJ, Arfe A, Garbe E, Herings R, Lucchi S, Picelli G, et al. Population-based analysis of non-steroidal anti-inflammatory drug use among children in four European countries in the SOS project: what size of data platforms and which study designs do we need to assess safety issues? BMC Pediatr 201313192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lee SH, Kang MJ, Yu HS, Hong K, Jung YH, Kim HY, Seo JH, Kwon JW, Kim BJ, Kim HJ, et al. Association between recent acetaminophen use and asthma: modification by polymorphism at TLR4. 2014;29:662–668. doi: 10.3346/jkms.2014.29.5.662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bisgaard H, Hermansen MN, Buchvald F, Loland L, Halkjaer LB, Bønnelykke K, Brasholt M, Heltberg A, Vissing NH, Thorsen SV, et al. Childhood asthma after bacterial colonization of the airway in neonates. N Engl J Med. 2007;357:1487–1495. doi: 10.1056/NEJMoa052632. [DOI] [PubMed] [Google Scholar]
  • 68.Kloepfer KM, Lee WM, Pappas TE, Kang TJ, Vrtis RF, Evans MD, Gangnon RE, Bochkov YA, Jackson DJ, Lemanske RF, Jr., et al. Detection of pathogenic bacteria during rhinovirus infection is associated with increased respiratory symptoms and asthma exacerbations. J Allergy Clin Immunol. 2014;133:1301–1307.e3. doi: 10.1016/j.jaci.2014.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Luoto R, Ruuskanen O, Waris M, Kalliomäki M, Salminen S, Isolauri E. Prebiotic and probiotic supplementation prevents rhinovirus infections in preterm infants: a randomized, placebo-controlled trial. J Allergy Clin Immunol. 2014;133:405–413. doi: 10.1016/j.jaci.2013.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lambrecht BN, Hammad H. Biology of lung dendritic cells at the origin of asthma. Immunity. 2009;31:412–424. doi: 10.1016/j.immuni.2009.08.008. [DOI] [PubMed] [Google Scholar]
  • 71.Monick MM, Yarovinsky TO, Powers LS, Butler NS, Carter AB, Gudmundsson G, Hunninghake GW. Respiratory syncytial virus up-regulates TLR4 and sensitizes airway epithelial cells to endotoxin. J Biol Chem. 2003;278:53035–53044. doi: 10.1074/jbc.M308093200. [DOI] [PubMed] [Google Scholar]
  • 72.Rate A, Upham JW, Bosco A, McKenna KL, Holt PG. Airway epithelial cells regulate the functional phenotype of locally differentiating dendritic cells: implications for the pathogenesis of infectious and allergic airway disease. J Immunol. 2009;182:72–83. doi: 10.4049/jimmunol.182.1.72. [DOI] [PubMed] [Google Scholar]
  • 73.Lemanske RF, Jr, Jackson DJ, Gangnon RE, Evans MD, Li Z, Shult PA, Kirk CJ, Reisdorf E, Roberg KA, Anderson EL, et al. Rhinovirus illnesses during infancy predict subsequent childhood wheezing. J Allergy Clin Immunol. 2005;116:571–577. doi: 10.1016/j.jaci.2005.06.024. [DOI] [PubMed] [Google Scholar]
  • 74.Jackson DJ, Evans MD, Gangnon RE, Tisler CJ, Pappas TE, Lee WM, Gern JE, Lemanske RF., Jr Evidence for a causal relationship between allergic sensitization and rhinovirus wheezing in early life. Am J Respir Crit Care Med. 2012;185:281–285. doi: 10.1164/rccm.201104-0660OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bacharier LB, Cohen R, Schweiger T, Yin-Declue H, Christie C, Zheng J, Schechtman KB, Strunk RC, Castro M. Determinants of asthma after severe respiratory syncytial virus bronchiolitis. J Allergy Clin Immunol. 2012;130:91–100.e3. doi: 10.1016/j.jaci.2012.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Escobar GJ, Ragins A, Li SX, Prager L, Masaquel AS, Kipnis P. Recurrent wheezing in the third year of life among children born at 32 weeks’ gestation or later: relationship to laboratory-confirmed, medically attended infection with respiratory syncytial virus during the first year of life. Arch Pediatr Adolesc Med. 2010;164:915–922. doi: 10.1001/archpediatrics.2010.177. [DOI] [PubMed] [Google Scholar]
  • 77.Kotaniemi-Syrjänen A, Vainionpää R, Reijonen TM, Waris M, Korhonen K, Korppi M. Rhinovirus-induced wheezing in infancy: the first sign of childhood asthma? J Allergy Clin Immunol. 2003;111:66–71. doi: 10.1067/mai.2003.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kusel MM, de Klerk NH, Kebadze T, Vohma V, Holt PG, Johnston SL, Sly PD. Early-life respiratory viral infections, atopic sensitization, and risk of subsequent development of persistent asthma. J Allergy Clin Immunol. 2007;119:1105–1110. doi: 10.1016/j.jaci.2006.12.669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kusel MM, Kebadze T, Johnston SL, Holt PG, Sly PD. Febrile respiratory illnesses in infancy and atopy are risk factors for persistent asthma and wheeze. Eur Respir J. 2012;39:876–882. doi: 10.1183/09031936.00193310. [DOI] [PubMed] [Google Scholar]
  • 80.Sigurs N, Bjarnason R, Sigurbergsson F, Kjellman B. Respiratory syncytial virus bronchiolitis in infancy is an important risk factor for asthma and allergy at age 7. Am J Respir Crit Care Med. 2000;161:1501–1507. doi: 10.1164/ajrccm.161.5.9906076. [DOI] [PubMed] [Google Scholar]
  • 81.Stein RT, Sherrill D, Morgan WJ, Holberg CJ, Halonen M, Taussig LM, Wright AL, Martinez FD. Respiratory syncytial virus in early life and risk of wheeze and allergy by age 13 years. Lancet. 1999;354:541–545. doi: 10.1016/S0140-6736(98)10321-5. [DOI] [PubMed] [Google Scholar]
  • 82.Carroll KN, Wu P, Gebretsadik T, Griffin MR, Dupont WD, Mitchel EF, Hartert TV. The severity-dependent relationship of infant bronchiolitis on the risk and morbidity of early childhood asthma. J Allergy Clin Immunol. 2009;123:1055–1061.e1. doi: 10.1016/j.jaci.2009.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Thomsen SF, van der Sluis S, Stensballe LG, Posthuma D, Skytthe A, Kyvik KO, Duffy DL, Backer V, Bisgaard H. Exploring the association between severe respiratory syncytial virus infection and asthma: a registry-based twin study. Am J Respir Crit Care Med. 2009;179:1091–1097. doi: 10.1164/rccm.200809-1471OC. [DOI] [PubMed] [Google Scholar]
  • 84.Voraphani N, Stern DA, Wright AL, Guerra S, Morgan WJ, Martinez FD. Risk of current asthma among adult smokers with respiratory syncytial virus illnesses in early life. Am J Respir Crit Care Med. 2014;190:392–398. doi: 10.1164/rccm.201311-2095OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Oh JW, Lee HB, Park IK, Kang JO. Interleukin-6, interleukin-8, interleukin-11, and interferon-gamma levels in nasopharyngeal aspirates from wheezing children with respiratory syncytial virus or influenza A virus infection. Pediatr Allergy Immunol. 2002;13:350–356. doi: 10.1034/j.1399-3038.2002.02018.x. [DOI] [PubMed] [Google Scholar]
  • 86.Lewis TC, Henderson TA, Carpenter AR, Ramirez IA, McHenry CL, Goldsmith AM, Ren X, Mentz GB, Mukherjee B, Robins TG, et al. Nasal cytokine responses to natural colds in asthmatic children. Clin Exp Allergy. 2012;42:1734–1744. doi: 10.1111/cea.12005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Molet S, Hamid Q, Davoine F, Nutku E, Taha R, Pagé N, Olivenstein R, Elias J, Chakir J. IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J Allergy Clin Immunol. 2001;108:430–438. doi: 10.1067/mai.2001.117929. [DOI] [PubMed] [Google Scholar]
  • 88.Mukherjee S, Lindell DM, Berlin AA, Morris SB, Shanley TP, Hershenson MB, Lukacs NW. IL-17-induced pulmonary pathogenesis during respiratory viral infection and exacerbation of allergic disease. Am J Pathol. 2011;179:248–258. doi: 10.1016/j.ajpath.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Barends M, de Rond LG, Dormans J, van Oosten M, Boelen A, Neijens HJ, Osterhaus AD, Kimman TG. Respiratory syncytial virus, pneumonia virus of mice, and influenza A virus differently affect respiratory allergy in mice. Clin Exp Allergy. 2004;34:488–496. doi: 10.1111/j.1365-2222.2004.01906.x. [DOI] [PubMed] [Google Scholar]
  • 90.Kim EY, Battaile JT, Patel AC, You Y, Agapov E, Grayson MH, Benoit LA, Byers DE, Alevy Y, Tucker J, et al. Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease. Nat Med. 2008;14:633–640. doi: 10.1038/nm1770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Chang YJ, Kim HY, Albacker LA, Baumgarth N, McKenzie AN, Smith DE, Dekruyff RH, Umetsu DT. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat Immunol. 2011;12:631–638. doi: 10.1038/ni.2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hong JY, Bentley JK, Chung Y, Lei J, Steenrod JM, Chen Q, Sajjan US, Hershenson MB. Neonatal rhinovirus induces mucous metaplasia and airways hyperresponsiveness through IL-25 and type 2 innate lymphoid cells. J Allergy Clin Immunol. 2014;134:429–439. doi: 10.1016/j.jaci.2014.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kato A, Favoreto S, Jr, Avila PC, Schleimer RP. TLR3- and Th2 cytokine-dependent production of thymic stromal lymphopoietin in human airway epithelial cells. J Immunol. 2007;179:1080–1087. doi: 10.4049/jimmunol.179.2.1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Perez GF, Pancham K, Huseni S, Preciado D, Freishtat RJ, Colberg-Poley AM, Hoffman EP, Rose MC, Nino G. Rhinovirus infection in young children is associated with elevated airway TSLP levels. Eur Respir J. 2014;44:1075–1078. doi: 10.1183/09031936.00049214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Krishnamoorthy N, Khare A, Oriss TB, Raundhal M, Morse C, Yarlagadda M, Wenzel SE, Moore ML, Peebles RS, Jr, Ray A, et al. Early infection with respiratory syncytial virus impairs regulatory T cell function and increases susceptibility to allergic asthma. Nat Med. 2012;18:1525–1530. doi: 10.1038/nm.2896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Rezaee F, DeSando SA, Ivanov AI, Chapman TJ, Knowlden SA, Beck LA, Georas SN. Sustained protein kinase D activation mediates respiratory syncytial virus-induced airway barrier disruption. J Virol. 2013;87:11088–11095. doi: 10.1128/JVI.01573-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Sajjan U, Wang Q, Zhao Y, Gruenert DC, Hershenson MB. Rhinovirus disrupts the barrier function of polarized airway epithelial cells. Am J Respir Crit Care Med. 2008;178:1271–1281. doi: 10.1164/rccm.200801-136OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kaiko GE, Loh Z, Spann K, Lynch JP, Lalwani A, Zheng Z, Davidson S, Uematsu S, Akira S, Hayball J, et al. Toll-like receptor 7 gene deficiency and early-life Pneumovirus infection interact to predispose toward the development of asthma-like pathology in mice. J Allergy Clin Immunol. 2013;131:1331–1339.e10. doi: 10.1016/j.jaci.2013.02.041. [DOI] [PubMed] [Google Scholar]
  • 99.Peebles RS, Jr, Sheller JR, Johnson JE, Mitchell DB, Graham BS. Respiratory syncytial virus infection prolongs methacholine-induced airway hyperresponsiveness in ovalbumin-sensitized mice. J Med Virol. 1999;57:186–192. doi: 10.1002/(sici)1096-9071(199902)57:2<186::aid-jmv17>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 100.Schneider D, Hong JY, Popova AP, Bowman ER, Linn MJ, McLean AM, Zhao Y, Sonstein J, Bentley JK, Weinberg JB, et al. Neonatal rhinovirus infection induces mucous metaplasia and airways hyperresponsiveness. J Immunol. 2012;188:2894–2904. doi: 10.4049/jimmunol.1101391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sorkness RL, Szakaly RJ, Rosenthal LA, Sullivan R, Gern JE, Lemanske RF, Jr, Sun X. Viral bronchiolitis in young rats causes small airway lesions that correlate with reduced lung function. Am J Respir Cell Mol Biol. 2013;49:808–813. doi: 10.1165/rcmb.2013-0096OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Dakhama A, Park JW, Taube C, Joetham A, Balhorn A, Miyahara N, Takeda K, Gelfand EW. The enhancement or prevention of airway hyperresponsiveness during reinfection with respiratory syncytial virus is critically dependent on the age at first infection and IL-13 production. J Immunol. 2005;175:1876–1883. doi: 10.4049/jimmunol.175.3.1876. [DOI] [PubMed] [Google Scholar]
  • 103.You D, Becnel D, Wang K, Ripple M, Daly M, Cormier SA. Exposure of neonates to respiratory syncytial virus is critical in determining subsequent airway response in adults. Respir Res. 2006;7:107. doi: 10.1186/1465-9921-7-107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Ilarraza R, Wu Y, Skappak CD, Ajamian F, Proud D, Adamko DJ. Rhinovirus has the unique ability to directly activate human T cells in vitro. J Allergy Clin Immunol. 2013;131:395–404. doi: 10.1016/j.jaci.2012.11.041. [DOI] [PubMed] [Google Scholar]
  • 105.Kuo C, Lim S, King NJ, Johnston SL, Burgess JK, Black JL, Oliver BG. Rhinovirus infection induces extracellular matrix protein deposition in asthmatic and nonasthmatic airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2011;300:L951–L957. doi: 10.1152/ajplung.00411.2010. [DOI] [PubMed] [Google Scholar]
  • 106.Bartlett NW, Walton RP, Edwards MR, Aniscenko J, Caramori G, Zhu J, Glanville N, Choy KJ, Jourdan P, Burnet J, et al. Mouse models of rhinovirus-induced disease and exacerbation of allergic airway inflammation. Nat Med. 2008;14:199–204. doi: 10.1038/nm1713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jartti T, Lehtinen P, Vuorinen T, Ruuskanen O. Bronchiolitis: age and previous wheezing episodes are linked to viral etiology and atopic characteristics. Pediatr Infect Dis J. 2009;28:311–317. doi: 10.1097/INF.0b013e31818ee0c1. [DOI] [PubMed] [Google Scholar]
  • 108.Rakes GP, Arruda E, Ingram JM, Hoover GE, Zambrano JC, Hayden FG, Platts-Mills TA, Heymann PW. Rhinovirus and respiratory syncytial virus in wheezing children requiring emergency care: IgE and eosinophil analyses. Am J Respir Crit Care Med. 1999;159:785–790. doi: 10.1164/ajrccm.159.3.9801052. [DOI] [PubMed] [Google Scholar]
  • 109.James KM, Gebretsadik T, Escobar GJ, Wu P, Carroll KN, Li SX, Walsh EM, Mitchel EF, Sloan C, Hartert TV. Risk of childhood asthma following infant bronchiolitis during the respiratory syncytial virus season. J Allergy Clin Immunol. 2013;132:227–229. doi: 10.1016/j.jaci.2013.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Blanken MO, Rovers MM, Molenaar JM, Winkler-Seinstra PL, Meijer A, Kimpen JL, Bont L Dutch RSV Neonatal Network. Respiratory syncytial virus and recurrent wheeze in healthy preterm infants. N Engl J Med. 2013;368:1791–1799. doi: 10.1056/NEJMoa1211917. [DOI] [PubMed] [Google Scholar]
  • 111.Simoes EA, Groothuis JR, Carbonell-Estrany X, Rieger CH, Mitchell I, Fredrick LM, Kimpen JL. Palivizumab prophylaxis, respiratory syncytial virus, and subsequent recurrent wheezing. J Pediatr. 2007;151:34–42.e1. doi: 10.1016/j.jpeds.2007.02.032. [DOI] [PubMed] [Google Scholar]
  • 112.Wenzel SE, Gibbs RL, Lehr MV, Simoes EA. Respiratory outcomes in high-risk children 7 to 10 years after prophylaxis with respiratory syncytial virus immune globulin. Am J Med. 2002;112:627–633. doi: 10.1016/s0002-9343(02)01095-1. [DOI] [PubMed] [Google Scholar]
  • 113.Yoshihara S, Kusuda S, Mochizuki H, Okada K, Nishima S, Simões EA C-CREW Investigators. Effect of palivizumab prophylaxis on subsequent recurrent wheezing in preterm infants. Pediatrics. 2013;132:811–818. doi: 10.1542/peds.2013-0982. [DOI] [PubMed] [Google Scholar]
  • 114.Chen CH, Lin YT, Yang YH, Wang LC, Lee JH, Kao CL, Chiang BL. Ribavirin for respiratory syncytial virus bronchiolitis reduced the risk of asthma and allergen sensitization. Pediatr Allergy Immunol. 2008;19:166–172. doi: 10.1111/j.1399-3038.2007.00610.x. [DOI] [PubMed] [Google Scholar]
  • 115.Stensballe LG, Ravn H, Kristensen K, Agerskov K, Meakins T, Aaby P, Simões EA. Respiratory syncytial virus neutralizing antibodies in cord blood, respiratory syncytial virus hospitalization, and recurrent wheeze. J Allergy Clin Immunol. 2009;123:398–403. doi: 10.1016/j.jaci.2008.10.043. [DOI] [PubMed] [Google Scholar]
  • 116.Piedimonte G, Walton C, Samsell L. Vertical transmission of respiratory syncytial virus modulates pre- and postnatal innervation and reactivity of rat airways. PLoS ONE. 2013;8:e61309. doi: 10.1371/journal.pone.0061309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Privolizzi R, Solari R, Johnston SL, McLean GR. The application of prophylactic antibodies for rhinovirus infections. Antivir Chem Chemother. 2014;23:173–177. doi: 10.3851/IMP2578. [DOI] [PubMed] [Google Scholar]
  • 118.Rezaee F, Gibson LF, Piktel D, Othumpangat S, Piedimonte G. Respiratory syncytial virus infection in human bone marrow stromal cells. Am J Respir Cell Mol Biol. 2011;45:277–286. doi: 10.1165/rcmb.2010-0121OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, Symington P, O’Toole S, Myint SH, Tyrrell DA, et al. Community study of role of viral infections in exacerbations of asthma in 9-11 year old children. BMJ. 1995;310:1225–1229. doi: 10.1136/bmj.310.6989.1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wu P, Dupont WD, Griffin MR, Carroll KN, Mitchel EF, Gebretsadik T, Hartert TV. Evidence of a causal role of winter virus infection during infancy in early childhood asthma. Am J Respir Crit Care Med. 2008;178:1123–1129. doi: 10.1164/rccm.200804-579OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Mello C, Aguayo E, Rodriguez M, Lee G, Jordan R, Cihlar T, Birkus G. Multiple classes of antiviral agents exhibit in vitro activity against human rhinovirus type C. Antimicrob Agents Chemother. 2014;58:1546–1555. doi: 10.1128/AAC.01746-13. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Respiratory and Critical Care Medicine are provided here courtesy of American Thoracic Society

RESOURCES