Skip to main content

Objectives, design and enrollment results from the Infant Susceptibility to Pulmonary Infections and Asthma Following RSV Exposure Study (INSPIRE)

Abstract

Background

Respiratory syncytial virus (RSV) lower respiratory tract infection (LRI) during infancy has been consistently associated with an increased risk of childhood asthma. In addition, evidence supports that this relationship is causal. However, the mechanisms through which RSV contributes to asthma development are not understood. The INSPIRE (Infant Susceptibility to Pulmonary Infections and Asthma Following RSV Exposure) study objectives are to: 1) characterize the host phenotypic response to RSV infection in infancy and the risk of recurrent wheeze and asthma, 2) identify the immune response and lung injury patterns of RSV infection that are associated with the development of early childhood wheezing illness and asthma, and 3) determine the contribution of specific RSV strains to early childhood wheezing and asthma development. This article describes the INSPIRE study, including study aims, design, recruitment results, and enrolled population characteristics.

Methods/design

The cohort is a population based longitudinal birth cohort of term healthy infants enrolled during the first months of life over a two year period. Respiratory infection surveillance was conducted from November to March of the first year of life, through surveys administered every two weeks. In-person illness visits were conducted if infants met pre-specified criteria for a respiratory illness visit. Infants will be followed annually to ages 3-4 years for assessment of the primary endpoint: wheezing illness. Nasal, urine, stool and blood samples were collected at various time points throughout the study for measurements of host and viral factors that predict wheezing illness. Nested case-control studies will additionally be used to address other primary and secondary hypotheses.

Discussion

In the INSPIRE study, 1952 infants (48% female) were enrolled during the two enrollment years and follow-up will continue through 2016. The mean age of enrollment was 60 days. During winter viral season, more than 14,000 surveillance surveys were carried out resulting in 2,103 respiratory illness visits on 1189 infants. First year follow-up has been completed on over 95% percent of participants from the first year of enrollment.

With ongoing follow-up for wheezing and childhood asthma outcomes, the INSPIRE study will advance our understanding of the complex causal relationship between RSV infection and early childhood wheezing and asthma.

Background

Prospective epidemiologic studies have consistently demonstrated that infants with respiratory syncytial virus (RSV) lower respiratory tract infections (LRIs) have an increased risk of early childhood wheezing and asthma [1-5]. There is also evidence from animal [6-10], human [8,11], and clinical studies [12,13] that mild RSV infections may reduce the risk of asthma development, presumptively by modulating pathogenic immunity through early activation and expansion of T regulatory cells. Intervention trials in humans demonstrating that preventing infant RSV illness prevents recurrent wheezing, and mechanistic studies in animal models provide proof of concept and plausible mechanisms through which RSV could cause asthma [14-17]. Additional research demonstrates that the strain of RSV is associated with the clinical severity of acute infection [18-20]. Thus, RSV may be an important organism that modifies the developing immune system, differently, depending on the severity or host response to the virus. This is important, as RSV prevention or modulating host response to viral infection could result in a new target for primary asthma prevention. The INSPIRE (Infant Susceptibility to Pulmonary Infections and Asthma Following RSV Exposure) study builds upon the known links between RSV LRI and wheezing illness to address unanswered questions about the role of mild RSV infection in decreasing the risk of childhood wheezing illness and host and viral characteristics associated with wheezing or asthma following infant RSV infection.

Because we cannot yet predict which infants with RSV LRI will ultimately develop childhood asthma and wheezing illnesses, or who might most benefit from preventing early viral infection [1,21,22], the INSPIRE study cohort was designed to help understand how the complete spectrum (mild to severe) of RSV illnesses contributes to early childhood wheezing and asthma. In particular, because there is a paucity of studies on outpatient RSV, our study captures mild RSV infections. This longitudinal cohort was specifically designed to 1) establish the relationship between the host phenotypic response to RSV infection in the first 6 months of life and the risk of recurrent wheeze and asthma; 2) identify the host immune response determinants of the RSV infection phenotype on the development of early childhood wheezing and asthma following RSV infection; and 3) determine the contribution of specific RSV strains to early childhood wheezing and asthma development. Figure 1 depicts the conceptual framework that the study is built upon.

Figure 1
figure 1

Hypothesis Testing: The epidemiologic triangle.

This paper details the study aims, procedures, and recruitment results from the INSPIRE study, a unique birth cohort of over 1,900 term, healthy infants followed longitudinally from infancy through early childhood for the outcomes of wheezing illnesses and asthma, with detailed characterization of respiratory illness during infancy during RSV season. The study will identify host, viral and environmental factors associated with RSV infection in infancy and the development of childhood asthma.

Methods

Primary objectives

The study was designed to test two primary hypotheses: 1) the severity of RSV infection during infancy is associated with recurrent wheezing and early childhood asthma in a severity-dependent manner and; 2) indices of host immune response and lung injury associated with infant RSV infection will differ among infants who do and do not develop recurrent wheezing and early childhood asthma.

Secondary objectives

The secondary objectives are to: 1) test the hypothesis that specific RSV strains will be associated with differential LRI severity, recurrent wheezing and early childhood asthma; 2) determine the association between all viral infections (URIs only (mild) any LRI (severe)) and allergic rhinitis (AR); 3) test the interactions between allergic rhinitis/laboratory atopy and smoking, family history of asthma, infant age at RSV infection with respiratory infection severity; 4) assess the relationship between LRI severity and allergic rhinitis within the LRI subgroup; 5) determine the association of RSV infection severity with recurrent wheezing and wheezing illness severity; 6) test the hypothesis that there are RSV strain differences in the pattern of cytokines in acute infection; 7) longitudinally characterize the airway microbiome in early infancy, during infant RSV acute respiratory illness (ARI), and at 2-3 years of age on a subset of children (n = 100); 8) determine whether the specific patterns of airway microbiome diversity in early infancy (prior to RSV ARI), during infant RSV ARI, and/or the change in pattern between health and RSV ARI are associated with respiratory outcomes of severity of RSV ARI in infancy and subsequent development of wheezing illness in early childhood; and 9) determine whether specific patterns of airway microbial diversity in early infancy (prior to RSV ARI), during infant RSV ARI, and/or the change in pattern between health and RSV ARI are associated with patterns T cell responses and innate cellular responses to RSV stimulation at 2-3 years on a subset of infants.

Study design

To address primary hypothesis 1, we designed a longitudinal observational birth cohort study of infants (the INSPIRE cohort), enrolled over a two year period, which included respiratory illness surveillance from November 1 to March 31 during the infant’s first year of life, and annual follow-up to the third or fourth birthday. Primary hypothesis 2 will be addressed through the use of a nested case-control study from participants within the cohort.

Study population and subject recruitment

We enrolled and are longitudinally following 1952 term, otherwise healthy infants from pediatric practices located in Middle Tennessee, or self-referral through research announcements. The recruitment area encompasses urban, suburban and rural areas. Infants meeting eligibility criteria outlined in Table 1 were approached at time of a pediatric visit to a participating practice. By design, infants whose birthdays fell between June 1st and December 31st were targeted for enrollment so that infants were on average younger than 6 months of age during winter virus season, when RSV infection is likely to both represent the first and most severe infection. To meet recruitment milestones and assure sufficient spectrum of disease severity during the 2 recruitment seasons, infants were also enrolled during acute illnesses. The protocol and informed consent documents were approved by the Institutional Review Board at Vanderbilt University Medical Center. One parent of each participant provided written informed consent for participation in this study. The informed consent document explained study procedures, use of data of and biospecimens for future studies, including a rider for genetic studies.

Table 1 Inclusion and Exclusion Criteria for Enrollment in INSPIRE Study

Study visits and procedures

Enrollment visit

At the baseline visit, trained study staff administered a baseline questionnaire. The components of the study visits are given in Table 2. The questionnaire included demographic characteristics, infant medical history, family history, secondhand smoke exposure, daycare attendance, growth trajectory and other factors known to be associated with childhood asthma and allergy development. Multiple bio-specimens were collected for different purposes at each time point and are described in Table 3. A urine sample was collected at enrollment, through use of an external bag, for markers of inflammation and lung injury. In year 2, infant stool samples and nasal samples were also collected at enrollment. The infant nasal samples at enrollment were collected by insertion of filter paper (Leukosorb™; Pall Corporation, Port Washington, NY) into each nostril, as described previously [23,24]. A maternal finger stick was also collected in year 2 to bank maternal blood samples for other assays, such as vitamin E isoforms.

Table 2 INSPIRE Main Study Schedule
Table 3 Specimen Repository for INSPIRE Study

Respiratory illness surveillance

Respiratory illness surveillance was conducted during the winter virus season (November to March) of each infant’s first year of life in order to capture and characterize infant acute respiratory illnesses during RSV season. Respiratory illness surveillance had three components: first, parents were instructed to call the research staff between November and March, at first onset of respiratory symptoms which included any one of the following: runny nose, congestion, wheezing, fever and/or cough at which time the screening respiratory illness survey was completed. Second, all enrolled infants who were seen for an acute visit at their pediatric practice were approached and administered the respiratory illness survey. Third, every two weeks, from November through March, parents received regular contact by an e-mail secure survey or telephone call to complete the respiratory illness survey. The respiratory illness survey is a series of questions about respiratory symptoms and time of onset.

Acute respiratory illness visits

Infants met our case definition of a respiratory illness visit based on responses to the respiratory illness surveillance questionnaires: a parent indicates ONE of the following major symptoms or diagnoses: wheezing, difficulty breathing, or told that your baby had a positive RSV test OR ANY TWO of the following minor symptoms or diagnoses: fever, runny nose/congestion/snotty nose, cough, ear infection (otitis media), or hoarse cry. Onset of symptoms must have been in the prior week (the past 5 days for the infants enrolled in year 1 of the study, and the prior 7 days for infants enrolled in year 2 of the study). This change in symptom/diagnosis window was made to accommodate missing respiratory visits because of no staffing on weekends.

If the infant met pre-specified criteria for a respiratory illness visit, an in-person visit was performed either in the clinic, the pediatric clinical research center, or the infant’s home. The visit included a brief physical exam with elements that measure the severity of respiratory infection: respiratory rate, oxygen saturation/heart rate, wheezing, and chest retractions. Chart review was used to supplement the research exam with information about doctor diagnosis, when a healthcare visit occurred. The respiratory symptom assessment detailed the symptoms present and the date of onset. In addition, a nasal wash and urine sample were collected at each respiratory illness visit. A nasal wash was conducted using 5 mL of sterile saline solution and immediately placed at 4 degrees Celsius. The nasal washes were aliquoted and snap frozen at -80 degrees within 24 hours of collection.

1-year study visit

The one year follow-up visit was conducted between 9 – 15 months of age and included questions about respiratory health and exposures, and a venipuncture for RSV serology. If the caregiver refused venipuncture or blood was not able to be drawn, research staff collected 250 μL of blood from a toe or finger stick. If consent was given for genetic studies, a buffy coat sample was banked for future DNA extraction. Participants were also asked to collect household dust samples by wiping the top of the doorframe of the room where the infant sleeps with 2 wipes (TefTex™, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada) and returning by mail [25]. These samples are to be used for endotoxin/lipopolysaccharide measurements [25,26].

Annual visits

Annual follow-up study contact is ongoing and will take place at year 2, 3 and 4. Year 2 and 4 follow-up is by telephone survey and includes the International Study of Asthma and Allergy in Children (ISAAC) [27] and other allergic disease and respiratory disease questionnaires. The third year visit is in-person and includes the above components, as well as skin prick testing to aeroallergens common to the region, or if skin testing cannot be performed, a blood draw for measurement of specific IgE to the same aeroallergens.

Study endpoints

The co-primary study endpoints are both recurrent wheezing and early childhood asthma. Recurrent wheezing is defined between ages 1-3 years, as two or more wheezing episodes at any time during that period. Early childhood asthma will be determined at age four using elements of the ISAAC questionnaire. Children who meet both of the following criteria at age four will be classified as definite early childhood asthmatics: (1) 12-month prevalence of asthma symptoms or exercise-induced wheeze or dry cough at night, AND (2) physician diagnosis by parental report or use/prescription of asthma medications [27-36]. Children who meet just one of the two criteria will be considered to have probable early childhood asthma.

Secondary endpoints

Respiratory illness severity at time of illness will be measured by an ordinal scale based on wheezing, respiratory rate, retractions, and oxygen saturation or heart rate, modified from scores derived for LRI [37]. Also, a dichotomous outcome, LRI versus URI will be used to measure severity. Allergic rhinitis (AR) diagnosis will also be made using elements of the ISAAC questionnaire. Children will be considered to have definite allergic rhinitis (AR) if each of three conditions are present at age four: (1) a history of nasal congestion, runny nose, itchy watery eyes, sneezing, or blocked nose; AND (2) substantial variability in symptoms over time or seasonality; AND (3) diagnosed as having AR by a physician or on medications for AR. Patients will be considered to have probable AR if they report symptoms as outlined above in (1) and (2) but have not been diagnosed or treated by a physician. Atopic dermatitis diagnosis will be confirmed through the ISAAC core questions on atopic dermatitis, which are based on a list of major and minor criteria proposed by Hanifin and Rajka in the 1970s [32,38,39]. Patients will be classified as definite atopic dermatitis if they report ever having an itchy rash that comes and goes for at least six months, AND being diagnosed with eczema by physician documentation. They will be classified as probable atopic dermatitis if they report one of the two above criteria. Atopy will be assessed by either a positive skin prick allergen test for aeroallergens (preferred) or specific IgE from blood test at age 3 years: 1) Positive skin prick testing is defined by a wheal (center raised part) of 3 mm or greater to one of the aeroallergens (when histamine control is positive and the saline control is negative [non-reactive], indicating that the patient is not dermatographic [i.e. reacting to irritation and not to specific allergens]), or 2) laboratory atopy will be defined as specific IgE > 0.1 PAU/L to at least one common aeroallergen performed on blood specimens collected at 3-years. Additional secondary endpoints will be defined by breaking down co-primary recurrent wheezing outcome into well-accepted asthma phenotypes [40], including: 1) transient early wheezing will be defined as wheezing episodes during the first three years of life [41]. 2) persistent wheezing will be defined as wheezing during the first three years and beyond. 3) Late onset wheezing will be defined as wheezing not present in the first three years, but with symptoms beginning between three and four years.

Primary exposure

Singleplex reverse-transcription real-time PCR assays for RSV, human rhinovirus, human enterovirus, and human ribonuclease P (RNAseP) in nasal washes were performed according to published protocols [42,43]. RNAseP served as an endogenous control for specimen quality, and specimens failing to demonstrate an RNAseP signal among multiple individually processed aliquots were deemed indeterminate for viruses not detected in repetitive testing. Efficiency of nucleic acid extraction was monitored through amplification of RNAseP in cultured human-cell lysates incorporated into each round of specimen processing. Results of amplification reactions were validated by expected findings for water blanks, negative controls, and positive controls derived from viral culture lysates (adjusted to concentrations representative of patient specimens) included in each PCR reaction plate. RSV IgG antibodies will be assessed at one year by four enzyme immunoassays (EIA). One EIA using RSV A2 (group A strain) lysate as the antigen, one using RSV B1 (group B strain) lysate, one using an RSV group A G protein peptide (RSV A2 aa 161-190 : NDFHFEVFNF VPCSICSNNP TCWAICKRIP), and one using an RSV group B G protein peptide (RSV B1 aa 161-190 : DDYHFEVFNF VPCSICGNNQ LCKSICKTIP). RSV serology at year one, coupled with viral identification of RSV by PCR at time of respiratory illness, will be used to determine whether infants were exposed to RSV.

Biomarkers analyzed during acute respiratory illness

Additional biomarkers analyzed during acute respiratory illness will also serve as primary endpoints for analyses. Lung injury and host immune response will be measured in urine samples collected during the acute respiratory illness and include cysteinyl leukotrienes, CC-16, KL-6, IL-18, TNF-α, HNE, prostacyclin, and isoprostanes. Immune function biomarkers assayed from nasal washes collected during acute respiratory illness include: VEGF, G-CSF, EGF, FGF, eotaxin, eotaxin-2, eotaxin-3, MCP-2, BCA-1, MCP-4, I-309, IL-16, TARC, X6CKine, LIF, TPO, SCF, TSLP, IL-33, IL-20, IL-21, IL-23, TRAIL, CTACK, SDF-1a-b, ENA-78, MIP-1d, IL-28A, IL-1B, IL-10, IL-13, IL-6, IL-12, RANTES, IL-17, MIP-1a, GM-CSF, MIP-1B, MCP-1, IL-15, IL-5, IFN-y, IFN-a, IL-1RA, TNF-a, IL-2, IL-7, IP-10, IL-2R, MIG, IL-4, IL-8, HGF and Muc5ac.

Planned statistical analyses

Descriptive analysis of infant demographic, clinical and biological measurements will be performed using means and standard deviations or medians and inter-quartile ranges for continuous variables, and proportions for categorical variables.

Cohort study

Our first hypothesis examines whether the severity of RSV infection is associated with risk of early childhood wheezing or early childhood asthma. RSV exposure is determined by confirmed RSV illness by PCR, and/or positive 1-year RSV serology. The co-primary endpoints will be recurrent wheezing and early childhood asthma as described previously. Logistic regression will be used to estimate the odds ratio (OR) and 95% confidence intervals [44] of developing childhood asthma or recurrent wheezing in children with severe or mild RSV infection relative to those who were not infected in infancy. Both crude ORs and ORs adjusted for confounding variables will be derived. Confounding variables for our multiple logistic regression models will include maternal age, infant age at first RSV infection, gestational age, secondhand smoke (SHS), infant sex, and family history of allergic diseases. Other confounding variables to be considered include further detail on home environment, including infant diet, use of acetaminophen or NSAIDs, study year, gender, and family history of asthma. A priori interaction terms will be included in our models and we will examine their effect on the change of model deviance. Additional analyses will be conducted to assess model fit. These will include scatter plots of squared standardized Pearson residuals against the estimated asthma probability for patients with each covariate pattern. Analyses will be conducted using SAS (SAS Institute Cary, NC) and R (http://www.r-project.org).

Nested case-control study

To test our hypothesis that measures of host immune response and lung injury will differ between infants with RSV illness who do and do not develop recurrent wheezing or early childhood asthma, we will conduct nested case-control studies of incident early childhood asthma cases following infant RSV infection to matched RSV-infected children who do not develop recurrent wheezing or asthma. Two controls will be selected for each case as follows: First, we will fit a logistic regression model in which asthma outcome is regressed against these matching covariates. The coefficients from this model will be used as weights to create an asthma risk score for each subject, which is the weighted sum of these risk covariates. Controls will then be frequency matched to cases based on their asthma risk score. This risk score will be derived from one of our prior analyses of an administrative data set that used a regression analysis [45].

Power calculations

Primary Hypothesis 1: Power calculations were performed before the study began. The primary outcome for this study is recurrent wheezing illness at age 3-4, rather than asthma, which requires more time for follow-up than allowed during the grant funding period. Our power calculations were based on a birth cohort of 1500 infants plus a total of 400 infants recruited at the time of RSV infection in infancy for a total of 1900 children. Two thirds of these 400 were assumed to have severe RSV disease necessitating hospitalization while one-third to have mild disease treated on an outpatient basis. We anticipated that 75% of the subjects will be successfully followed up to measure asthma outcomes at 4-6 years of age. We have powered our primary hypothesis with RSV exposure levels of 20% of infants with LRI, 40% URI and 40% with no RSV infection in their first year of life. We further assumed a conservative estimate of 11% infants who were not exposed to RSV to develop asthma at ages 4-6 years. We performed power calculations based on a 2-sided test of the null hypothesis that the asthma prevalence was not affected by infant RSV exposure, with a Type I error probability of α = 0.05. In Figure 2, the red curve shows the power for different asthma odds ratios for infants with RSV LRI relative to those with no infection. We expect that the prevalence of asthma among infants with URI (mild) will be 7% and the dashed blue curve in Figure 2 shows the asthma odds ratios for infants with no exposure vs. mild infection. The prevalence of recurrent wheezing is expected to be much higher than the asthma prevalence shown, thus we expect to have ample power for our analysis. We anticipate that 30% of the entire cohort will have recurrent wheezing at three years of age. This will provide us with considerably more power to detect different odds ratios for wheezing than we will attain for asthma, as shown in Figure 2. These power calculations show we will have at least 80% power to detect recurrent wheezing relative risks greater than 1.65 or less than 0.61 in subgroups of infants defined by RSV exposure and assuming 75% follow up.

Figure 2
figure 2

Power for primary hypothesis 1 to assess the association between RSV exposure and asthma based on 1900 infants with 25% attrition (α = 0.05).

Primary Hypothesis 2, Nested Case-Control Study: Preliminary data from previous studies provide estimates of selected biomarkers, including leukotrienes, CC-16, KL-6, IL-18, TNF-α, and HNE. Estimates of minimal detectable differences (two sided α = 0.05) are given in Table 4, for different numbers of asthma cases identified [46-50].

Table 4 Nested case-control study, minimal detectable differences of biomarkers under different hypothetical assumptions with 1:2 case-control matching

Recruitment results

Screening and enrollment

An overview of the recruitment is presented in Figure 3. 1952 infants were enrolled: 861 infants in the first year and 1091 in the second year. We approached 83% of eligible infants (3559/4381) for enrollment with 55% agreed to participate (1952/3559). The most common reason participants provided for not enrolling was that they were “not interested” (63%; n = 685/1085). Five hundred and twenty-two parents received brochures and study information, but decided not to enroll at the time of visit. In this situation, we were not able to establish contact again, despite a continued presence at their pediatrician’s office and do not know the reason for non-enrollment. At the time of this publication, 82 participants have withdrawn from the study, most commonly due to an unexpected move out of Middle Tennessee (N = 21). The second most common reason was discomfort with the 1-year blood draw (N = 18). For those willing to participate after moving, a blood sample will be collected, processed and shipped, and the annual case report form will be administered by telephone.

Figure 3
figure 3

Flowchart of INSPIRE Study, through December 15, 2014.

Demographic characteristics of the sample are shown in Table 5. Approximately 93% percent of our participants were enrolled from 8 pediatric practices, where staff was located on-site throughout the study. The remaining visits were performed at the Vanderbilt Pediatric Clinical Research Center (PCRC). Eight percent (164/1952) of infants had an acute respiratory infection at the time of enrollment. Among those age-eligible for the 1-year visit, 85% have been completed as of December 15, 2014.

Table 5 Demographic Characteristics of Infants in the INSPIRE Study

Acute respiratory illness surveillance

Over the two respiratory seasons between November through March, 14,285 respiratory surveillance contacts were completed through e-mail, telephone or in-person on 1,943 individuals (9 individuals never responded). The median time between contacts was 14 days with an interquartile range (IQR) of 12.6-15.6 days. The median number of responses per individual was 8 (IQR: 6-10.5). 5,200 survey respondents reported symptoms within the past 5 days; however, only 2,516 met criteria for an acute respiratory illness visit by either 1 major symptom or 2 minor symptoms.

Respiratory illness visits

2,103 respiratory illness visits were completed on 1,189 infants, with 2,036 nasal washes collected and stored. Of these 61% of infants had one or more respiratory illness visits. 682 infants had 1 visit, 314 had 2 visits, 149 had 3 visits and 78 had 4 or more visits. Urine was collected during 1652 (79%) respiratory illness visits. 385 (19%) infant respiratory illness visits were RSV positive based on PCR of nasal washes. Blood was banked for RSV serology testing on 1-year infant samples.

Longitudinal follow-up

At the time of this publication, 2-year telephone calls are ongoing for those recruited in the first year of enrollment and 1-year visits are ongoing for infants enrolled in the second year. We have completed approximately 95% of the 1-year follow-up visits for first year of enrollment, after excluding withdrawals (760/805).

Discussion

Currently, INSPIRE is the largest population based prospective epidemiologic study where the spectrum of severity of RSV infection illnesses are assessed in relationship to recurrent wheezing illnesses and asthma onset. Asthma is a multifactorial disease with onset, course and severity reflecting the combined effects of development-specific exposures in utero and post-natally, and the host responses to those exposures. RSV is one of these ubiquitous exposures, which as an infectious agent likely works through differentially modifying the developing immune system and lung, based on specific strains, severity of infection, and host response to infection. The first primary study hypothesis uses the entire cohort to determine the relationship between the spectrum of RSV exposure to severe infection on risk of wheezing illness in childhood. While severe LRI has been linked to development of wheezing or asthma [1-5,51], little is known about the role of mild infection.

The INSPIRE study was designed to address gaps in our knowledge about the role of the host immune response and lung injury to RSV infection in asthma inception, and to understand how viral strain differences contribute to infant morbidity and asthma risk. This will be done through the second study hypothesis that utilizes a nested case-control design within the total cohort. This design will identify host immune and lung injury markers that are differentially associated with outcomes of wheezing illnesses at ages 3-4, and eventually childhood asthma at age 6. Our hypothesis is based on evidence that host responses are associated with increased risk of severe RSV LRI. Among infants with severe RSV infection, TNF-α and eosinophil active cytokines are elevated compared to non-ill infants [52] or mildly ill infants [53,54] and correlate with the severity of illness [55]. After RSV LRI, cysteinyl leukotrienes and IL-17 remain elevated in nasal epithelium [53,56], and IL10 production and genetic variants IL-13, IL-19 and IL-20 are associated with post-bronchial wheeze [57-60]. RSV infection may also cause harmful remodeling of infant lungs [40]. Infant lungs are not fully developed at birth; changes in airway epithelium and alveolarization continue through the first two years of life [61]. Animal studies have shown increased airway hyper-responsiveness and decreased neuronal relaxation after experimental RSV infection [62]. The INSPIRE study will bridge what is known about the immune response at time of infection and extend to 3-4 year outcomes of wheezing illnesses. RSV strains from infants in INSPIRE will be sequenced to determine if strain differences are associated with acute infection morbidity in infancy and later wheezing outcomes.

Prior studies of infant RSV infection have focused almost exclusively on the 3-5% of infants hospitalized with RSV LRI. Most infants, however, have milder RSV infection, and the majority of those who do develop RSV LRI do not develop asthma. While severe LRI has been clearly linked to the development of asthma, an important unanswered question is whether mild RSV infection confers intermediate risk or may actually protect against the inception of childhood asthma. If mild RSV infection primes the immune system or confers a protective effect against future wheezing illnesses, this has important implications for the development of RSV vaccines and RSV immunoprophylaxis. Thus, there are compelling reasons to focus on the development of new approaches to understanding why certain infants are predisposed to developing viral LRI, and which children with RSV LRI develop subsequent childhood asthma.

Strengths of our study include biweekly surveillance to capture the full spectrum of respiratory infections, including even mild infections by regular surveillance intervals, and serologic confirmation of infant infection to identify those who are infected and those who are unexposed. The location of research staff at pediatric clinics allowed frequent contact with participants throughout winter virus season. We also have a large repository of banked urine samples during enrollment and acute illness, 1-year plasma samples, DNA, stool samples, acute illness nasal washes, and baseline enrollment nasal filter paper samples.

This study has several limitations that are important to consider. First, the study sample was not selected from the general population, but instead was recruited from participating pediatric clinics in the city of Nashville and surrounding counties; however, we attempted enrollment of all births at these clinics meeting criteria. Our study cohort demographics represent a varied population demographic that reflects the diversity of the community from which the cohort is recruited. Surveillance for respiratory illnesses occurred during a full 5-month period. However, surveillance was by biweekly surveys or self-reporting, and illnesses may have been missed or not reported. In addition, the respiratory illness visits may have occurred up to 7 days after onset of symptoms, thus characterization of the illness severity at the visit may not reflect the time when the illness was most severe. Although we decided to include an enrichment sample of infants with acute respiratory infections at the emergency room or hospital admission, this was not a major source of enrollment because many of these participants were not able to fulfill the longitudinal study requirements primarily due to large distances to our study site.

This study was funded by a National Institute of Allergy and Infectious Diseases (NIAID) Asthma and Allergic Diseases Cooperative Research Center (AADCRC) grant that also supports three related projects that will investigate key disease mechanisms, and utilize samples from this cohort. (1) The first project aims at identifying the molecular basis for RSV strain differences associated with mild and severe RSV respiratory infection. To do this, collaborators at Emory University School of Medicine (MLM, LJA) and J. Craig Venter Institute (SRD) will genotype RSV isolates of the INSPIRE cohort in order to identify RSV genotypes associated with bronchiolitis severity, childhood wheeze, and the development of asthma. (2) A second project will study the role of PGI2 in a mouse model of respiratory syncytial virus (RSV) infection (RSP). The ultimate goal is to determine if targeted therapies of the host immune response could prevent or treat severe RSV bronchiolitis and the subsequent development of asthma. (3) A third project is the longitudinal characterization of the infant and early childhood microbiome during health and acute respiratory illness, and the association of microbial diversity with both infant acute respiratory illness severity and childhood wheezing outcomes (TVH, CRS, and SRD).

In summary, this ongoing study will answer important questions about the mechanisms through which RSV causes acute and long term respiratory morbidity. This methodology paper outlines the study questions addressed, how the birth cohort (INSPIRE) was designed to address these questions, and provides preliminary enrollment data. Our unique study design targets infants born from June to December, such that they are on average 6 months of age during RSV circulation. With well-characterized respiratory infections during infancy, we are poised to address important questions about the causal role of RSV infection in the development of childhood wheezing and early childhood asthma. In addition to a carefully phenotyped cohort, we have a rich biospecimen repository. Our future plans include following this cohort to childhood asthma outcomes at age 6, characterizing the host responses to infection which are associated with protection from or risk of asthma, and understanding other associated risk factors including host genetics.

Abbreviations

ARI:

Acute Respiratory Infection

INSPIRE:

Infant Susceptibility to Pulmonary Infections and Asthma Following RSV Exposure

LRI:

Lower Respiratory Tract Infection

OR:

Odds Ratio

URI:

Upper Respiratory Tract Infection

PCRC:

Pediatric Clinical Research Center

PBMC:

Peripheral Blood Mononuclear Cells

RSV:

Respiratory Syncytial Virus

References

  1. Stein RT, Sherrill D, Morgan WJ, Holberg CJ, Halonen M, Taussig LM, et al. Respiratory syncytial virus in early life and risk of wheeze and allergy by age 13 years. Lancet. 1999;354:541–5.

    Article  CAS  PubMed  Google Scholar 

  2. Weber MW, Milligan P, Giadom B, Pate MA, Kwara A, Sadiq AD, et al. Respiratory illness after severe respiratory syncytial virus disease in infancy in The Gambia. J Pediatr. 1999;135:683–8.

    Article  CAS  PubMed  Google Scholar 

  3. 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;161:1501–7.

    Article  CAS  PubMed  Google Scholar 

  4. Henderson J, Hilliard TN, Sherriff A, Stalker D, Al Shammari N, Thomas HM. Hospitalization for RSV bronchiolitis before 12 months of age and subsequent asthma, atopy and wheeze: a longitudinal birth cohort study. Pediatr Allergy Immunol. 2005;16:386–92.

    Article  PubMed  Google Scholar 

  5. Stensballe LG, Kristensen K, Simoes EA, Jensen H, Nielsen J, Benn CS, et al. Atopic disposition, wheezing, and subsequent respiratory syncytial virus hospitalization in Danish children younger than 18 months: a nested case-control study. Pediatrics. 2006;118:e1360–8.

    Article  PubMed  Google Scholar 

  6. Peebles Jr RS, Sheller JR, Collins RD, Jarzecka AK, Mitchell DB, Parker RA, et al. Respiratory syncytial virus infection does not increase allergen-induced type 2 cytokine production, yet increases airway hyperresponsiveness in mice. J Med Virol. 2001;63:178–88.

    Article  CAS  PubMed  Google Scholar 

  7. Smit JJ, Lindell DM, Boon L, Kool M, Lambrecht BN, Lukacs NW. The balance between plasmacytoid DC versus conventional DC determines pulmonary immunity to virus infections. PLoS One. 2008;3, e1720.

    Article  PubMed Central  PubMed  Google Scholar 

  8. Bartz H, Buning-Pfaue F, Turkel O, Schauer U. Respiratory syncytial virus induces prostaglandin E2, IL-10 and IL-11 generation in antigen presenting cells. Clin Exp Immunol. 2002;129:438–45.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Smit JJ, Boon L, Lukacs NW. Respiratory virus-induced regulation of asthma-like responses in mice depends upon CD8 T cells and interferon-gamma production. Am J Pathol. 2007;171:1944–51.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  10. Smit JJ, Rudd BD, Lukacs NW. Plasmacytoid dendritic cells inhibit pulmonary immunopathology and promote clearance of respiratory syncytial virus. J Exp Med. 2006;203:1153–9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  11. Gill MA, Palucka AK, Barton T, Ghaffar F, Jafri H, Banchereau J, et al. Mobilization of plasmacytoid and myeloid dendritic cells to mucosal sites in children with respiratory syncytial virus and other viral respiratory infections. J Infect Dis. 2005;191:1105–15.

    Article  PubMed  Google Scholar 

  12. Illi S, Von ME, Lau S, Bergmann R, Niggemann B, Sommerfeld C, et al. Early childhood infectious diseases and the development of asthma up to school age: a birth cohort study. BMJ. 2001;322:390–5.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Von ME, Illi S, Hirsch T, Leupold W, Keil U, Weiland SK. Frequency of infections and risk of asthma, atopy and airway hyperresponsiveness in children. Eur Respir J. 1999;14:4–11.

    Article  Google Scholar 

  14. Wu P, Hartert TV. Evidence for a causal relationship between respiratory syncytial virus infection and asthma. Expert Rev Anti Infect Ther. 2011;9:731–45.

    Article  PubMed Central  PubMed  Google Scholar 

  15. Stensballe LG, Simonsen JB, Thomsen SF, Larsen AM, Lysdal SH, Aaby P, et al. The causal direction in the association between respiratory syncytial virus hospitalization and asthma. J Allergy Clin Immunol. 2009;123:131–7.

    Article  PubMed  Google Scholar 

  16. Wu P, Dupont WD, Griffin MR, Carroll KN, Mitchel EF, Gebretsadik T, et al. Evidence of a causal role of winter virus infection during infancy on early childhood asthma. Am J Respir Crit Care Med. 2008;178:1123–9.

    Article  PubMed Central  PubMed  Google Scholar 

  17. Krishnamoorthy N, Khare A, Oriss TB, Raundhal M, Morse C, Yarlagadda M, et al. Early infection with respiratory syncytial virus impairs regulatory T cell function and increases susceptibility to allergic asthma. Nat Med. 2012;18(10):1525–30.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Walsh EE, McConnochie KM, Long CE, Hall CB. Severity of respiratory syncytial virus infection is related to virus strain. J Infect Dis. 1997;175:814–20.

    Article  CAS  PubMed  Google Scholar 

  19. Tran DN, Pham TM, Ha MT, Tran TT, Dang TK, Yoshida LM, et al. Molecular epidemiology and disease severity of human respiratory syncytial virus in Vietnam. PLoS One. 2013;8, e45436.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Martinello RA, Chen MD, Weibel C, Kahn JS. Correlation between respiratory syncytial virus genotype and severity of illness. J Infect Dis. 2002;186:839–42.

    Article  PubMed  Google Scholar 

  21. Sigurs N, Aljassim F, Kjellman B, Robinson PD, Sigurbergsson F, Bjarnason R, et al. Asthma and allergy patterns over 18 years after severe RSV bronchiolitis in the first year of life. Thorax. 2010;65:1045–52.

    Article  PubMed  Google Scholar 

  22. Sigurs N, Gustafsson PM, Bjarnason R, Lundberg F, Schmidt S, Sigurbergsson F, et al. Severe respiratory syncytial virus bronchiolitis in infancy and asthma and allergy at age 13. Am J Respir Crit Care Med. 2005;171:137–41.

    Article  PubMed  Google Scholar 

  23. Chawes BL, Edwards MJ, Shamji B, Walker C, Nicholson GC, Tan AJ, et al. A novel method for assessing unchallenged levels of mediators in nasal epithelial lining fluid. J Allergy Clin Immunol. 2010;125:1387–9.

    Article  CAS  PubMed  Google Scholar 

  24. Folsgaard NV, Chawes BL, Rasmussen MA, Bischoff AL, Carson CG, Stokholm J, et al. Neonatal cytokine profile in the airway mucosal lining fluid is skewed by maternal atopy. Am J Respir Crit Care Med. 2012;185:275–80.

    Article  PubMed  Google Scholar 

  25. Konya T, Ewaze JO, Urch B, Shu H, Curran J, Speck M, et al. Development and validation of the TefTex electrostatic wipe for indoor dust sampling. Healthy Buildings. 2012;4H9:1–5.

    Google Scholar 

  26. Thorne PS, Metwali N, Avol E, McConnell RS. Surface sampling for endotoxin assessment using electrostatic wiping cloths. Ann Occup Hyg. 2005;49:401–6.

    Article  CAS  PubMed  Google Scholar 

  27. Asher I. ISAAC International Study of Asthma and Allergies in Childhood. Pediatr Pulmonol. 2007;42:100.

    Article  PubMed  Google Scholar 

  28. Bauer BA, Reed CE, Yunginger JW, Wollan PC, Silverstein MD. Incidence and outcomes of asthma in the elderly. A population-based study in Rochester, Minnesota. Chest. 1997;111:303–10.

    Article  CAS  PubMed  Google Scholar 

  29. Beard CM, Yunginger JW, Reed CE, O’Connell EJ, Silverstein MD. Interobserver variability in medical record review: an epidemiological study of asthma. J Clin Epidemiol. 1992;45:1013–20.

    Article  CAS  PubMed  Google Scholar 

  30. Silverstein MD, Reed CE, O’Connell EJ, Melton III LJ, O’Fallon WM, Yunginger JW. Long-term survival of a cohort of community residents with asthma. N Engl J Med. 1994;331:1537–41.

    Article  CAS  PubMed  Google Scholar 

  31. Hartert TV, Togias A, Mellen BG, Mitchel EF, Snowden MS, Griffin MR. Underutilization of controller and rescue medications among older adults with asthma requiring hospital care. J Am Geriatr Soc. 2000;48:651–7.

    Article  CAS  PubMed  Google Scholar 

  32. Asher MI, Keil U, Anderson HR, Beasley R, Crane J, Martinez F, et al. International Study of Asthma and Allergies in Childhood (ISAAC): rationale and methods. Eur Respir J. 1995;8:483–91.

    Article  CAS  PubMed  Google Scholar 

  33. Pearce N, Weiland S, Keil U, Langridge P, Anderson HR, Strachan D, et al. Self-reported prevalence of asthma symptoms in children in Australia, England, Germany and New Zealand: an international comparison using the ISAAC protocol. Eur Respir J. 1993;6:1455–61.

    CAS  PubMed  Google Scholar 

  34. Worldwide variation in prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and atopic eczema: ISAAC. The International Study of Asthma and Allergies in Childhood (ISAAC) Steering Committee. Lancet. 1998;351:1225–32.

    Article  Google Scholar 

  35. ISAAC. ISAAC Manual (2nd Ed). Auckland/Münster. 1993

  36. Ellwood P, Asher MI, Beasley R, Clayton TO, Stewart AW, on behalf of the ISAAC Steering Committee and the ISAAC Phase Three Study Group. ISAAC Phase Three Manual. Auckland. July 2000. ISBN 0-473-06910-5.

  37. McCallum GB, Morris PS, Wilson CC, Versteegh LA, Ward LM, Chatfield MD, et al. Severity scoring systems: are they internally valid, reliable and predictive of oxygen use in children with acute bronchiolitis? Pediatr Pulmonol. 2013;48:797–803.

    Article  PubMed  Google Scholar 

  38. Hanifin JM, Rajka G. Diagnostic features of atopic dermatitis. Acta Derm Venereol. 1980;92:44–7.

    Google Scholar 

  39. Diepgen TL, Fartasch M, Hornstein OP. Evaluation and relevance of atopic basic and minor features in patients with atopic dermatitis and in the general population. Acta Derm Venereol Suppl (Stockh). 1989;144:50–4.

    CAS  Google Scholar 

  40. Martinez FD, Wright AL, Taussig LM, Holberg CJ, Halonen M, Morgan WJ. Asthma and wheezing in the first six years of life. The Group Health Medical Associates. N Engl J Med. 1995;332:133–8.

    Article  CAS  PubMed  Google Scholar 

  41. Morgan WJ, Stern DA, Sherrill DL, Guerra S, Holberg CJ, Guilbert TW, et al. Outcome of asthma and wheezing in the first 6 years of life: follow-up through adolescence. Am J Respir Crit Care Med. 2005;172:1253–8.

    Article  PubMed Central  PubMed  Google Scholar 

  42. Kodani M, Yang G, Conklin LM, Travis TC, Whitney CG, Anderson LJ, et al. Application of TaqMan low-density arrays for simultaneous detection of multiple respiratory pathogens. J Clin Microbiol. 2011;49:2175–82.

    Article  PubMed Central  PubMed  Google Scholar 

  43. Emery SL, Erdman DD, Bowen MD, Newton BR, Winchell JM, Meyer RF, et al. Real-time reverse transcription-polymerase chain reaction assay for SARS-associated coronavirus. Emerg Infect Dis. 2004;10:311–6.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  44. Dupont W. Statistical modeling for biomedical researchers. 1st ed. Cambridge: Cambridge University Press; 2002.

    Google Scholar 

  45. Carroll KN, Wu P, Gebretsadik T, Griffin MR, Dupont WD, Mitchel EF, et al. The Severity-Dependent Relationship of Infant Bronchiolitis on the Risk and Morbidity of Early Childhood Asthma. J Allergy Clin Immunol. 2009;123:1055–61.

    Article  PubMed Central  PubMed  Google Scholar 

  46. Kawasaki Y, Aoyagi Y, Abe Y, Go H, Imamura T, Kaneko M, et al. Serum KL-6 levels as a biomarker of lung injury in respiratory syncytial virus bronchiolitis. J Med Virol. 2009;81:2104–8.

    Article  CAS  PubMed  Google Scholar 

  47. Imai T, Takase M, Takeda S, Kougo T. Serum KL-6 levels in pediatric patients: reference values for children and levels in pneumonia, asthma, and measles patients. Pediatr Pulmonol. 2002;33:135–41.

    Article  PubMed  Google Scholar 

  48. Asano K, Lilly CM, O’Donnell WJ, Israel E, Fischer A, Ransil BJ, et al. Diurnal variation of urinary leukotriene E4 and histamine excretion rates in normal subjects and patients with mild-to-moderate asthma. J Allergy Clin Immunol. 1995;96:643–51.

    Article  CAS  PubMed  Google Scholar 

  49. Sadeghi M, Daniel V, Naujokat C, Wiesel M, Hergesell O, Opelz G. Strong inflammatory cytokine response in male and strong anti-inflammatory response in female kidney transplant recipients with urinary tract infection. Transpl Int. 2005;18:177–85.

    Article  CAS  PubMed  Google Scholar 

  50. Smith CJ, Fischer TH, Heavner DL, Rumple MA, Bowman DL, Brown BG, et al. Urinary thromboxane, prostacyclin, cortisol, and 8-hydroxy-2′-deoxyguanosine in nonsmokers exposed and not exposed to environmental tobacco smoke. Toxicol Sci. 2001;59:316–23.

    Article  CAS  PubMed  Google Scholar 

  51. Blanken MO, Rovers MM, Molenaar JM, Winkler-Seinstra PL, Meijer A, Kimpen JL, et al. Respiratory syncytial virus and recurrent wheeze in healthy preterm infants. N Engl J Med. 2013;368:1791–9.

    Article  CAS  PubMed  Google Scholar 

  52. Choi J, Callaway Z, Kim HB, Fujisawa T, Kim CK. The role of TNF-alpha in eosinophilic inflammation associated with RSV bronchiolitis. Pediatr Allergy Immunol. 2010;21:474–9.

    Article  PubMed  Google Scholar 

  53. Faber TE, Groen H, Welfing M, Jansen KJ, Bont LJ. Specific increase in local IL-17 production during recovery from primary RSV bronchiolitis. J Med Virol. 2012;84:1084–8.

    Article  CAS  PubMed  Google Scholar 

  54. Faber TE, Schuurhof A, Vonk A, Koppelman GH, Hennus MP, Kimpen JL, et al. IL1RL1 gene variants and nasopharyngeal IL1RL-a levels are associated with severe RSV bronchiolitis: a multicenter cohort study. PLoS One. 2012;7, e34364.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  55. Vieira RA, Diniz EM, Ceccon ME. Correlation between inflammatory mediators in the nasopharyngeal secretion and in the serum of children with lower respiratory tract infection caused by respiratory syncytial virus and disease severity. J Bras Pneumol. 2010;36:59–66.

    Article  PubMed  Google Scholar 

  56. Da DL, Callegaro S, Carraro S, Andreola B, Corradi M, Baraldi E. Nasal lavage leukotrienes in infants with RSV bronchiolitis. Pediatr Allergy Immunol. 2007;18:100–4.

    Article  Google Scholar 

  57. Schuurhof A, Janssen R, De GH, Hodemaekers HM, De KA, Kimpen JL, et al. Local interleukin-10 production during respiratory syncytial virus bronchiolitis is associated with post-bronchiolitis wheeze. Respir Res. 2011;12:121.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. Bont L, Heijnen CJ, Kavelaars A, van Aalderen WM, Brus F, Draaisma JT, et al. Monocyte IL-10 production during respiratory syncytial virus bronchiolitis is associated with recurrent wheezing in a one-year follow-up study. Am J Respir Crit Care Med. 2000;161:1518–23.

    Article  CAS  PubMed  Google Scholar 

  59. Ermers MJ, Hoebee B, Hodemaekers HM, Kimman TG, Kimpen JL, Bont L. IL-13 genetic polymorphism identifies children with late wheezing after respiratory syncytial virus infection. J Allergy Clin Immunol. 2007;119(5):1086–91.

    Article  CAS  PubMed  Google Scholar 

  60. Ermers MJ, Janssen R, Onland-Moret NC, Hodemaekers HM, Rovers MM, Houben ML, et al. IL10 family member genes IL19 and IL20 are associated with recurrent wheeze after respiratory syncytial virus bronchiolitis. Pediatr Res. 2011;70:518–23.

    Article  CAS  PubMed  Google Scholar 

  61. Holt PG, Upham JW, Sly PD. Contemporaneous maturation of immunologic and respiratory functions during early childhood: implications for development of asthma prevention strategies. J Allergy Clin Immunol. 2005;116:16–24.

    Article  PubMed  Google Scholar 

  62. Larsen GL, Colasurdo GN. Neural control mechanisms within airways: disruption by respiratory syncytial virus. J Pediatr. 1999;135:21–7.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This study was supported by National Institute of Allergy and Infectious Diseases (NIAID) grant AI U19-AI-095277 (RSP, PI), and Vanderbilt Institute for Clinical and Translational Research Grant (UL1 TR000445 from NCATS/NIH). We thank Dr. Alkis Togias, Dr. Peter Gergen, and Ms. Joy Panza Laurienzo for thoughtful feedback on our study protocol and its implementation, as well as, careful review of our study materials. Study data were collected and managed using REDCap electronic data capture tools hosted at Vanderbilt. We also gratefully acknowledge the enthusiasm and support of parents and children in our study.

The following INSPIRE team members contributed to data collection. Vanderbilt: Sandy Alvarez-Calvillo, Margaret Bender, Teresa Chipps, Alexandra Connolly, Kaitlin Costello, Marcia Freudenthal, Rebecca Gammell, Jeff Horner, Ashudee Kirk, Anca Ifrim, Lina Keene, Zhouwen Liu, Kelsie McMurtry, Sara Reiss, Theresa Rogers, Patty Russell, Stephanie Steen, and Kimberly B. Woodward.

The following INSPIRE Collaborators contributed to the care of our participants:

Columbia Pediatrics: Anthony L. Smith; Goodlettsville Pediatrics: M. Catherine Dundon; Pediatrics Associates of Franklin: A. Scott Brooks; Rivergate Pediatrics: Elizabeth P. Pierce; University Pediatrics: Rachel L. Mace; Vanderbilt Integrated Providers at Gallatin, Lafayette and White House: Ronald V. Miller; Vanderbilt University: Barron L. Patterson (Data and Safety Monitoring), Sandra A. Moutsios.

Author information

Authors and Affiliations

Authors

Consortia

Corresponding author

Correspondence to Tina V Hartert.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

EKL participated in the study design, coordination, analysis, and manuscript preparation. MLM, LJA, JC, RSP, SD, CRS and FP participated in design of select study components. TG, WD participated in the design of the study and data analysis. PAM, PEM, RV, and DHA participated in study coordination. TVH conceived of the idea, designed the study, obtained study funding, supervised acquisition and analysis of patient and biospecimen data and prepared the manuscript. All authors read and approved the final manuscript.

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Larkin, E.K., Gebretsadik, T., Moore, M.L. et al. Objectives, design and enrollment results from the Infant Susceptibility to Pulmonary Infections and Asthma Following RSV Exposure Study (INSPIRE). BMC Pulm Med 15, 45 (2015). https://doi.org/10.1186/s12890-015-0040-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12890-015-0040-0

Keywords