You are on page 1of 9

Clinical Commentary Review

Rhinovirus Infections and Their Roles in Asthma:


Etiology and Exacerbations
David J. Jackson, MRCP, MSc, PhDa,b, and James E. Gern, MDc London, United Kingdom; and Madison, Wis

INFORMATION FOR CATEGORY 1 CME CREDIT AMA PRA Category 1 CreditÔ. Physicians should claim only the credit
commensurate with the extent of their participation in the activity.
Credit can now be obtained, free for a limited time, by reading the
review articles in this issue. Please note the following instructions. List of Design Committee Members: David J. Jackson, MRCP, MSc,
PhD, and James E. Gern, MD (authors); Robert S. Zeiger, MD, PhD
Method of Physician Participation in Learning Process: The core (editor)
material for these activities can be read in this issue of the Journal or
online at the JACI: In Practice Web site: www.jaci-inpractice.org/. The Learning objectives:
accompanying tests may only be submitted online at www.jaci-
inpractice.org/. Fax or other copies will not be accepted. 1. To predict the subsequent asthma risk for preschool children who
wheeze with rhinoviruses (RVs).
Date of Original Release: March 1, 2022. Credit may be obtained for
these courses until February 28, 2023. 2. To recognize that respiratory viruses and in particular RVs are the
dominant drivers of exacerbations in asthma.
Copyright Statement: Copyright Ó 2022-2024. All rights reserved.
3. To explain the relationship between deficiencies in antiviral immune
Overall Purpose/Goal: To provide excellent reviews on key aspects of responses and RV-induced morbidity in asthma.
allergic disease to those who research, treat, or manage allergic disease.
4. To discuss how controlling underlying allergic and type 2 (T2)
Target Audience: Physicians and researchers within the field of immune pathways could reduce RV-induced morbidity in asthma.
allergic disease.
Recognition of Commercial Support: This CME has not received
Accreditation/Provider Statements and Credit Designation: The external commercial support.
American Academy of Allergy, Asthma & Immunology (AAAAI) is
accredited by the Accreditation Council for Continuing Medical Edu- Disclosure of Relevant Financial Relationships with Commercial
cation (ACCME) to provide continuing medical education for physi- Interests: All authors and reviewers reported no relevant financial
cians. The AAAAI designates this journal-based CME activity for 1.00 relationships.

Rhinovirus infections can cause wheezing illnesses in all age groups. airway obstruction and exacerbations in children and adults.
In preschool children, rhinovirus infections frequently initiate acute Paradoxically, for most individuals, rhinovirus infections
wheezing illnesses. Children who wheeze with rhinoviruses are at commonly cause cold symptoms with little or no involvement of the
increased risk to go on to develop asthma. Once asthma is lower airways. This paradox has led investigators to identify specific
established, rhinovirus infections are potent triggers for acute risk factors and mechanisms for rhinovirus wheezing, and this
review will outline progress in 3 main areas. First, the 3 species of
a
Guy’s Severe Asthma Centre, Guy’s & St Thomas’ NHS Trust, London, United rhinoviruses have different patterns of infection and virulence.
Kingdom Second, personal factors such as lung function and immunity
b
School of Immunology & Microbial Sciences, King’s College London, London, influence lower respiratory outcomes of rhinovirus infection. The
United Kingdom
c
Departments of Pediatrics and Medicine, University of Wisconsin School of
mucosal immune response is critical, and the quality of the
Medicine and Public Health, Madison, Wis interferon response and allergic inflammation interacts to
This work was funded by the National Institutes of Health (grant nos. UM1 determine the risk for rhinovirus wheezing. Finally, rhinovirus
AI160040-01, U19 AI104317, and UH3 OD023282). infections can promote pathogen-dominated airway microbiota
Conflicts of interest: D. J. Jackson has received speaker fees and consultancy fees
that increase the risk for wheezing. Although specific antivirals for
from AstraZeneca, GlaxoSmithKline, Sanofi, Teva, and Chiesi. J. E. Gern is a paid
consultant to AstraZeneca, Gossamer Bio, and Meissa Vaccines, has stock options rhinovirus are still not available, identifying risk factors for
in Meissa Vaccines, and has patents on methods for producing rhinoviruses. wheezing illnesses has provided several other potential targets and
Received for publication November 17, 2021; revised manuscript received and strategies for reducing the risk of rhinovirus-induced wheezing and
accepted for publication January 6, 2022. exacerbations of asthma. Ó 2022 The Authors. Published by
Available online January 22, 2022.
Corresponding author: David J. Jackson, MRCP, MSc, PhD, Guy’s Severe Asthma
Elsevier Inc. on behalf of the American Academy of Allergy, Asthma
Centre, School of Immunology & Microbial Sciences, King’s College London, & Immunology. This is an open access article under the CC BY
London SE1 9RT, UK. E-mail: David.Jackson@gstt.nhs.uk. license (http://creativecommons.org/licenses/by/4.0/). (J Allergy
2213-2198 Clin Immunol Pract 2022;10:673-81)
Ó 2022 The Authors. Published by Elsevier Inc. on behalf of the American Academy
of Allergy, Asthma & Immunology. This is an open access article under the CC Key words: Rhinovirus; Infection; Asthma; Exacerbation; Type 2
BY license (http://creativecommons.org/licenses/by/4.0/).
inflammation
https://doi.org/10.1016/j.jaip.2022.01.006

673
674 JACKSON AND GERN J ALLERGY CLIN IMMUNOL PRACT
MARCH 2022

immune antiviral responses and stimulate the secretion of che-


Abbreviations used mokines and growth factors that recruit and activate leuko-
COVID-19- coronavirus disease 2019 cytes.9,10 Notably, interferon (IFN) responses are lower in young
ICS- inhaled corticosteroid infants, which may partially explain the greater severity of RV
IFN- interferon
illnesses in this age group. Infections cause increases in airway
ILC2- type 2 innate lymphoid cell
neutrophils and mononuclear cells during the acute phase, along
RV- rhinovirus
type 2- T2 with smaller numbers of eosinophils during the recovery period.
IFNs produced by epithelial cells and mononuclear cells have
potent anti-RV activity.11 A brisk IFN response at the beginning
of infection likely limits viral replication and prevents significant
illnesses.12 If antiviral defenses do not control viral replication
and spread during the early stages of infection, high-level viral
INTRODUCTION replication can induce pronounced IFN and neutrophilic re-
Rhinovirus (RV)-induced wheezing illnesses are a prominent sponses that likely add to the severity of illness.13,14 Analysis of
feature of asthma in children and adults. Although several res- nasal epithelial cell transcriptional responses suggests that some
piratory viruses can cause wheezing illnesses and exacerbations of illnesses are accompanied by marked activation of IFN pathways,
asthma, RV infections are the most common cause of wheezing whereas others are IFN-low and instead increase expression of
in children older than 2 years. Children who experience RV- proinflammatory cytokines and growth factors.15
induced wheezing and develop respiratory allergies at an early RV infections of the upper and lower airway can induce
age are quite likely to develop persistent childhood asthma. Once cellular inflammation, proinflammatory mediators, shedding of
asthma is established, RV infections continue to be a major cause dead cells, and mucus production, which can contribute to res-
of acute wheezing illnesses. The contrast between RV infections piratory symptoms. In addition, lower airway infection can in-
causing severe wheezing in asthma compared with mild upper crease airway responsiveness, which correlates with the induction
respiratory illnesses in the general population has led to extensive of proinflammatory cytokines such as IL-6.16 These pathophys-
studies to identify risk factors for more severe RV illnesses in iologic responses to the virus can lead to airway obstruction in
asthma. This review will summarize current concepts to explain preschool children and both children and adults with asthma.
how RV infections and illnesses in early childhood relate to the
subsequent onset of asthma. We will also discuss mechanisms
RV INFECTIONS AND THE ONSET OF ASTHMA
linking RV infections to acute exacerbations of asthma. Finally,
Several factors related to the virus, host, and environment are
we will explore opportunities for the prevention and treatment of
associated with the risk of developing more severe RV illnesses.
more severe RV wheezing illnesses.
Children who wheeze with RV infections are more likely to
develop asthma, and risk factors for this transition have also been
RV INFECTIONS AND IMMUNE RESPONSES identified (Figure 1).
RV infections are the most common illness of childhood.
During the first few years of life, nearly all children are infected Viral factors
multiple times. The multitude of RV infections in children is due RV-A and RV-C species viruses are more commonly associ-
to the large number of viral serotypes (w170) that induce ated with significant illnesses and wheezing compared with RV-B
antibody responses with little or no cross-neutralization among species viruses.17 RV-B infections generally cause either mild
known serotypes.1 Epidemiologic studies demonstrate that symptoms or are asymptomatic but can contribute to exacerba-
approximately 20 RV types can circulate in a given geographic tions in patients with more severe or unstable asthma.17,18 The
area during the spring or fall season.2 This broad circulation of increased virulence of the A and C species may be related to
viruses, which are spread most efficiently by young children, faster rates of viral replication and eliciting greater inflammatory
likely contributes to infection rates that peak when children re- responses compared with the RV-B spcies.19 A pooled analysis of
turn to school following summer or spring vacations.3 RV in- RV epidemiology from multiple studies suggests that there may
fections are widespread and typically produce mild illnesses in be certain RV types that are consistently more common and that
most children. Although only a subset of RV infections cause virulence may vary by type even within species.17
lower respiratory infections and wheezing, RV-induced
morbidity and health care costs are immense due to the sheer Host factors
number of infections. Several host factors are related to a greater risk for severe RV
RV types are classified into 3 species on the basis of sequence illnesses and wheezing. First, young age predisposes to wheezing
similarity.4 The A and B species were first identified decades ago, or febrile illnesses, especially with RV-C infection.17 Second,
whereas the RV-C species was first recognized in 2006.5 The genetic factors can increase the risk for RV wheezing. For
delay in finding RV-C was because it binds to a unique receptor example, single nucleotide polymorphisms in the 17q12-21 re-
(cadherin-related family member 3), which is not expressed in gion are associated with acute RV wheezing and asthma devel-
cell lines commonly used for viral culture.6 These viruses grow in opment in children who experience RV wheezing episodes.20
ciliated airway epithelial cells in the upper airways and the large The polymorphisms most closely linked to asthma risk also
and medium-size lower airways.7 Infections are generally limited regulate expression of gasdermin-B,21 a protein that regulates a
to the airway, although the virus can sometimes be detected in cell death and inflammatory process known as pyroptosis. When
the bloodstream of young children infected with RV-C.8 a cell senses an intracellular pathogen, pyroptosis and other cell
The immune response to the virus begins in the airway death pathways may help to limit the spread of viruses to other
epithelial cell. Sensors for viral capsid and RNA initiate innate cells.22 For RV-C, a single nucleotide polymorphism in the
J ALLERGY CLIN IMMUNOL PRACT JACKSON AND GERN 675
VOLUME 10, NUMBER 3

bacteria that contribute to the severity of illness, as has been


demonstrated in school-age children.2
Conversely, airway microbiomes dominated by commensals
were associated with reduced risk for viral wheezing episodes.
RV-C wheezing episodes, febrile illnesses, and colonization with
Streptococcus were all associated with subsequent risk for
asthma.34 In addition, the combination of a pathogen-dominated
microbiome during acute illnesses and allergic sensitization was
associated with persistent wheeze in early childhood.35 The
Childhood Origins of Asthma birth cohort study compared the
etiology of wheezing illnesses and changes in the airway micro-
biota through the age of 2 years to subsequent asthma diagnoses
FIGURE 1. Risk factors for RV wheezing illnesses and for pro-
through age 18 years. This analysis identified 2 risk factors for
gression from RV wheezing to childhood asthma. sIgE, Specific
persistent asthma: (1) an airway microbiome dominated by
IgE.
Staphylococcus aureus in the first 6 months of life, and (2)
detection of RV and Moraxella-dominated microbiome during
wheezing illnesses.36 An analysis of upper airway secretions
cadherin-related family member 3 (RV-C receptor) gene regu- during RV bronchiolitis using an integrated “omics” approach
lates how much protein is expressed on the surface of airway identified 4 different phenotypes. The phenotype associated with
epithelial cells,23 and increases RV-C binding and replica- RV-C infection, Moraxella-dominated microbiome, and
tion.24,25 The single nucleotide polymorphism (rs6967330) increased type 2 (T2) cytokine response had the highest inci-
associated with greater cell surface expression also increases the dence of childhood asthma by age 5 years.37
risk of RV-C infections during childhood,26 wheezing illnesses These studies suggest that in early life, the balance between airway
during infancy and childhood,17,23 and early-onset childhood commensals and pathogens in airway secretions influences the risk
asthma.23 The association of genetic risk factors for RV wheezing for wheezing illnesses. Underlying mechanisms may be related to the
with childhood asthma suggests that injury due to repeated RV ability of pathogens to induce inflammation or disturb epithelial cell
lower respiratory infections in early life could promote the function. Defining mechanisms for bacterial pathogens to interact
development of asthma. with viruses, allergic inflammation, and epithelial cell function may
There may be bidirectional relationships between lung func- lead to new strategies for asthma prevention.
tion and RV lower respiratory illnesses. Low lung function in-
creases the risk of wheezing during infections with RV and other Do RV wheezing illnesses cause asthma?
respiratory viruses. For example, van der Zalm et al27 reported Longitudinal studies have established that children with RV
that airway resistance in the first 2 months of life was related to wheezing illnesses are at increased risk for developing asthma,
the risk of RV wheezing illness in the first year of life. In addi- leading to speculation that this relationship is causal. Consider-
tion, RV wheezing illnesses in early childhood are associated with able data from experimental models support this theory. As
reduced FEV1 and FEV1/forced vital capacity measured by discussed above, RV lower respiratory infection can damage the
spirometry at age 5 to 8 years.28 Given that RV infections can airway epithelium and induce secretion of factors that influence
induce factors involved in airway remodeling,29 repeated RV repair and promote pathologic remodeling of the airways. In
lower respiratory infections could promote progressive airway mouse models, RV infection at an earlier age produces long-
obstruction. lasting effects on airway physiology.38 However, proving cau-
Allergy and evidence of type 2 inflammation are important sality would require a clinical trial with an effective antiviral
risk factors for RV wheeze. Preschoolers sensitized to aero- intervention. For example, treatment with palivizumab effec-
allergens or who have other atopic features such as eczema or tively prevents wheezing episodes due to respiratory syncytial
eosinophilia are at increased risk for RV wheeze.30 Family history virus and has enabled studies to test for effects on incident
of allergies or asthma in first-degree relatives also increases the asthma. Although palivizumab treatment reduces the risk of
risk for RV wheeze. Furthermore, the combination of atopy and recurrent wheeze through the preschool years, it has not
RV wheeze in the first few years of life strongly predicts pro- consistently reduced subsequent asthma.39,40 Similar studies are
gression to asthma.31,32 needed to determine whether inhibiting RV-induced preschool
wheezing can reduce the risk of asthma.

Environmental factors RV INFECTIONS AND EXACERBATIONS OF


Birth cohort studies have identified the airway microbiome as ASTHMA
an important factor in determining the risk for viral wheeze and It is now almost 50 years since Minor et al41 from Madison,
asthma. In the Copenhagen Prospective Study on Asthma in Wisconsin, first described a clear association between RV infec-
Childhood birth cohort, pharyngeal aspirates that tested positive tion and exacerbations of asthma. In this early report, an exac-
for bacterial pathogens were associated with recurrent wheezing erbation was observed in 21 of 23 patients with severe viral
and early childhood asthma.33 In the Childhood Asthma Study respiratory tract infections, of which 14 of 21 (66.7%) were
conducted in Perth, Australia, acute illnesses were related to identified as having infections caused by RV. The significance
detecting respiratory viruses and transient pathogen-dominated and seriousness of this link was further underscored a few years
airway microbiomes.34 These studies suggest that viral respira- later with the first case report of a fatal outcome of a young child
tory infections can cause a temporary increase in pathogenic with an RV infection exacerbating their asthma.42
676 JACKSON AND GERN J ALLERGY CLIN IMMUNOL PRACT
MARCH 2022

FIGURE 2. The pathogenesis of an RV-induced asthma exacerbation. RV infects the respiratory epithelium, leading to release of the
epithelial-derived cytokines IL-25, IL-33, and TSLP, which drives downstream release of IL-4, IL-5, and IL-13 from TH2 cells and ILC2s. IL-5
plays a key role in driving airway eosinophilia and is itself released by eosinophils; IL-13 is central for mucus hypersecretion and airway
hyperresponsiveness and is additionally eosinophil-derived. pDCs are important sources of antiviral type 1 IFN, and their responsiveness
to RV infection is impaired in the presence of allergic inflammation. ILC2 production of T2 cytokines is partly inhibited by type 1 IFN. pDC,
Plasmocytoid dendritic cell; TSLP, thymic stromal lymphopoietin.

Since then, several larger analyses have been conducted for exacerbations in patients with poor asthma control in the real
including the Origins of Asthma study, which recorded respira- world.49-51
tory viruses in more than two-thirds of 217 exacerbations in In combination with a substantial body of work using both
children, with RV again identified as the most common experimental mouse models and human in vitro studies, the
precipitant.43 human in vivo data to emerge from experimental RV infection of
As described earlier, a clear seasonal pattern of pediatric subjects with asthma have highlighted 2 critical interconnected
asthma exacerbations is well established, with asthma hospitali- areas of dysregulated immunity underpinning the relationship
zation rates in children peaking each year in autumn and early between RV infection and asthma exacerbations.47,52-55 The first
winter, hypothesized to be mediated by viral transmission.3 relates to an exaggerated activation of T2 immune pathways in
Although this pattern is slightly more variable in adults, a individuals with asthma following infection with RV; the second
strong link between asthma exacerbations and respiratory virus relates to a deficiency and/or delay in the antiviral immune
infections remains, with RV and influenza A standing out as the response to RV infection (Figure 2). These mechanisms will each
main culprits.44,45 This is true even in the most severe cases, as be discussed in the following sections.
evidenced by a recent analysis of adults with near-fatal asthma
exacerbations requiring extracorporeal membrane oxygenation in
which respiratory viruses were the trigger in more than 50% of RV-INDUCED T2 INFLAMMATION IN ASTHMA
patients, with RV again the most common pathogen.46 Upregulated T2 immune pathways represent the dominant
A significant step forward in understanding the mechanisms of inflammatory phenotype seen in asthma, with levels directly
RV-induced exacerbations of asthma followed the development relating to both disease severity and disease control.50,56,57 RV
of human models of experimental infection with RV. Clinically, has been shown to induce a wide range of factors relevant to this
experimental infection with RV-16 results in significant declines pathway, including activation of the key transcription factor
in lung function and increased lower respiratory symptoms in signal transducer and activator of transcription 6, and induction
subjects with asthma compared with healthy subjects.47 More- of the T-cell and eosinophil chemokines CCL11, CCL17,
over, the level of asthma control at the time of experimental CCL22, and CCL26, the epithelial-derived IL-25 and IL-33, as
infection directly relates to the severity of the lower respiratory well as the cytokines IL-4, IL-5, IL-13, and prostaglandin
symptoms,48 a finding that accurately reflects the increased risk D2.47,54,58-61
J ALLERGY CLIN IMMUNOL PRACT JACKSON AND GERN 677
VOLUME 10, NUMBER 3

However, others have not seen a difference in viral titers be-


tween subjects with asthma and healthy controls,65 which may
reflect differences in the baseline level of asthma control and
severity between studies.
Although much of the data to date relate to bronchial
epithelial cells as the primary site of infection, the range of cells
involved in the dysregulated antiviral immune response in
asthma continues to expand. Wirz et al66 recently investigated a
potential new function of B cells comparing the response of
circulating B cells upon experimental RV infection in healthy
versus asthmatic subjects. Costimulation of B cells with RV and
IFN-a upregulated proinflammatory cytokine expression;
notably, subjects with asthma showed dysregulation of antiviral
gene expression. Jansen et al67 recently investigated the effects of
RV infection on regulatory T cells by isolating regulatory T cells
from subjects with asthma and controls after experimental
infection with the RV. The authors demonstrated that RV
induced a strong antiviral response in regulatory T cells; however,
in subjects with asthma, the inflammatory response was dysre-
gulated compared with that observed in the controls. In addition,
subjects with asthma failed to upregulate several immunosup-
pressive molecules such as CTLA4 and CD69.67
FIGURE 3. Potentially modifiable risk factors for RV-induced
wheezing and exacerbations of asthma.
RELATIONSHIP BETWEEN IMPAIRED ANTIVIRAL
Early in the T2 cascade, IL-33 (synthesized in response to RESPONSES AND T2 INFLAMMATION
bronchial epithelial infection) appears to be a particularly critical Although first considered an independent finding and distinct
mediator in both TH2 cell and T2 innate lymphoid cell (ILC2)- from the exaggerated T2 inflammation triggered by RV in asthma,
driven responses to RV.47 Levels of IL-33 during infection the deficient antiviral immune response is increasingly considered
correlate with asthma symptom severity, with IL-5 and IL-13 tightly connected. Altman et al12 reported that the ratio of T2 to
induction as well as with viral load. Blocking the IL-33 recep- T1 IFN gene expression was predictive in determining the risk for
tor ST2 completely inhibits RV-induced TH2 polarization, a future exacerbation; individuals in the highest quartile for this
making the prospect of IL-33 inhibition an exciting one.47 expression ratio were associated with a significantly shorter time to
ILC2s are important sources of T2 cytokines including IL-5, exacerbation than were individuals in the lower 3 quartiles. This
IL-9, and IL-13, leading to the recruitment and activation of finding very much supports the clinical association between the
eosinophils and mast cells, and goblet cellemediated mucus routinely used T2 biomarkers (blood eosinophil count and frac-
production, respectively. In subjects with asthma, experimental tional exhaled nitric oxide) and exacerbation risk.68,69
RV infection leads to an increase in airway eosinophilia47 as well Using mouse models of infection, Duerr et al70 elegantly
as ILC2 numbers, with the ILC2:ILC1 ratio correlating with demonstrated that deficiency in type 1 IFN signaling led to
exacerbation severity and duration of RV infection.62 dysregulated activation of ILC2s and infection-associated T2
Somewhat counterintuitively, airway neutrophilia is also immunopathology. They showed that IFN-b acted directly on
increased during virus-induced exacerbations and may have an ILC2s in vivo and that cytokine secretion by human ILC2s was
important role in exacerbation pathogenesis by activating significantly decreased by the addition of IFN-b. The authors
neutrophil extracellular traps (NETosis). However, rather than also observed that IFN-b inhibited the IL-33emediated increase
this process being independent of T2 pathways, NETosis boosts in pulmonary eosinophilia, and that IL-33einduced airway
T2 responses, further driving exacerbation severity.63 hyperresponsiveness was significantly suppressed by IFN-b.70
The reverse has also been demonstrated, with IL-33 shown to
have a role in dampening innate and adaptive TH1-like responses
EVIDENCE FOR A DEFICIENT ANTIVIRAL including suppressing IFN-b.71
RESPONSE IN ASTHMA A fascinating piece of evidence corroborating the relationship
IFNs represent the first-line defense against respiratory viruses between allergic inflammation and antiviral responses has been
such as RV and are a critical component of the innate immune the observation that impaired IFN responses to RV in children
response. Three classes of IFNs have been described: types 1 with asthma correlate with increased FcεRI expression on plas-
(IFN-a and IFN-b), 2 (IFN-g), and 3 (IFN-l). To date, a macytoid dendritic cells (a key cellular source of type 1 IFN) as
substantial number of studies using various cell types including well as reduced IFN responses in the presence of FcεRI cross-
human bronchial epithelial cells, bronchoalveolar lavage cells, linking.72 Subsequently, a landmark study using the anti-IgE
PBMCs, and dendritic cells have demonstrated a delay and/or therapy omalizumab reduced the infection-related seasonal
deficiency in IFN induction following RV infection in patients peaks in exacerbations.73 In the controlled setting of an experi-
with asthma.52,53,64 During experimental infection with RV, this mental RV challenge, Heymann et al74 administered omalizu-
antiviral deficiency manifests in vivo as increased virus load in mab or matched placebo in a blinded fashion to 20 subjects with
subjects with asthma compared with healthy subjects.47,62 mild asthma, demonstrating both subjective symptom benefit
678 JACKSON AND GERN J ALLERGY CLIN IMMUNOL PRACT
MARCH 2022

and a blunting of the RV-induced blood eosinophilia. Another As a natural progression to the in vitro studies of human
mechanism that could contribute to the effectiveness of anti-IgE primary bronchial epithelial cells highlighting that exogenous
may relate to a potential role of RV-specific IgE in mediating IFNs reduce virus shedding of RV, Djukanovic et al86 conducted
RV-triggered asthma exacerbations.75 However, data supporting a clinical trial of inhaled IFN-b initiated at the onset of a
this possibility are limited at present. community-acquired respiratory virus infection in subjects with
asthma. Although the overall study was negative, it was possible
THERAPEUTIC APPROACHES TO LIMIT VIRUS- to demonstrate modest efficacy in the more severe, poorly
INDUCED MORBIDITY controlled subgroup of patients.86 Although this treatment
Identifying risk factors for more severe RV wheezing illnesses approach has not progressed further in asthma, it is now being
and exacerbations of asthma suggest several potential approaches investigated in the management of severe coronavirus disease
to reducing RV-related illnesses and respiratory morbidity. Risk 2019 (COVID-19) infection.87
factors that are potentially modifiable include T2 inflammation, The macrolide azithromycin has been intensively investigated
impaired antiviral responses, and pathogen-dominated micro- for nonantibiotic properties, and there is some promising data to
biome (Figure 3). Apart from preventing the immediate harm suggest it may possess some antiviral effects. Gielen et al88
from an exacerbation, these measures are important because observed that azithromycin enhanced viral-induced type I and
repeated RV-induced exacerbations may drive an irreversible loss III IFN, leading to reduced RV replication and release following
of lung function through induction of airway remodeling factors in vitro infection of primary human bronchial epithelial cells.
including TGF-b, and vascular endothelial growth factor.29 Although azithromycin was able to reduce exacerbations by
approximately 1 per year in the AMAZES study, it has not been
Targeting T2 inflammation demonstrated that this was due to the inhibition of a virus-
The remarkable success of the current generation of biologic induced exacerbation.89
agents targeting mediators and cells of the T2 cascade in pre-
Modifying the airway microbiome
venting asthma exacerbations has provided overwhelming evi-
Growing evidence relating wheezing illnesses to combinations
dence of the central role of this pathway in virus-induced asthma
of viral and bacterial pathogens raises the possibility that modi-
exacerbations. How much of this relates to the direct inhibition
fying the airway microbiome could help to prevent acute
of these mediators or the indirect augmentation and normaliza-
wheezing illnesses. Probiotics are now being developed to restore
tion of antiviral pathways remains unclear. However, it most
the physiology of the epithelial environment on the skin for
likely is a combination of the 2. In addition to the results of the
patients with atopic dermatitis and populate the gastrointestinal
anti-IgE therapy omalizumab described earlier, some of the most
tract with commensal bacteria, promote the development of
impressive results have been seen in the controlled and real-world
tolerance mechanisms, and reduce the risk for developing allergy.
studies of the eosinophil targeting antieIL-5/5R mAbs mepoli-
A Cochrane review of oral probiotics concluded that there is low-
zumab and benralizumab, in which most patients become
quality evidence that treatment of children with oral probiotics
completely exacerbation-free, despite a high probability that
can reduce rates of respiratory illness.90 In a study involving
most encounter at least 1 seasonal respiratory virus during the
experimental RV inoculation, probiotic supplementation influ-
autumn or winter months.76-81 Interestingly, the reductions in
enced airway immune responses to RV but did not have
exacerbation rates also seen in the placebo arms of the biologic
demonstrable antiviral effects.91 Studies in murine models
trials are likely to reflect the impact of improved adherence to the
demonstrated that nasal administration of commensal bacteria
T2-inhibiting inhaled corticosteroids (ICSs)76,81,82 these patients
could reduce respiratory virus-induced pathology,92 suggesting
are also prescribed and is in keeping with the general effectiveness
that clinical studies of nasal probiotics are warranted. Other
of even short-term ICSs in preventing exacerbations.83
approaches to altering the nasal microbiota could include
Following the identification of the annual September
vaccination against airway pathogens. In addition, oral bacterial
epidemic of asthma exacerbations in children returning to school,
extracts (eg, OM-85) have shown promise to prevent severe
Johnston et al84 performed a randomized, placebo-controlled
wheezing illnesses in children.93 Finally, 2 high-quality studies
trial of the addition of the antileukotriene montelukast to
have shown evidence that starting azithromycin treatment at the
usual asthma therapy between September and mid-October with
onset of respiratory illness can prevent severe wheezing illnesses
the aim of curtailing this trend. Although a less potent anti-T2
in preschool children with a history of recurrent wheeze.94,95
therapy compared with ICSs, of the 194 children with asthma
Enthusiasm for this approach is dampened by concerns about
who participated in the study, 22 (4 montelukast vs 18 placebo)
overuse of antibiotics leading to resistant organisms, and also by
required an unscheduled physician visit, representing a 78%
studies linking antibiotic use in early life to increased risk for
reduction in those receiving montelukast.84
developing asthma.96 Additional studies are warranted to deter-
Augmenting antiviral pathways mine whether there are subgroups of young children who would
Unfortunately, there are currently no approved antiviral agents benefit from antibiotic treatment for recurrent wheeze.
for the treatment of RV infection. Several approaches have been
evaluated in clinical trials, including the small-molecule capsid- LESSONS LEARNED FROM THE COVID-19
binding inhibitor pleconaril, the 3C protease inhibitor rupin- PANDEMIC
trivir, as well as soluble intercellular adhesion molecule 1.85 All On the background of the well-established relationship be-
have failed to progress either because of safety concerns, limited tween asthma exacerbations and respiratory viruses, the medical
efficacy, or potential drug interactions. The hope of a vaccine has community and patients with asthma alike faced the COVID-19
been additionally hampered by the large number of antigenically pandemic with a heightened sense of concern and trepidation.
distinct RV serotypes. Yet, almost 2 years into the pandemic, the question of whether or
J ALLERGY CLIN IMMUNOL PRACT JACKSON AND GERN 679
VOLUME 10, NUMBER 3

not severe acute respiratory syndrome coronavirus 2 actually capable of dampening T2 inflammation including ICSs and
causes a conventional asthma exacerbation in the way that RV or anti-T2 biologics potentially correct many of these antiviral
influenza does remains a contentious point of ongoing debate.97 deficiencies for us. However, even the dramatic responses to anti-
In contrast to RV, seasonal coronaviruses have not generally been T2 biologics appear to be eclipsed by the success of non-
reported as a prominent exacerbation trigger in asthma.44 In pharmacological interventions including mask wearing and hand
essence, this suggests a fundamental difference in how corona- hygiene in preventing virus-induced exacerbations made evident
viruses and RVs interact with, and stimulate the asthmatic host by the COVID-19 pandemic.
immune system. Indeed, rather than the eosinophilia that
frequently accompanies an RV-induced exacerbation of
REFERENCES
asthma,47 one of the earliest findings in the blood counts of 1. Cooney MK, Kenny GE, Tam R, Fox JP. Cross relationships among 37 rhi-
patients admitted with COVID-19 has been their eosinopenia. noviruses demonstrated by virus neutralization with potent monotypic rabbit
At present, some data suggest that compared with the general antisera. Infect Immun 1973;7:335-40.
population, patients with asthma with more severe disease and 2. Kloepfer KM, Lee WM, Pappas TE, Kang TJ, Vrtis RF, Evans MD, et al.
Detection of pathogenic bacteria during rhinovirus infection is associated with
those with frequent exacerbations (ie, poor disease control) increased respiratory symptoms and asthma exacerbations. J Allergy Clin
appear more likely to be admitted to hospital with COVID-19 Immunol 2014;133. 1301-7.e3.
and more likely to require intensive care unit admission, after 3. Johnston NW, Johnston SL, Norman GR, Dai J, Sears MR. The September
accounting for major risk factors.98 However, the data remain epidemic of asthma hospitalization: school children as disease vectors.
J Allergy Clin Immunol 2006;117:557-62.
difficult to interpret in light of its retrospective nature as well as
4. Simmonds P, Gorbalenya AE, Harvala H, Hovi T, Knowles NJ, Lindberg AM,
other confounding factors. Importantly, Bloom et al98 demon- et al. Recommendations for the nomenclature of enteroviruses and rhinovi-
strated that these same patients with asthma had a significantly ruses. Arch Virol 2020;165:793-7.
higher risk of pneumonia and influenza-related admissions, with 5. Arden KE, McErlean P, Nissen MD, Sloots TP, Mackay IM. Frequent
similar levels of relative risk to COVID-19. detection of human rhinoviruses, paramyxoviruses, coronaviruses, and boca-
virus during acute respiratory tract infections. J Med Virol 2006;78:1232-40.
Perhaps one of the most remarkable outcomes of the 6. Bochkov YA, Watters K, Ashraf S, Griggs TF, Devries MK, Jackson DJ, et al.
pandemic has been an overall fall in asthma exacerbations. Cadherin-related family member 3, a childhood asthma susceptibility gene
Several factors could explain this, including a fall in urban air product, mediates rhinovirus C binding and replication. Proc Natl Acad Sci U
pollution levels and/or a change in patient behavior leading to an S A 2015;112:5485-90.
7. Mosser AG, Vrtis R, Burchell L, Lee WM, Dick CR, Weisshaar E, et al.
improvement in adherence to ICSs.99 However, it is possible that
Quantitative and qualitative analysis of rhinovirus infection in bronchial tis-
the nonpharmacologic interventions of increased hand washing sues. Am J Respir Crit Care Med 2005;171:645-51.
and mask wearing, aimed at reducing transmission of severe 8. Lu X, Schneider E, Jain S, Bramley AM, Hymas W, Stockmann C, et al.
acute respiratory syndrome coronavirus 2, will ultimately prove Rhinovirus viremia in patients hospitalized with community-acquired pneu-
to be the most effective intervention against seasonal virus- monia. J Infect Dis 2017;216:1104-11.
9. Slater L, Bartlett NW, Haas JJ, Zhu J, Message SD, Walton RP, et al. Co-
induced asthma exacerbations since the introduction of ICSs ordinated role of TLR3, RIG-I and MDA5 in the innate response to rhinovirus
almost half a century ago. For example, in New Zealand during in bronchial epithelium. PLoS Pathog 2010;6:e1001178.
the first wave of the pandemic, the usual winter influenza surge 10. Bochkov YA, Hanson KM, Keles S, Brockman-Schneider RA, Jarjour NN,
was essentially eliminated, with a 99.9% reduction in influenza Gern JE. Rhinovirus-induced modulation of gene expression in bronchial
epithelial cells from subjects with asthma. Mucosal Immunol 2010;3:69-80.
virus detections, and a 74.6% reduction in RV infections as
11. Becker TM, Durrani SR, Bochkov YA, Devries MK, Rajamanickam V,
compared with previous years. When the nonpharmacologic Jackson DJ. Effect of exogenous interferons on rhinovirus replication and
interventions were relaxed after lockdown, the incidence of RV airway inflammatory responses. Ann Allergy Asthma Immunol 2013;111:
increased rapidly.100 Using weekly national hospital admission 397-401.
data in New Zealand, Fairweather et al101 observed that asthma 12. Altman MC, Gill MA, Whalen E, Babineau DC, Shao B, Liu AH, et al.
Transcriptome networks identify mechanisms of viral and nonviral asthma
admissions initially fell during the period of more stringent re- exacerbations in children. Nat Immunol 2019;20:637-51.
strictions, only to sharply rise in parallel with RV detection rates 13. Schwantes EA, Manthei DM, Denlinger LC, Evans MD, Gern JE, Jarjour NN,
immediately on dropping the restrictions. Similar falls in asthma et al. Interferon gene expression in sputum cells correlates with the Asthma
exacerbation rates have been reported in several other countries as Index Score during virus-induced exacerbations. Clin Exp Allergy 2014;44:
813-21.
well.102,103 Critically, this raises the question as to whether
14. Miller EK, Hernandez JZ, Wimmenauer V, Shepherd BE, Hijano D, Libster R,
improved advice around nonpharmacological measures of hand et al. A mechanistic role for type III IFN-lambda1 in asthma exacerbations
washing and face masks should be routinely included in national mediated by human rhinoviruses. Am J Respir Crit Care Med 2012;185:
and international asthma guidelines long after the COVID-19 508-16.
pandemic has subsided. 15. Khoo SK, Read J, Franks K, Zhang G, Bizzintino J, Coleman L, et al. Upper
airway cell transcriptomics identify a major new immunological phenotype
with strong clinical correlates in young children with acute wheezing.
J Immunol 2019;202:1845-58.
CONCLUSIONS 16. Parikh V, Scala J, Patel R, Corbi C, Lo D, Bochkov YA, et al. Rhinovirus C15
The relationship between RV infection, asthma inception, and induces airway hyperresponsiveness via calcium mobilization in airway
smooth muscle. Am J Respir Cell Mol Biol 2020;62:310-8.
exacerbation pathogenesis continues to be an area of intense
17. Choi T, Devries M, Bacharier LB, Busse W, Camargo CA Jr, Cohen R, et al.
study. Researchers over the last 3 decades have revealed several Enhanced neutralizing antibody responses to rhinovirus C and age-dependent
key insights in asthma that underlie this association, including a patterns of infection. Am J Respir Crit Care Med 2021;203:822-30.
genetic predisposition, multiple deficiencies in the antiviral IFN 18. Esquivel A, Busse WW, Calatroni A, Togias AG, Grindle KG, Bochkov YA,
response, as well as the inappropriate activation of the T2 in- et al. Effects of omalizumab on rhinovirus infections, illnesses, and exacer-
bations of asthma. Am J Respir Crit Care Med 2017;196:985-92.
flammatory cascade in response to infection. Although specific 19. Nakagome K, Bochkov YA, Ashraf S, Brockman-Schneider RA, Evans MD,
antiviral approaches have largely failed, the close interrelationship Pasic TR, et al. Effects of rhinovirus species on viral replication and cytokine
between IFN and T2 pathways has preordained that treatments production. J Allergy Clin Immunol 2014;134:332-41.
680 JACKSON AND GERN J ALLERGY CLIN IMMUNOL PRACT
MARCH 2022

20. Caliskan M, Bochkov YA, Kreiner-Moller E, Bonnelykke K, Stein MM, Du G, 42. Las Heras J, Swanson VL. Sudden death of an infant with rhinovirus infection
et al. Rhinovirus wheezing illness and genetic risk of childhood-onset asthma. complicating bronchial asthma: case report. Pediatr Pathol 1983;1:319-23.
N Engl J Med 2013;368:1398-407. 43. Lemanske RF Jr. The childhood origins of asthma (COAST) study. Pediatr
21. Ober C, McKennan CG, Magnaye KM, Altman MC, Washington C III, Allergy Immunol 2002;13:38-43.
Stanhope C, et al. Expression quantitative trait locus fine mapping of the 44. Edwards MR, Bartlett NW, Hussell T, Openshaw P, Johnston SL. The
17q12-21 asthma locus in African American children: a genetic association microbiology of asthma. Nat Rev Microbiol 2012;10:459-71.
and gene expression study. Lancet Respir Med 2020;8:482-92. 45. Coleman AT, Jackson DJ, Gangnon RE, Evans MD, Lemanske RF, Gern JE.
22. Orning P, Lien E, Fitzgerald KA. Gasdermins and their role in immunity and Comparison of risk factors for viral and nonviral asthma exacerbations.
inflammation. J Exp Med 2019;216:2453-65. J Allergy Clin Immunol 2015;136. 1127-9.e4.
23. Bonnelykke K, Sleiman P, Nielsen K, Kreiner-Moller E, Mercader JM, 46. Patel S, Shah NM, Malhotra AM, Lockie C, Camporota L, Barrett N, et al.
Belgrave D, et al. A genome-wide association study identifies CDHR3 as a Inflammatory and microbiological associations with near-fatal asthma
susceptibility locus for early childhood asthma with severe exacerbations. Nat requiring extracorporeal membrane oxygenation. ERJ Open Res 2020;6:
Genet 2014;46:51-5. 00267-2019.
24. Basnet S, Bochkov Y, Brockman-Schneider R, Kuipers I, Aesif SW, 47. Jackson DJ, Makrinioti H, Rana BM, Shamji BW, Trujillo-Torralbo MB,
Jackson DJ, et al. CDHR3 asthma-risk genotype affects susceptibility of Footitt J, et al. IL-33-dependent type 2 inflammation during rhinovirus-induced
airway epithelium to rhinovirus C infections. Am J Resp Cell Mol Biol 2019; asthma exacerbations in vivo. Am J Respir Crit Care Med 2014;190:1373-82.
61:450-8. 48. Jackson DJ, Trujillo-Torralbo MB, del-Rosario J, Bartlett NW, Edwards MR,
25. Everman JL, Sajuthi S, Saef B, Rios C, Stoner AM, Numata M, et al. Func- Mallia P, et al. The influence of asthma control on the severity of virus-induced
tional genomics of CDHR3 confirms its role in HRV-C infection and child- asthma exacerbations. J Allergy Clin Immunol 2015;136:497-500.e3.
hood asthma exacerbations. J Allergy Clin Immunol 2019;144:962-71. 49. Wang E, Wechsler ME, Tran TN, Heaney LG, Jones RC, Menzies-Gow AN,
26. Bonnelykke K, Coleman AT, Evans MD, Thorsen J, Waage J, Vissing NH, et al. Characterization of severe asthma worldwide. Chest 2020;157:790-804.
et al. Cadherin-related family member 3 genetics and rhinovirus C respiratory 50. Jackson DJ, Busby J, Pfeffer PE, Menzies-Gow A, Brown T, Gore R, et al.
illnesses. Am J Respir Crit Care Med 2018;197:589-94. Characterisation of patients with severe asthma in the UK Severe Asthma
27. van der Zalm MM, Uiterwaal CS, Wilbrink B, Koopman M, Verheij TJ, van Registry in the biologic era. Thorax 2021;76:220-7.
der Ent CK. The influence of neonatal lung function on rhinovirus-associated 51. Yang F, Busby J, Heaney LG, Menzies-Gow A, Pfeffer PE, Jackson DJ, et al.
wheeze. Am J Respir Crit Care Med 2011;183:262-7. Factors associated with frequent exacerbations in the UK Severe Asthma
28. Guilbert TW, Singh AM, Danov Z, Evans MD, Jackson DJ, Burton R, et al. Registry. J Allergy Clin Immunol Pract 2021;9. 2691-701.e1.
Decreased lung function after preschool wheezing rhinovirus illnesses in 52. Contoli M, Message SD, Laza-Stanca V, Edwards MR, Wark PA, Bartlett NW,
children at risk to develop asthma. J Allergy Clin Immunol 2011;128. 532-538. et al. Role of deficient type III interferon-lambda production in asthma exac-
e1-538. erbations. Nat Med 2006;12:1023-6.
29. Leigh R, Oyelusi W, Wiehler S, Koetzler R, Zaheer RS, Newton R, et al. 53. Zhu J, Message SD, Mallia P, Kebadze T, Contoli M, Ward CK, et al.
Human rhinovirus infection enhances airway epithelial cell production of Bronchial mucosal IFN-alpha/beta and pattern recognition receptor expression
growth factors involved in airway remodeling. J Allergy Clin Immunol 2008; in patients with experimental rhinovirus-induced asthma exacerbations.
121:1238-45. J Allergy Clin Immunol 2019;143. 114-25.e4.
30. Jartti T, Kuusipalo H, Vuorinen T, Soderlund-Venermo M, Allander T, 54. Hansel TT, Tunstall T, Trujillo-Torralbo M-B, Shamji B, Del-Rosario A,
Waris M, et al. Allergic sensitization is associated with rhinovirus-, but not Dhariwal J, et al. A comprehensive evaluation of nasal and bronchial cytokines
other virus-, induced wheezing in children. Pediatr Allergy Immunol 2010;21: and chemokines following experimental rhinovirus infection in allergic
1008-14. asthma: increased interferons (IFN-g and IFN-l) and type 2 inflammation (IL-
31. Jackson DJ, Gangnon RE, Evans MD, Roberg KA, Anderson EL, Pappas TE, 5 and IL-13). EBioMedicine 2017;19:128-38.
et al. Wheezing rhinovirus illnesses in early life predict asthma development in 55. Nikonova A, Khaitov M, Jackson DJ, Traub S, Trujillo-Torralbo M-B,
high-risk children. Am J Respir Crit Care Med 2008;178:667-72. Kudlay DA, et al. M1-like macrophages are potent producers of anti-viral
32. Kusel MM, de Klerk NH, Kebadze T, Vohma V, Holt PG, Johnston SL, et al. interferons and M1-associated marker-positive lung macrophages are
Early-life respiratory viral infections, atopic sensitization, and risk of subse- decreased during rhinovirus-induced asthma exacerbations. EBioMedicine
quent development of persistent asthma. J Allergy Clin Immunol 2007;119: 2020;54. 102734.
1105-10. 56. Heaney LG, Perez de Llano L, Al-Ahmad M, Backer V, Busby J,
33. Bisgaard H, Hermansen MN, Buchvald F, Loland L, Halkjaer LB, Canonica GW, et al. Eosinophilic and noneosinophilic asthma: an expert
Bonnelykke K, et al. Childhood asthma after bacterial colonization of the consensus framework to characterize phenotypes in a global real-life severe
airway in neonates. N Engl J Med 2007;357:1487-95. asthma cohort. Chest 2021;160:814-30.
34. Teo SM, Mok D, Pham K, Kusel M, Serralha M, Troy N, et al. The infant 57. Price DB, Rigazio A, Campbell JD, Bleecker ER, Corrigan CJ, Thomas M,
nasopharyngeal microbiome impacts severity of lower respiratory infection et al. Blood eosinophil count and prospective annual asthma disease burden: a
and risk of asthma development. Cell Host Microbe 2015;17:704-15. UK cohort study. Lancet Respir Med 2015;3:849-58.
35. Teo SM, Tang HHF, Mok D, Judd LM, Watts SC, Pham K, et al. Airway 58. Beale J, Jayaraman A, Jackson DJ, Macintyre JDR, Edwards MR, Walton RP,
microbiota dynamics uncover a critical window for interplay of pathogenic et al. Rhinovirus-induced IL-25 in asthma exacerbation drives type 2 immunity
bacteria and allergy in childhood respiratory disease. Cell Host Microbe 2018; and allergic pulmonary inflammation. Sci Transl Med 2014;6. 256ra134.
24:341-352.e5. 59. Farne H, Jackson DJ, Johnston SL. Are emerging PGD2 antagonists a prom-
36. Tang HHF, Lang A, Teo SM, Judd LM, Gangnon R, Evans MD, et al. ising therapy class for treating asthma? Expert Opin Emerg Drugs 2016;21:
Developmental patterns in the nasopharyngeal microbiome during infancy are 359-64.
associated with asthma risk. J Allergy Clin Immunol 2021;147:1683-91. 60. Williams TC, Jackson DJ, Maltby S, Walton RP, Ching YM, Glanville N, et al.
37. Raita Y, Camargo CA Jr, Bochkov YA, Celedon JC, Gern JE, Mansbach JM, Rhinovirus-induced CCL17 and CCL22 in asthma exacerbations and differ-
et al. Integrated-omics endotyping of infants with rhinovirus bronchiolitis and ential regulation by STAT6. Am J Respir Cell Mol Biol 2021;64:344-56.
risk of childhood asthma. J Allergy Clin Immunol 2021;147:2108-17. 61. Farne H, Glanville N, Johnson N, Kebadze T, Aniscenko J, Regis E, et al.
38. Hong JY, Bentley JK, Chung Y, Lei J, Steenrod JM, Chen Q, et al. Neonatal Effect of CRTH2 antagonism on the response to experimental rhinovirus
rhinovirus induces mucous metaplasia and airways hyperresponsiveness infection in asthma: a pilot randomised controlled trial. Thorax. Published
through IL-25 and type 2 innate lymphoid cells. J Allergy Clin Immunol 2014; online October 29, 2021. https://doi.org/10.1136/thoraxjnl-2021-217429
134:429-39. 62. Dhariwal J, Cameron A, Wong E, Paulsen M, Trujillo-Torralbo B, Del
39. Mochizuki H, Kusuda S, Okada K, Yoshihara S, Furuya H, Simoes EA. Rosario A, et al. Pulmonary innate lymphoid cell responses during rhinovirus-
Palivizumab prophylaxis in preterm infants and subsequent recurrent induced asthma exacerbations in vivo: a clinical trial. Am J Respir Crit Care
wheezing: 6 year follow up study. Am J Respir Crit Care Med 2017;196:29-38. Med 2021;204:1259-73.
40. Simoes EA, Carbonell-Estrany X, Rieger CH, Mitchell I, Fredrick L, 63. Toussaint M, Jackson DJ, Swieboda D, Guedán A, Tsourouktsoglou T-D,
Groothuis JR. The effect of respiratory syncytial virus on subsequent recurrent Ching YM, et al. Host DNA released by NETosis promotes rhinovirus-induced
wheezing in atopic and nonatopic children. J Allergy Clin Immunol 2010;126: type-2 allergic asthma exacerbation. Nat Med 2017;23:681-91.
256-62. 64. Jackson DJ, Glanville N, Trujillo-Torralbo MB, Shamji BW, Del-Rosario J,
41. Minor TE, Dick EC, DeMeo AN, Ouellette JJ, Cohen M, Reed CE. Viruses as Mallia P, et al. Interleukin-18 is associated with protection against rhinovirus-
precipitants of asthmatic attacks in children. JAMA 1974;227:292-8. induced colds and asthma exacerbations. Clin Infect Dis 2015;60:1528-31.
J ALLERGY CLIN IMMUNOL PRACT JACKSON AND GERN 681
VOLUME 10, NUMBER 3

65. Kennedy JL, Shaker M, McMeen V, Gern J, Carper H, Murphy D, et al. 84. Johnston NW, Mandhane PJ, Dai J, Duncan JM, Greene JM, Lambert K, et al.
Comparison of viral load in individuals with and without asthma during in- Attenuation of the September epidemic of asthma exacerbations in children: a
fections with rhinovirus. Am J Respir Crit Care Med 2014;189:532-9. randomized, controlled trial of montelukast added to usual therapy. Pediatrics
66. Wirz OF, Jansen K, Satitsuksanoa P, van de Veen W, Tan G, Sokolowska M, 2007;120:e702-12.
et al. Experimental rhinovirus infection induces an antiviral response in 85. Hayden FG, Herrington DT, Coats TL, Kim K, Cooper EC, Villano SA, et al.
circulating B cells which is dysregulated in patients with asthma. Allergy 2022; Efficacy and safety of oral pleconaril for treatment of colds due to picorna-
77:130-42. viruses in adults: results of 2 double-blind, randomized, placebo-controlled
67. Jansen K, Wirz OF, van de Veen W, Tan G, Mirer D, Sokolowska M, et al. trials. Clin Infect Dis 2003;36:1523-32.
Loss of regulatory capacity in Treg cells following rhinovirus infection. 86. Djukanovic R, Harrison T, Johnston SL, Gabbay F, Wark P, Thomson NC, et al.
J Allergy Clin Immunol 2021;148:1016-1029.e16. The effect of inhaled IFN-b on worsening of asthma symptoms caused by viral
68. Jackson DJ, Humbert M, Hirsch I, Newbold P, Garcia Gil E. Ability of serum infections. A randomized trial. Am J Respir Crit Care Med 2014;190:145-54.
IgE concentration to predict exacerbation risk and benralizumab efficacy for 87. Monk PD, Marsden RJ, Tear VJ, Brookes J, Batten TN, Mankowski M, et al.
patients with severe eosinophilic asthma. Adv Ther 2020;37:718-29. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for
69. Busse WW, Wenzel SE, Casale TB, FitzGerald JM, Rice MS, Daizadeh N, treatment of SARS-CoV-2 infection: a randomised, double-blind, placebo-
et al. Baseline FeNO as a prognostic biomarker for subsequent severe asthma controlled, phase 2 trial. Lancet Respir Med 2021;9:196-206.
exacerbations in patients with uncontrolled, moderate-to-severe asthma 88. Gielen V, Johnston SL, Edwards MR. Azithromycin induces anti-viral re-
receiving placebo in the LIBERTY ASTHMA QUEST study: a post-hoc sponses in bronchial epithelial cells. Eur Respir J 2010;36:646-54.
analysis. Lancet Respir Med 2021;9:1165-73. 89. Gibson PG, Yang IA, Upham JW, Reynolds PN, Hodge S, James AL, et al.
70. Duerr CU, McCarthy CD, Mindt BC, Rubio M, Meli AP, Pothlichet J, et al. Efficacy of azithromycin in severe asthma from the AMAZES randomised
Type I interferon restricts type 2 immunopathology through the regulation of trial. ERJ Open Res 2019;5:00056-2019.
group 2 innate lymphoid cells. Nat Immunol 2016;17:65-75. 90. Hao Q, Dong BR, Wu T. Probiotics for preventing acute upper respiratory tract
71. Ravanetti L, Dijkhuis A, Dekker T, Sabogal Pineros YS, Ravi A, Dierdorp BS, infections. Cochrane Database Syst Rev 2015. CD006895.
et al. IL-33 drives influenza-induced asthma exacerbations by halting innate 91. Lehtinen MJ, Hibberd AA, Mannikko S, Yeung N, Kauko T, Forssten S, et al.
and adaptive antiviral immunity. J Allergy Clin Immunol 2019;143: Nasal microbiota clusters associate with inflammatory response, viral load, and
1355-1370.e16. symptom severity in experimental rhinovirus challenge. Sci Rep 2018;8:11411.
72. Durrani SR, Montville DJ, Pratt AS, Sahu S, Devries MK, Rajamanickam V, 92. Fujimura KE, Demoor T, Rauch M, Faruqi AA, Jang S, Johnson CC, et al.
et al. Innate immune responses to rhinovirus are reduced by the high-affinity House dust exposure mediates gut microbiome Lactobacillus enrichment and
IgE receptor in allergic asthmatic children. J Allergy Clin Immunol 2012; airway immune defense against allergens and virus infection. Proc Natl Acad
130:489-95. Sci U S A 2014;111:805-10.
73. Busse WW, Morgan WJ, Gergen PJ, Mitchell HE, Gern JE, Liu AH, et al. 93. Sly PD, Galbraith S, Islam Z, Holt B, Troy N, Holt PG. Primary prevention of
Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children. severe lower respiratory illnesses in at-risk infants using the immunomodulator
N Engl J Med 2011;364:1005-15. OM-85. J Allergy Clin Immunol 2019;144. 870-2.e11.
74. Heymann PW, Platts-Mills TAE, Woodfolk JA, Borish L, Murphy DD, 94. Bacharier LB, Guilbert TW, Mauger DT, Boehmer S, Beigelman A,
Carper HT, et al. Understanding the asthmatic response to an experimental Fitzpatrick AM, et al. Early administration of azithromycin and prevention of
rhinovirus infection: exploring the effects of blocking IgE. J Allergy Clin severe lower respiratory tract illnesses in preschool children with a history of
Immunol 2020;146:545-54. such illnesses: a randomized clinical trial. JAMA 2015;314:2034-44.
75. Tam JS, Grayson MH. IgE and antiviral immune response in asthma. J Allergy 95. Stokholm J, Chawes BL, Vissing NH, Bjarnadottir E, Pedersen TM,
Clin Immunol 2017;139:1717. Vinding RK, et al. Azithromycin for episodes with asthma-like symptoms in
76. Chupp GL, Bradford ES, Albers FC, Bratton DJ, Wang-Jairaj J, Nelsen LM, young children aged 1-3 years: a randomised, double-blind, placebo-controlled
et al. Efficacy of mepolizumab add-on therapy on health-related quality of life trial. Lancet Respir Med 2016;4:19-26.
and markers of asthma control in severe eosinophilic asthma (MUSCA): a 96. Patrick DM, Sbihi H, Dai DLY, Al Mamun A, Rasali D, Rose C, et al.
randomised, double-blind, placebo-controlled, parallel-group, multicentre, Decreasing antibiotic use, the gut microbiota, and asthma incidence in chil-
phase 3b trial. Lancet Respir Med 2017;5:390-400. dren: evidence from population-based and prospective cohort studies. Lancet
77. Kavanagh JE, d’Ancona G, Elstad M, Green L, Fernandes M, Thomson L, Respir Med 2020;8:1094-105.
et al. Real-world effectiveness and the characteristics of a “super-responder” to 97. Smith SJ, Busby J, Heaney LG, Pfeffer PE, Jackson DJ, Yang F, et al. The
mepolizumab in severe eosinophilic asthma. Chest 2020;158:491-500. impact of the first COVID-19 surge on severe asthma patients in the UK.
78. Kavanagh JE, Hearn AP, Dhariwal J, d’Ancona G, Douiri A, Roxas C, et al. Which is worse: the virus or the lockdown? ERJ Open Res 2020:00768-2020.
Real-world effectiveness of benralizumab in severe eosinophilic asthma. Chest 98. Bloom CI, Cullinan P, Wedzicha JA. Asthma phenotypes and COVID-19 risk:
2021;159:496-506. a population-based observational study. Am J Respir Crit Care Med 2022;205:
79. Menzies-Gow A, Gurnell M, Heaney LG, Corren J, Bel EH, Maspero J, et al. 36-45.
Oral corticosteroid elimination via a personalised reduction algorithm in adults 99. Dhruve H, d’Ancona G, Holmes S, Dhariwal J, Nanzer AM, Jackson DJ.
with severe, eosinophilic asthma treated with benralizumab (PONENTE): a Prescribing patterns and treatment adherence in patients with asthma
multicentre, open-label, single-arm study. Lancet Respir Med 2022;10:47-58. during the COVID-19 pandemic. J Allergy Clin Immunol Pract 2022;10.
80. Kavanagh JE, Hearn AP, d’Ancona G, Dhariwal J, Roxas C, Green L, et al. 100-7.e2.
Benralizumab after sub-optimal response to mepolizumab in severe eosino- 100. Huang QS, Wood T, Jelley L, Jennings T, Jefferies S, Daniells K, et al. Impact
philic asthma. Allergy 2021;76:1890-3. of the COVID-19 nonpharmaceutical interventions on influenza and other
81. Busse WW, Bleecker ER, Fitzgerald JM, Ferguson GT, Barker P, Sproule S, respiratory viral infections in New Zealand. Nat Commun 2021;12:1001.
et al. Long-term safety and efficacy of benralizumab in patients with severe, 101. Fairweather SM, Chang CL, Mansell CJ, Shafuddin E, Hancox RJ. Impact of
uncontrolled asthma: 1-year results from the BORA phase 3 extension trial. COVID-19 pandemic restrictions on the cardio-respiratory health of New
Lancet Respir Med 2019;7:46-59. Zealanders. Respirology 2021;26:1041-8.
82. D’Ancona G, Kavanagh J, Roxas C, Green L, Fernandes M, Thomson L, et al. 102. Davies GA, Alsallakh MA, Sivakumaran S, Vasileiou E, Lyons RA,
Adherence to corticosteroids and clinical outcomes in mepolizumab therapy Robertson C, et al. Impact of COVID-19 lockdown on emergency asthma
for severe asthma. Eur Respir J 2020;55:1902259. admissions and deaths: national interrupted time series analyses for Scotland
83. O’Byrne PM, FitzGerald JM, Bateman ED, Barnes PJ, Zheng J, Gustafson P, and Wales. Thorax 2021;76:867-73.
et al. Effect of a single day of increased as-needed budesonide-formoterol use 103. Wee LE, Conceicao EP, Tan JY, Sim JXY, Venkatachalam I. Reduction in
on short-term risk of severe exacerbations in patients with mild asthma: a post- asthma admissions during the COVID-19 pandemic: consequence of public
hoc analysis of the SYGMA 1 study. Lancet Respir Med 2021;9:149-58. health measures in Singapore. Eur Respir J 2021;57. 2004493.

You might also like