You are on page 1of 9

Rhinovirus Respiratory Infections and Asthma

James E. Gern, MD

R
Viral infections, particularly respiratory illnesses hinoviruses are the most frequent cause of viral
caused by rhinovirus, are the most common cause of respiratory infections (VRIs), or the common
asthma exacerbations in children and contribute in cold, and have been shown to be responsible for
large part to asthma morbidity in adults. Epidemio- more than 50% of colds during the peak fall season.1–3 Of
logic studies and increasingly sophisticated viral de-
even greater clinical significance, however, is the growing
tection methodologies have helped to define the role
of rhinovirus as a potential causative agent in asthma
body of evidence implicating rhinovirus-induced respi-
exacerbations. Rhinovirus-induced lung disease is ratory infections as a cause of asthma exacerbations
multifaceted and can be characterized in terms of a in children and adults.3–5 Rhinovirus infections of the
variety of physiologic, immunologic, and viral pro- upper and lower respiratory tracts in individuals with
cesses. The precise direct and indirect mechanisms asthma have important implications for the epidemiol-
of viral contribution to exacerbations must still be ogy, associated risk factors, pathogenesis, and therapeutic
elucidated. Understanding them will have an impact management of this disease.
on the design of future treatment modalities. Am J
Med. 2002;112(6A):19S–27S. © 2002 by Excerpta Med- Epidemiology of Asthma and Rhinovirus
ica, Inc. Infections
Use of Reverse Transciption Polymerase
Chain Reaction to Identify Presence of Viruses
Early studies attempting to establish the connection be-
tween respiratory pathogens and wheezing or related
breathing disorders were limited by the use of relatively
insensitive standard cell culture techniques or methods to
detect increasing titers of virus-specific antibody. The de-
velopment of reverse transcription polymerase chain re-
action (RT-PCR) assays has helped to clarify the role of
viruses, and rhinovirus in particular, in asthma exacerba-
tions.3–5 Although RT-PCR detection is more sensitive
and rapid than culture, currently this method is primarily
a research tool for identification of rhinoviruses.
Rhinovirus Isolation in Epidemiologic Studies
in Children and Adults with Asthma
Using PCR and standard viral diagnostic tests, Johnston
et al3 reported that viral infection was associated with
80% to 85% of asthma exacerbations (as defined by re-
ported symptomatology and reduced peak expiratory
flow rates) in school-aged children aged 9 to 11 years who
had a history of asthma. In this study, viruses were de-
tected in 226 of 292 reported episodes of respiratory ill-
ness or reduced peak expiratory flow: picornaviruses
(which include the subgroups rhinoviruses and enterovi-
ruses) accounted for about 66% (147 of 226) of detected
viruses.
Nicholson et al5 found that among 138 adults who had
asthma, colds were reported in 80% (223 of 280) of epi-
sodes with symptoms of wheeze, chest tightness, or
breathlessness, and 89% (223 of 250) of colds were asso-
From the Department of Pediatrics, University of Wisconsin–Madison, ciated with asthma symptoms. Viruses were identified in
Madison, Wisconsin, USA. 57% of subjects with symptomatic colds and asthma ex-
Requests for reprints should be addressed to James E. Gern, MD,
Department of Pediatrics, University of Wisconsin–Madison, K4/918 acerbations, with exacerbations defined as decreased pul-
CSC, 600 Highland Avenue, Madison, Wisconsin 53792-9988. monary function (ie, greater than 50 L/min reduction in

© 2002 by Excerpta Medica, Inc. 0002-9343/02/$20.00 19S


All rights reserved. PII S0002-9343(01)01060-9
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

Table 1. Risk Factors for More Severe Rhinovirus Infections IgE and Atopy
● Low neutralizing antibody titers Researchers at the University of Virginia conducted a
● Asthma study to evaluate potential interactions between rhinovi-
—Allergy rus infections and age, atopic status, and eosinophil
—IgE markers. In a cross-sectional emergency department
—Cytokine production study of 70 children with wheezing and 59 control sub-
IFN-␥ jects,7 respiratory viruses were detected in more than
IL-5 80% of wheezing children 2 months to 16 years of age.
● Chronic lung diseases Rhinoviruses were the predominant pathogens in wheez-
● Extremes in age
ing children older than 2 years of age and accounted for
IFN ⫽ interferon; Ig ⫽ immunoglobulin; IL ⫽ interleukin. 71% of respiratory illnesses. After 2 years of age, the
Adapted from Clin Microbiol Rev.4 strongest odds for wheezing were observed among those
who had a positive PCR test for rhinovirus plus positive
radioallergosorbent tests, nasal eosinophilia, or elevated
mean peak expiratory flow rate). Again, rhinovirus was nasal eosinophil cationic protein (odds ratios ⫽ 17, 21,
the most frequent virus identified by PCR in these epi- and 25, respectively). These findings suggest that there
sodes, isolated from 76 (33%) of the 229 nose and throat may be synergistic interactions between rhinovirus infec-
swabs from patients with symptoms of asthma or colds, tions and allergic airway inflammation to increase the
or both. Together, these results demonstrate that a large risk of wheezing illnesses.
majority of emergent wheezing illnesses in both children
and adults can be linked to infection with common cold Cytokine Production
viruses such as rhinovirus. The pattern of cytokine production in patients with
asthma may also confer risk of an exacerbation in the
Rhinovirus Seasonality presence of rhinovirus infection. Recent experimental in-
Further evidence for rhinovirus involvement with oculation studies with the rhinovirus RV16 have related
asthma is based on the seasonality of exacerbations. Rhi- cytokine production to clinical symptoms and to the de-
novirus infections occur year round with peaks in fall and gree of viral replication.8,9
spring.1–3 Johnston et al6 conducted a time-trend analysis To test the hypothesis that rhinovirus-induced im-
comparing the seasonal patterns of respiratory infections mune responses influence the outcome of rhinovirus in-
and hospital admissions for asthma among adults and fections, 22 seronegative subjects who had either asthma
children. Strong correlations were found between the or allergic rhinitis were experimentally infected with
seasonal patterns of upper respiratory infections (URIs) RV16.8 Before the cold, the peripheral blood mononu-
and hospital admissions for asthma (r ⫽ 0.72; clear cell response to inoculation was assessed by mea-
P ⬍0.0001). This relationship was stronger for pediatric surement of levels of rhinovirus-induced IFN-␥ from the
(r ⫽ 0.68; P ⬍0.0001) than for adult admissions (r ⫽ culture supernatants. Peak rhinovirus shedding mea-
0.53; P ⬍0.01). URIs and admissions for asthma were sured during the most symptomatic part of the illness was
more frequent in the fall and spring when children went inversely related to IFN-␥ production. That is, subjects
back to school after vacations (P ⬍0.001). Rhinoviruses who produced adequate levels of IFN-␥ tended to shed
were the major pathogens implicated in these infections. less virus during their cold.
The authors speculated that school attendance and During the acute illness, mRNA for both IFN-␥ and in-
crowding of children in classrooms facilitate spread of the terleukin-5 (IL-5) was measured in the sputum to deter-
virus. mine the relationship between virus-induced Th1 and Th2
cytokine production and effects of viral infection.9 The ratio
Risk Factors for Rhinovirus-induced of IFN-␥ to IL-5 mRNA from sputum was also evaluated
Exacerbations of Asthma (Figure 1) as an immune-response indicator. Th1-like cyto-
A number of factors might place asthma patients at in- kines such as IFN-␥ are frequently produced in response to
creased risk for more severe rhinovirus infections result- viral infection, whereas Th2 cytokines such as IL-5 may pro-
ing in lower respiratory manifestations (Table 1).4 mote allergic inflammation. Therefore, a high IFN-␥–IL-5
Because bronchospasm during asthma results from mRNA ratio may represent a strong antiviral response. In
airway inflammation and hyperresponsiveness, the ef- this study, there was an inverse relationship between this
fects of infection on these conditions are of special inter- ratio and peak symptom scores. That is, individuals who
est. The finding that rhinovirus infections frequently tended to have a more Th1-like response (ie, patients with
exacerbate asthma suggests specific interactions between high IFN-␥ and low IL-5) tended to have fewer symptoms
viral infections and respiratory allergies or the disease during the peak of the cold.9
itself on airway biology and immunology. The duration of viral shedding was also assessed. The

20S April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A)
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

Figure 1. Sputum interferon-␥/interleukin-5 (IFN-␥/IL-5) ratio and respiratory symptoms. A strong Th1-like cytokine response
(high IFN-␥ and low IL-5) in the airway may play an important role in limiting the amount of viral replication and respiratory
symptoms in colds caused by rhinovirus. In this study, there was an inverse relation between this ratio and peak symptom scores. That
is, individuals who tended to have a more Th1-like response tended to have fewer symptoms during the peak of the cold. mRNA ⫽
messenger RNA. (Reprinted with permission from Am J Respir Crit Care Med.12)

IFN-␥ –IL-5 ratio was compared with the presence of Direct and/or Remote Effects of Viral Infection
rhinovirus in nasal secretions and sputum at 7 and 14 Direct and/or remote effects of viral infection may have a
days. At 14 days, rhinovirus RNA was found in the spu- role in provoking asthma.4 First, it is possible that the
tum of approximately 50% of individuals. In those who VRI, which clearly involves the upper airway, also in-
had cleared the virus at 14 days, there was a significantly volves lower airway epithelium and that viral replication
higher IFN-␥–IL-5 mRNA ratio compared with those in the lower airway causes inflammation and provokes
who had not cleared the virus.9 When considered to- bronchospasm, airway obstruction, and wheezing. It is
gether, these findings suggest that a predominant Th1 also possible that the virus is largely confined to the upper
response (ie, high IFN-␥–IL-5 ratio) is associated with a airway and that remote, indirect mechanisms provoke
more powerful antiviral response, more rapid clearing of asthma. These remote effects might be neural reflexes or a
the virus, and shorter duration of illness. Additional stud- virus-induced systemic immune stimulation that affects
ies are needed to determine whether reduced interferon the lung if there is pre-existing inflammation. A variety of
response to viral infection contributes to more severe immune factors are associated with lower airway symp-
lower respiratory manifestations in patients with asthma. toms in individuals with asthma.
Finally, because cytokine dysregulation has also been as-
sociated with allergic sensitization, it will be of interest to The Association Between Circulating
determine whether these risk factors for more severe Neutrophils and Symptoms
manifestations of rhinovirus infection are concordant Studies have noted that rhinovirus infections, like other
and associated or instead have independent effects. viral infections, cause a transient increase in the number
of circulating neutrophils that correspond with the peak
Pathogenesis of Viral Infection and symptoms. Peak symptom scores and the magnitude of
Asthma the change in neutrophils also are related.10,11 Our group
The studies discussed suggest that there may be certain reported on possible action of rhinovirus on certain local
pathologic mechanisms, particularly in asthma but also factors that could, in turn, act on the bone marrow to
in allergic disease in general, that may promote more se- increase peripheral blood neutrophils.9,10 Seronegative
vere VRIs in individuals with allergy or asthma. The un- individuals with allergic rhinitis or asthma were inocu-
resolved question is how viral infections, particularly lated with RV16. On days 1 and 2 after the inoculation, we
with rhinoviruses, provoke asthma. performed nasal lavage for viral cultures and cell counts

April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A) 21S
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

and obtained blood specimens for interleukin-8 (IL-8) responsiveness, as measured by PC20 (the concentration
and granulocyte colony-stimulating factor (G-CSF). IL-8 of histamine that causes a 20% reduction in lung func-
is known to be an important chemoattractant for neutro- tion), is an indicator of lower airway sensitivity to non-
phils, and IL-8 levels generally correlate with neutrophil specific irritants, which is a consistent feature of asthma.
increases in the nose. G-CSF is known to stimulate the In the RV16-treated patients there was an increased
production of neutrophils in the bone marrow. sensitivity to histamine at day 4, which was most pro-
G-CSF and IL-8 increased significantly in the nasal se- nounced in those with a severe cold (P ⫽ 0.01). In addi-
cretions during the acute phase of the cold, correlating tion, IL-8 levels increased in the RV16 group at days 2 and
with peak symptoms after experimental inoculation. Fur- 9 (P ⬍0.001), and the increase in nasal IL-8 at day 2 cor-
thermore, the amount of G-CSF in the nose correlated related significantly with the change in PC20 at day 4 (r ⫽
with serum G-CSF (correlation coefficient 0.95). Serum ⫺0.48, P ⫽ 0.04). These results suggest that more severe
G-CSF also significantly correlated with changes in the colds may be required to disturb lower airway physiology
number of blood neutrophils, suggesting that there is a and, by extension, to cause exacerbations of asthma. Fur-
systemic immune response to the viral infection and that thermore, this study was able to relate changes in an up-
increased mediators in the serum in turn act on the bone per airway inflammatory response (increased IL-8) to an
marrow. increase in lower airway sensitivity to histamine. Whether
There was also a positive correlation between serum this occurred by means of systemic immune stimulation
G-CSF and symptoms (r ⫽ 0.72, P ⬍0.05) and between or was an indicator of concurrent increases in lower air-
serum neutrophils and symptom scores (r ⫽ 0.86, P way IL-8 could not be determined by this study design.
⬍0.005). These findings suggest that rhinovirus infects Does rhinovirus infection of the lower airway cause
the respiratory epithelium locally, and the generation of exacerbations of asthma? Studies to determine the
G-CSF in the nose also increases levels in the serum (Fig- mechanisms by which viruses cause lower airway disease
ure 2). Then, serum G-CSF affects the bone marrow, re- are hampered by difficulty in sampling lower airways, and
sulting in an increase in neutrophil synthesis and release. efforts to culture rhinovirus from the lower airway after
G-CSF may also affect neutrophil attraction and recruit- experimental inoculation have met with limited success.4
ment into the airway by means of mechanisms that have However, studies using advanced technologies to detect
not yet been defined. virus, such as RT-PCR and in situ hybridization, have
The true nature of the relationship between rhinovirus detected rhinovirus in the lower airway.14,15
infection, increased neutrophil numbers, and the gener- Gern et al14 reported that the use of RT-PCR with
ation of respiratory symptoms remains to be determined. bronchoscopy consistently identified virus in patients af-
Are the increased neutrophil numbers merely a reflection ter inoculation. Furthermore, Papadopoulos et al15 used
of more severe infections, or do they participate in the in situ hybridization for RV16 on bronchial biopsy sam-
pathogenesis of respiratory symptoms and in asthma air- ples taken before infection (baseline), during the peak of
way obstruction? The answer to this question awaits the the infection, and 6 to 8 weeks after infection to obtain
development of specific inhibitors of neutrophil recruit- definitive evidence that rhinovirus infection occurs in the
ment (eg, IL-8 antagonist) or activation. human lung. These researchers showed that virus, at least
Rhinovirus Infections and Lower Airway under some conditions, can actively replicate in the lower
Physiology airway.
One of the cardinal features of asthma is airway hyperre- Effect of temperature on viral replication. Most rhino-
sponsiveness, which is defined as an increased propensity viruses preferentially grow at temperatures cooler than
to bronchoconstriction in response to such irritants as 37⬚ C (98.6⬚ F), and this has been cited as a potential
histamine, tobacco smoke, or allergens. It has long been barrier to growth in the lower airway. Direct thermal
recognized that VRIs can transiently increase airway re- mapping of the airways has revealed, however, that tem-
sponsiveness, and several studies have been conducted to peratures in the lower airways are conducive to the
determine the mechanisms for this effect.12 growth of rhinoviruses.
One such study, by Grunberg et al,13 examined the For example, McFadden et al16 found that during quiet
effect of RV16 infection on airway hypersensitivity to his- breathing at rest, at room temperature, the carina had a
tamine and on IL-8 in nasal lavage. This placebo-con- temperature of 32⬚ C inspiratory and 33⬚ C expiratory,
trolled, parallel study involved 27 nonsmoking, atopic which is perfect for rhinovirus replication (Figure 3). It
subjects with mild asthma who were given a dose of RV16 was not until the temperature probe reached peripheral
or placebo administered nasally.13 Symptom scores were airways that recorded temperatures were high enough to
measured throughout, and histamine challenges were inhibit rhinovirus replication. Additional studies have
performed at entry and on days 4 and 11 after inoculation demonstrated that rhinoviruses can grow in bronchial
to assess changes in airway responsiveness. Airway hyper- epithelial cells.17 Therefore, it appears that the environ-

22S April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A)
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

Figure 2. Rhinovirus (RV)-induced recruitment of polymorphonuclear leukocytes (PMN). The RV infects the respiratory epithe-
lium locally, causing an increase of granulocyte colony-stimulating factor (G-CSF) first in the nasal secretions and then in the blood.
Increased G-CSF affects the bone marrow, resulting in an increase in neutrophil synthesis and release. Interleukin-8 (IL-8), and
possibly G-CSF, then increase neutrophil recruitment into the airway.

Figure 3. Thermal mapping of the airways. In the large airways of the lung, inspiratory and expiratory temperatures are sufficiently
low to allow rhinovirus replication to occur. (Adapted from J Appl Physiol.16)

ment is appropriate, at least in the large airways, for rep- rus-infected epithelium releases a host of cytokines and
lication of rhinovirus. mediators that regulate airway inflammation. In particu-
Although the presence of rhinovirus in the lower air- lar, such factors as IL-8 (regulated upon activation, nor-
ways has been confirmed, the quantity present is unclear. mal T-cell expressed and secreted) and G-CSF are se-
It is uncertain whether levels of rhinovirus are sufficiently creted during acute colds. These cytokines increase the
high to cause local inflammation resulting in the effects of synthesis of leukocytes, recruit them into the airway, and
asthma. help to activate the inflammatory functions of neutro-
Cellular Mechanisms Contributing to phils, macrophages, and lymphocytes. Plasma leakage oc-
Rhinovirus-induced Asthma curs very early during the cold, which contributes to
The potential cellular mechanisms that contribute to rhi- edema and secretions in the airway that can cause ob-
novirus-induced asthma are illustrated in Figure 4.4 Vi- struction.18 After the plasma leakage, there is mucus hy-

April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A) 23S
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

Figure 4. Cellular mechanisms contributing to rhinovirus-induced asthma. Virus-infected epithelium releases cytokines and
mediators that can recruit additional cells to the airway. Plasma leakage occurs, which contributes to edema and secretions in the
airway that can cause obstruction. Then mucus hypersecretion causes thick, viscous secretions that could occlude the airway. Other
effects that may contribute to airway hyperresponsiveness and bronchospasm include neural stimulation by virus that could promote
bronchospasm and cough during these episodes. (Reprinted with permission from Clin Microbiol Rev.4)

persecretion, thick, viscous secretions that could occlude tion (acute cold BL2 vs BL1). Typically, this is the time
the airway. Other effects also may contribute to airway when changes in airway responsiveness are observed.13
hyperresponsiveness and bronchospasm. For example, Studies, such as this one, using experimental rhinovi-
neural stimulation and neurogenic inflammatory re- rus inoculation have begun to establish the sequence of
sponses to virus could result in bronchospasm and cough neutrophilic inflammation in different body compart-
during these episodes. ments. For example, the greatest changes in blood neu-
The neutrophil is the primary cell recruited to the up- trophils have been found 1 day after inoculation. Upper
per airway during VRIs, and recent studies indicate that airway symptoms and inflammatory responses tend to
this also occurs in lower airways. In a study by Jarjour et peak 2 to 3 days after experimental inoculation. Inflam-
al10 to assess changes in lower airways, 8 subjects who had mation in the lower airway and changes in airway respon-
a history of allergic asthma were experimentally inocu- siveness occur 4 to 7 days after inoculation. This suggests
that infection may progress from the upper airway to the
lated with RV16, and lower airway secretions were evalu-
lower airway and lead to asthma exacerbations.
ated using bronchoscopy and lavage. Before the cold,
bronchoscopy itself was associated with small increases in
lavage neutrophils (pre-cold BL1 vs BL2; Figure 5). How- Implications for Therapy
ever, during the acute cold there was a significant increase The immune response to VRIs contributes to airway ob-
in bronchial lavage neutrophils 96 hours after inocula- struction and respiratory symptoms by inducing produc-

24S April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A)
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

Figure 5. The effect of rhinovirus 16 (RV16) infection on bronchial lavage (BL) neutrophils (n ⫽ 8). Bronchoscopy and BL were
conducted twice before inoculation and twice after inoculation with RV16. Before inoculation, few neutrophils were found in BL
fluid. During the acute cold, there were few neutrophils 2 days after inoculation (acute cold BL1). However, 4 days after inoculation
(acute cold BL2), there was a significant increase in BL neutrophils from the initial values (acute cold BL1) and from both pre-cold
values (pre-cold BL1 and BL2). *P ⬍0.05 vs BL values (pre-cold and cold); †P ⬍0.05 vs BL2 pre-cold. (Reprinted with permission
from J Allergy Clin Immunol.11)

tion of proinflammatory cytokines and mediators. The rhinovirus colds developed in 1.3% of the treated group
inflammatory cascade, along with direct effects of the vi- and in 15.1% of the placebo group.19
rus, activates neural mechanisms to trigger airway hyper- The clinical utility of IFN-␣ is limited, however, in that
responsiveness and promote airway obstruction. It is it causes local nasal irritation, and it is generally ineffec-
likely that the immune response to the viral infection can tive if administration is delayed until after the onset of
enhance the pre-existing airway inflammation in asthma, symptoms. In addition, IFN-␣ administered to patients
leading to clinical exacerbations. This model suggests op- with chronic respiratory disease (including asthma) after
portunities for the design of therapeutic interventions close contact with people who have upper respiratory
(Figure 6).4 symptoms does not prevent cold-related lower respira-
Vaccination is not a viable therapeutic option, because tory symptoms.20
there are more than 100 serotypes of human rhinovirus. Several new antiviral agents have been developed for
However, several antiviral medications are in various the treatment of rhinovirus infection, including soluble
stages of development. Rhinoviruses bind to specific re- intercellular adhesion molecule–1 and capsid-binding
ceptors (90% of rhinovirus serotypes bind to intercellular agents, which either hinder rhinovirus binding to cellular
adhesion molecule–1) on the epithelial cell in order to receptors or inhibit uncoating of the virus and release of
replicate; the multiple generations of rhinovirus in the RNA inside the cell.21,22 In addition, inhibitors of rhino-
cell trigger secretion of cytokines and chemokines. Sev- virus 3C protease inhibit rhinovirus replication in vitro.23
eral antiviral agents have been developed to inhibit spe- Some of these compounds have been tested in clinical
cific steps in the virus replication cycle. trials and have demonstrable antiviral activity.24 Whether
Interferon-␣ (IFN-␣) was the first antirhinovirus these agents can be used at the first sign of a cold to pre-
agent to be tested in clinical trials. One such study com- vent asthma exacerbations or are better used prophylac-
pared intranasal ␣-2 IFN with placebo begun within 48 tically during peak rhinovirus seasons has yet to be deter-
hours of the onset of illness to prevent respiratory illness mined.
in healthy contacts of ill family members. Respiratory ill- Once viral replication occurs, epithelial cells and leu-
ness developed in 52 of 222 persons in the placebo group, kocytes in the airway produce pro-inflammatory cyto-
compared with 32 of 226 in the interferon group (P ⫽ kines, which are likely to contribute to common cold
0.02). In the 2-week period during and after spraying, symptoms and perhaps airway obstruction in asthma. In-

April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A) 25S
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

Figure 6. Potential therapeutic interventions include specific antiviral medications targeted to the rhinovirus, mechanisms that
interfere with signaling pathways for cellular activation, and agonists and antagonists that moderate the host inflammatory response.
(Reprinted with permission from Clin Microbiol Rev.4)

vestigators are now defining intracellular mechanisms by Virus-induced exacerbations of asthma tend to be re-
which viruses activate cytokine genes.4 Methods to block sistant to treatment with bronchodilators and inhaled
the signaling pathways for cellular activation are potential corticosteroids.27 Oral corticosteroids, although partially
therapeutic targets to reduce virus-induced inflamma- effective, can be associated with significant side effects.
tion and symptoms. Short bursts of oral corticosteroids These findings suggest that development of antirhinovi-
early during the course of a VRI in patients with asthma rus medications or specific inhibitors of rhinovirus-in-
have been effective in reducing the severity and duration duced inflammation could have a major impact on re-
of asthma exacerbations.25 However, although effective duction of the severity, and perhaps frequency, of asthma
in reducing lower airway symptoms, they do not lessen exacerbations.
the cold symptoms.26 Furthermore, systemic steroids
have undesirable side effects. Therefore, corticosteroids
may not be the optimal therapeutic approach. Because REFERENCES
1. Arruda E, Pitkäranta A, Witek TJ, Doyle CA, Hayden FG.
many of the involved inflammatory cells, cytokines, and Frequency and natural history of rhinovirus infections in
mediators have overlapping effects, therapeutic ap- adults during autumn. J Clin Microbiol. 1997;35:2864 –
proaches may need to inhibit multiple inflammatory 2868.
pathways in order to reverse the effects of rhinovirus on 2. Gwaltney JM Jr, Hendley JO, Simon G, Jordan WS Jr.
airway function. Rhinovirus infections in an industrial population. I. The oc-
currence of illness. N Engl J Med. 1966;275:1261–1268.
3. Johnston SL, Pattemore PK, Sanderson G, et al. Commu-
Conclusions nity study of role of viral infections in exacerbations of
asthma in 9 –11 year old children. BMJ. 1995;310:1225–
Rhinovirus infection can trigger severe episodes of
1229.
asthma exacerbations. The immune response to VRIs, 4. Gern JE, Busse WW. Association of rhinovirus infections
which is crucial to clearing the virus, may also contribute with asthma. Clin Microbiol Rev. 1999;12:9 –18.
to airway obstruction and respiratory symptoms. Al- 5. Nicholson KG, Kent J, Ireland DC. Respiratory viruses and
though the mechanisms for these effects are not fully elu- exacerbations of asthma in adults. BMJ. 1993;307:982–
cidated, they appear to be associated with the ability of 986.
respiratory viruses to induce the production of proin- 6. Johnston SL, Pattemore PK, Sanderson G, et al. The rela-
tionship between upper respiratory infections and hospital
flammatory cytokines and mediators. In the context of admissions for asthma: a time-trend analysis. Am J Respir
asthma, the immune response to rhinoviruses is likely to Crit Care Med. 1996;154:654 – 660.
enhance pre-existing inflammation in the lower airway, 7. Rakes GP, Arruda E, Ingram JM, et al. Rhinovirus and
leading to clinical exacerbations. respiratory syncytial virus in wheezing children requiring

26S April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A)
A Symposium: Rhinovirus Respiratory Infections and Asthma/Gern

emergency care. IgE and eosinophil analyses. Am J Respir 17. Schroth MK, Grimm E, Frindt P, et al. Rhinovirus replication
Crit Care Med. 1999;159:785–790. causes RANTES production in primary bronchial epithelial
8. Parry DE, Busse WW, Sukow KA, Dick CR, Swenson CA, cells. Am J Respir Cell Mol Biol. 1999;20:1220 –1228.
Gern JE. Rhinovirus-induced peripheral blood mononuclear 18. Igarashi Y, Skoner DP, Doyle WJ, White MV, Fireman P,
cell responses and outcome of experimental infection in Kaliner MA. Analysis of nasal secretions during experimen-
allergic subjects. J Allergy Clin Immunol. 2000;105:692– tal rhinovirus upper respiratory infections. J Allergy Clin
698. Immunol. 1993;92:722–731.
9. Gern JE, Vrtis R, Grindle KA, Swenson C, Busse WW. 19. Hayden FG, Albrecht JK, Kaiser DL, Gwaltney JM Jr. Pre-
Relationship of upper and lower airway cytokines to out- vention of cold by contact prophylaxis with intranasal alpha
come of experimental rhinovirus infection. Am J Respir Crit 2-interferon. N Engl J Med. 1986;314:71–75.
Care Med. 2000;162:2226 –2231. 20. Wiselka MJ, Nicholson KG, Kent J, Cookson JB, Tyrrell
10. Jarjour NN, Gern JE, Kelly EA, Swenson CA, Dick CR, DAJ. Prophylactic intranasal alpha2 interferon and viral ex-
Busse WW. The effect of an experimental rhinovirus 16 acerbations of chronic respiratory disease. Thorax. 1991;
infection on bronchial lavage neutrophils. J Allergy Clin 46:706 –711.
Immunol. 2000;105:1169 –1177. 21. Rotbart HA. Pleconaril treatment of enterovirus and rhino-
11. Levandowski RA, Weaver CW, Jackson GG. Nasal secre- virus infections. Infect Med. 2000;17:488 – 494.
tion leukocyte populations determined by flow cytometry 22. Huguenel ED, Cohn D, Dockum DP, et al. Prevention of
during acute rhinovirus infection. J Med Virol. 1988;25:423– rhinovirus infection in chimpanzees by soluble intercellular
432. adhesion molecule-1. Am J Respir Crit Care Med. 1997;
12. Folkerts G, Busse WW, Nijkamp FP, Sorkness R, Gern JE. 155:1206 –1210.
State of the art: virus-induced airway hyperresponsiveness 23. Dragovich PS, Prins TJ, Zhou R, et al. Structure-based
and asthma. Am J Respir Crit Care Med. 1998;157:1708 – design, synthesis, and biological evaluation of irreversible
1720. human rhinovirus 3C protease inhibitors. 3. Structure-ac-
13. Grunberg K, Timmers MC, Smits HH, et al. Effect of exper- tivity studies of ketomethylene-containing peptidomimet-
imental rhinovirus 16 colds on airway hyperresponsiveness ics. J Med Chem. 1999;42:1203–1212.
to histamine and interleukin-8 in nasal lavage in asthmatic 24. Turner RB. The treatment of rhinovirus infections: progress
subjects in vivo. Clin Exp Allergy. 1997;27:36 – 45. and potential. Antiviral Res. 2001;49:1–14.
14. Gern JE, Galagan DM, Jarjour NN, Dick EC, Busse WW. 25. Brunette MG, Lands L, Thibodeau LP. Childhood asthma
Detection of rhinovirus RNA in lower airway cells during prevention of attacks with short-term corticosteroid treat-
experimentally induced infection. Am J Respir Crit Care ment of upper respiratory tract infection. Pediatrics. 1988;
Med. 1997;155:1159 –1161. 81:624 – 629.
15. Papadopoulos NG, Bates PJ, Bardin PG, et al. Rhinovi- 26. Gustafson LM, Proud D, Hendley JO, Hayden FG, Gwaltney
ruses infect the lower airways. J Infect Dis. 2000;181:1875– JM. Oral prednisone therapy in experimental rhinovirus in-
1884. fections. J Allergy Clin Immunol. 1996;97:1009 –1014.
16. McFadden ER Jr, Pichurko BM, Bowman HF, et al. Thermal 27. Reddel H, Ware S, Marks G, Salome C, Jenkins C, Wool-
mapping of the airways in humans. J Appl Physiol. 1985; cock A. Differences between asthma exacerbations and
58:564 –570. poor asthma control. Lancet. 1999; 353:364 –369.

April 22, 2002 THE AMERICAN JOURNAL OF MEDICINE威 Volume 112 (6A) 27S

You might also like