ACP medicine, 3rd Edition

Infectious Disease

Respiratory Viral Infections

Frederick G. Hayden MD, FACP1

Professor of Internal Medicine and Pathology

Michael G. Ison MD, MSC2

Director

1University of Virginia School of Medicine

2Transplant Infectious Diseases, Northwestern University Feinberg School of Medicine

Frederick G. Hayden, M.D., F.A.C.P., has received grant or research support from Abbott Laboratories, Biota, Inc., Loders Croklaan, MedImmune, Inc., Perlan Therapeutics, Inc., Pfizer, Inc., and ViroPharma, Inc.; he is a consultant for Biota, Inc., Immuno-Rx, Inc., Perlan Therapeutics, Roche, and ViroPharma, Inc.; and he participates in the speakers' bureaus for MedImmune and Roche.

Michael G. Ison, M.D., M.Sc., is a consultant for Roche.

June 2006

The respiratory tract can be infected by a diverse group of viruses that produce syndromes ranging in severity from mild colds to fulminant pneumonias. Respiratory viral infections are a leading cause of morbidity, hospitalization, and mortality throughout the world; influenza and pneumonia were the most prevalent infectious causes of death during the 20th century in the United States.1 Respiratory viral infections are also the single most common cause of acute illness and physician visits in the United States and, ironically, a leading cause of prescriptions for antibiotics.2

Epidemiology

The high prevalence of respiratory viral infections results from the large number of infectious agents and serotypes and their efficiency of transmission from person to person; from incomplete immunity, with frequent reinfections by some respiratory viruses; and, in the instance of influenza viruses, from frequent changes in viral antigenicity. The frequency of viral respiratory illness is highest in children up to 4 years of age, gradually declines in teenagers, rises again in parents exposed to children, and is generally lowest in the elderly.3 In families, the average adult contracts two to four acute respiratory illnesses annually, about one quarter of which lead to physician contact. This age-related pattern reflects the gradual acquisition of immunity to some agents and the important role that children play in the transmission of infection. The major reservoir for most respiratory viruses is schoolchildren, who acquire infection in the classroom and introduce it into their homes.

Respiratory viral infections have strong seasonal patterns [see Table 1], but sporadic cases or nosocomial outbreaks can occur year-round. The presence or absence of a lipid-containing envelope affects viral survival in the environment. In temperate areas, the enveloped viruses, such as influenza virus, respiratory syncytial virus (RSV), and coronavirus, are characteristically prevalent during midwinter periods, whereas nonenveloped ones, such as rhinoviruses, are found most often in spring through fall. Although respiratory viruses spread from person to person, the relative importance of different routes depends on the virus. Large-droplet spread over short distances (< 1 m) appears common for many viruses. Viruses such as influenza virus and adenovirus spread efficiently in small-particle aerosols. Others, including rhinovirus and RSV, can spread by hand contact with contaminated skin and fomites, with subsequent inoculation onto the nasal mucosa or conjunctiva. A number of respiratory viruses, mainly RSV, influenza virus, and parainfluenza viruses (PIV), cause outbreaks of infection in closed populations, including hospitals, day care centers, and nursing homes. These outbreaks are associated with a high mortality in frail elderly or immunocompromised hosts.

Table 1 Epidemiologic Features of Principal Human Respiratory Viruses

Virus

Incubation Period (days)

Principal Mode of Spread*

Peak Seasonality

Nosocomial Pathogen

Adenovirus

4–7

Aerosol, direct

Summer

Yes

Coronavirus

2–5

Droplet

Winter

Minor

Influenza A and B viruses

1–4

Aerosol

Winter

Major

Parainfluenza virus

3–6

Droplet

Types 1 and 2, fall; type 3, spring and summer

Yes

Respiratory syncytial virus

2–8

Hands, droplet

Late fall to spring

Major

Rhinovirus

1–5

Hands, droplet

Early fall, late spring

Yes

*Multiple routes of transmission (i.e., direct, fomites, large droplets, and small-particle aerosols) are possible for most respiratory viruses.

Pathophysiology

Viruses that infect the respiratory tract can be divided into primary respiratory pathogens, whose transmission and replication are generally restricted to the human respiratory tract, and pathogens that affect the respiratory tract as part of a systemic or, sometimes, locally reactivated infection. The latter group includes three types of viruses: (1) those that cause pneumonia occasionally in healthy persons but more often in immunocompromised or pregnant persons, such as measles virus and varicella-zoster virus (VZV) [see 7:XXVI Herpesvirus Infections]; (2) those that primarily affect immunocompromised hosts, such as cytomegalovirus (CMV) [see 7:XXVI Herpesvirus Infections], herpes simplex virus (HSV) [see 7:XXVI Herpesvirus Infections], and, less frequently, human herpesvirus type 6; and (3) several viruses that cause uncommon but severe zoonotic infections, such as severe acute respiratory syndrome coronavirus (SARS-CoV), avian influenza virus, hantaviruses, and zoonotic paramyxoviruses (e.g., Hendra and Nipah viruses) [see 7:XXXI Viral Zoonoses]. The primary human respiratory viruses (adenovirus, coronavirus, influenza virus, PIV, human metapneumovirus [hMPV], RSV, and rhinovirus) share global distribution, mucosal sites of infection, and person-to-person transmission, but they differ in many important features, including viral composition, seasonality, pathogenesis of disease, and availability of vaccines and specific antiviral agents.

Pathogenesis

Although the extent of viral replication correlates well with severity of illness for most respiratory viruses, the pathogenesis of infection differs for the various groups. Rhinovirus and coronavirus infections are largely limited to the upper respiratory tract, whereas influenza virus, RSV, PIV, SARS-CoV, and adenovirus commonly also infect lower airways. With all respiratory viruses, progression to severe lower respiratory tract disease is more common in patients with impaired immune systems. Respiratory viruses may also produce intra-alveolar inflammatory exudate with hemorrhage, hyaline membrane formation, and differing amounts of epithelial damage. Tracheobronchitis and damage to the respiratory epithelium are typical of influenza virus. RSV infection in young infants is associated with epithelial damage and desquamation, bronchial edema with inflammatory cells, and plugging of small airways. In contrast, damage to the nasal mucosa is modest during rhinovirus or coronavirus colds. Disease manifestations are associated with respiratory tract damage caused by the virus and with host responses to infection, including specific immunologic responses, release of host inflammatory mediators, and neurogenic reflexes. For example, increased levels of histamine, leukotriene C4, eosinophilic cationic protein, and virus-specific IgE occur in the respiratory secretions of children with RSV bronchiolitis. Influenza is associated with increases in the levels of interleukin-6 (IL-6), interferon gamma (IFN-γ), and tumor necrosis factor-α (TNF-α) in the nasal mucosa and the blood; correlations exist between viral replication, symptom expression, and IL-6 production.4,5 Severe avian influenza infections in humans have been associated with elevated serum levels of TNF-α, soluble IL-2 receptor, IL-6, and IFN-γ.6 In persons with colds from rhinoviruses, nasal secretions have higher concentrations of bradykinin, IL-1, IL-6, and IL-8. In addition, respiratory viral infections may alter bacterial colonization patterns, increase bacterial adherence to respiratory epithelium, reduce mucociliary clearance, and alter bacterial phagocytosis by host cells. The impairment of host defenses may foster bacterial infection of areas that are normally sterile, such as the paranasal sinuses, middle ear, and lower respiratory tract.

For many respiratory viruses, the presence of neutralizing antibody in serum and respiratory secretions correlates with protection against infection. Generally, immunity is longer lasting and reinfection less common with virus groups that have many serotypes (e.g., rhinovirus and adenovirus) than in those with only a few serotypes. Although reinfection is frequent with PIV and RSV, severity generally decreases with each episode. In addition to humoral responses, cell-mediated immunity appears to be important for recovery from infection by certain respiratory viruses. In patients who have undergone bone marrow or solid-organ transplantation and in other highly immunocompromised hosts (e.g., patients with acute leukemia or AIDS), many respiratory viruses can cause prolonged infection and severe pulmonary disease, graft dysfunction, and increased mortality.7

Diagnosis

CLINICAL MANIFESTATIONS

In general, respiratory viruses cause acute, spontaneously resolving illnesses, although involvement of the lower airways and certain complications (e.g., otitis media, sinusitis, and exacerbations of asthma or chronic obstructive pulmonary disease [COPD]) are common. Clinical diagnosis is difficult because there are a number of respiratory viruses that can cause a variety of overlapping clinical syndromes. Consequently, clinical diagnosis is accurate only under certain circumstances, such as during an influenza epidemic or outbreaks of RSV bronchiolitis. The limited correlation between virus and syndrome emphasizes the importance of rapid virologic techniques for making a specific etiologic diagnosis, particularly in hospitalized or severely affected patients [see Table 2]. Improvement in the accuracy of clinical diagnosis will require data from community-based surveillance programs, such as those that track which respiratory viral pathogens are circulating within a community.

Table 2 Laboratory Methods for Diagnosis of Respiratory Viral Infections

Virus

Specimen Source

Time to Isolation in Cell Culture (days)

Rapid Detection Methods and Molecular Techniques

Common Serologic Assays

Adenovirus

Respiratory tract, throat, conjunctiva, urine, stool, blood

2–10

EIA, IF, RT-PCR, IC

CF

Coronavirus

Nasopharynx

Not routine

Not routine, RT-PCR, IF

Not routine (ELISA)

Influenza A and B viruses

Nasopharynx, throat, lower respiratory tract

2–5

EIA, IF, neuraminidase assay, RT-PCR

CF, HAI, ELISA

Parainfluenza virus

Nasopharynx, lower respiratory tract

3–14

IF, RT-PCR

CF, HAI, ELISA

Respiratory syncytial virus

Nasopharynx, lower respiratory tract

3–14

EIA, IF, RT-PCR

CF, ELISA, NT

Rhinovirus

Nasopharynx

2–7

Not routine, RT-PCR

Not routine (NT)

CF—complement fixation  EIA—enzyme immunoassay  ELISA—enzyme-linked immunosorbent assay  HAI—hemagglutination inhibition  IC—immunochromatography
IF—immunofluorescence  NT—neutralization  RT-PCR—reverse transcriptase-polymerase chain reaction

LABORATORY TESTS

Most respiratory viruses can be isolated from nose and throat swabs, nasal washes or aspirates, sputum, and other lower respiratory samples. Tests done on nasopharyngeal samples taken early in illness provide the most sensitive results, although early samples in SARS may be falsely negative. In addition, some respiratory viruses are slow growing [see Table 2], and most are present in lower titers in adults than in children, so samples from adults may require longer periods for isolation or have lower yields on rapid diagnostic tests. Because temperature fluctuations and freezing may cause loss of infectivity, samples should be transported at refrigerator temperatures (4° C) or on wet ice. Cell culture inoculation (shell vials) followed by antigen detection has a sensitivity of 80% or greater at 1 to 2 days for adenovirus, influenza virus, PIV, and RSV.8

Immunofluorescence testing of exfoliated respiratory cells and enzyme immunoassay for viral antigens are reasonably sensitive, specific, and rapid techniques for detecting some respiratory viruses, particularly RSV and influenza virus.9 Nucleic acid amplification techniques appear to be the most sensitive direct detection methods for most viruses, including SARS-CoV. However, false negative results are possible (during the first days of infection, SARS-CoV may be undetectable even by polymerase chain reaction), and positive results must be confirmed to ensure accuracy. For simultaneous detection of respiratory viruses (influenza virus types A and B; RSV; and PIV types 1, 2, and 3), multiplex reverse transcriptase PCR (RT-PCR) has good sensitivity,10 and a commercial assay is available. Real-time quantitative RT-PCR tests offer a sensitive means of detection of many respiratory viruses and, potentially, of monitoring responses to treatment.

Serologic diagnosis requires paired serum specimens in most instances and is intrinsically slower than direct methods. A variety of serologic techniques are used to measure antibodies, including neutralization, hemagglutination inhibition, complement fixation, and enzyme-linked immunosorbent assay (ELISA). Measurement of complement-fixation antibodies is generally less sensitive than the other methods and does not provide a serotype-specific diagnosis. Immunocompromised hosts often fail to develop diagnostic increases in antibody titers. The combined use of cultures, antigen detection, nucleic acid detection, and serology provides the most comprehensive approach for identification of important respiratory viral pathogens in hospitalized adults.

Clinical Syndromes

COMMON COLDS

Diagnosis

The common cold is generally a mild illness of the upper respiratory tract, primarily affecting the nasopharynx and paranasal sinuses. Rhinoviruses, which cause about 50% of colds, and coronaviruses, which cause 10% to 20%, are the most important pathogens. RSV, PIV, influenza virus, adenovirus, and some enteroviruses also cause colds.

Adults with rhinovirus colds typically have prominent upper respiratory tract symptoms (i.e., sneezing, nasal discharge, nasal obstruction, sore or scratchy throat, and cough) and often have headache but little fever or few systemic complaints other than malaise. Physical findings are nonspecific and include nasal discharge and mucosal erythema. Although generally absent in adults, lower respiratory tract manifestations (e.g., cough, sputum, and wheezing) do occur in about 60% of cases in elderly persons, and lower respiratory tract illness and fever are common in young children infected with rhinovirus.

The differential diagnosis of the common cold includes allergic rhinitis, bacterial nasopharyngitis in infants, and, in rare instances, nasal diphtheria or bacterial nasal infection in adults. Because nearly 50% of colds last longer than 1 week and 25% last up to 2 weeks, it is often unclear whether a complicating viral or bacterial sinusitis has occurred. More than 80% of patients with uncomplicated colds have reversible sinus abnormalities demonstrable on computed tomography,11 and sinusitis is an inherent part of rhinovirus colds. Fever, purulent nasal drainage, cough productive of purulent sputum, increasing symptoms or malaise after 1 week, localized facial pain, or maxillary toothache suggests a bacterial sinusitis.12 Otitis media complicates approximately 2% of colds in adults and 5% of colds in children.13

Treatment

Treatment of the common cold is directed toward specific symptoms. Saline nose drops may be helpful for relief from obstructing secretions, particularly in young children, and warm saline gargles reduce sore throat. Topical decongestants such as phenylephrine and the longer-acting oxymetazoline provide prompt relief of nasal obstruction but may be associated with rebound congestion and possibly throat irritation. Oral pseudoephedrine also provides partial relief of nasal obstruction but may cause anxiousness and insomnia in some patients. The oral decongestant phenylpropanolamine was removed from the market because of increased risk of hemorrhagic stroke. It is uncertain whether topical or oral vasoconstrictors alter the risk of bacterial complications. First-generation oral antihistamines, such as chlorpheniramine and clemastine, reduce sneezing and, to a lesser extent, rhinorrhea, but they are often associated with excess sedation. Nonsedating antihistamines such as loratadine and astemizole are not effective for colds in nonallergic persons. Intranasal ipratropium, a quaternary anticholinergic, reduces cold-associated rhinorrhea by about 30% and may also reduce sneezing, but excess nasal drying may result in blood-tinged mucus.14 Cough suppressants with codeine or dextromethorphan may be used if the cough is severe or disturbs sleep, but they should be used cautiously in patients with underlying chronic obstructive disease. Specific antiviral therapy is not currently available. Two intranasal antiviral agents from different classes, the 3-C protease inhibitor ruprintrivir and the capsid inhibitor pleconaril, are being investigated. Although effective in treating rhinovirus colds, oral pleconaril was not approved by the Food and Drug Administration because of concerns about safety and drug interactions, most notably with oral contraceptives.15 A number of other remedies (e.g.,Echinacea preparations, zinc lozenges or nasal gels, and hot-air inhalation) are of unproven or doubtful value.16,17,18

PHARYNGITIS AND LARYNGITIS

Pharyngitis usually is associated with the same viruses that cause common colds and occurs concurrently with the common cold, but it is also often a prominent complaint of persons with adenovirus and influenza virus infections. Enteroviruses are also important causes of fever and pharyngitis during the summer and early fall.

Diagnosis

Pharyngitis may be associated with characteristic clinical findings, such as palate vesicles and ulcers caused by coxsackievirus-induced herpangina, acute ulcerative stomatitis and pharyngitis caused by HSV, or exudative pharyngitis related to Epstein-Barr virus mononucleosis. Pharyngitis occurs with primary HIV infection and may be associated with mucosal erosions and lymphadenopathy. Pharyngitis caused by group A streptococci and other bacteria (anaerobes, Corynebacterium diphtheriae, Chlamydophila pneumoniae, andMycoplasma pneumoniae) are generally not associated with acute rhinorrhea [see 7:XIX Bacterial Infections of the Upper Respiratory Tract].

Treatment

Treatment for most cases of viral pharyngitis is symptomatic. However, HSV stomatitis, particularly in immunocompromised patients, should be treated with specific antiviral therapy (e.g., acyclovir, famciclovir, or valacyclovir).

Hoarseness caused by laryngitis frequently occurs during acute viral infections and is more common during infections caused by viruses that invade the lower airways. Treatment is primarily voice rest.

ACUTE BRONCHITIS

Diagnosis

Acute bronchitis manifests as a severe or prolonged cough that is usually nonproductive or productive of scant mucoid sputum. Bronchitis can follow infection by any of the respiratory viruses and is a hallmark of influenza. Viral infections commonly cause exacerbation of preexisting airway disease, including chronic bronchitis, emphysema, asthma, and cystic fibrosis. Rhinoviruses are the most frequently implicated viral pathogens.19 HSV can cause bronchospasm and sometimes ulcerative tracheobronchitis in immunocompromised persons and, less often, in immunocompetent persons.

Cough production in acute bronchitis results from direct viral damage to the respiratory mucosa, release of inflammatory mediators, stimulation of airway irritant receptors, and increased production and decreased clearance of respiratory secretions. Infection may also enhance airway hyperreactivity, characterized by increased sensitivity to cold air and pollutants such as smoke.

The differential diagnosis of acute viral bronchitis includes bacterial, mycoplasmal, chlamydial, and pertussis infections, as well as noninfectious entities such as asthma.

Treatment

Treatment of acute bronchitis in otherwise healthy persons is directed at suppression of the cough with codeine or dextromethorphan. However, these agents should be used cautiously in patients with underlying chronic obstructive disease. Antibiotics are generally not indicated.20

INFLUENZA SYNDROME

Diagnosis

The influenza syndrome is characterized by the rapid onset of constitutional and respiratory tract symptoms. Constitutional symptoms, which include fever, chills, prostration, muscle aches, and headache, tend to dominate during the first several days of illness. Respiratory complaints are sore throat, coryza, hoarseness, and, especially, cough. Persistent nonproductive cough, easy fatigability, and asthenia are common in the second week of illness.

Epidemic influenza A and B viruses are the principal causes of the influenza syndrome, but other respiratory viruses, including RSV, PIV, adenovirus, and, sometimes, rhinovirus, can cause influenzalike illness.

Treatment

Treatment for the influenza syndrome is usually symptomatic and includes bed rest, oral hydration, antipyretics, and antitussives. Specific antiviral therapy for influenza virus infection is available [see Influenza Virus, below].21,22,23 Fever should be treated in patients in whom it would contribute to disease, such as children with previous febrile convulsions or patients with preexisting cardiac disease. Aspirin should be avoided in pediatric patients because of its association with Reye syndrome.

CROUP

Diagnosis

Croup, or laryngotracheobronchitis, is a characteristic pediatric syndrome characterized by brassy or barking cough, inspiratory stridor, dyspnea, and hoarseness. Symptoms are often preceded by several days of upper respiratory tract illness and typically become worse at night. Croup is seen primarily in children younger than 5 years and is associated most closely with PIV infections and less often with RSV, hMPV, influenza viruses, adenoviruses, and rhinoviruses. Measles virus is an important cause of severe croup. Acute spasmodic croup is characterized by recurrent attacks precipitated by viral infections, allergies, and, possibly, other factors. The differential diagnosis of croup includes foreign-body aspiration or trauma, bacterial epiglottitis or tracheitis, localized abscess, and angioedema.

Inflammation of the larynx and trachea with subglottic narrowing causes localized obstruction; this obstruction is enhanced by inspiration, because of the extrathoracic location of the obstruction and the compliance of the airway walls in children. In addition, inflammatory changes occur throughout the lower respiratory tract. Symptoms usually abate in 3 to 4 days.

Treatment

Most cases of croup can be managed at home, with moist air. Correction of hypoxemia is essential for hospitalized patients. Aerosolized racemic epinephrine provides transient benefit for those with persistent stridor. Systemic glucocorticoids appear to be effective in moderating the severity of illness and shortening hospitalization; inhaled steroids are beneficial in less severe cases.

BRONCHIOLITIS

Diagnosis

Bronchiolitis is an acute inflammatory disorder of the small airways that is characterized by airway obstruction with wheezing, hyperinflation, and atelectasis. It is associated most closely with RSV infection, which is detected in about 75% of infants hospitalized with bronchiolitis, and less often with rhinoviruses, hMPV, PIV, influenza viruses, and adenoviruses. The peak incidence is in infants from 2 to 6 months of age, and over 80% of cases occur in the first year of life. Premature infants and young children with underlying pulmonary conditions are at high risk for RSV bronchiolitis or pneumonia. After several days of coryza and possibly fever, patients experience acute respiratory distress with wheezing, cough, and, often, inspiratory rales; apnea may occur in babies. Characteristic radiologic features are atelectasis and signs of hyperinflation. In mild cases, bronchiolitis symptoms resolve within several days, but prolonged or severe disease may occur in premature infants or those with underlying cardiopulmonary or immunodeficiency disorders. The major differential diagnosis is asthma, which is uncommon in children younger than 2 years.

The bronchiolitis syndrome results from viral destruction of the respiratory epithelium and associated peribronchiolar mononuclear inflammation, which is perhaps in part immunologically mediated. In infants, because of their disproportionately narrow airways, bronchiolar obstruction causes distal collapse or air trapping and abnormal gas exchange.

Treatment

Correction of hypoxemia is the most important aspect of managing RSV bronchiolitis. Bronchodilators, glucocorticoids, and routine antibiotics are not of proven value. Aerosolized ribavirin therapy is available for selected infants hospitalized with RSV bronchiolitis or pneumonia, but it is an expensive intervention of uncertain value.24

REACTIVE AIRWAY DISEASE EXACERBATION

Infection with respiratory viruses, most commonly rhinoviruses,19 can precipitate exacerbations of chronic respiratory illness. Respiratory viral infections, half of which are rhinovirus infections, can worsen asthma in patients of all ages but are associated with up to 85% of asthma exacerbations in schoolchildren.25 Young children hospitalized with wheezing illness are in most cases infected with rhinoviruses, RSV, hMPV, or PIV. Exacerbations of COPD often result from respiratory viral infections. Approximately 25% of hospitalizations for COPD exacerbations have been associated with a documented respiratory tract viral infection; this percentage increases to 45% of COPD patients admitted between December and March.26

PNEUMONIA

Viral infections have been associated with at least 10% to 15% of community-acquired pneumonias in hospitalized adults and with as many as half of cases in hospitalized infants.27 The relative importance of different viruses depends heavily on the geographic location of the outbreak, the season, and patient age and immune status. Influenza A and, less often, influenza B viruses are the most common causes of community-acquired viral pneumonia in adults and the elderly. In children younger than 3 years, RSV accounts for about 50% of all hospitalizations for pneumonia28; PIV type 3 is also prominent. PIV, hMPV, adenovirus, rhinovirus, and non-SARS respiratory coronavirus infections have been implicated as causes of pneumonia in children and adults.

Respiratory viruses reach the lung by contiguous spread from the upper respiratory tract or inhalation of small-particle aerosols. Depending on the agent, nonrespiratory viruses infect the lung parenchyma by hematogenous dissemination (e.g., CMV, VZV, HSV, and measles) or, less often, by contiguous spread from the tracheobronchial tree (e.g., HSV).

SARS

SARS is a unique form of viral pneumonia. In contrast to most other viral pneumonias, upper respiratory symptoms are usually absent in SARS, although cough and dyspnea occur in most patients. Typically, patients present with a nonspecific illness manifesting fever, myalgia, malaise, and chills or rigors; watery diarrhea may occur as well [see SARS Coronavirus, below].29

Diagnosis

The clinical and radiographic features of most viral pneumonias are not distinctive. Unless a typical exanthem of measles or varicella is present, identification of a specific pathogen requires a careful epidemiologic history and appropriate virologic studies. The clinical and radiologic appearance of viral pneumonia is often indistinguishable from that of bacterial infection, and mixed infections are common. Differentiation between pure viral pneumonias, mixed viral-bacterial pneumonias, and viral infections of the tracheobronchial tree with complicating bacterial pneumonia is usually not possible on clinical grounds.

When taking the history, the clinician should ask about risk factors for exposure, including illness in contacts, institutional residence during an outbreak of illness, travel history, and contact with animals associated with zoonotic transmission. Knowledge of local patterns of viral circulation (e.g., epidemic influenza) and of the patient's immune status are also important in diagnosis. SARS should be suspected in any patient with radiographically confirmed pneumonia who has one or more epidemiologic risk factors for the syndrome [see SARS Coronavirus,below].30

Treatment

Management of viral pneumonia is primarily supportive; ventilatory support has improved the prognosis for patients with extensive viral pneumonia. Specific antiviral therapy for respiratory viruses is limited, and there are no controlled data regarding the use of these agents in pneumonia. Antiviral agents are available to treat influenza31,32 [see Influenza Virus, below]. Inhaled ribavirin is available for RSV and other viral infections but is of uncertain value; it has been used intravenously for adenovirus, hantavirus, measles, PIV, and influenza virus infections. Intravenous acyclovir is effective in HSV or VZV pneumonia.

Pneumonia in Immunocompromised Hosts

Immunocompromised hosts, particularly patients undergoing organ transplantation, experiencing chemotherapy-induced neutropenia and immunosuppression, or with HIV-related immunosuppression, are at risk for reactivation of latent viruses (e.g., HSV, CMV, or adenovirus) or acquisition of community respiratory viruses. Bronchoalveolar lavage yields viruses in up to half of immunocompromised patients with acute pneumonia.33 CMV is the most frequently recovered, accounting for more than 80% of isolates; fewer than 10% of isolates are HSV or various respiratory viruses.34 In immunosuppressed patients, influenza viruses, PIV, RSV, hMPV, rhinovirus, and adenoviruses can cause severe pneumonia35 that is often nosocomially acquired and usually associated with a preceding upper respiratory tract illness.7 It seems prudent to delay chemotherapy or transplantation in the presence of an upper respiratory tract illness.

Intravenous acyclovir is recommended for patients with HSV or VZV pneumonia. Ganciclovir with immunoglobulin appears to be effective in reducing the mortality associated with CMV pneumonia in bone marrow transplant (BMT) recipients and has been used as monotherapy in other patient groups.36 Aerosolized ribavirin and RSV-specific antibodies (RSV immunoglobulin [RespiGam] or palivizumab), if given early, may be effective for preventing progression of RSV pneumonia or treating established disease in such patients [see Respiratory Syncytial Virus, below]. Aerosolized and intravenous ribavirin have been used for adenovirus37 or severe measles. Intravenous cidofovir has been effective in the management of severe adenoviral infection in immunocompromised patients but may cause nephrotoxicity.38

Infections Caused by Specific Agents

ADENOVIRUS

Adenoviruses cause a variety of respiratory tract syndromes, ranging from pharyngoconjunctival fever (often contracted while swimming in contaminated water) to severe pneumonia in infants, immunosuppressed patients, and, uncommonly, healthy adults. Epidemics of acute respiratory disease and pneumonia caused by adenovirus have been recognized for decades in United States military recruits and are expected to continue, given the cessation of immunization programs in the military. However, a new manufacturer for adenovirus vaccine has been identified, and vaccination of military personnel is expected to resume.39

Classification and Pathogenesis

Adenoviruses are medium-sized (65 to 80 nm), nonenveloped, double-stranded DNA viruses, of which 49 antigenic types (divided into six subgroups) are associated with human infections. The protein coat of the virus is composed of hexagonal and pentagonal subunits (capsomers) with long fibers at each vertex, which are the sites of host cell attachment. Type-specific antigens, which give rise to neutralizing antibody, are present on the hexons and fibers of the capsid. The hexons also contain a group-specific antigen with cross-reactivity among most adenoviruses. Only about one half of serotypes cause disease. Immunity correlates with the presence of type-specific neutralizing antibody.

Epidemiology and Transmission

Adenovirus infections may be endemic or epidemic. Lower-numbered serotypes routinely infect infants and young children, although types 3, 4, and 7 can also be acquired later in life and are typically associated with epidemic respiratory disease. In addition to transmission by direct contact with respiratory secretions or infectious aerosols, fecal-oral transmission occurs. Infection may be acquired by pharyngeal or conjunctival inoculation from contaminated water. Adenovirus infection occurs throughout the year but is often recognized during the summer in association with outbreaks of pharyngitis or bronchitis. Adenovirus probably accounts for about 5% of total acute respiratory infections in civilian adults. Nosocomial transmission, including clusters of pneumonia in long-term care facilities and large outbreaks of epidemic keratoconjunctivitis, is well documented.40,41 Persistent infections occur in the tonsils and gastrointestinal (GI) tract, and prolonged viral shedding is common in immunocompromised hosts and in the GI tract of children.

Diagnosis

Clinical features

The incubation period for adenovirus infection of the respiratory tract is usually 4 to 7 days. Adenovirus respiratory infection typically causes a moderate to severe pharyngitis, which is sometimes exudative, and tracheobronchitis. Fever and systemic symptoms are often prominent, and rhinitis, cervical adenitis, and follicular conjunctivitis are common. Pharyngoconjunctival fever, usually associated with serotypes 3, 4, and 7, tends not to be accompanied by lower respiratory tract symptoms. In infants and young children, infections also cause a pertussislike syndrome, croup, and bronchiolitis.

Adenovirus pneumonia is similar to other viral pneumonias; it usually begins with pharyngitis, rhinitis, fever, and cervical adenopathy—conjunctivitis may or may not be present—and then spreads to involve the lower airways and lungs. Bilateral interstitial or alveolar infiltrates are common, although focal consolidation or effusion also occurs.

Laboratory findings

The presence of cells with large basophilic, intranuclear inclusions (smudge cells) in lower respiratory tract biopsy specimens may provide early histopathologic diagnosis. A variety of serotypes have been recovered from stool, urine, and lower respiratory tract specimens of immunocompromised patients.42 Rapid methods of virus detection include enzyme immunoassay (EIA), immunofluorescence, antigen immunochromatography,43 and RT-PCR. Quantitative PCR to measure blood adenoviral DNA levels is emerging as a useful marker for predicting the risk of progression of disease and for monitoring responses to antiviral therapy in immunocompromised patients.

Complications

Acute complications, which occur mainly in children and in immunocompromised hosts, are leukopenia, lymphocytopenia, thrombocytopenia, rhabdomyolysis, disseminated intravascular coagulation, renal failure, and bacterial infection. Survivors may acquire restrictive lung disease or bronchiectasis; children in particular are at risk for bronchiolitis obliterans. Mortality in BMT recipients with adenovirus pneumonia is as high as 60%.44

Adenoviruses are also associated with extrarespiratory infections, including epidemic keratoconjunctivitis; hepatitis and genitourinary infections in immunosuppressed hosts; myocarditis; arthritis; meningoencephalitis; and, especially in children, hemorrhagic cystitis, mesenteric adenitis, intussusception, and gastroenteritis. Adenovirus commonly complicates hematopoietic stem cell transplantation, especially in children, and is often associated with invasive disease, which is associated with reduced survival.45

Treatment

Antiviral therapy of proven value is not available, but intravenous ribavirin, ganciclovir, cidofovir, and immunoglobulin have been used to treat severe adenoviral infections. In invasive adenoviral infections, including pneumonia, mortality appears to be lower in patients who have received cidofovir than in those who have received intravenous ribavirin, but further studies are needed.33,46,47,48 Treatment of invasive adenoviral infections is otherwise supportive.

Prevention

Live attenuated vaccines for adenovirus types 4 and 7 were used in the military but are not licensed for civilian use.39,40 Other control measures are swimming-pool chlorination, careful hand washing, and, in the instance of nosocomial keratoconjunctivitis, use of gloves and proper sterilization of equipment.

CORONAVIRUSES

Classification

Coronaviruses are moderate-sized (100 to 150 nm), enveloped, single-stranded RNA viruses named for their distinctive club-shaped surface projections, which resemble a crown. Three major groups of human respiratory corona-viruses (group 1 [229E and NL63 or NH], group 2 [OC43 and HKU1], and group 3 [SARS-CoV]) have been found to cause disease in humans.49

Epidemiology and Transmission

Coronaviruses are the second most frequently documented cause of common colds. Coronaviruses account for 4% to 15% of cases of acute respiratory disease annually and as many as 35% during peak periods. Most coronavirus infections occur in winter and early spring, but infections have been detected throughout the year. Immunity to infection is incomplete, and reinfection is common. SARS-CoV causes a clinical syndrome unique among the coronaviruses and is discussed separately [see SARS Coronavirus, below].

Diagnosis

Clinical features

The incubation period of coronavirus colds ranges from 2 to 5 days, longer than that of rhinovirus colds. Infections produce a typical coryzal illness that is indistinguishable from colds caused by other viruses. Lower respiratory tract manifestations include pneumonia in children, military recruits, and possibly immunocompromised hosts; exacerbation of asthma in children; and exacerbation of chronic airway disease in adults. In one study, a strong association between HCoV-NL63 infection and croup was reported.50 Reported associations with GI disease, including necrotizing enterocolitis in infants, are controversial. A possible association between NL63 and Kawasaki disease51 requires confirmation, as other studies have not found such an association.52

Laboratory findings

Virus isolation is insensitive; infections are usually identified by serology or by detection of coronavirus RNA. Culture of virus in human embryonic trachea cells is the most sensitive cell culture system for recovering human respiratory tract coronaviruses but is impractical.

Treatment

Effective antivirals or vaccines are unavailable, although intranasal IFN-α is protective against experimental coronavirus colds. Treatment is symptomatic.

SARS CORONAVIRUS

Epidemiology

SARS-CoV is a novel coronavirus identified as the cause of a clinical syndrome, SARS, that was first recognized as an unusual atypical pneumonia in Guangdong Province, China, in November 2002. Over the ensuing 6 months, the virus spread to multiple areas of the world and resulted in over 8,000 illnesses and 774 deaths. China, Hong Kong, Singapore, Taiwan, and Toronto, Canada, were most severely affected. The virus is of animal origin, but the primary reservoir remains uncertain. It possibly originated from Himalayan palm civets found in live-animal markets, but it is infectious for a wide variety of species, including civets, raccoon dogs, ferrets, cats, mice, and monkeys.

Molecular epidemiology has shown that SARS viruses from outbreaks in Hong Kong, Vietnam, Singapore, Toronto, and Taiwan are clonally related to the index case from Hong Kong, whereas viruses from Guangdong Province are genetically more diverse. It is possible that different strains of SARS-CoV have different characteristics, including efficiency of transmission and virulence. Future SARS outbreaks may differ from the outbreak of 2002–2003. Unlike diseases from other respiratory coronaviruses, SARS does not have any recognized seasonality.

Transmission

SARS-CoV spreads efficiently between humans. The primary mode of transmission appears to be through close personal contact: infectious droplets from the cough or sneeze of an infected person travel through the air a short distance and land on respiratory or, possibly, conjunctival mucous membranes or are deposited there indirectly. Other possible modes of spread include fomites, with hand contamination and subsequent self-inoculation; small-particle aerosols; and perhaps fecal-oral transmission. Transmission may be amplified by certain aerosol-generating procedures, such as endotracheal intubation, bronchoscopy, and treatment with aerosolized medications. Recently, modeling was used to substantiate airborne spread of SARS as the cause of the large outbreak at the Amoy Gardens housing complex in Hong Kong.53 Person-to-person spread has been especially common in health care and hospital settings, but transmissions have occurred in homes and hotels, in the workplace, on aircraft, and in taxis. Laboratory transmission of SARS-CoV and presumed acquisition from animals have also occurred.54 No transmission of SARS has been documented before the onset of symptoms or more than 10 days after resolution of fever.

Diagnosis

SARS is diagnosed on the basis of epidemiologic evidence, clinical suspicion, and diagnostic testing. The current case definition of SARS, as well as the most current recommendations on diagnosis, can be found at the Centers for Disease Control and Prevention Web site (http://www.cdc.gov/ncidod/sars).

Epidemiologic evidence

SARS should be included in the differential diagnosis of pneumonia in patients of all ages who have one or more of the following risk factors:

  • A history of recent travel to mainland China, Hong Kong, or Taiwan or close contact (within 10 days before symptom onset) with ill persons with a history of recent travel to such areas
  • Employment in an occupation that involves particular risk for SARS-CoV exposure, including health care with direct patient contact or work in a laboratory that contains live SARS-CoV
  • Association with a cluster of cases of atypical pneumonia without an alternative diagnosis during periods of known SARS circulation

Clinical features

The incubation period of SARS typically ranges from 2 to 7 days but may be as long as 10 days or even, in rare cases, 14 days. Patients present with fever, myalgia, malaise, and chills and may have a nonproductive cough. Other upper respiratory symptoms are usually absent. Fever may be absent, particularly in the elderly, in the initial phase of the illness. Diarrhea may occur.

Laboratory findings

Lymphocytopenia is common in SARS, as are elevations in D-dimer levels and activated partial thromboplastin time. Chest radiographs show evidence of pneumonia in 60% to 100% of patients. Distinctive radiographic findings include a predominant peripheral lung involvement; initial unilateral (typically, in the right lung) focal air-space opacity; and absence of cavitation, lymphadenopathy, and pleural effusion.55High-resolution CT scans are abnormal in up to 67% of patients with normal chest radiographs.

Current diagnostic tests for SARS include RNA detection by RT-PCR, virus culture, and serology. Virus is detectable in respiratory secretions (from the nasopharynx, throat, nose, and lower respiratory tract), as well as in feces, serum, and plasma. Initial RT-PCR techniques were associated with poor sensitivity, particularly in early infection, but improved extraction methods and real-time RT-PCR assays have been developed that allow for more sensitive testing during the first days of infection.56 Any positive test result must be confirmed by testing another clinical sample, reextracting and testing the original specimen, or retesting with an assay that targets different parts of the viral genome. Measurement of specific antibodies at 21 days or later after illness onset using whole-virus immunofluorescence or ELISA remains the gold standard for retrospective confirmation of SARS-CoV infection, because a negative RNA assay does not rule out infection.

Treatment

Initial treatment in patients with suspected SARS is with empirical antimicrobials directed against the typical pathogens in community-acquired pneumonia. There have been no prospective studies of antiviral agents in the treatment of SARS. Ribavirin, lopinavir-ritonavir, IFN-α, and traditional Chinese medicines were used during the 2002–2003 outbreak. In vitro studies, animal testing, and limited human experience suggest that IFN-α has activity against SARS-CoV; IFN-β also has efficacy in vitro.57 Ribavirin is not active in vitro at clinically achievable concentrations and was associated with significant toxicity and no obvious antiviral effects in SARS. Neutralizing antibodies, including recently described humanized monoclonal antibodies, are active in animal models58 and are promising for prophylaxis and perhaps early treatment.

Lung biopsies in patients with SARS suggest a possible immune component; the samples reveal diffuse alveolar damage with desquamation of pneumocytes, inflammatory infiltrates, edema, and hyaline membrane formation. Consequently, high-dose methylprednisolone has been used to modify the disease course and has produced short-term reductions in fever and infiltrates. However, steroid therapy offers uncertain overall benefit and poses such risks as enhanced viral replication, superinfection, and late aseptic necrosis. Thymic peptides, intravenous immune globulin (IVIg), and plasma from patients in the convalescent phase of illness are likewise of unclear benefit. Neutralizing human monoclonal antibodies against S protein are active in experimental animal infection and may prove useful in prophylaxis or possibly early treatment.59

Prevention of exposure is the key to limiting future outbreaks. All patients with suspected SARS should be isolated for at least 10 days; in patients with documented SARS, isolation should be continued for at least 10 days after defervescence. Quarantine of contacts has been used in some outbreaks. Current recommendations regarding appropriate infection control practices can be found at the CDC Web site (http://www.cdc.gov/ncidod/sars). In health care settings, precautions against transmission through airborne respiratory droplets and contact should be strictly enforced, with enhanced airborne precautions used during aerosol-generating procedures.

Prognosis

One third of patients with SARS defervesce and improve clinically. The remainder experience progressive illness, with diarrhea and increasing tachypnea, shortness of breath, and abnormal findings on pulmonary examination and radiography; 20% to 30% of patients require admission to an intensive care unit, usually for mechanical ventilation, and approximately 10% succumb. Death typically results from respiratory failure, multiple organ failure, sepsis, or intercurrent medical illness. More severe disease and higher mortality occur in older adults (≥ 60 years) and those with underlying medical conditions.

HUMAN METAPNEUMOVIRUS

hMPV is a respiratory pathogen that causes infections ranging from colds to severe bronchiolitis and pneumonia. The first metapneumovirus associated with human infection, hMPV was discovered in 2001 when viruses that had been isolated from children presenting with RSV-like illnesses failed to be identified by standard techniques. After random-primer RT-PCR was used, the virus was identified as a new metapneumovirus related to turkey tracheitis virus. Serologic studies demonstrated that by 5 years of age, all children were seropositive and that sera originally collected in 1958 were also positive for hMPV antibodies.60 Two genetic clusters of hMPV that correspond to two different serotypes of hMPV have been recognized on every continent.61

Epidemiology

Infections by hMPV occur worldwide and year-round, with a winter predominance. The season peaks between December and February in the Northern Hemisphere, with a longer season in temperate climates. hMPV can be isolated from 5.5% to 6.5% of children hospitalized with respiratory symptoms. The peak incidence is between 4 and 6 months of age, with most children being younger than 5 years. Coinfection with hMPV and RSV has been documented frequently, and several cases of concomitant hMPV and SARS-CoV infection have been documented.

Asymptomatic or mild illness appears to be much more common with hMPV. hMPV has been detected in 20% of ambulatory young children presenting with respiratory illness in whom no other cause was found. Most cases occurred between December and April.62 In infirm elderly patients, the incidence is 6.5%. The incidence of hMPV in general-community persons presenting with influenzalike illness is only 2%, whereas hMPV can be isolated from 3% to 6.6% of the general community with acute respiratory illnesses.

Diagnosis

Clinical features

hMPV causes clinical syndromes indistinguishable from RSV, including bronchiolitis, croup, asthma exacerbation, and pneumonia. Studies attempting to differentiate hMPV from other respiratory viruses have found that hMPV infection appears to occur at a slightly older age and causes slightly milder symptoms than RSV. Infections in adults manifest as colds, influenzalike illness, bronchitis, exacerbations of underlying airway disease, or, uncommonly, pneumonia. hMPV infections in immunocompromised patients can be severe and have been associated with at least three deaths.

Laboratory tests

RT-PCR has become the standard method of detecting hMPV in respiratory samples. Methods that use primers targeted at the polymerase (L) and fusion (F) protein genes have shown adequate sensitivities. The presence of antibodies is not diagnostic by itself, although seroconversion or a greater than fourfold rise in titer is indicative of recent infection. The virus can be grown in cell culture, but the cytopathic effect may not be seen for up to 14 days.

Treatment

In vitro data suggest that ribavirin has activity against hMPV similar to that against RSV. Likewise, neutralizing antibody titers against hMPV in IVIg are similar to titers against RSV. Clinical studies are required to determine the possible clinical efficacy of ribavirin and IVIg.63

INFLUENZA VIRUS

Classification and Pathogenesis

Influenza viruses belong to the orthomyxovirus family and consist of types A, B, and C. These medium-sized (80 to 120 nm), enveloped, single-stranded RNA viruses contain eight gene segments (seven for influenza C). The segmented nature of the genome allows reassortment of RNA segments between two influenza viruses during dual infection and facilitates antigenic variation. Surface glycoprotein spikes possess either hemagglutinin or neuraminidase activity. Hemagglutinin mediates cell attachment and fusion of virus and cell membranes. By cleaving terminal sialic acid residues and destroying the receptors recognized by hemagglutinin, neuraminidase promotes release of virus from infected cells and spread within the respiratory tract.

Influenza A viruses are further classified into subtypes on the basis of their surface proteins (15 hemagglutinins and nine neuraminidases are recognized in animal influenza viruses). Three A hemagglutinin subtypes (H1, H2, and H3) and two neuraminidases (N1, N2) have caused extensive human infections. Influenza C viruses have seven gene segments and lack a neuraminidase. Influenza viruses are named by the type; location of isolation; isolate number; year of recovery; and, for influenza A type viruses, the subtype (e.g., A/Texas/36/91[H1N1]).

The surface glycoproteins induce host humoral and cellular immune responses and are responsible for the changing antigenicity of influenza viruses. Two major types of antigenic change can occur: drift and shift. Antigenic drift refers to relatively minor changes in hemagglutinin and, less often, neuraminidase antigenicity that occur frequently (usually every few years) and sequentially in the setting of selective immunologic pressure in the population. Drift results from point mutations of the corresponding RNA segment. Antigenic shift occurs only in influenza A viruses and results from acquisition of a new gene segment for hemagglutinin with or without one for neuraminidase. This may occur through genetic reassortment during dual infections with human and animal influenza type A viruses; by the reintroduction of a virus that has not circulated recently in the human population; or by direct transmission to humans of an animal influenza virus that is capable of efficient human-to-human transmission.

Aquatic fowl are the main reservoir for influenza A viruses in nature, although some subtypes also infect other species, including swine, horses, and marine mammals. Swine are susceptible to infection with viruses from both birds and humans and may serve as a so-called mixing vessel, providing an opportunity for the generation of new pathogenic viruses.

Epidemiology and Transmission

Influenza viruses A and B cause annual outbreaks of illness affecting approximately 5% to 10% of adults, with higher rates in children. In the United States, influenza causes an average of over 36,000 deaths64 and 130,000 to 170,000 hospitalizations during each epidemic.21The appearance of a new strain for which most of the population lacks immunity can result in worldwide outbreaks, or pandemics. Pandemic strains are associated with global spread over months and with high attack rates. Three such pandemics occurred during the 20th century; the most severe was the Spanish influenza pandemic in 1918 and 1919, which caused 20 to 40 million deaths worldwide and over 500,000 deaths in the United States.

Epidemic influenza occurs annually in temperate areas, typically between the months of December and March in the Northern Hemisphere, and follows the reintroduction of virus each year. Influenza activity usually occurs in May through August in the Southern Hemisphere and can be year-round in the tropics. Regional outbreaks caused by a particular strain are usually short (6 to 8 weeks), although successive waves of infection by different influenza viruses can occur. Influenza activity in the community is marked by increased medical contacts for febrile respiratory illness, increased absenteeism from school and the workplace, subsequent increased hospitalizations for pneumonia and other cardiopulmonary disorders, and increased mortality. Pneumonia hospitalizations increase by two to five times in high-risk patients. Persons 65 years of age and older constitute nearly 50% of excess hospitalizations and over 85% of deaths from influenza.65

Influenza viruses are transmitted principally via large and small aerosolized particles. Direct transmission of influenza A virus to humans from animals has been documented.66 Although animal-to-human transmission has typically been from swine, direct transmission from birds has resulted in human infections by avian H5, H7, and H9 viruses. At present, an unprecedented epizootic of avian influenza A subtype H5N1 (A/H5N1) virus is causing mortality in some migratory waterfowl and many millions of poultry in Asia, with recent expansion to the Middle East, the Balkans, sub-Saharan Africa, and Europe. Initially recognized in Hong Kong when poultry die-offs and six deaths among 18 affected humans occurred, the virus has been recovered from healthy migratory birds that presumably have been the vehicle for geographic dissemination and infection of domestic poultry. In addition, multiple sublineages of A/H5N1 viruses have become established in poultry in Southeast Asia.67 Sporadic human infections have occurred in China, Thailand, Vietnam, Cambodia, Indonesia, Turkey, and Iraq. Direct contact with infected domestic poultry is the principal route of human infection, although exposure to contaminated environmental sources may account for some infections. Isolated instances of human-to-human transmission have been reported, but there is no evidence of sustained human transmission.

Recognized cases of avian influenza have manifested as severe viral pneumonia and sometimes multiorgan dysfunction. Fatality for patients with confirmed A/H5N1 infection averages about 50% (ranging from 30% to over 80%),68 with many fatalities occurring in otherwise healthy, young individuals.

Avian influenza A subtype H9N2 virus infection has been documented in several children, and a large outbreak of avian influenza A subtype H7N7 virus in the Netherlands was associated with at least one death.69 H7 subtype viruses have also caused outbreaks, predominantly of conjunctivitis, in British Columbia, Canada,69 and infected an immunocompromised man in New York. Influenza A/H5N1 and other avian viruses pose a pandemic threat70; the likelihood of such a pandemic depends on the extent to which viral adaptation through either reassortment with human viruses or mutational evolution might facilitate efficient human-to-human transmission of the virus.71

Diagnosis

Clinical features

The incubation period for influenza virus is short, averaging 2 days (range, 1 to 4 days). Classic influenza is distinguished by abrupt onset of prominent systemic symptoms, including fever, chills, headache, myalgia, malaise, and anorexia. Fever usually lasts for an average of 3 days in adults. Sore throat, dry cough, photophobia, and pain on eye movement occur frequently early in the illness. Mild conjunctivitis, clear nasal discharge, pharyngeal injection, and small, tender cervical lymph nodes are also common. As systemic illness abates, respiratory symptoms become more apparent. The most troubling respiratory symptom is protracted cough, which results from viral tracheobronchitis. Airway hyperactivity and abnormalities in pulmonary function may last from weeks to several months in previously healthy persons. Exacerbations of asthma and other types of preexisting airway disease are often severe. Infections may be subclinical or cause milder illness, including colds. In comparison with typical human influenza, patients with A/H5N1 infections have a higher frequency of diarrhea (perhaps related to direct gastrointestinal infection in some patients) and lower frequency of upper respiratory complaints during the initial phase of illness.68 Most patients develop cough and dyspnea by the end of the first week of illness, and they present with radiographically documented pneumonia. Uncommon presentations have included febrile diarrhea and encephalopathic illness.72

Primary influenza virus pneumonia is a heterogeneous condition, ranging from mild disease with patchy infiltrates to rapidly fatal infection. Severe pneumonia generally accounts for 2% of influenza-associated pneumonia, but during pandemics, it can account for up to 20%; severe pneumonia is the usual presentation of avian A/H5N1 infection. Human influenza A viruses cause more than 90% of cases.73 As many as 40% of patients with influenza pneumonia have no prior underlying disease. Pneumonia usually begins with typical influenza, followed within 1 to 3 days by rapidly progressive dyspnea, cyanosis, diffuse rales, and wheezing. Pleuritic chest pain and blood-tinged sputum or frank hemoptysis occur. Patients with influenza virus pneumonia have a high mortality.

Laboratory tests

Multiple rapid assays are commercially available in the United States to detect influenza A and B antigens or neuraminidase activity; some of these assays differentiate between influenza A and B, and several can be performed by clinical personnel at the point of care [see Table 2]. The specificity of these assays is good to excellent, but the sensitivity varies between approximately 60% and 90%, depending on the sample type, the age of the patient, and the duration of the illness.70,74 Diagnosis can also be made by viral culture or by RT-PCR; virus cultures should be performed only in high-containment laboratories. Rapid antigen assays are less sensitive in detecting A/H5N1 infections than are RT-PCR assays. Nucleic acid amplification assays can detect viral RNA in respiratory tract, blood, and stool samples; pharynx sampling appears to have a higher yield than nasal sampling.68 The Influenza A/H5 (Asian lineage) Virus Real-time RT-PCR Primer and Probe Set can detect the presence of the H5 strain in suspected influenza samples within 4 hours. Further testing can identify the specific H5 subtype.75 When viral pneumonia is present, Gram stain of sputum shows few to many polymorphonuclear leukocytes, but only rarely does it show bacteria. The chest radiograph shows bilateral infiltrates that may be in the form of diffuse interstitial infiltrates, perihilar pulmonary edema, or dense opacifications. On blood counts, leukocytosis with a left shift is variably present.

A definitive diagnosis of influenza can have a significant impact on medical management. In a pediatric population, detection of influenza A antigen resulted in a decrease in antibiotic use, a decrease in duration of antibiotic use in hospitalized patients, and an increase in antiviral use.76

Complications

Secondary bacterial pneumonia should be suspected when fever, increasing cough, and sputum production develop after several days of improvement. Streptococcus pneumoniae is the most common bacterial pathogen, but Staphylococcus aureus, including community-acquired methicillin-resistant strains, causes up to 25% of cases and is associated with high mortality. S. aureus and certain other bacteria produce proteolytic enzymes that activate influenza hemagglutinin and enhance viral replication. Haemophilus influenzae and S. pyogenes are also recognized as causes of bacterial complications. Bacterial infections of the sinuses and ears are frequent. Toxic-shock syndrome and invasive meningococcal disease have also been known to complicate influenza.

Uncommon complications are myositis with rhabdomyolysis, renal failure, disseminated intravascular coagulopathy, myocarditis, pericarditis, myelitis, Guillain-Barré syndrome, and Reye syndrome. Neurologic complications, including encephalopathy or encephalitis, are unusual and occur mainly in children.66,77

Treatment

A variety of antiviral agents are available for treatment of influenza [see Table 3]. The M2 inhibitors amantadine and rimantadine are active against influenza A only, although almost all A/H3N2 viruses and many human isolates of avian A/H5N1 viruses are resistant to these agents.70,78,79 Oral amantadine and rimantadine reduce the duration of fever and symptoms of uncomplicated influenza A virus infection by 1 to 2 days and provide more rapid overall functional recovery. Effectiveness in preventing complications or treating severe illness in hospitalized patients is uncertain. Resistant viruses may arise during treatment and are transmissible to close contact as well as in the community.80 Side effects of amantadine are GI upset and minor, reversible central nervous system symptoms (e.g., insomnia, dizziness, and difficulty with concentration). Rimantadine appears to be as effective as amantadine and is associated with a lower risk of CNS side effects.

Table 3 Agents Used to Prevent and Treat Influenza123,124

Drug

Dosage

Efficacy for Documented Influenza

Dosage Adjustment

Prophylaxis*

Treatment

Prophylaxis

Treatment

State

Dosage

Amantadine

100 mg b.i.d.

100 mg b.i.d.

63% efficacy95

Shorter duration of fever and symptoms

CCr 30–50
CCr 15–30
CCr < 15
Hemodialysis
Elderly

100 mg q.d.
100 mg q.o.d.
100 q.wk.
100 mg q.wk.
≤ 100 mg q.d.

Rimantadine

100 mg b.i.d. or 200 mg q.d.

100 mg b.i.d. or 200 mg q.d.

72%–83% efficacy95

Shorter duration of fever and symptoms

Severe hepatic dysfunction
Elderly
CCr < 10

100 mg q.d.
100 mg q.d.
100 mg q.d.

Zanamivir

2 puffs q.d.

2 puffs b.i.d.

67%–84% efficacy96

1 day faster recovery from illness (up to 2.5 day improvement in high-risk patients)
Reduction in severity of illness, number of nights of disturbed sleep, and use of relief medications
Reduction in complications that require antibiotic
More rapid return to normal function

No dosage adjustments

Oseltamivir

75 mg q.d.

75 mg b.i.d.

87% efficacy96

24 to 36 hr faster recovery from illness
Reduction in severity of illness, number of nights of disturbed sleep, and use of relief medications
Reduction in complications that require antibiotic

CCr < 30
Age 1–13 yr < 15 kg 15–23 kg 23–40 kg > 40 kg

Treatment, 75 mg q.d.
Prophylaxis, 75 mg q.o.d.
30 mg b.i.d. 45 mg b.i.d. 60 mg b.i.d. 75 mg b.i.d.

*Duration of prophylaxis depends on the clinical circumstances: 7–10 days after close contact for postexposure prophylaxis; 2 wk after immunization of an adult; 4–6 wk after a community outbreak; and at least 1 wk (preferably 2 wk) after the last case in outbreak control.
First dose should be given within 48 hr of the onset of illness to be effective. Therapy should be continued for 5 days.
The Food and Drug Administration has not approved zanamivir for this indication.
CCr—creatinine clearance

The neuraminidase inhibitors, zanamivir and oseltamivir, are active against influenza A and B viruses,32 including the avian A/H5N1 strain. Resistance to oseltamivir may emerge during treatment; it may also occur in immunocompromised hosts and in some patients with A/H5N1 infection that is associated with treatment failure.81,82 Zanamivir is administered by inhalation and may, in rare cases, cause bronchospasm, which can be severe. Oseltamivir is administered orally and is associated with self-limited GI upset in about 10% to 15% of treated patients; this can be reduced by taking the medication with food. Treatment of acute uncomplicated influenza in adults with inhaled zanamivir or oral oseltamivir provides symptomatic relief, reduces time to functional recovery, and decreases the likelihood of lower respiratory tract complications leading to antibiotic use.22,23 In children 1 to 12 years of age, oseltamivir provides symptomatic relief and also reduces the likelihood of otitis media.21 Oseltamivir treatment has been associated with reduced risk of hospitalization in both previously healthy and high-risk or elderly adults.83

Influenza pneumonia

Treatment of influenza virus pneumonia is primarily supportive. Improvements in assisted ventilation techniques have raised the survival rate above 50%, although pulmonary fibrosis develops in some patients. M2 inhibitors, NA inhibitors, and aerosolized or I.V. ribavirin, which is active against influenza A and B viruses, have been used with uncertain benefit. Combination therapies (e.g., an M2 inhibitor plus a neuraminidase inhibitor for influenza A) show enhanced activity in animal models. Although unstudied, the use of neuraminidase inhibitors, either alone or in combination with other agents, seems reasonable in the treatment of influenza virus pneumonia.

Prevention

Chemoprophylaxis

In the past, antiviral chemoprophylaxis with amantadine or rimantadine was about 70% to 90% effective in preventing illness caused by influenza A virus. Unfortunately, the emergence of resistance to M2 inhibitors in the majority of circulating influenza A viruses suggests that amantadine and rimantadine should not currently be used for prophylaxis.78,84

The neuraminidase inhibitors are also effective for prophylaxis of influenza A and B infections. Chemoprophylaxis can provide several weeks' protection to patients immunized after influenza A activity has begun; it can be given throughout the influenza season to those who cannot receive influenza vaccine (e.g., those with egg allergy) or who are unlikely to respond to the vaccine. Chemoprophylaxis also can be used for protection of high-risk persons at times when the epidemic strain diverges significantly from the vaccine antigens. Both inhaled zanamivir85,86 and oral oseltamivir87,88 prevent influenza when used for seasonal prophylaxis or after exposure (e.g., for family or nursing home contacts), but only oseltamivir has been approved by the Food and Drug Administration for this indication. Oseltamivir has been found to be safe for the treatment of children as young as 1 year, as well as for prophylaxis in these patients89; inhaled zanamivir has been found to be safe in children as young as 5 years. Of note, both the M2 and the neuraminidase inhibitors may reduce the efficacy of live attenuated influenza vaccine, if the vaccine and the antiviral agent are given together.

Immunization

Influenza vaccines are available in two forms: (1) an intramuscular preparation containing formalin-inactivated virus and purified surface antigen and (2) an intranasal spray containing live attenuated viruses.21 Both are made from egg-grown virus. Vaccine composition is reviewed annually and adjusted to reflect changes in antigenicity and anticipated circulation of viral strains; current vaccines contain one influenza B and two influenza A (H1 and H3 subtypes) antigens.21 The efficacy of these vaccines is approximately 70% to 90% in young adults, especially when the vaccine antigen and the circulating strain are closely matched. Immunization in healthy working adults is associated with fewer upper respiratory illnesses and fewer visits to physicians' offices.90,91 The intranasal vaccine is currently approved only for healthy persons 5 to 49 years of age; it appears to be less effective in the elderly but possibly superior to inactivated vaccine in young children. Immunization with inactivated vaccine reduces influenza-related hospitalizations, including acute cardiovascular events and COPD exacerbations,92 and reduces mortality in ambulatory elderly patients by 40% to 60%.93 Wide-scale immunization of schoolchildren may reduce influenza-related mortality in older adults.94 Although inactivated vaccine is less effective in infirm elderly patients, it provides partial protection against pneumonia and death.

The highest priority for vaccination should be given to persons at increased risk for influenza-related complications and their contacts [seeTable 4]. Administration of the inactivated vaccine is associated with soreness at the injection site in as many as one third of recipients, but fever or systemic reactions are uncommon in adults. Influenza vaccination does not adversely affect CD4+ T cell counts or accelerate progression to AIDS or death in HIV-infected patients.95

Table 4 Indications for Influenza Vaccination*21

PERSONS AT INCREASED RISK FOR COMPLICATIONS

    All adults 65 yr of age and older
    Residents of nursing homes and other long-term care facilities that house persons of any age who have chronic medical conditions
    Adults and children with chronic cardiopulmonary disorders, including asthma
    Adults and children who have required regular medical follow-up or hospitalization during the preceding year because of chronic metabolic diseases (including diabetes), renal dysfunction, hemoglobinopathies, or immunosuppression (including immunosuppression caused by medication or by HIV)
    Children and teenagers (6 mo to 18 yr of age) who are receiving long-term aspirin therapy and therefore may be at risk for Reye syndrome
    Women who will be pregnant during the influenza season
    Children 6–23 mo of age
PERSONS 50–64 YR OF AGE
PERSONS WHO CAN TRANSMIT INFLUENZA TO THOSE AT HIGH RISK
    Physicians, nurses, and other personnel in both hospital and outpatient care settings, including medical emergency response workers (e.g., paramedics and emergency medical technicians)
    Employees of nursing homes and long-term care facilities who have contact with patients or residents
    Employees of assisted-living and other residences for persons in groups at high risk
    Home care providers for persons at high risk
    Household contacts of high-risk persons

*Both the inactivated influenza vaccine and live, attenuated influenza vaccine (LAIV) can be used to reduce the risk of influenza. Healthy persons 5–49 yr of age who are not contacts of severely immunosuppressed persons can receive either LAIV or inactivated influenza vaccine; all other persons should receive inactivated influenza vaccine. In addition to persons for whom annual influenza vaccination is recommended, influenza vaccine should be given to any person who wishes to reduce the likelihood of becoming ill with influenza, if vaccine availability permits.
Vaccination is recommended for persons 50–64 yr of age because of the increased prevalence of persons with high-risk conditions in this group.

The intranasal vaccine causes transient coryza and sore throat. Vaccine viruses are recoverable from nasal samples in low titers for up to 1 week in adults and are genetically stable. Although viral transmission is rare between children and has not been documented in adults to date, vaccine recipients should avoid close contact with highly immunocompromised hosts (e.g., stem cell transplant recipients) for 1 week after immunization.21 Veterinary vaccines for A/H5N1 infection are available. Various strategies are being pursued to develop human vaccines, although the H5 hemagglutinin appears to be poorly immunogenic in humans, such that at least two doses, preferably combined with an adjuvant agent, will be required.

PARAINFLUENZA VIRUSES

PIV infections are the most commonly recognized cause of croup, accounting for up to 75% of cases with a documented viral etiology, and they are a leading cause of lower respiratory tract disease resulting in hospitalization of infants. Infection with PIV induces partial immunity that moderates disease severity; reinfection in older children and adults produces milder respiratory illness and colds.

Classification and Pathogenesis

Human PIVs are medium-sized (120 to 150 nm), enveloped RNA viruses belonging to the Paramyxoviridae family and are classified into four antigenically stable types (1, 2, 3, and 4); type 4 is further classified into subtypes A and B. PIV replicates in epithelial cells of the upper and lower respiratory tract, and antibodies to the two major envelope surface glycoproteins—namely, the hemagglutinin-neuraminidase protein and the fusion protein—confer protection against infection. These proteins are necessary for attachment of virus to host cell receptors and membrane-fusing activity. PIV type 1 and type 2, which cause croup and laryngitis, probably replicate principally in the major airways, whereas PIV-3 causes lower respiratory tract infections.

Epidemiology and Transmission

PIV infections occur initially during childhood; PIV-3 infections often occur during infancy and are an important cause of illness in infants younger than 6 months, as well as in immunocompromised persons. The incidence of croup and respiratory disease caused by PIV-1 or PIV-2 infections is highest between the ages of 6 months and 5 years. PIV-1 typically causes epidemics lasting several months, peaking in October or November of alternating years, whereas PIV-2 activity is more sporadic. PIV-3 causes infections throughout the year, with outbreaks during the spring and summer.

PIVs are transmitted from person to person, either by direct contact with respiratory secretions or by large aerosol droplets. Transmission occurs readily in families, and reinfections in older children and adults are common. Outbreaks occur in nurseries, day care centers, and hospitals, with attack rates of 40% or higher in susceptible patients.

Diagnosis

Clinical features

The incubation period of PIVs is approximately 3 to 6 days. Virus replication is generally limited to the respiratory tract, although viremia and CNS infections96 have been rarely documented. Initial infections cause fever, rhinitis, pharyngitis, laryngitis, croup, and bronchitis in children.

Laboratory tests

Immunofluorescence testing of respiratory secretions and RT-PCR have been used for rapid PIV detection. Culture is generally slow, requiring days to weeks. Serologic assays, including complement fixation, ELISA, and hemagglutination assays, are used for retrospective diagnosis.

Treatment

Specific antiviral therapy of proven value has not been established. Early treatment with aerosol ribavirin may benefit some immunocompromised patients with serious infections, and intravenous ribavirin has been used for treatment of PIV pneumonia in BMT patients.97

Prevention

An effective PIV vaccine is not yet available, but live attenuated vaccines derived from either human or bovine PIV have been developed and appear to be safe and immunogenic in children. Infection control measures, with an emphasis on exclusion of sick caregivers and isolation protocols, may reduce the risk of nosocomial acquisition in compromised hosts.

Complications

In adults and older children, reinfections are often asymptomatic but may cause serious lower respiratory tract disease, including exacerbations of chronic airway disease and pneumonia. During periods of circulation, about 2% to 3% of cases of community-acquired pneumonia in adults are linked to PIV infection.98 Severe pneumonia, sometimes in the absence of upper respiratory tract illness, occurs in immunosuppressed adults and children, particularly after BMT.5

RESPIRATORY SYNCYTIAL VIRUS

Classification and Pathogenesis

RSV is a paramyxovirus with surface glycoproteins that have neither hemagglutinin nor neuraminidase activity. The F protein is responsible for fusion of the viral envelope with the host cell membranes. Antibody against this protein neutralizes RSV infectivity and blocks syncytial cell formation. The G protein is responsible for attachment. Two major subgroups (A and B) are distinguished primarily by antigenic differences in the G glycoprotein, and multiple strains are recognized within each subgroup.

Epidemiology and Transmission

RSV is the major cause of lower respiratory tract disease in infants and young children, accounting for 45% to 90% of bronchiolitis cases and up to 40% of pneumonia cases. Approximately 40% of infections in the first year of life cause lower respiratory tract illness, and 1% to 2% result in hospitalization; about 10% of hospitalized infants require ventilatory support.

As many as 100,000 hospitalizations and 510 deaths of infants and children are attributed to RSV each year in the United States.99 RSV is also increasingly recognized as a cause of lower respiratory tract disease in older adults and immunocompromised persons.100,101 Mortality is usually less than 1% in previously healthy infants but is much higher in persons with primary immunodeficiency, persons undergoing cancer chemotherapy, and persons with preexisting pulmonary and heart disease.102 The number of RSV-related deaths in the elderly is estimated to be 10-fold higher than that in infants.103 In elderly persons, it is estimated that RSV is associated with 2% to 9% of hospitalizations or deaths caused by pneumonia100 and about 10% of hospitalizations for acute cardiopulmonary conditions.99 Each year, RSV is responsible for an estimated 11,321 deaths in the United States.64

RSV causes prolonged outbreaks of infection from late fall to spring annually in temperate climates.94 Most outbreaks peak between January and March in the Northern Hemisphere, but sporadic cases can occur year-round. Epidemics are associated with increased pediatric hospitalizations and deaths from lower respiratory tract illness. Essentially all children are infected within several years after birth. Immunity is incomplete, and reinfections in children and adults are common. Higher titers of circulating and mucosal antibody occur with successive infections and appear to be associated with milder illness.

RSV appears to be spread by large-particle aerosols during close personal contact and by hand contamination with self-inoculation of the eye or nose. RSV is a major nosocomial pathogen, and high attack rates occur during outbreaks in hospitals, transplantation units, day care centers, and geriatric homes.104 Illness rates are commonly 20% to 50% among hospital staff and patients during epidemics. In households, secondary infection occurs in about one third of adults.

Diagnosis

Clinical features

The incubation period of RSV averages 4 to 5 days but ranges up to 1 week. In infants and young children, almost all primary infections are symptomatic, and 40% or more are associated with bronchiolitis or pneumonia. Febrile upper respiratory tract illness and otitis media are common.

Most recurrent infections in adults are associated with upper respiratory tract illness. Adults typically experience coryza, pharyngitis, and cough, often with low-grade fever. Bronchitis, influenzalike illness, pneumonia, and exacerbations of asthma and chronic bronchitis also occur. In the elderly, the clinical manifestations and outcomes of RSV infections are similar to those of influenza.99,105 Elderly adults hospitalized with RSV commonly have dyspnea, wheezing, and sputum production. Viral bronchopneumonia or secondary bacterial pneumonia appears to complicate a high proportion of RSV infections in such patients.

RSV causes severe lower respiratory tract disease in immunosuppressed patients, particularly BMT recipients and acute leukemia patients receiving chemotherapy.6 Most cases are nosocomial; mortality exceeds 60% when pneumonia develops. Rhinitis with sinusitis or otitis often precedes the development of pneumonia by several days and may provide a clue to diagnosis.

Laboratory tests

Testing of bronchoalveolar lavage samples with RSV antigen or RNA detection provides rapid diagnosis in pneumonia cases. Commercially available ELISAs are sensitive to RSV in children, but nasopharyngeal samples are an insensitive means of detecting RSV antigen in adults, who have low titers of virus in the upper respiratory tract. RT-PCR is a reasonably sensitive technique to detect RSV infection, whereas culture of virus is slow and less sensitive. Serology is more sensitive than cell culture for diagnosis in adults but is slow because of the need for paired sera.99

Treatment

The value of aerosolized ribavirin in the management of RSV disease in hospitalized children remains controversial, and studies of its value in mechanically ventilated infants have yielded conflicting results.23 Aerosolized ribavirin should be considered for infants hospitalized with RSV infections who are at high risk for severe or complicated illness (e.g., those with congenital heart disease, bronchopulmonary dysplasia, cystic fibrosis, and immunodeficiency), for those who are already severely ill, and for those who are younger than 6 weeks. Treatment with polyclonal or monoclonal RSV antibodies does not provide clear therapeutic benefit for RSV-infected infants and young children. Aerosol ribavirin may reduce progression from upper to lower respiratory tract disease in highly immunocompromised hosts. Early treatment with aerosolized ribavirin plus IVIg, RSV immunoglobulin, or palivizumab appears to benefit immunosuppressed patients with RSV pneumonia.106

Prevention

Passive immunoprophylaxis by monthly administration of high-titer anti-RSV immunoglobulin or anti-F monoclonal antibody (palivizumab) reduces the risk of lower respiratory tract RSV disease and hospitalization in high-risk infants and children. Palivizumab is approved for prophylaxis in premature infants and children younger than 2 years with bronchopulmonary dysplasia and is appropriate for infants and young children with hemodynamically significant congenital heart disease.107 A phase I trial has found that palivizumab appears to be safe and well tolerated in recipients of hematopoietic stem cell transplants.108 No vaccine against RSV is available yet, but studies of intranasal live-attenuated vaccine in children and injected subunit vaccine in elderly persons are ongoing.

Interruption of nosocomial transmission may be facilitated by thorough hand washing,109 decontamination of fomites, early identification and rapid isolation of infected infants and other potential RSV cases,110 and cohorting of staff with infected infants.111 Regular use of gowns, gloves, disposable eye-nose goggles, and possibly masks by hospital staff caring for infected children may further reduce the risk of nosocomial RSV spread.

RHINOVIRUS

Classification and Pathogenesis

Rhinoviruses are small (30 nm), nonenveloped viruses that belong to the Picornaviridae family. They have a single-stranded RNA genome and are composed of four structural proteins. Three of the proteins induce neutralizing antibodies and form the basis for serotyping. The large number of serotypes (> 110) makes the development of an effective vaccine unlikely. Nearly 90% of rhinovirus serotypes use intercellular adhesion molecule-1 as a common cellular receptor. Although replication of rhinovirus was thought to be limited to the upper respiratory tract, increasing data suggest that rhinovirus can replicate and cause disease in the lower airways.112

Epidemiology and Transmission

Rhinoviruses cause approximately one infection per person per year in adults, and rates are even higher in children. Rhinovirus causes about 50% of colds in adults each year and up to 90% during the fall months.113 Immunity to rhinovirus is serotype specific and relatively enduring after infection, although reinfection can occur. Rhinoviruses cause infections year-round but are most prevalent in early fall and late spring. The relative importance of fomite spread with hand contamination and self-inoculation into the eyes or nose and aerosols in rhinovirus transmission is uncertain under natural conditions.

Diagnosis

Clinical features

The incubation period of rhinoviruses averages about 2 days, with a range of 1 to 5 days. Rhinovirus colds vary in severity from mild episodes characterized by 1 to 2 days of coryza or scratchy throat to illnesses with profuse rhinorrhea, pharyngitis, and tracheobronchitis. Symptoms usually peak on the second and third days of illness, and the median duration is 1 week.

Rhinovirus infections are associated with more frequent lower respiratory tract illness in the elderly and with bronchospasm and protracted cough in persons with preexisting airway disease.114,115 Rhinovirus is the major infectious cause of asthma exacerbations in children and in adults. In addition, rhinovirus infections are associated with acute sinusitis,116 otitis media,117 exacerbations of chronic bronchitis, and lower respiratory tract disease in infants and young children. In BMT recipients, rhinovirus infection has been associated with pneumonia and poor prognosis.1

Laboratory tests

No rapid diagnostic test for rhinovirus infection is routinely available; detection is usually by virus isolation and, more recently, by RT-PCR.118

Treatment

No antiviral therapy of proven clinical value is currently available, but several approaches are under investigation. Oral administration of the capsid-binding antirhinoviral compound pleconaril reduces the severity and duration of uncomplicated rhinovirus colds in adults but is associated with a risk of drug interactions.119 Nonselective regimens such as high-dose vitamin C, Echinacea preparations, inhalation of heated air, and zinc lozenges are of doubtful value.

Prevention

Barrier control measures may be beneficial, although tissues impregnated with virucides have proved disappointing in this regard. Hand washing and avoidance of finger-to-nose and finger-to-eye contact should reduce the risk of infection. Because of the risk of nosocomial transmission of rhinovirus infections, adherence to infection control precautions in cases of upper respiratory tract illness is important.

BOCAVIRUS

With the use of a virus screening library, a novel parvovirus—human bocavirus (HBoV)—was recently discovered.120 One study detected the virus in 3.1% of nasopharyngeal aspirates collected in children hospitalized with acute respiratory illness. Half of the children had an underlying history of asthma. All had some degree of respiratory distress and fever at the time of presentation.120 Another study documented HBoV infection in 5.6% of children presenting to a hospital with respiratory symptoms in the winter months; interestingly, half of the patients with HBoV had evidence of coinfection with another virus.121 Further studies are needed to fully identify the epidemiology, clinical presentation, and preventive and therapeutic options for this new virus.

HANTAVIRUS

Hantavirus causes hantavirus cardiopulmonary syndrome (HCPS).122 Most HCPS cases have occurred in young to middle-aged adults with no underlying disease. The largest number of cases have occurred in New Mexico, Arizona, and California, but cases have also been identified throughout western North America and sporadically in the eastern United States. HCPS is typically a zoonosis; the principal animal reservoir is the deer mouse. Human infections occur by inhalation of aerosols of infectious excreta. Detailed discussion of HCPS is provided elsewhere [see 7:XXXI Viral Zoonoses].

INTERNET RESOURCES

A listing of Internet resources with information about respiratory viral infections is provided [see Sidebar, Internet Resources for Respiratory Viral Infections].

Internet Resources for Respiratory Viral Infections

  • Adenovirus

o   National Respiratory and Enteric Virus Surveillance System http://www.cdc.gov/ncidod/dvrd/revb/respiratory/eadfeat.htm

  • SARS

o   Centers for Disease Control and Prevention http://www.cdc.gov/ncidod/sars

o   World Health Organization http://www.who.int/csr/sars/en/

  • Influenza Virus

o   World Health Organization http://www.who.int/csr/disease/influenza/en

o   Centers for Disease Control and Prevention http://www.cdc.gov/ncidod/diseases/flu/fluvirus.htm

o   National Foundation for Infectious Diseases http://www.nfid.org/library/influenza

o   American Lung Association http://www.lungusa.org/site/pp.asp?c=dvLUK9O0E&b=35426

  • Avian Influenza

o   World Health Organization http://www.who.int/csr/disease/avian_influenza/en/index.html

o   Centers for Disease Control and Prevention http://www.cdc.gov/flu/avian/index.htm

o   U.S. Department of Health & Human Services http://www.pandemicflu.gov

  • Parainfluenza Viruses

o   National Respiratory and Enteric Virus Surveillance System http://www.cdc.gov/ncidod/dvrd/revb/respiratory/hpivfeat.htm

  • Respiratory Syncytial Virus

o   National Respiratory and Enteric Virus Surveillance System http://www.cdc.gov/ncidod/dvrd/revb/respiratory/rsvfeat.htm

o   American Lung Association http://www.lungusa.org/site/pp.asp?c=dvLUK9O0E&b=35695

  • Hantavirus

o   Centers for Disease Control and Prevention http://www.cdc.gov/ncidod/diseases/hanta/hps/index.htm

o   Emerging Viruses Research Center Hantavirus Reference Laboratory http://www.hsc.unm.edu/pathology/research/Hjellelab

o   American Lung Association http://www.lungusa.org/site/pp.asp?c=dvLUK9O0E&b=35428

References

  1. Armstrong GL, Conn LA, Pinner RW: Trends in infectious disease mortality in the United States during the 20th century. JAMA 281:61, 1999
  2. Mainous AG, Hueston WJ: The cost of antibiotics in treating upper respiratory tract infections in a medical population. Arch Fam Med 7:45, 1998
  3. Monto AS: Studies of the community and family: acute respiratory illness and infection. Epidemiol Rev 16:351, 1994
  4. Kaiser L, Fritz RS, Straus SE, et al: Symptom pathogenesis during acute influenza: interleukin-6 and other cytokine responses. J Med Virol 64:262, 2001
  5. Hayden FG, Fritz R, Lobo MC, et al: Local and systemic cytokine responses during experimental human influenza A virus infection: relation to symptom formation and host defense. J Clin Invest 101:643, 1998
  6. To KF, Chan PK, Chan KF, et al: Pathology of fatal human infection associated with avian influenza A H5N1 virus. J Med Virol 63:242, 2001
  7. Sable CA, Hayden FG: Orthomyxoviral and paramyxoviral infections in transplant patients. Infect Dis Clin North Am 9:987, 1995
  8. Olsen MA, Shuck KM, Sambol AR, et al: Isolation of seven respiratory viruses in shell vials: a practical and highly sensitive method. J Clin Microbiol 31:422, 1993
  9. Hindiyeh M, Goulding C, Morgan H, et al: Evaluation of BioStar Flu OIA assay for rapid detection of influenza A and B viruses in respiratory specimens. J Clin Virol 17:119, 2000
  10. Grondahl B, Puppe W, Hoppe A, et al: Rapid identification of nine microorganisms causing acute respiratory tract infections by single-tube multiplex reverse transcription—PCR: feasibility study. J Clin Microbiol 37:1, 1999
  11. Gwaltney JM Jr, Phillips CD, Miller RD, et al: Computed tomographic study of the common cold. N Engl J Med 330:25, 1994
  12. Anon JB, Jacobs MR, Poole MD, et al: Antimicrobial treatment guidelines for acute bacterial rhinosinusitis. Sinus and Allergy Health Partnership. Otolaryngol Head Neck Surg 130(1 suppl):1, 2004
  13. Arola M, Ruuskanen O, Ziegler T, et al: Clinical role of respiratory virus infection in acute otitis media. Pediatrics 86:848, 1990
  14. Hayden FG, Diamond L, Wood PB, et al: Effectiveness and safety of intranasal ipratropium bromide in common colds: a randomized, double-blind, placebo-controlled trial. Ann Intern Med 125:89, 1996
  15. Romero JR: Pleconaril: a novel antipicornaviral drug. Expert Opin Investig Drugs 10:369, 2001
  16. Melchart D, Linde K, Fischer P, et al: Echinaceafor preventing and treating the common cold. Cochrane Database Syst Rev (2):CD000530, 2000
  17. Marshall I: Zinc for the common cold. Cochrane Database Syst Rev (2):CD001364, 2000
  18. Vickers AJ, Smith C: Homeopathic Oscillococcinum for preventing and treating influenza and influenza-like syndromes. Cochrane Database Syst Rev (2):CD001957, 2000
  19. Hayden FG: Rhinovirus and the lower respiratory tract. Rev Med Virol 14:17, 2004
  20. Gonzales R, Bartlett JG, Besser RE, et al: Principles of appropriate antibiotic use for treatment of uncomplicated acute bronchitis: background. Ann Intern Med 134:521, 2001
  21. Harper SA, Fukuda K, Uyeki TM, et al: Prevention and control of influenza: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 53 (RR-6):1, 2004
  22. Kaiser L, Keene ON, Hammond JMJ, et al: Impact of zanamivir on antibiotic use for respiratory events following acute influenza in adolescents and adults. Arch Intern Med 160:3234, 2000
  23. Treanor JJ, Hayden FG, Vrooman PS, et al: Efficacy and safety of the oral neuraminidase inhibitor oseltamivir in treating acute influenza: a randomized controlled trial. US Oral Neuraminidase Study Group. JAMA 283:1016, 2000
  24. Reassessment of the indications for ribavirin therapy in respiratory syncytial virus infections. American Academy of Pediatrics Committee on Infectious Diseases. Pediatrics 97:137, 1996
  25. Folkerts G, Busse WW, Nijkamp FP, et al: Virus-induced airway hyperresponsiveness and asthma. Am J Respir Crit Care Med 157:1708, 1998
  26. Glezen WP, Greenberg SB, Atmar RL, et al: Impact of respiratory virus infections on persons with chronic underlying conditions. JAMA 283:499, 2000
  27. Greenberg SB: Viral pneumonia. Infect Dis Clin North Am 5:603, 1991
  28. Loda FA, Clyde WA Jr, Glezen WP, et al: Studies on the role of viruses, bacteria, and M. pneumoniaeas causes of lower respiratory tract infections in children. J Pediatr 72:161, 1968
  29. Peiris JSM, Phil D, Yue KY, et al: The severe acute respiratory syndrome. N Engl J Med 349:2431, 2003
  30. Clinical guidance on the identification and evaluation of possible SARS-CoV disease among persons presenting with community-acquired illness. Centers for Disease Control and Prevention, Atlanta, 2004 http://www.cdc.gov/ncidod/sars/clinicalguidance.htm
  31. Gubareva LV, Kaiser L, Hayden FG: Influenza virus neuraminidase inhibitors. Lancet 355:827, 2000
  32. Hayden FG, Osterhaus AD, Treanor JJ, et al: Efficacy and safety of the neuraminidase inhibitor zanamivir in the treatment of influenza virus infections. N Engl J Med 337:874, 1997
  33. Connolly MG Jr, Baugham RP, Dohn MN, et al: Recovery of viruses other than cytomegalovirus from bronchoalveolar lavage fluid. Chest 105:1775, 1994
  34. Baughman RP: The lung in the immunocompromised patient: infectious complications part 1. Respiration 66:95, 1999
  35. Rabella N, Rodriguez P, Labeaga R, et al: Conventional respiratory viruses recovered from immunocompromised patients: clinical considerations. Clin Infect Dis 28:1043, 1999
  36. Duggan JM, Farrehi J, Duderstadt S, et al: Treatment with ganciclovir of adenovirus pneumonia in a cardiac transplant patient. Am J Med 103:439, 1997
  37. Shetty AK, Gans HA, So S, et al: Intravenous ribavirin therapy for adenovirus pneumonia. Pediatr Pulmonol 29:69, 2000
  38. Safrin S, Cherrington J, Jaffe HS: Clinical uses of cidofovir. Rev Med Virol 7:145, 1997
  39. Ludwig SL, Brundage JF, Kelley PW, et al: Prevalence of antibodies to adenovirus serotypes 4 and 7 among unimmunized US Army trainees: results of a retrospective nationwide seroprevalence survey. J Infect Dis 178:1776, 1998
  40. Jernigan JA, Lowry BS, Hayden FG, et al: Adenovirus type 8 epidemic keratoconjunctivitis in an eye clinic: risk factors and control. J Infect Dis 167:1307, 1993
  41. Uemura T, Kawashitam T, Ostuka Y, et al: A recent outbreak of adenovirus type 7 infection in a chronic inpatient facility for the severely handicapped. Infect Control Hosp Epidemiol 21:559, 2000
  42. Hierholzer JC: Adenoviruses in the immunocompromised host. Clin Microbiol Rev 5:262, 1992
  43. Tsutsumi H, Ouchi K, Ohsaki M, et al: Immunochromatography test for rapid diagnosis of adenovirus respiratory tract infections: comparison with virus isolation in tissue culture. J Clin Microbiol 37:2007, 1999
  44. Couch RB, Englund JA, Whimbey E: Respiratory viral infections in immunocompetent and immunocompromised persons. Am J Med 102:2, 1997
  45. Howard DS, Phillips GL II, Reece DE, et al: Adenovirus infections in hematopoietic stem cell transplant recipients. Clin Infect Dis 29:1494, 1999
  46. Dagan R, Schwartz RH, Insel RA, et al: Severe diffuse adenovirus 7a pneumonia in a child with combined immunodeficiency: possible therapeutic effect of human immune serum globulin containing specific neutralizing antibody. Pediatr Infect Dis 3:246, 1984
  47. DeVincenzo J: Pre-emptive ribavirin for “RSV” in BMT. Bone Marrow Transplant 26:113, 2000
  48. Goetz MB, Mathisen GE: Clinical course and treatment of adults with severe measles pneumonitis. Clin Infect Dis 21:443, 1995
  49. Kahn JS: The widening scope of coronaviruses. Curr Opin Pediatr 18:42, 2006
  50. van der Hoek L, Sure K, Ihorst G, et al: Croup is associated with the novel coronavirus NL63. PloS Med 2:e240, 2005
  51. Esper F, Shapiro ED, Weibel C, et al: Association between a novel human coronavirus and Kawasaki disease. J Infect Dis 191:499, 2005
  52. Chang LY, Chiang BL, Kao CL, et al: Lack of association between infection with a novel human coronavirus (HCoV), HCoV-NH, and Kawasaki disease in Taiwan. J Infect Dis 193:283, 2006
  53. Yu IT, Li Y, Wong TW, et al: Evidence of airborne transmission of the severe acute respiratory syndrome virus. N Engl J Med 350:1731, 2004
  54. Lim PL, Kurup A, Gopalakrishna G, et al: Laboratory-acquired severe acute respiratory syndrome. N Engl J Med 350:1740, 2004
  55. Wong KT, Antonio GE, Hui DS, et al: Severe acute respiratory syndrome: radiographic appearances and pattern of progression in 138 patients. Radiology 228:401, 2003
  56. Poon LL, Chan KH, Wong OK, et al: Early diagnosis of SARS coronavirus infection by real time RT-PCR. J Clin Virol 28:233, 2003
  57. Tan EL, Ooi EE, Lin CY, et al: Inhibition of SARS coronavirus infection in vitro with clinically approved antiviral drugs. Emerg Infect Dis 10:581, 2004
  58. ter Meulen J, Bakker AB, van den Brink EN, et al: Human monoclonal antibody as prophylaxis for SARS coronavirus infection in ferrets. Lancet 363:2139, 2004
  59. Traggiai E, Becker S, Subbarao K, et al: An efficient method to make human monoclonal antibodies from memory B cells: potent neutralization of SARS coronavirus. Nat Med 10:871, 2004
  60. van den Hoogen BG, de Jong JC, Groen J, et al: A newly discovered human pneumovirus isolated from young children with respiratory tract disease. Nature Medicine 7:719, 2001
  61. van den Hoogen BG, Osterhaus ADME, Fouchier RAM: Clinical impact and diagnosis of human metapneumovirus infection. Pediatr Infect Dis J 23(1 suppl):S25, 2004
  62. Williams JV, Harris PA, Tollefson SJ, et al: Human metapneumovirus and lower respiratory tract disease in otherwise healthy infants and children. N Engl J Med 350:443, 2004
  63. Wyde PR, Chetty SN, Jewell AM, et al: Comparison of the inhibition of human metapneumovirus and respiratory syncytial virus in ribavirin and immune serum globulin in vitro. Antiviral Res 60:51, 2003
  64. Thompson WW, Shay DK, Weintraub E, et al: Mortality associated with influenza and respiratory syncytial virus. JAMA 289:179, 2003
  65. Lui KJ, Kendal AP: Impact of influenza epidemics on mortality in the United States from October 1972 to May 1985. Am J Public Health 77:712, 1987
  66. Kaiser L, Hayden FG: Hospitalizing influenza in adults. Curr Clin Top Infect Dis 19:112, 1999
  67. Chen H, Smith GJD, Li KS, et al: Establishment of multiple sublineages of H5N1 influenza virus in Asia: implications for pandemic control. Proc Natl Acad Sci USA 103:2845, 2006
  68. Beigel JH, Farrar J, Han AM, et al: Avian influenza A (H5N1) infection in humans. N Engl J Med 353:1374, 2005
  69. Koopmans M, Wilbrink B, Conyn M, et al: Transmission of H7N7 avian influenza A virus to human beings during a large outbreak in commercial poultry farms in the Netherlands. Lancet 363:587, 2004
  70. Nicholson KG, Wood JM, Zambon M: Influenza. Lancet 362:1733, 2003
  71. Lewis DB: Avian flu to human influenza. Annu Rev Med 57:139, 2006
  72. de Jong MD, Bach VC, Phan TQ, et al: Fatal avian influenza A (H5N1) in a child presenting with diarrhea followed by coma. N Engl J Med 352:686, 2005
  73. Oliveira EC, Marik PE, Colice G: Influenza pneumonia: a descriptive study. Chest 119:1717, 2001
  74. Leonardi GP, Leib H, Birkhead GS, et al: Comparison of rapid detection methods for influenza A virus and their value in health-care management of institutionalized geriatric patients. J Clin Microbiol 32:70, 1994
  75. FDA approves new laboratory test to detect human infections with avian influenza A/H5 viruses. U. S. Department of Health and Human Services, Washington, D.C., February 3, 2006 http://www.hhs.gov/news/press/2006pres/20060203.html
  76. Noyola DE, Demmler GJ: Effect of rapid diagnosis on management of influenza A infections. Pediatr Infect Dis J 19:303, 2000
  77. Fujimoto S, Kobayashi M, Uemura O, et al: PCR on cerebrospinal fluid to show influenza-associated acute encephalopathy or encephalitis. Lancet 352:873, 1998
  78. Bright RA, Shay DK, Shu B, et al: Adamantane resistance among influenza A viruses isolated early during the 2005-2006 influenza season in the United States. JAMA 295:891, 2006
  79. Bright RA, Medina MJ, Xu X, et al: Incidence of adamantane resistance among influenza A (H3N2) viruses isolated worldwide from 1994 to 2005: a cause for concern. Lancet 366:1175, 2005
  80. Hayden FG: Amantadine and rimantadine: clinical aspects. Antiviral Drug Resistance. Richman DD, Ed. John Wiley & Sons, 1996, p 59
  81. Le QM, Kiso M, Someya K, et al: Avian flu: isolation of drug-resistant H5N1 virus. Nature 437:1108, 2005
  82. de Jong MD, Thanh TT, Khanh TH, et al: Oseltamivir resistance during treatment of influenza A (H5N1) infection. N Engl J Med 353:2667, 2005
  83. Kaiser L, Wat C, Mills T, et al: Impact of oseltamivir treatment on influenza-related lower respiratory tract complications and hospitalizations. Arch Intern Med 163:1667, 2003
  84. High levels of adamantane resistance among influenza A (H3N2) viruses and interim guidelines for use of antiviral agents—United States, 2005–06 influenza season. MMWR Morb Mortal Wkly Rep 55:1, 2006
  85. Monto AS, Robinson DP, Herlocher ML, et al: Zanamivir in the prevention of influenza among healthy adults: a randomized controlled trial. JAMA 282:31, 1999
  86. Hayden FG, Gubareva LV, Monto AS, et al: Inhaled zanamivir for the prevention of influenza in families. N Engl J Med 343:1282, 2000
  87. Hayden FG, Atmar RL, Schilling M, et al: Use of the selective oral neuraminidase inhibitor oseltamivir to prevent influenza. N Engl J Med 341:1336, 1999
  88. Munoz FM, Galasso GJ, Gwaltney JM Jr, et al: Current research on influenza and other respiratory viruses: II international symposium. Antiviral Res 46:91, 2000
  89. Whitley RJ, Hayden FG, Reisinger KS, et al: Oral oseltamivir treatment of influenza in children. Pediatr Infect Dis J 20:127, 2001
  90. Nichol KL, Lind A, Margolis KL, et al: The effectiveness of vaccination against influenza in healthy, working adults. N Engl J Med 333:889, 1995
  91. Nichol KL, Mendelman PM, Mallon KP, et al: Effectiveness of live, attenuated intranasal influenza virus vaccine in healthy, working adults: a randomized controlled trial. JAMA 282:137, 1999
  92. Nichol KL: The efficacy, effectiveness and cost-effectiveness of inactivated influenza virus vaccines. Vaccine 21:1769, 2003
  93. Gross PA, Hermogenes AW, Sacks HS, et al: The efficacy of influenza vaccine in elderly persons: a meta-analysis and review of the literature. Ann Intern Med 123:518, 1995
  94. Reichert TA, Sugaya N, Fedson DS, et al: The Japanese experience with vaccinating schoolchildren against influenza. N Engl J Med 344:889, 2001
  95. Sullivan PS, Hanson DL, Dworkin MS, et al: Effect of influenza vaccination on disease progression among HIV-infected persons. AIDS 14:2781, 2000
  96. Arisoy ES, Demmler GJ, Thakar S, et al: Meningitis due to parainfluenza virus type 3: report of two cases and review. Clin Infect Dis 17:995, 1993
  97. Ison MG, Hayden FG: Viral infections in immunocompromised patients: what's new with respiratory viruses? Curr Opin Infect Dis 15:355, 2002
  98. Marx A, Gary HE Jr, Marston BJ, et al: Parainfluenza virus infection among adults hospitalized for lower respiratory tract infection. Clin Infect Dis 29:134, 1999
  99. Shay DK, Holman RC, Roosevelt GE, et al: Bronchiolitis-associated mortality and estimates of respiratory syncytial virus-associated deaths among US children 1979–1997. J Infect Dis 183:16, 2001
  100. Falsey AR, Cunningham CK, Barker WH, et al: Respiratory syncytial virus and influenza A infections in the hospitalized elderly. J Infect Dis 172:389, 1995
  101. Han LL, Alexander JP, Anderson LJ: Respiratory syncytial virus pneumonia among elderly: an assessment of disease burden. J Infect Dis 179:25, 1999
  102. Holberg CJ, Wright AL, Martinez FD, et al: Risk factors for respiratory syncytial virus—associated lower respiratory illnesses in the first year of life. Am J Epidemiol 133:1135, 1991
  103. Navas L, Wang E, de Carvalho V, et al: Improved outcome of respiratory syncytial virus infection in a high-risk hospitalized population of Canadian children. Pediatric Investigators Collaborative Network on Infections in Canada. J Pediatr 121:348, 1992
  104. Falsey AR, McCann RM, Hall WJ, et al: Acute respiratory tract infection in daycare centers for older persons. J Am Geriatr Soc 43:30, 1995
  105. Respiratory syncytial virus activity—United States, 2000–01 season. MMWR Morb Mortal Wkly Rep 51:26, 2002
  106. Whimbey E, Champlin RE, Englund JA, et al: Combination therapy with aerosolized ribavirin and intravenous immunoglobulin for respiratory syncytial virus disease in adult bone marrow transplant recipients. Bone Marrow Transplant 16:393, 1995
  107. Revised indications for the use of palivizumab and respiratory syncytial virus immune globulin intravenous for the prevention of respiratory syncytial virus infections. American Academy of Pediatrics Committee on Infectious Diseases and Committee on Fetus and Newborn. Pediatrics 112:1442, 2003
  108. Boeckh M, Berrey MM, Bowden RA, et al: Phase 1 evaluation of the respiratory syncytial virus—specific monoclonal antibody palivizumab in recipients of hematopoietic stem cell transplants. J Infect Dis 184:350, 2001
  109. Contreras PA, Sami IR, Darnell ME, et al: Inactivation of respiratory syncytial virus by generic hand dishwashing detergents and antibacterial hand soaps. Infect Control Hosp Epidemiol 20:57, 1999
  110. Karanfil LV, Conlon M, Lykens K, et al: Reducing the rate of nosocomially transmitted respiratory syncytial virus. Am J Infect Control 27:91, 1999
  111. Madge P, Paton JY, McColl JH, et al: Prospective controlled study of four infection-control procedures to prevent nosocomial infection with respiratory syncytial virus. Lancet 340:1079, 1992
  112. Ison MG, Hayden FG, Kaiser L, et al: Rhinovirus infections in hematopoietic stem cell transplant recipients with pneumonia. Clin Infect Dis 36:1139, 2003
  113. Arruda E, Pitkaranta A, Witek TJ, et al: Frequency and natural history of rhinovirus infections in adults during autumn. J Clin Microbiol 35:2864, 1997
  114. Wald TG, Shult P, Krause P, et al: A rhinovirus outbreak among residents of a long-term care facility. Ann Intern Med 123:588, 1995
  115. Nicholson KG, Kent J, Hammersley V, et al: Acute viral infections of upper respiratory tract in elderly people living in the community: comparative, prospective, population based study of disease burden. BMJ 315:1060, 1997
  116. Pitkaranta A, Arruda E, Malmberg H, et al: Detection of rhinovirus in sinus brushings of patients with acute community-acquired sinusitis by reverse transcription-PCR. J Clin Microbiol 35:1791, 1997
  117. Pitkaranta A, Virolainen A, Jero J: Detection of rhinovirus, respiratory syncytial virus, and coronavirus infections in acute otitis media by reverse transcriptase polymerase chain reaction. Pediatrics 102:291, 1998
  118. Andeweg AC, Bestebroer TM, Huybreghs M, et al: Improved detection of rhinoviruses in clinical samples by using a newly developed nested reverse transcription-PCR assay. J Clin Microbiol 37:524, 1999
  119. Hayden FG, Herrington DT, Coats TL, et al: Efficacy and safety of oral pleconaril for treatment of colds due to picornaviruses in adults: results of 2 double-blind, randomized, placebo-controlled trials. Clin Infect Dis 36:1523, 2003
  120. Allander T, Tammi MT, Eriksson M, et al: Cloning of a human parvovirus by molecular screening of respiratory tract samples. Proc Natl Acad Sci USA 102:12891, 2005
  121. Sloots TP, McErlean P, Speicher DJ, et al: Evidence of human coronavirus HKU1 and human bocavirus in Australian children. J Clin Virol 35:99, 2006
  122. Update: Hantavirus pulmonary syndrome—United States, 1999. MMWR Morb Mortal Wkly Rep 48:521, 1999
  123. Jefferson TO, Demicheli V, Deeks JJ, et al: Amantadine and rimantadine for preventing and treating influenza A in adults. Cochrane Database Syst Rev (2):CD001169, 2000
  124. Jefferson T, Demicheli V, Deeks J, et al: Neuraminidase inhibitors for preventing and treating influenza in healthy adults. Cochrane Database Syst Rev (2):CD001265, 2000

Editors: Dale, David C.; Federman, Daniel D.



If you find an error or have any questions, please email us at admin@doctorlib.info. Thank you!