Introduction
The clinical microbiology laboratory plays a critical role in diagnosis and management of patients with lower respiratory tract infections. By providing pathogen detection and identification and susceptibility testing the laboratory provides the basis of optimal empirical antimicrobial therapy and individually tailored regimens.1 The microbiology laboratory also provides epidemiologic data that assist the hospital epidemiologist in the prevention, detection, investigation, and termination of nosocomial outbreaks.2 When correctly and promptly used, the information provided by the clinical microbiology laboratory improves clinical outcomes, reduces unnecessary utilization of antibiotics, and prevents nosocomial transmissions.3, 4
The primary aim of this chapter is to assist clinicians in efficient and effective utilization of the resources of the clinical microbiology laboratory in diagnosis of the causes of infections of the lower respiratory tract. This chapter assumes that clinical laboratories are using validated methods and reporting quality-assured results and does not delve into technical or operational aspects of the clinical microbiology laboratory. For additional information on laboratory operation, the reader is referred to the latest edition of the Manual of Clinical Microbiology (American Society for Microbiology).5
Preanalytic Principles
Principles of Testing
The decision to order a diagnostic test should hinge on whether the result is likely to affect the clinician's treatment decisions. If the clinician is certain the patient has a disease based on clinical presentation and prevalence (high pretest probability), then the decision to treat will likely not be altered by the test result and testing should not be ordered. Similarly, testing should not be ordered if the clinician has a high degree of a priori certainty that the patient does not have a disease, because the decision not to treat will likely not be altered by the test result. Testing is most useful when the clinician is uncertain about the probability of disease and the result can sway the physician's decision about treatment. In addition to the pretest probability, several factors affect this decision. For example, if therapy comes at a low harm (in terms of toxicity, dollar cost, and selection of resistance), then treating all patients without testing may be appropriate. If the diagnostic has a low sensitivity (i.e., the test is positive in a low percentage of patients with disease), then testing may lead to an inappropriate decision not to treat. Similarly, if a diagnostic has a low specificity (i.e., the test is positive in a high percentage of patients without disease), then testing may lead to unnecessary treatment. The determination that clinical suspicion is uncertain enough to benefit from a particular diagnostic involves the interplay of the cost and accuracy of the diagnostic test, the pretest probability of the disease, and the benefit and harm of treatment.
Infection Prevention
The clinician plays a critical role in notifying the microbiology laboratory (and the hospital infection control epidemiologist) when virulent and transmissible agents are suspected as the cause of disease. Alerting laboratory staff reduces the exposure risk of laboratory staff handling specimens and cultures harboring highly virulent pathogens. A list of such pathogens is shown Table 17-1 . Not all specimens from patients with infectious diseases should be handled by the on-site laboratory. According to guidelines developed by local and national public health officials, specimens potentially containing selected high-risk agents such as Bacillus anthracis spores, Francisella tularensis, Yersinia pestis, variola major, hemorrhagic fever viruses, or Clostridium botulinum toxin are directly sent to the public health laboratories, where appropriate containment facilities and diagnostic tools are applied to make a diagnosis. Other pathogens that are handled by the on-site laboratory but still require laboratory notification include Coccidioides and Brucella species, because cultures of these are associated with a high risk for laboratory-associated infection. Although the technologists are expected to handle all specimens and microbiologic cultures using universal precautions, accidental exposures can happen, especially if the findings are unexpected. Therefore laboratory notification serves to alert the staff to protect themselves from potential exposure to highly transmissible agents.
Table 17-1.
ORGANISM |
|
Syndromic Order Sets
The diversity of etiologic agents of lower respiratory tract infection poses a number of diagnostic challenges to the clinician. First the provider must formulate a comprehensive yet pragmatic differential diagnosis that takes into account the clinical presentation, immune status, and the exposure history of the patient. Then the clinician must order the correct set of laboratory tests and ensure collection of the appropriate specimens and their placement in correct transport containers as well as their transport to the laboratory under permissive conditions for testing. Because improper test selection and specimen collection could reduce the analytic sensitivity and specificity of assays performed in the laboratory, syndromic order sets have been designed that consider the most common pathogens for the specific syndrome. Syndromic order sets incorporate general guidelines for the types of specimen required, collection and transport, and available assays for pathogens expected in a given clinical setting or syndrome. By prioritizing diagnostics that maximize yield and avoiding the need to repeat invasive procedures, these order sets also serve to minimize risk to the patient and to lower health care costs. However, it is the responsibility of the clinician to ensure that specimen requirements are met and the most critical tests are prioritized, especially when the amount of specimen material obtained is limited and multiple tests are ordered. Table 17-2, Table 17-3, Table 17-4 show syndromic order sets for community-acquired pneumonia (CAP), hospital-acquired and ventilator-associated pneumonia, and immunocompromised host pneumonia, respectively. Order sets developed to address local epidemiologic characteristics and preanalytic practices may be tailored to serve each institution. Clinicians also should familiarize themselves with local sample storage practices in case additional tests need to be performed.
Table 17-2.
Syndrome/Organisms | Testing Uses/Indications | Appropriate Specimens | Available Testing |
---|---|---|---|
TYPICAL BACTERIA | |||
Haemophilus influenzae Moraxella catarrhalis Staphylococcus aureus Streptococcus pneumoniae Streptococcus pyogenes Aerobic gram-negative bacilli |
Outpatients: microbiologic studies optional Inpatients:
|
Sputum Bronchoscopic specimen Tissue |
Gram stain Aerobic culture |
Blood | Aerobic culture | ||
LESS COMMON BACTERIA | |||
Chlamydophila pneumoniae Chlamydia psittaci Coxiella burnetii Legionella pneumophila serogroup 1 Legionella spp.—other Mycobacterium tuberculosis Mycoplasma pneumoniae |
Mycoplasma and C. pneumoniae: outbreaks and familial transmission C. psittaci: exposure to psitaccines |
Nasopharyngeal swab, throat swab or washings Sputum Bronchoscopic specimen Bronchoalveolar lavage Tissue (including FFPE) |
NAT (species specific): M. pneumoniae; C. pneumoniae; C. psittaci NAT: 16S rRNA sequencing (tissue only) DFA: C. pneumoniae |
Serum | IgM, IgG: M. pneumoniae; C. pneumoniae; C. psittaci IgM, IgA, IgG: C. burnetii |
||
Legionella: outbreaks, travel-associated, lack of response to cell wall–active antibiotics, severe illness | Sputum Bronchoscopic specimen Tissue (including FFPE) |
BCYE culture NAT: Legionella species NAT: 16S rRNA sequencing (tissue only) DFA: L. pneumophila |
|
Urine | L. pneumophila serogroup 1 antigen | ||
M. tuberculosis complex: appropriate epidemiology | Sputum Bronchoscopic specimen Tissue Pleural fluid |
Acid-fast stain Mycobacterial culture NAT |
|
VIRUSES | |||
Influenza A/B Adenovirus Parainfluenza 1/2/3 Respiratory syncytial virus Human metapneumovirus Varicella-zoster virus Hantaviruses Novel coronaviruses Novel influenza viruses |
Viral testing may provide justification for discontinuing antibiotics Seasonal epidemiology |
Nasopharyngeal swab Nasal aspirates or washes Bronchoscopic specimen Tissue |
NAT |
ASPIRATION PNEUMONIA | |||
Mixed anaerobic infections | Anaerobes typically already covered by broad-spectrum antibiotics; anaerobic culture rarely changes management | Pleural fluid Bronchoscopic specimen using protected specimen brush Tissue |
Gram stain Aerobic culture Anaerobic culture |
Pleural fluid Tissue |
NAT | ||
INVASIVE FUNGI | |||
Dimorphic mold | |||
Blastomyces dermatitidis Coccidioides immitis Coccidioides posadasii Histoplasma capsulatum Paracoccidioides brasiliensis |
From area of high endemicity | Sputum Bronchoscopic specimen Tissue |
Fungal stain Fungal culture |
Tissue | Histology | ||
Tissue (including FFPE) Pleural fluid |
NAT: species specific NAT: rRNA locus sequencing |
||
Serum | Antigen: H. capsulatum; B. dermatitidis IgG (complement fixation, EIA): H. capsulatum; C. immitis; B. dermatitidis IgM (immunodiffusion, latex agglutination, EIA): C. immitis |
||
Urine | Antigen: H. capsulatum | ||
Cryptococcus C. neoformansC. gattii |
Serum |
Cryptococcal antigen test |
|
Tissue | Fungal stain Culture |
||
PARASITES | |||
Strongyloides stercoralis Paragonimus spp. |
From area of high endemicity | Sputum Bronchoscopic specimen Tissue |
Microscopic examination |
BCYE, buffered charcoal yeast extract; DFA, direct fluorescent antibody; EIA, enzyme immunoassay; FFPE, formalin-fixed paraffin-embedded; ICU, intensive care unit; Ig, immunoglobulin; NAT, nucleic acid test.
Table 17-3.
Syndrome/Organisms | Testing Uses/Indications | Appropriate Specimens | Available Testing |
---|---|---|---|
TYPICAL BACTERIA | |||
Aerobic Gram-Positive Cocci | |||
Staphylococcus aureus Streptococcus pneumoniae Aerobic Gram-Negative Bacilli Acinetobacter species Enterobacter species Escherichia coli Klebsiella pneumoniae Pseudomonas aeruginosa Stenotrophomonas maltophilia Anaerobes Mixed anaerobic species |
Refractoriness to antibiotics Clinically ill patients with suspicious respiratory or chest radiograph findings Anaerobes typically already covered by broad-spectrum antibiotics; anaerobic culture rarely changes management |
Sputum Endotracheal aspirate Bronchoalveolar lavage Bronchoscopic specimen using protected specimen brush Tissue |
Gram stain Aerobic culture Anaerobic culture |
Tissue (including FFPE) | NAT: 16S rRNA sequencing | ||
Blood | Aerobic culture | ||
ATYPICAL BACTERIA | |||
Legionella pneumophila serogroup 1 Legionella species—other |
Legionella outbreaks Refractory to β-lactams or AGs Immunocompromised Pneumonia plus GI symptoms |
Induced sputum Bronchoscopic specimen |
BCYE culture Legionella spp. NAT DFA |
Urine | L. pneumophila serogroup 1 urine antigen | ||
Tissue (including FFPE) | NAT: 16S rRNA sequencing | ||
VIRUSES | |||
Influenza A, B Adenovirus Parainfluenza 1, 2, 3 Respiratory syncytial virus |
Circulating in community/seasonality Unvaccinated host Outbreak/cluster Pneumonia despite broad-spectrum antibiotics |
Nasopharyngeal swab Nasal aspirates or washes Endotracheal aspirate Bronchoscopic specimen Bronchoscopic specimen using protected specimen brush |
NAT |
INVASIVE FUNGI | |||
Aspergillus species Mucorales Mold species—other |
Pulmonary cavity disease Environmental exposure/outbreak Immunocompromised |
Endotracheal aspirate Bronchoalveolar lavage Bronchoscopic specimen using protected specimen brush Tissue |
Fungal stain Fungal culture NAT: species-specific NAT: rRNA locus sequencing (tissue only) |
Tissue (including FFPE) | Histology NAT: 18S rRNA sequencing |
||
Bronchoalveolar lavage Serum |
Galactomannan (1→3) β-d-glucan |
AGs, aminoglycosides; BCYE, buffered charcoal yeast extract; DFA, direct fluorescent antibody; FFPE, formalin-fixed paraffin-embedded; GI, gastrointestinal; NAT, nucleic acid test; spp., species.
Table 17-4.
Syndrome/Organisms | Testing Uses/Indications | Appropriate Specimens | Available Testing |
---|---|---|---|
BACTERIA | |||
CAP and HAP/VAP bacteria | See Tables 17-2 and 17-3 | See Tables 17-2 and 17-3 | See Tables 17-2 and 17-3 |
Burkholderia cepacia complex | Cystic fibrosis, CGD | Sputum Bronchoscopic specimen |
Aerobic culture |
Aerobic Actinomycetes | |||
Nocardia species Rhodococcus species Actinomycetes—other |
Soil/environmental exposure | Sputum Bronchoscopic specimen Tissue (including FFPE) |
Gram stain Modified acid-fast stain Aerobic culture including BCYE plate NAT: 16S rRNA sequencing (tissue only) |
Mycobacteria | |||
M. tuberculosis complex M. avium-intracellulare complex M. kansasii M. xenopi M. haemophilum |
From area of high endemicity Known exposure/outbreak Bronchiectasis Appropriate epidemiology |
Expectorated sputum Bronchoscopic specimen Tissue (including FFPE) |
Cytology Acid-fast stain Mycobacterial culture NAT: M. tuberculosis–specific NAT: nontuberculous mycobacteria–specific NAT: 16S rRNA sequencing (tissue only) |
M. abscessus M. chelonae—other |
Tissue | Histology | |
VIRUSES | |||
CAP and HAP/VAP viruses | See Tables 17-2 and 17-3 | See Tables 17-2 and 17-3 | See Tables 17-2 and 17-3 |
Cytomegalovirus Herpes simplex virus Varicella-zoster virus |
CMV 1-4 months after transplant Serodiscordant donor/recipient Skin lesions |
Bronchoscopic specimen Tissue |
Cytology NAT Shell vial culture: CMV; HSV |
Tissue (fresh and FFPE) | Histology Immunohistochemistry: CMV; HSV NAT |
||
Plasma | NAT | ||
FUNGI | |||
Pneumocystis jirovecii | Sputum Bronchoalveolar lavage Bronchoscopic specimen |
DFA Fungal stain NAT |
|
Cryptococcus neoformans | Serum |
Cryptococcal antigen test |
|
Cryptococcus gattii | Tissue | Fungal stain Culture |
|
Monomorphic molds Aspergillus fumigatus Other Aspergillus species |
Sputum Bronchoscopic specimen |
Fungal stain Fungal culture |
|
Tissue |
Histology |
||
Tissue (fresh and FFPE) Pleural fluid |
NAT: species specific NAT: rRNA locus sequencing |
||
Serum | Antigen: galactomannan Antigen: (1→3) β-d-glucan |
||
Dimorphic molds | See Table 17-2 | See Table 17-2 | See Table 17-2 |
PARASITES | |||
Toxoplasma gondii | Cat exposure Raw meat consumption From area of high endemicity Lymphadenopathy |
Induced sputum Bronchoscopic specimen Tissue |
Giemsa stain NAT |
Serum | IgM | ||
Strongyloides stercoralis | From area of high endemicity | Induced sputum Bronchoscopic specimen Stool |
Microscopy for larvae Strongyloides culture |
Tissue | Histology |
BCYE, buffered charcoal yeast extract; CAP, community-acquired pneumonia; CGD, chronic granulomatous disease; CMV, cytomegalovirus; DFA, direct fluorescent antibody; FFPE, formalin-fixed paraffin-embedded; HAP, hospital-acquired pneumonia; HSV, herpes simplex virus; IgM, immunoglobulin M; NAT, nucleic acid test; VAP, ventilator-acquired pneumonia.
Specimen Selection, Collection, and Transport
In general, sterile specimens such as tissue samples and aspirates are the most valuable diagnostically because the absence of contamination with commensal organisms ensures that any organism detected likely represents a true pathogen. Histopathologic examination of tissue also provides information on the immunopathologic characteristics of the infectious process. However, a major diagnostic challenge of lower respiratory tract infection is that lower respiratory tract secretions are usually obtained through the oropharynx, which normally contains 1010 to 1012 colony-forming units (CFU) of aerobic and anaerobic bacteria per milliliter. Therefore lower respiratory tract secretions collected for microbiologic examination are commonly contaminated with diverse bacteria (Table 17-5 ),6 some of which, such as Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, and Neisseria meningitidis, can also be pathogens of the lower respiratory tract.7, 8, 9 The oropharynx can also contain Mycoplasma pneumoniae 10 and aerobic actinomycetes including Nocardia and nontuberculous mycobacteria in the absence of disease.11 In addition, aspiration of even minute amounts (0.1 to 1 µL) of oropharyngeal secretions can deliver a bolus of 109 CFU to the tracheobronchial tree. The distinction in such cases between colonization of the upper respiratory tract and pneumonia cannot be easily made by sputum examination and culture. Another challenge is that oropharyngeal secretions, which normally contain only a few gram-negative bacilli (such as Enterobacteriaceae, Pseudomonas, Acinetobacter), often become colonized with as many as 107 CFU of gram-negative bacilli per milliliter in seriously ill patients requiring intensive care,12 patients treated with antibiotics after hospitalization for acute pulmonary inflammatory disease,13 chronic alcoholic and diabetic patients,14 institutionalized older adults and chronically ill patients,15 and hospitalized patients with acute leukemia.16 Lastly, Aspergillus spores present in the environment are commonly deposited in the lower respiratory tract and may be recovered from sputum in the absence of disease, although in immunocompromised patients it is best to consider this finding seriously.17 In summary, because lower respiratory tract secretions collected through the oropharynx are nearly always contaminated with resident microflora of the oral cavity and definitive diagnosis would require sterile lung tissue with demonstration of parenchymal invasion, appropriate steps must be taken to obtain specimens of highest quality for microbiologic testing.
Table 17-5.
Commonly Present | Less Commonly Present, Transiently Present, or Present Only in Specific Contexts |
---|---|
Actinomyces, Corynebacterium, Eikenella corrodens, Enterococcus, Haemophilus, Moraxella catarrhalis, Neisseria, Staphylococcus, Streptococcus, Candida | Enterobacteriaceae, Pseudomonas aeruginosa, Acinetobacter baumannii, nontuberculous mycobacteria |
Expectorated sputum is the specimen most frequently obtained for the laboratory diagnosis of lower respiratory tract infection.5 The importance of proper sputum collection was documented by Laird18 100 years ago in studies on the yield of Mycobacterium tuberculosis according to the appearance and cellular composition of the sputum examined. The first requirement for collection of a good-quality sputum specimen is an alert and cooperative patient who can be instructed to rinse out his or her mouth with water or even brush his or her teeth before producing a lower respiratory tract specimen. The patient then must be encouraged to cough deeply to expectorate a specimen of lower respiratory tract secretions. With some infections such as tuberculosis (TB), a larger sample volume can improve the sensitivity of culture.19 Specimens are to be collected in sterile, leakproof, screw-capped containers. Containers should be transported in a watertight plastic biohazard bag.
Although a single sputum specimen may be sufficient for establishing the diagnosis of an acute bacterial process, collection of a series of two or three sputum specimens obtained on one or two days is recommended for patients suspected of having mycobacterial infections.20 In patients with nonproductive cough or suspected mycobacterial, fungal, or Pneumocystis jirovecii infections,21 it may be helpful to induce sputum production with an inhaled aerosol of hypertonic salt solution (3% to 10%).
Once collected, the specimens should be rapidly delivered to the laboratory for processing to avoid overgrowth by contaminating flora, which can compromise microscopic detection and isolation of pathogenic bacteria.22, 23 Penn and Silberman23 found that organisms observed microscopically on Gram-stained smears of sputum specimens and their relative numbers in cultures changed dramatically between processing within an hour of collection and processing after overnight refrigeration. Although there were no significant differences in the culture results between the immediate and delayed cultures in this study, the loss of reliable microscopic features had significant impact on the interpretation of culture results. Processing delay is particularly important for culture recovery of slow-growing mycobacteria.24 Specimens that are not sent to the laboratory for processing within 2 hours should be refrigerated for no more than 5 days. If refrigeration is not possible, samples should be treated first with equal volume of 0.6% cetylpyridinium bromide or 1% cetylpyridinium chloride in 2% sodium chloride, which reduces the survival of contaminating microorganisms while preserving the viability of M. tuberculosis for up to 8 days.24, 25, 26 Although the recovery of fungi is optimal from cultures of fresh specimens, most clinically significant fungi appear to survive storage of 16 days or longer.27 Specimens for viral cultures should be shipped refrigerated but not frozen, whereas specimens for chlamydial culture should be placed into sucrose phosphate medium and shipped frozen.
Although there is no universal agreement on the value of anaerobic culture,28 protected catheter brushes may be used to obtain samples for culture and identification of organisms causing anaerobic pleuropulmonary disease.29 It is essential to transport samples in an anaerobic vial to preserve the viability of anaerobic organisms.
For detection of respiratory viruses, nasopharyngeal specimens are preferred, although lower respiratory tract specimens may be necessary to detect viral infection of the lower respiratory tract.30 There are a number of methods for the collection of nasopharyngeal specimens, which includes flocked and traditional swabs, as well as aspirates and washes. Flocked swabs contain perpendicular arrangements of fibers with an open structure to create a highly absorbent thin layer capable of efficient uptake of respiratory samples and elution into viral transport media. Nasopharyngeal flocked swabs have been shown to be more sensitive for the detection of respiratory viruses than traditional swabs.31, 32 In turn, nasopharyngeal aspirates or washes have been shown to be more sensitive than nasopharyngeal flocked swabs.33, 34, 35 However, the modest gains in sensitivity for detection of most respiratory viruses using aspirates or washes may be offset by the ease of nasopharyngeal specimen collection using flocked swabs. Oropharyngeal specimens are less sensitive than nasopharyngeal specimens, though the combination may increase respiratory virus detection.36, 37, 38, 39 Oropharyngeal swabs may also be used for detecting Chlamydophila pneumoniae, 40, 41, 42, 43 M. pneumoniae, 40, 44, 45 and Legionella species.40
In patients who are critically ill, immunocompromised, or who cannot produce expectorate, one or more invasive approaches may be necessary to obtain diagnostic samples. Specimens may include endotracheal aspirates, pleural fluids, bronchoalveolar lavage (BAL), percutaneous lung aspirate, or lung biopsies.5, 46 The use of BAL has also been expanded to include diagnosis of bacterial pneumonia, especially for nosocomial cases.47, 48, 49 In patients with CAP requiring admission to the hospital, use of protected catheter brush and BAL has been shown to provide microbiologic diagnoses that are not obtainable by noninvasive means,50 although there is little support for using these procedures to diagnose CAP.51 Although the results of cultures from protected catheter brushes and BAL specimens are quantitatively similar, Meduri and Baselski50 concluded that BAL specimens provided a larger and more representative sample of lower respiratory tract secretions than the protected catheter brushes, allowing microscopic analysis of the cytocentrifuged BAL fluid to identify the type of bacteria present and to demonstrate the presence of neutrophils with intracellular organisms. These procedures may also yield additional pathogens not obtainable by noninvasive approaches. Much work has also been done with the use of BAL for the diagnosis of ventilator-associated pneumonia,49, 50, 51, 52 (see Chapter 34).
In children under 7 years of age with suspected TB, gastric aspirate is used as a surrogate for respiratory samples. Historically it has been recommended that the pH of gastric aspirate be neutralized with sodium bicarbonate before transport to the laboratory; however, a recent study suggests that neutralization of gastric aspirate may reduce the recovery of M. tuberculosis.53 Nasopharyngeal aspirates have also been used for diagnosis of TB, although the sensitivity of culture-confirmed TB is lower compared to induced sputum.54 Stool samples in children with pulmonary TB may become the specimen of choice if processing methods can be optimized to concentrate the tubercle bacilli.55, 56
Other specimen types that may aid in diagnosis of lower respiratory tract infection include whole blood for blood culture, serum for antibody and antigen testing, and urine for antigen testing. Blood culture is recommended in cases of severe pneumonia57 but is positive only up to 37% in CAP and in less than 25% in nosocomial pneumonia.57, 58, 59, 60, 61 It is important to note that a large blood volume (60 mL or three sets of blood culture bottles in adults) is necessary to maximize sensitivity of blood culture.62, 63 Although routine blood culture systems have been shown to be highly sensitive for detection of candidemia and cryptococcemia, automated blood culture systems are insensitive for cultivation of monomorphic and dimorphic molds. Isolation of molds (and fastidious bacteria) from blood requires the lysis-centrifugation method (Isolator)64, 65, 66 or the use of enriched fungal medium bottles.67, 68
S. pneumoniae can be recovered from urine cultures in as many as 38% of patients with pneumococcal pneumonia.69 Urine may be tested for the presence of pneumococcal70 and Legionella pneumophila serogroup 171 antigens. Fungal antigen tests of urine are also available for diagnosis of histoplasmosis and blastomycosis.72, 73 Antigen assays are discussed later in this chapter.
Specimen Adequacy
Clinical laboratories are mandated by accrediting agencies to monitor specimen quality and quantity, and to enforce rejection criteria when sample requirements are not met. Common causes for rejection include insufficient sample quantity, poor sample quality, and mislabeling of samples. For bacterial cultures, microscopic examination of sputum and endotracheal aspirate with Gram stain is used to screen samples for adequate quality.18, 74 The presence of excessive squamous epithelial cells (>10 to 25 per low-power field) is indicative of oropharyngeal contamination and therefore grounds for rejection for bacterial culture (Fig. 17-1 ). Although earlier criteria for the adequacy of sputum specimens for bacterial cultures also required the presence of polymorphonuclear leukocytes (neutrophils), the number of neutrophils in a sample is no longer used to evaluate specimen adequacy.75 Endotracheal aspirates are rejected if the screening Gram-stained smears show no organisms.74, 76 For mycobacterial, fungal, and viral cultures, cytologic screening to determine specimen acceptability is not enforced, because contamination with commensals does not interfere with interpretation of the culture results. However, the presence of respiratory columnar epithelial cells has been shown to improve respiratory virus detection by direct fluorescent antibody (DFA) testing.77
Microbiologic Assays
The clinical microbiology laboratory offers a broad range of assays for diagnosis of lower respiratory tract infection. For any particular pathogen, multiple assays may be available, and therefore it is the responsibility of the clinician to choose the assay with the best performance characteristic for a particular specimen type. Table 17-6 summarizes the accuracy of assays used in the diagnosis of lower respiratory tract infections caused by bacteria, fungi, and parasites. In addition, the clinician must be familiar with the turnaround time for each assay to optimize use of the results in managing the patient.
Table 17-6.
Organism | Diagnostic Target | Testing Method | Sample Type | Sensitivity (%) | Specificity (%) | References |
---|---|---|---|---|---|---|
BACTERIA | ||||||
Chlamydia species (excluding C. trachomatis) | Antibody (IgG) | Microimmunofluorescence | Serum | 65–92 | 30–51 | 256 |
Antibody (IgM) | Microimmunofluorescence | Serum | 43–75 | 67–84 | 256 | |
Antibody (IgG and IgM) | Microimmunofluorescence | Serum | 87–100 | 22–40 | 256 | |
Antibody (IgM) | Enzyme immunoassay | Serum | 100 | 92.9 | 257 | |
DNA | PCR—enzyme immunoassay | Nasopharyngeal swab | 55–83 | 91–99 | 256, 258 | |
Coxiella burnetii | Antibody (IgG, IgA, and IgM) | Microimmunofluorescence | N/A | N/A | N/A | * |
DNA | PCR | N/A | N/A | N/A | * | |
Francisella tularensis | DNA | PCR | Swab/tissue | 73–78 | 97 | 259 |
Antibody | Enzyme immunoassay | Serum | 93.9 | 96.1 | 260 | |
Antibody | Latex microagglutination | Serum | 81.8 | 98.0 | 260 | |
Legionella species | Antibody | Direct fluorescent antibody | Respiratory samples | 25–66 | 94 | 95 |
Antigen | Enzyme immunoassay | Urine | 37.9–85.7 | N/A | 261 | |
Antigen | Lateral flow immunoassay | Urine | 80 | 97–100 | 262 | |
Legionella pneumophila serotype 1 | Antigen | Immunoassay | Urine | 74 | 99.1 | 192 |
DNA | PCR | Urine/serum | 64–73 | 100 | 263 | |
DNA | PCR | Throat swab | 88.2 | 100 | 264 | |
Mycoplasma pneumoniae | Antibody | Complement fixation | Serum | 65 | 97 | 265 |
Antibody (IgG and IgM) | Enzyme immunoassay | Serum | 35–77 | 49–100 | 265 | |
DNA | PCR | Throat swab | 62 | 96 | 266 | |
Nocardia asteroides group | DNA | PCR | Tissue/sputum/BAL | 100 | 100 | 267 |
Streptococcus pneumoniae | Antigen | Lateral flow immunoassay | Urine | 67–82 | 93–99.8 | 189 |
DNA | PCR | Plasma or sputum | 26–100 | 58–99 | 268 | |
Streptococcus pyogenes | Antigen | Enzyme immunoassay | Throat swab | 70–90 | 90–100 | 269 |
MYCOBACTERIA | ||||||
M. tuberculosis complex | DNA | NAT | Smear negative | 33.3–92.9 | N/A | 91 |
DNA | NAT | Smear positive | 85.7–94.6 | 98 | 91 | |
Organism | Microscopy | Carbolfuchsin | 32–94 | N/A | 90 | |
Organism | Microscopy | Fluorochrome (HIV−) | 52–97 | N/A | 90 | |
Organism | Microscopy | Fluorochrome (HIV+) | 26–100 | N/A | 90 | |
INVASIVE FUNGI | ||||||
Aspergillus species | DNA | PCR | Serum | 80 | 100 | 270 |
Antigen | Galactomannan enzyme immunoassay | Serum | 71 | 89 | 203 | |
Blastomyces dermatitidis | Antigen | Enzyme immunoassay | Urine | 80.7–92.9 | 77–79† | 271, 272 |
Antigen | Enzyme immunoassay | Serum | 81.8 | 100† | 272 | |
Antibody | Immunodiffusion | Serum | 28 | 100 | 273 | |
Antibody | Enzyme immunoassay | Serum | 77–100 | 86–96 | 273 | |
Antibody | Complement fixation | Serum | 9 | 100 | 273 | |
Organism | Microscopy | Body fluid/tissue | 38–97 | N/A | 274 | |
Coccidioides species | DNA | Real-time PCR | Respiratory sample | 92.9–100 | 98.1–98.4 | 275 |
Antibody (IgG) | Complement fixation | Serum | 67–75 | N/A | 237 | |
Serum (IC patients) | 33–100 | N/A | 237 | |||
Antibody (IgG and IgM) | Immunodiffusion | Serum | 53–73 | N/A | 237 | |
Serum (IC patients) | 0–75 | N/A | 237 | |||
Antibody (IgG and IgM) | Enzyme immunoassay | Serum | 75–92.6 | 84.6–98.3 | 276 | |
Serum (IC patients) | 25–90 | N/A | 237 | |||
Cryptococcus neoformans (and Cryptococcus gattii) | Antigen | Latex agglutination | Serum | 83–91.1 | 92.9–100 | 277, 278 |
Antigen | Latex agglutination | Urine | N/A | 100 | 277 | |
Antigen | Latex agglutination | CSF | 93–100 | 93–98 | 278 | |
Antigen | Lateral flow assay | Serum | 90.1–100 | 92.9–100 | 279 | |
Antigen | Lateral flow assay | Urine | 70.3–94.4 | 100 | 277 | |
Antigen | Enzyme immunoassay | Serum | 94.1–100 | 93–100 | 278, 279 | |
Antigen | Enzyme immunoassay | Urine | 92% | Unknown | 279 | |
Histoplasma capsulatum | Antibody (IgG) | Complement fixation | Serum/urine | 72.8–94.3 | 70–80 | 280 |
Antibody (IgM) | Microimmunodiffusion | Serum | 70–100 | 100 | 280, 281 | |
Antibody (IgG) | Enzyme immunoassay | Serum | 91–100 | 66–97 | 280, 281 | |
Serum (AIDS, disseminated) | 69.2 | N/A | 199 | |||
Serum (other IC patients, disseminated) | 84.2 | N/A | 199 | |||
Serum (non-IC patients, disseminated) | 85.7 | N/A | 199 | |||
Serum (pulmonary subacute infection) | 92.3 | N/A | 199 | |||
Antigen | Enzyme immunoassay | Urine | 30.4–100 | Variable | 199 | |
Urine (AIDS, disseminated) | 92.1 | N/A | 199 | |||
Urine (other IC patients, disseminated) | 93.5 | N/A | 199 | |||
Urine (non-IC patients, disseminated) | 63.6 | N/A | 199 | |||
Urine (pulmonary subacute infection) | 38.9 | N/A | 199 | |||
Antibody | Latex agglutination | Serum | 65–97 | 39 | 280 | |
Pneumocystis jirovecii | DNA | PCR | Respiratory samples | 93 | 90 | 158 |
Organism | Microscopy with silver stain | Induced sputum/BAL | 86–92 | 92–97 | 98 | |
Antigen | Direct fluorescent antibody | Induced sputum/BAL | 90–97 | 85–90 | 98 | |
Antigen | Indirect fluorescent antibody | Induced sputum/BAL | 86–97 | 100 | 98 | |
Organism | Diff-Quik stain | Induced sputum/BAL | 81–92 | 97–100 | 98 | |
Antigen | β-d-glucan assay | Serum/plasma | 78–100 | 70–100 | 218 | |
Other (fungi excluding P. jirovecii) | Antigen | β-d-glucan assay | Serum/plasma | 76.8 | 85.3 | 218 |
PROTOZOA | ||||||
Toxoplasma gondii | Antibody (IgG) | Sabin-Feldman dye test Enzyme immunoassay Immunofluorescence antibody IgG avidity |
Serum | N/A | N/A | * |
Antibody (IgG and IgM) | Agglutination | Serum | N/A | N/A | * | |
DNA | PCR | Serum/CSF/aqueous humor/BAL | 15–85 | 95 | 282 |
AIDS, acquired immunodeficiency syndrome; BAL, bronchoalveolar lavage; CSF, cerebrospinal fluid; HIV, human immunodeficiency virus; IC, immunocompromised; Ig, immunoglobulin; N/A, not available; NAT, nucleic acid test; PCR, polymerase chain reaction.
Testing for acute Q fever and toxoplasmosis should be done using a battery of tests that must be interpreted together because sensitivity and specificity of individual tests are not available.
Cross reaction is seen with Histoplasma capsulatum.
Microscopy
Microscopic examination of lower respiratory tract specimens offers a rapid approach to detection and identification of many pathogens. However, as discussed earlier, a major limitation of microscopic examination is that it cannot distinguish between infection, colonization, and contamination when the specimen is collected through the oropharynx.78, 79, 80 In addition, microscopy lacks sensitivity in specimens with less than 104 CFU per milliliter. Microscopy does routinely provide valuable information on the quality of specimen and the type of inflammatory response present. Specimens demonstrating a preponderance of polymorphonuclear leukocytes, ciliated columnar epithelial cells, or alveolar macrophages with few, if any, squamous epithelial cells (<10 per low-power field) represent lower respiratory tract secretions. The presence of alveolar macrophages is a more specific marker of lower respiratory tract secretions than neutrophils and is more likely to be associated with a significantly lower incidence of oropharyngeal contamination.81 The finding of neutrophils with intracellular organisms is considered indicative of an active infectious process.82, 83, 84
The Gram-stained smear is an essential and necessary part of evaluation of sputum and tracheal aspirates for determining the quality and acceptability of specimens for bacterial culture74, 76 and for providing a rapid assessment of the most likely etiologic agent of the pneumonia. Although Gram stains might also suggest the presence of mycobacteria, fungi, and parasites, special stains should be ordered when those pathogens are suspected. The Gram stain also stains squamous epithelial cells, ciliated columnar epithelial cells, neutrophils, and alveolar macrophages, which are used for assessment of specimen quality and inflammatory response. Table 17-7 shows criteria used by the laboratory to interpret findings on Gram stain and report them to the physician. Although it is impossible to correlate every staining pattern to a particular pathogen, several Gram stain patterns are pathognomonic for a particular pathogen or clinical entity (Table 17-8 ).
Table 17-7.
Number of Bacteria Found per Field under Oil Immersion 100× Objective | Bacterial Quantity Reported to Clinician |
---|---|
0 | Negative |
<1 | 1+ (rare) |
1–5 | 2+ (few) |
6–30 | 3+ (moderate) |
>30 | 4+ (heavy) |
Table 17-8.
Pattern Reported | Pathogen or Entity Suggested |
---|---|
Intracellular organisms | Active infection |
Gram-positive cocci in pairs (lancet-shaped diplococci) and short chains | Streptococcus pneumoniae |
Pleomorphic gram-negative coccobacilli | Haemophilus influenzae |
Gram-negative diplococci | Moraxella catarrhalis* |
Gram-positive cocci in clusters | Staphylococcus aureus |
Mixed morphotypes of gram-positive and gram-negative rods, cocci, and coccobacilli | Aspiration pneumonia |
Beaded gram-positive or gram-variable rods | Actinomycetales order, which includes genera Mycobacterium, Actinomyces, Corynebacterium |
Filamentous branching gram-positive or gram-variable rods | Nocardia, Actinomyces |
Cannot be distinguished from Neisseria meningitidis.
The accuracy of Gram stain for detection of infection depends on the stringency of criteria. In assessing patients with acute CAP, Rein and colleagues85 found that three or more gram-positive lancet-shaped diplococci (Fig. 17-2 ) correctly predicted the presence of pneumococci in corresponding cultures in 90% of cases, with a sensitivity and specificity of 62% and 85%, respectively. As expected, improving the sensitivity of the Gram stain examination by lowering the criteria for positivity resulted in reduced specificity in the diagnosis of pneumococcal infection. Similar levels of sensitivity of Gram-stained smears have been reported by others in identifying pneumococci as well as H. influenzae (Fig. 17-3 ) in sputum specimens from patients with acute CAP.86, 87 For the diagnosis of pneumococcal pneumonia, a combination of Gram-stained smear and culture of sputum can yield the correct diagnosis in more than 80% of patients who received less than 24 hours of effective antibiotic therapy.88
Direct examination of sputum for the identification of other organisms that can either be commensals or causes of acute bacterial pneumonia in adults, such as H. influenzae, M. catarrhalis, and N. meningitidis, may also be problematic.7, 78, 80, 89 Their role as etiologic agents of pneumonia is strongly suggested by the finding of large numbers of gram-negative coccobacilli (H. influenzae) and diplococci (M. catarrhalis and N. meningitidis) located within and outside of neutrophils in sputum specimens.82, 83, 84 Because sputum microscopy (and subsequent culture) can give misleading results, the diagnosis of Haemophilus, Moraxella, or meningococcal pneumonia can be confirmed only by invasive techniques. Because invasive techniques are infrequently performed to identify the etiologic agents of acute CAP, the sensitivity and specificity of sputum Gram-stained smears cannot be determined reliably in such cases. A similar challenge is faced when trying to distinguish between colonization and infection by gram-negative bacilli. For example, up to 108 CFU of gram-negative bacilli per milliliter may be found in respiratory secretions of patients on mechanical ventilation in intensive care units without evidence of pneumonia.12
Acid-fast staining is the method of choice for visualization of mycobacteria in respiratory specimens. Laboratories may use either carbolfuchsin or fluorochrome to stain mycobacteria. Table 17-9 shows criteria used by the laboratory to report findings on the acid-fast stain. In most studies the carbolfuchsin-based stain has a sensitivity of 60% or lower compared to culture.90 The fluorochrome stain, which uses fluorescent dye such as auramine or auramine-rhodamine to highlight mycobacteria, is on average 10% more sensitive than carbolfuchsin90 and is therefore the method recommended by the World Health Organization for screening sputum smears for mycobacteria. The sensitivity of acid-fast microscopy depends on the bacillary burden, sample volume, host immune status, staining method, and other variables.91 The major limiting factor is that approximately 104 acid-fast bacilli per milliliter of sputum must be present to be visualized under light microscopy using an acid-fast stain.91 The insensitivity of smear microscopy for TB therefore necessitates the use of more sensitive methods such as culture and nucleic acid tests.92, 93 In addition, although acid-fast stains have high specificity, they cannot distinguish between M. tuberculosis complex and nontuberculous mycobacteria, and carbolfuchsin stains also stain Legionella micdadei (also known as Tatlockia micdadei).94 Modified acid-fast stain, a modified carbolfuchsin stain, is used for direct staining of partially acid-fast–positive organisms such as Nocardia, Tsukamurella, Rhodococcus, and Gordonia.5
Table 17-9.
No. of AFB Found under Oil Immersion 100× Objective (Carbolfuchsin Stain) | No. of AFB Found under 10× Objective (Fluorochrome Stain) | Bacterial Quantity Reported to Clinician |
---|---|---|
0 per 300 fields | 0 per 30 fields | Negative |
1-2 per 300 fields | 1-2 per 30 fields | ± (suspicious) |
1-9 per 100 fields | 1-9 per 10 fields | 1+ |
1-9 per 10 fields | 1-9 per field | 2+ |
1-9 per field | 10-90 per field | 3+ |
>9 per field | >90 per field | 4+ |
AFB, acid-fast bacilli.
Adapted from David HL: Bacteriology of the mycobacterioses, Atlanta, GA, DHEW Publication No. (CDC) 76-8316, 1976:153.
Immunofluorescence examination by DFA staining is an alternative method for direct visualization of organisms. The diagnosis of legionellosis is usually made by a combination of direct immunofluorescence examination and culture of respiratory specimens, and antigen and antibody testing. DFA staining can be performed on sputum, endotracheal aspirate, bronchial washing, and lung tissue specimens, with sensitivities ranging from 25% to 66% for the diagnosis of L. pneumophila pneumonia and specificities of more than 94%95 (Fig. 17-4 ). Both clinical and technical variables account for the broad range of sensitivity of this test, and the accuracy of this method for detection of pneumonia due to other Legionella species is less precisely known. In the absence of other supporting evidence, a positive DFA result is generally not accepted as sufficient for the diagnosis of Legionella infection, and other confirmatory measures should be undertaken.
DFA testing has also been found to be sensitive and specific in detecting Chlamydia trachomatis in nasopharyngeal specimens from infants with pneumonia and has also been applied to sputum and BAL for detection of P. jirovecii, the causative agent of Pneumocystis pneumonia.96, 97 Silver stain, direct immunofluorescence, indirect immunofluorescence, and Diff-Quik (a modified Giemsa stain) have all been found to have greater than 90% sensitivity for detecting P. jirovecii in induced sputum and BAL samples from human immunodeficiency virus (HIV)-infected patients98 (Fig. 17-5 ). All of these staining techniques have lower sensitivity in patients who are not infected with HIV, but DFA is consistently more sensitive than the other staining techniques.99, 100
DFA has also been applied to respiratory secretions for the diagnosis of respiratory virus infections. DFA testing can be performed in 1 to 4 hours and is typically more sensitive than rapid antigen tests.77, 101 However, DFA testing for respiratory viruses requires a high level of technical and interpretive expertise, is difficult to adapt to the high throughput required for pandemic or high-volume testing, and remains less sensitive than real-time polymerase chain reaction (PCR).77, 102 DFA for viral detection has been phased out in many clinical laboratories as rapid, sensitive, multiplexed molecular diagnostic respiratory virus tests have become available.
The visualization of fungal elements in respiratory secretions requires the use of special stains. Historically, potassium hydroxide was used to degrade host tissue and visualize fungal elements (Fig. 17-6 ). Calcofluor white stain, a fluorochrome that binds to chitin and cellulose present in the fungal cell wall, is now commonly added to potassium hydroxide or used alone to provide better delineation of fungal elements (Fig. 17-7 ).103, 104 Table 17-10 lists staining patterns that are suggestive of certain fungal pathogens. It is important to note that identification of fungi based on microscopic appearance of fungal elements in lung tissue or secretions is subject to error, and definitive identification must be deferred to culture.105
Table 17-10.
Key Patterns Reported from Respiratory Specimens | Pathogens the Findings Suggest |
---|---|
Broad-based budding yeast (see Fig. 17-6) | Blastomyces dermatitidis |
Narrow-based budding yeast (Fig. 17-8) | Candida, Cryptococcus, Histoplasma capsulatum, Sporothrix schenckii |
Spherule (Fig. 17-9) | Coccidioides immitis, Coccidioides posadasii |
Nonseptate hyphal element (Fig. 17-10) | Zygomycetes (cannot be predicted from formalin-fixed paraffin-embedded tissue) |
Septate hyphal element (Fig. 17-11) | Monomorphic hyaline and dematiaceous molds, including Aspergillus |
The identification of pulmonary parasites such as Strongyloides stercoralis and Paragonimus spp. is typically made by microscopic examination of respiratory secretions. S. stercoralis larvae and rarely eggs can be seen on most stains but are sufficiently large that they are more likely to be found using a low-power objective (Fig. 17-12 ).106 The diagnosis of microfilariae causing tropical pulmonary eosinophilia requires peripheral blood parasite examination of nightly blood samples because these parasites typically circulate in the blood only at night, which coincides with activity of its insect vector.107 Echinococcus cysts may be detected in pulmonary cyst fluid, and Entamoeba histolytica may be seen in association with pleural disease, if an amebic liver abscess erodes through the diaphragm.
Culture
Microbiologic culture of respiratory specimens allows definitive identification of the suspected pathogens and permits determination of bacterial, mycobacterial, and yeast susceptibility to antimicrobial agents. For cultivation of particular groups or species of microorganisms, laboratories must inoculate processed samples on one or more culture media supplemented with nutrients suitable for cultivation of the desired microorganisms and inhibitors for selective inhibition of undesirable organisms. The clinician must therefore be aware that, although many organisms do grow on routine aerobic and anaerobic cultures, a number of respiratory pathogens such as Mycoplasma, Legionella, Mycobacterium, Nocardia, Rhodococcus, and Histoplasma capsulatum require pathogen-specific growth conditions (growth supplements, temperature and carbon dioxide requirements, and incubation times), which have to be specified when fastidious pathogens are part of the differential diagnosis.5 For example, Legionella species require culture media supplemented with l-cysteine and α-ketoglutarate (buffered charcoal yeast extract),95 slow-growing mycobacteria require media enriched with a lipid extract and antimicrobials to limit the growth of oral commensals,91 and H. capsulatum requires extended incubation time up to 4 weeks.108 Although most Nocardia species grow well on mycobacterial culture media as well as on ordinary bacterial and fungal culture media, optimal recovery from clinical specimens is obtained by using the same buffered charcoal yeast extract culture medium as that for the isolation of Legionella. 109
Laboratories commonly use a semiquantitative culturing method and report the number of colonies present in consecutive streaked quadrants using a 1+ to 4+ grading system. Table 17-11 lists criteria for reporting of semiquantitative cultures. It is important to note that with the semiquantitative culture method, the volume of specimen cultured is not standardized. Quantitative cultures are also performed on certain respiratory specimens such as protected catheter brush specimens and BAL fluid.110 Nonetheless, a randomized trial found that in mechanically ventilated patients with pneumonia, the use of quantitative BAL and nonquantitative endotracheal aspirate cultures resulted in similar clinical outcomes, although patients known to be colonized or infected with Pseudomonas aeruginosa or methicillin-resistant Staphylococcus aureus were excluded and antibiotics were withheld until the results of culture were available.52 Bacteria present in quantities greater than 103 CFU/mL in cultures of protected catheter brushes48 and in quantities greater than 104 CFU/mL in cultures of BAL fluids50 are defined by the laboratory as positive and identified and tested for their susceptibility to appropriate antimicrobial agents.111 The diagnostic value of break points of bacterial growth (i.e., 103 to 105 CFU/mL) depends not only on the type of microbiologic processing used but also on the relationship of two variables: the concentration of pathogens present in the BAL fluid and the degree of contamination of the bronchoscopic channel through which lavage fluid was injected and aspirated. Other variables affecting the sensitivity of BAL specimens include antibiotic administration and the volume of lavage fluid injected and the volume of fluid retrieved.112 Diagnostic specificity depends greatly on techniques used to minimize contamination of the specimen by upper respiratory flora, such as discarding the first aliquot of aspirated fluid.113
Table 17-11.
NO. OF COLONIES PRESENT IN CONSECUTIVE STREAKED QUADRANTS |
|||
---|---|---|---|
Grade | First | Second | Third |
1+ | <10 | 0 | 0 |
2+ | >10 | <5 | 0 |
3+ | >10 | >5 | <5 |
4+ | >10 | >5 | >5 |
Based on data from Waites KB, Saubolle MA, Talkington DF, et al: Cumitech 10A: laboratory diagnosis of upper respiratory tract infections (Sharp SF, coordinating editor), Washington, DC, 2006, ASM Press.
Because many pathogens of the lower respiratory tract are also members of the oropharyngeal flora, culture results must be correlated with the Gram stain findings, including the presence or absence of polymorphonuclear leukocytes. Respiratory pathogens seen on Gram stain to be predominant with typical morphologic characteristics both within and outside polymorphonuclear leukocytes are reported and identified to the species level. Upper respiratory colonization with potentially pathogenic microorganisms, such as gram-negative bacilli, may not be related to the actual etiologic agents of lower respiratory tract infection. Sputum specimens contaminated with Enterobacteriaceae or S. aureus from oropharyngeal secretions may obscure the diagnosis of pneumococcal pneumonia, anaerobic pleuropulmonary infection, or even TB.114, 115, 116, 117 Except for Cryptococcus, yeasts are considered to be of upper respiratory origin and are not routinely identified.117
The sensitivity and specificity of cultures depend on the pathogen burden, specimen type, collection method, the cytologic screening criteria applied to ensure sampling of lower respiratory tract secretions, and the threshold of colony count to distinguish infection from contamination. In 1971 Barrett-Connor114 showed that only 45% of patients with bacteremic pneumococcal pneumonia had pneumococci isolated from their sputum cultures, whereas 27% of patients had moderate to heavy growth of another potential pathogen in these cultures. In contrast, fungal cultures of respiratory specimens were positive in approximately 85% of cases with disseminated or chronic pulmonary histoplasmosis.118, 119 We now better understand how careful specimen collection, cytologic screening of specimens to discard those contaminated with oropharyngeal secretions, and use of the results of the Gram-stained smear to guide identification of isolates in culture all contribute to the diagnostic value of sputum culture in acute pneumococcal pneumonia.
It is important for the clinician to have knowledge of the turnaround time of all tests, including cultures. The time to detection of positive culture results is dependent on the number of organisms in the inoculum and the replication rate of the pathogen.108 Table 17-12 shows typical turnaround times for lower respiratory tract pathogens.
Table 17-12.
Key Respiratory Pathogens | Time to Detection in Culture |
---|---|
Acinetobacter baumannii, Aspergillus, Coccidioides immitis, Coccidioides posadasii, Cryptococcus, Escherichia coli, Haemophilus influenzae, Klebsiella pneumoniae, Moraxella catarrhalis, Mycobacterium abscessus group, Mycobacterium chelonae, Neisseria meningitidis, Nocardia,*Pseudomonas aeruginosa, Staphylococcus aureus, Streptococcus pneumoniae, Zygomycetes | 1–3 days |
Actinomyces, Legionella,*Sporothrix schenckii | 3–5 days |
Blastomyces dermatitidis, Histoplasma capsulatum, Mycobacterium avium-intracellulare complex, Mycobacterium tuberculosis complex, Mycoplasma pneumoniae | 1–4 weeks |
May take longer.
Adapted from Hove MG, Woods GL: Duration of fungal culture incubation in an area endemic for Histoplasma capsulatum. Diagn Microbiol Infect Dis 28:41, 1997.
Fecal culture may be useful in cases of suspected pulmonary involvement with S. stercoralis when sputum cytologic examination fails to identify larvae. The agar plate method, which looks for tracking of bacteria by the motile larvae, is a useful adjunct to standard microscopic fecal examination and may be up to six times more sensitive.120, 121, 122, 123
Viral culture techniques previously played an important role in the diagnosis of respiratory virus infections.124 However, traditional viral culture is laborious, requires significant technical and interpretive expertise, allows the isolation of only a limited range of disease-causing viruses, and has a long turnaround time that limits clinical utility.125 Shell vial culture is an improved method in which a sample is centrifuged onto a layer of cells, with subsequent detection of viral antigen. Shell vial cultures, particularly those using a mixture of mink lung and A549 cells, provide equivalent to improved sensitivity compared to traditional culture, with more rapid turnaround time.126, 127, 128 Similar to respiratory virus DFA, routine respiratory viral cultures are being phased out in many clinical laboratories with the widespread availability of respiratory virus molecular diagnostic tests. However, in BAL samples from transplant recipients and other immunocompromised patients, shell vial cultures using human fibroblast cell lines may be clinically useful for detection of cytomegalovirus (CMV)129, 130 and traditional viral cultures may be useful for recovery of herpes simplex virus.131, 132, 133
Antimicrobial Susceptibility Testing
Antimicrobial susceptibility testing is performed to assist clinicians with the selection of appropriate targeted antibiotic therapy to optimize clinical outcomes. There are several aspects of this in vitro testing that are important to understand. First of all, testing methods and interpretation of results must be done according to accepted standards, such as the Clinical and Laboratory Standards Institute (CLSI), the U.S. Food and Drug Administration (FDA), or the European Committee on Antimicrobial Susceptibility Testing (EUCAST) for the various categories of organism. Second, the selection of antibiotics to test and report is determined in collaboration with CLSI/EUCAST guidelines, the hospital formulary, infectious disease specialists, the pharmacy, and the infection prevention committee. Third, antimicrobial susceptibility testing should not be performed when the pathogen has a predictable susceptibility profile (e.g., all Streptococcus pyogenes are currently susceptible to penicillin), nor is susceptibility testing needed for a specific antibiotic when an organism has intrinsic resistance to that antibiotic (e.g., Enterobacter spp., Klebsiella spp., Citrobacter spp., and Serratia spp. are all intrinsically resistant to ampicillin). The bacterial pathogens from the lower respiratory tract, for which the susceptibility profile is not predictable and thus antimicrobial susceptibility testing is commonly performed, are S. pneumoniae, S. aureus, H. influenzae, M. catarrhalis, the Enterobacteriaceae, P. aeruginosa, and other nonfermenting gram-negative rods. Fourth, because antimicrobial susceptibility testing measures in vitro activity, other factors must be considered when determining in vivo activity, including antimicrobial pharmacokinetics and pharmacodynamics and patient-specific factors such as immune status.
Phenotypic susceptibility testing methods used by the clinical laboratory consist of the disk diffusion method, which generates a qualitative result based on the zone size of bacterial growth inhibition, and the dilution method, which generates a quantitative result: the minimum inhibitory concentration. Results generated by each method are reported as: “susceptible,” “intermediate,” or “resistant.” Susceptible implies the organism will likely respond to treatment with the antibiotic at a standard dosage. Intermediate may be effective if higher dosing can be used (e.g., β-lactams) or the antibiotic being used concentrates at the site of infection (e.g., fluoroquinolones for urinary tract infections). Resistant implies the organism is not likely to respond to therapy with that antibiotic. The interpretation criteria (susceptible, intermediate, resistant) are specific to each organism-drug combination as well as to pharmacokinetic (e.g., peak serum levels, protein binding, and clearance rate of the drug) and pharmacodynamic (e.g., whether the rate of bacterial killing is concentration dependent) characteristics of each drug. Therefore, because the measurement of the minimum inhibitory concentration alone does not capture these multiple considerations, simply choosing a drug on the basis of the lowest minimum inhibitory concentration in a susceptibility report is not recommended.
Genotypic susceptibility testing is also possible for certain pathogen-drug combinations when a monogenic mutation accurately predicts a resistant phenotype. Examples include the mecA gene for methicillin resistance in Staphylococcus, 134 vanA and vanB for vancomycin resistance in Enterococcus, 135 and specific rpoB mutations in rifampin resistance in M. tuberculosis.136
Not all pathogens isolated in the laboratory can be reliably tested for antimicrobial susceptibility. For organisms such as Chlamydophila, Mycoplasma, Legionella, nontuberculous mycobacteria, and molds, there are currently no standard test methods or interpretive criteria. For these pathogens, clinical experience, use of consensus guidelines, and careful assessment of patient response to antimicrobial therapy is most valuable for optimal patient management.
Nucleic Acid Tests
In recent years, technological advances in nucleic acid testing (NAT) and instrument automation have revolutionized the simplicity, speed, and accuracy of detecting fastidious pathogens directly from respiratory specimens.136, 137, 138 In many laboratories these tests have replaced conventional, less sensitive, more laborious methods for routine use. With the unique capabilities of molecular diagnostic tests and the need for rapid, sensitive detection of respiratory pathogens, molecular assays will continue to gain an increasing role in the diagnosis and management of patients with opportunistic and community-acquired pneumonia. Most NATs are based on amplification and detection of nucleic acid targets specific to the pathogens of interest. NATs offer several advantages over conventional direct examination, microbiologic cultures, and serologic assays. First, NATs have the ability to detect and identify pathogens rapidly (in hours). Second, NATs provide the only means of detection for some microorganisms that are difficult or impossible to grow in culture. Certain viruses, for example, are very difficult to cultivate by conventional culture-based methods, and NATs enable detection of these organisms in clinical specimens.139 Third, NATs make possible detection of pathogens, such as M. tuberculosis, in resource-limited settings where laboratory infrastructure for culture is lacking.140, 141 Finally, for some pathogens, NATs allow determination of antimicrobial susceptibility directly from respiratory tract specimens.
A number of commercial and in-house NATs are available for direct detection of M. tuberculosis in sputum. These assays can yield results in 2 to 8 hours. For example, in comparison to conventional cultures for M. tuberculosis, NATs have high sensitivities (86% to 97%) and specificities (98%) in smear-positive respiratory samples.91 In smear-negative, culture-positive specimens, NATs can confirm the presence of M. tuberculosis in 33% to 96% of samples, weeks earlier than culture.91 The sensitivity further improves with smear-negative sputa if the assay is performed on one to two additional samples.136 Detection of M. tuberculosis susceptibility to first- and second-line drugs can also be accomplished directly from sputum.142, 143 Commercial NATs detect rifampin and isoniazid resistance with sensitivity of 94% to 99% and 88% to 95%, respectively.91, 143, 144, 145 Based on the improved performance of NATs over smear microscopy and the rapid turnaround time compared to culture, the U.S. Centers for Disease Control and Prevention (CDC) recommended that NAT “testing should be performed on at least one respiratory specimen from each patient with signs and symptoms of pulmonary TB for whom a diagnosis of TB is being considered but has not yet been established, and for whom the test result would alter case management or TB control activities.”93 In addition, the World Health Organization has recommended the use of NATs to screen for multidrug-resistant TB. Three such NATs are commercially available in Europe and elsewhere, and one has been approved by the FDA as of 2014.
Diagnostic methods have also been described for detecting the nucleic acids of C. pneumoniae,7, 41, 42 M. pneumoniae, 40, 44, 45 L. pneumophila, 40 dimorphic fungi,146 monomorphic fungi,147, 148, 149, 150 P. jirovecii, 151, 152 Toxoplasma gondii, 153 respiratory viruses,139 herpes simplex virus,132 and CMV154, 155 directly in respiratory specimens of normal and immunocompromised hosts. Accuracy studies have indicated that most of these assays are at least comparable to, if not better than, conventional culture, direct antigen, and/or serologic detection methods, especially when examining respiratory specimens that contain low numbers of pathogens. For example, a rapid PCR assay has been applied for the diagnosis of an outbreak of Chlamydia psittaci that resulted from transmission of this infectious agent to humans from birds purchased in stores; in this outbreak, PCR detected 50% more cases than did culture.156 Compared to culture and serologic tests, PCR was also shown to be the most sensitive method for detection of C. pneumoniae during an outbreak of CAP.157 However, NAT results should be interpreted with caution because NATs currently cannot differentiate between organisms that inhabit the upper airway without causing disease and those that are responsible for the patient's illness. For example, as mentioned, S. pneumoniae, C. pneumoniae, and M. pneumoniae may inhabit the upper airway without causing disease.6, 10 Similarly, the lowered diagnostic specificity of P. jirovecii NATs is likely due to the detection of P. jirovecii nucleic acid from cysts that are present in low numbers and in a latent state in pulmonary tissues of asymptomatic patients.158, 159, 160
In patients with culture-negative infection, or when cultures were not performed on a tissue biopsy sample before fixation, a broad-range PCR assay coupled with amplicon sequencing can be used for detection and identification of bacterial and fungal DNA from fresh or formalin-fixed paraffin-embedded (FFPE) specimens.161, 162, 163, 164 The bacterial 16S ribosomal RNA gene and the fungal ribosomal RNA operon (encoding 5.8S, 18S, 28S ribosomal subunit genes with the internal transcribed spacer regions [ITS1 and ITS2]) are the most reliable and frequently used targets for identifying bacterial and fungal sequences, respectively.165 As with all NATs, sensitivity is higher from fresh tissue than from FFPE specimens: for example, fungal sequencing was successfully completed on 97% of specimens when performed on fresh specimens compared to 63% to 70% when performed on FFPE specimens.163 Despite the many advantages of direct bacterial and fungal identification by sequencing, clinicians must be aware that the success of this method is dependent on the amount of specimen submitted (e.g., open biopsy versus needle biopsy), the pathogen burden, and whether fresh versus FFPE tissue is tested. It is imperative that testing is strictly limited to samples obtained from sterile sources because contamination of the sample with commensal organisms or environmental spores could yield false-positive results and lead to mismanagement of the patient.163, 164 Sequence results must always be correlated with clinical, histopathologic, and ancillary test results (antibody or antigen detection) to ensure the clinical accuracy of sequence results.
The diagnosis of respiratory virus infections (Table 17-13 ) has been revolutionized by nucleic acid amplification testing.139 These tests are now considered more sensitive than all other current methods of virus detection, including viral culture and DFA testing, discussed earlier, as well as rapid antigen tests, discussed later. Following the 2009 influenza A H1N1 pandemic, there was a tremendous increase in the availability and widespread implementation of real-time PCR assays for influenza and other respiratory viruses. These real-time PCR assays include those endorsed by the CDC, as well as numerous laboratory-developed and commercially produced tests.139 However, in-house real-time PCR tests are of high complexity, requiring experienced and highly skilled staff, as well as specialized molecular diagnostic laboratory facilities. Furthermore, these tests are generally batched, thereby prolonging the turnaround time and reducing clinical utility. Finally, real-time PCR allows only a moderate level of multiplexing (i.e., use of primers and probes for detecting multiple targets in the same reaction), a potential issue for detecting a broad range of respiratory viruses with a limited number of reactions. To address these issues, a variety of commercial test systems have been developed, though no single system provides comprehensive detection of clinically relevant respiratory viruses with optimal sensitivity in a format that can be performed at or near the point of care. Three groups of molecular testing now exist, with different levels of technical and personnel demands (referred to as moderate or high complexity, as established by the Clinical Laboratory Improvement Amendments).
Table 17-13.
STANDARD RESPIRATORY VIRUS TEST PANEL |
Influenza A, B |
Respiratory syncytial virus |
Human Metapneumovirus |
Parainfluenza 1, 2, 3 |
Adenovirus |
EXPANDED RESPIRATORY VIRUS PANEL (MAY INCLUDE ONE OR MORE) |
Rhinovirus |
Enterovirus* |
Human Coronavirus (229E, HKU1, OC43, NL63) |
Human bocavirus (HBoV)† |
ADDITIONAL VIRUSES TO CONSIDER IN THE IMMUNE COMPROMISED |
Cytomegalovirus |
Herpes simplex virus |
Varicella-zoster virus |
Human herpesvirus 6 |
OTHER VIRUSES THAT CAUSE LOWER RESPIRATORY TRACT INFECTION |
Hantavirus |
Measles virus |
Parechovirus |
Parainfluenza virus 4 |
Influenza C |
Polyomavirus (BK, WU, K1) |
EMERGING SEVERE ACUTE RESPIRATORY VIRUSES |
Avian influenza (H5N1, H7N9) |
Human coronavirus—severe acute respiratory syndrome |
Human coronavirus—Middle Eastern respiratory syndrome |
Commercial multiplex nucleic acid amplification tests may not distinguish between Rhinovirus and Enterovirus.
Human bocavirus detection in respiratory specimens is of uncertain clinical significance, though this virus is included in several commercial multiplex nucleic acid amplification tests.
One group of molecular tests includes limited respiratory virus panels of moderate complexity on FDA-cleared, sample-to-answer platforms that combine the speed and simplicity of rapid antigen tests with the sensitivity of nucleic acid amplification. These tests include Xpert Flu (Cepheid; Sunnyvale, CA), which detects influenza A and B in about 1 hour, and Verigene Respiratory Virus Plus (Nanosphere; Northbrook, IL), which detects and subtypes influenza A and B, and detects and types respiratory syncytial virus (RSV) in less than hours. Though more sensitive than traditional respiratory virus diagnostics, Xpert Flu is slightly less sensitive than in-house real-time PCR assays.101, 166, 167, 168, 169, 170, 171 Performance of the Verigene Respiratory Virus Plus appears comparable to in-house real-time PCR.172 These assays have not yet been compared to each other.
A second group of tests are the high-complexity, FDA-cleared, comprehensive respiratory virus panels that require separate extraction and PCR amplification steps before multiplex nucleic acid detection. These tests include the xTAG Respiratory Virus Panels (Luminex; Austin, TX): xTAG RVPv1 and xTAG RVP FAST, as well as the eSensor Respiratory Virus Panel (GenMark Diagnostics; Carlsbad, CA). The xTAG RVPs achieve high-level multiplexing through the combination of target amplicon labeling with universal, minimally cross-hybridizing, complementary oligonucleotide sequences and flow cytometric detection using a solution-phase array composed of spectrally distinct microspheres.173 In approximately 8 hours, RVPv1 detects influenza A, including subtyping H1 and H3, influenza B, RSV A, RSV B, parainfluenza virus 1, 2, and 3, human metapneumovirus, adenovirus, and rhinovirus/enterovirus. Compared to RVPv1, RVP FAST has a shorter time to result (about 6 hours); however, it does not detect the parainfluenza viruses, and it does not distinguish between RSV types. Several studies have compared the clinical performance of the xTAG RVPs and found that both the xTAG RVPv1 and RVP FAST panels are more sensitive than traditional respiratory virus testing methodologies, less sensitive than in-house real-time PCR, and that RVPv1 is more sensitive than RVP FAST.174, 175, 176, 177, 178, 179, 180 Of note, these assays demonstrate low sensitivity for adenoviruses and do not distinguish between the closely related picornaviruses, rhinovirus, and enterovirus.
In this same group the eSensor RVP achieves high-level multiplexing through competitive hybridization and electrochemical detection using a microfluidic device containing an array of single-stranded oligonucleotide capture probes. In approximately 8 hours the eSensor RVP detects influenza A, including H1, 2009 H1, and H3 subtyping, influenza B, RSV A, RSV B, parainfluenza virus 1, 2, and 3, human metapneumovirus, adenovirus C, adenovirus B/E, and rhinovirus. The eSensor RVP demonstrates comparable performance to in-house real-time PCR, including detection of adenovirus and rhinovirus, and at this time appears to be the most sensitive of the multiplex respiratory virus assays.180, 181 Both the xTAG and eSensor RVP assays have research-only versions or versions approved outside of the United States that offer detection of parainfluenza 4, human coronaviruses 229E, HKU1, OC43, and NL63 and, for xTAG RVP, bocavirus and the severe acute respiratory syndrome coronavirus. Determination of the clinical utility of these expanded panels requires additional study.
The third group of molecular respiratory virus tests are of moderate complexity and combine the simplicity and speed of rapid antigen testing with the sensitivity and multiplexing capability of the xTAG or eSensor RVP. At present, one assay is available in this category: the FilmArray Respiratory Panel (BioFire Diagnostics; Salt Lake City, UT), which integrates sample preparation, nested-PCR amplification, real-time fluorescent detection, and analysis in a single assay pouch.182 In about 1 hour, including less than 5 minutes of hands-on time, this assay detects influenza A, including H1, 2009 H1, and H3 subtyping, influenza B, RSV, parainfluenza virus 1, 2, 3, and 4, human metapneumovirus, adenovirus, rhinovirus/enterovirus, and human coronaviruses 229E, HKU1, OC43, and NL63. In addition, it detects three bacterial species: Bordetella pertussis, C. pneumoniae, and M. pneumoniae. Clinical performance is similar to xTAG RVPv1, demonstrating higher sensitivity than traditional respiratory virus testing methodologies and lower sensitivity compared to in-house real-time PCR.177, 179, 180, 183, 184, 185, 186, 187, 188 The current Film Array Respiratory Panel also has low sensitivity for adenoviruses, does not distinguish between rhinovirus and enterovirus, and has low throughput, because only one test can be performed on an instrument at a time. However, its rapid turnaround and ease of use allows most laboratories to perform multiplexed molecular respiratory virus testing. In the future it is expected that assays in this category will improve sensitivity, specificity, and throughput to create optimal, rapid multiplex molecular respiratory virus tests.
Given the variety of nucleic acid tests that are currently available for the diagnosis of respiratory virus infection and the rapid development of new NATs, it is important to communicate with the laboratory to confirm the viruses included in the local panel, the expected turnaround time, and local test performance characteristics. Furthermore, note that nasopharyngeal swabs and aspirate/washes are the only specimen types that have received FDA clearance so far, so it is also important to verify that the local laboratory has validated the use of lower respiratory tract specimens in their test system before sending such samples for testing.
Antigen Testing
The diagnosis of lower respiratory tract infections can be aided by detection of pathogen-specific antigens in serum or other body fluids. Antigen detection offers an alternative to direct examination of infected tissue and may play a role in the detection of pathogens that grow poorly, or not at all, in culture.
Urinary antigen assays may have value for adults with S. pneumoniae and L. pneumophila infections. In a meta-analysis of 27 studies using a composite of culture tests as the reference standard, the pooled sensitivity for direct antigen detection of S. pneumoniae in the urine of adults with CAP was 74% (95% confidence interval [CI], 67% to 82%) and specificity was 97% (95% CI, 93% to 100%).189 In children, pneumococcal antigen in urine is less specific for invasive infection, because it was detected in 43% of children with only nasopharyngeal colonization.190 Similarly, in children with pneumonia, detection of H. influenzae type B antigens in urine is of potential diagnostic value, but transient antigenuria may follow immunization with H. influenzae type B conjugate vaccine.191 In the evaluation of adults for legionnaires' disease, urine L. pneumophila serogroup 1 antigen has a high negative predictive value with pooled sensitivity of 74% (95% CI, 68% to 81%) and specificity of 99% (95% CI, 98% to 100%).192 Urine antigen for L. pneumophila is detectable in 80% to 89% of patients with legionnaires' disease beginning with the first 3 days of symptoms and continuing for at least 14 days; the duration of antigenuria was reduced by antibiotic therapy and was detectable for up to 42 days, especially in immunocompromised patients.193 The urinary antigen assays are limited to detection of infections due to L. pneumophila serogroup 1 and not other Legionella serogroups or species.94
Detection of fungal antigen in serum, urine, and other body fluids is used as an aid in the diagnosis of infections due to Cryptococcus neoformans, Aspergillus, H. capsulatum, and P. jirovecii. Assays to detect capsular polysaccharides of C. neoformans in serum or cerebrospinal fluid are essential for rapid diagnosis of cryptococcosis. Commercially available assays show sensitivity ranging from 83% to 97% and specificity from 93% to 100%.194 Cryptococcal antigen may also be detectable in pleural fluid and BAL fluid of patients with cryptococcal pneumonia.195, 196 Serial measurement of serum antigen titers over time is not useful for management of patients with pulmonary cryptococcosis.197 Cryptococcal antigen may be falsely positive due to the presence of rheumatoid factor or heterophile antibodies (i.e., antibodies produced to poorly defined antigens with weak affinity and multispecific activities) and falsely negative due to the prozone effect (antigen excess), localized infection, infection with a poorly encapsulated strain, or low organism burden.194
Several commercial H. capsulatum antigen assays with variable accuracies are available for diagnosis of histoplasmosis.198 The polysaccharide antigen of H. capsulatum can be detected in urine in approximately 90% of patients with disseminated disease and 75% with diffuse acute pulmonary histoplasmosis.118, 119 A recent multicenter study identified antigenuria in 91.8% of 158 patients with disseminated histoplasmosis, 83.3% of 6 patients with acute histoplasmosis, 30.4% of 46 patients with subacute infection, and 87.5% of 8 patients with chronic histoplasmosis; antigenemia was detected in 100% of 31 patients with disseminated infection.199 Urinary Histoplasma antigen levels persist during ongoing active infection, become undetectable with successful therapy, and rise with relapse of infection. The specificity of the Histoplasma antigen assay was 99% in patients with nonfungal infections and in healthy controls199; however, the assay is known to yield positive results in patients with disseminated infections caused by Blastomyces dermatitidis, Coccidioides immitis, Paracoccidioides brasiliensis, Aspergillus, and Penicillium marneffei. 200, 201 Cross reactivity of the assay has not been observed in patients with invasive candidiasis, cryptococcosis, or other opportunistic systemic mycoses.200
At least one commercial laboratory offers antigen tests for the diagnosis of coccidioidomycosis and blastomycosis, but both tests exhibit significant cross reaction with H. capsulatum. 202
Aspergillus galactomannan is a major cell wall component of the fungus, and detection of this antigen has been studied in many different clinical situations. Galactomannan detected in serum by enzyme immunoassay can aid in the early diagnosis of invasive pulmonary aspergillosis, with pooled diagnostic sensitivity of 71% (95% CI, 68% to 74%) and specificity of 89% (95% CI, 88% to 90%) for proven cases of invasive aspergillosis.203 The reported sensitivity and specificity range from 40% to 100% and 56% to 100%, respectively, in various patient groups17, 147, 204, 205, 206 The Platelia Aspergillus test (Bio-Rad Laboratories, Hercules, CA), a commercially available assay that detects galactomannan from A. fumigatus, A. flavus, A. niger, A. versicolor, and A. terres, has been shown to yield positive results at an early stage of infection, with positive and negative predictive values of more than 90% in high-risk patients who were tested biweekly.204 However, this assay may cross react with Histoplasma, P. brasiliensis, Penicillium, Paecilomyces, Alternaria, and Cryptococcus, 206, 207, 208 and false-positive galactomannan antigen assay results can be observed in patients receiving certain foods or intravenous piperacillin-tazobactam, amoxicillin, or ticarcillin.208, 209, 210, 211 Cross reactivity with Listeria monocytogenes has also been reported.212 BAL fluid specimens from lung transplant recipients have also been evaluated for testing by Aspergillus galactomannan detection assay, with sensitivity of 82% and specificity of 96%.213 Of note, the Plasma-Lyte (Baxter International Inc., Deerfield, IL) solution commonly used to perform BAL has been found to yield false-positive results with the Platelia Aspergillus test.214 Overall, assays for galactomannan show promise, but currently a single positive result has limited value and cross reactivity is common.
Another fungal antigen used for the diagnosis of invasive fungal infection is (1→3)-β-d-glucan, which is a component of the outer cell wall of saprophytic and pathogenic fungi except Zygomycetes (Mucor and Rhizopus species) and Cryptococcus species204, 215 This antigen has been detected in serum or other body fluids of patients with invasive aspergillosis, invasive candidiasis, and infections caused by Fusarium, Acremonium, Trichosporum, Scedosporium, Saccharomyces, and P. jirovecii.149, 216, 217 A meta-analysis for diagnosis of invasive fungal infection showed a pooled sensitivity of 77% (95% CI, 67% to 84%), and specificity of 85% (95% CI, 80% to 90%).218 Use of different assay cutoff values may result in differences in sensitivity and specificity among the various commercially available assays for detecting this antigen.215, 219 For the diagnosis of Pneumocystis pneumonia in HIV-positive patients, the sensitivity of the (1→3)-β-d-glucan assay was 92% and specificity was 65%.220 In pneumocystis pneumonia, β-d-glucan levels do not correlate with organism burden, Pneumocystis pneumonia severity, or response to therapy.220 Cross reactivity of β-d-glucan assays has been reported with the use of cotton gauzes, swabs, packs, pads, or sponges for wound care or surgery; cellulose filters in hemodialysis patients; and various antimicrobial agents including piperacillin-tazobactam.215, 221
Rapid antigen tests for influenza A and B are commonly used in both ambulatory and inpatient settings. Rapid detection of influenza is critical to allow prompt treatment with antiviral agents, to reduce the risk for further transmission through implementation of infection control practices, and to reduce inappropriate use of antibiotics.222, 223 A meta-analysis evaluating 159 studies and 26 different rapid influenza antigen tests reported a sensitivity of 62% (95% CI, 58% to 67%) and a specificity of 98% (95% CI, 98% to 99%).223, 224 Because of this low sensitivity, physicians should consider following negative rapid influenza antigen tests with more sensitive testing, particularly if specimens are collected during times of high influenza prevalence.225 Rapid antigen tests for RSV are also available and may similarly aid in patient triage, infection control, and antibiotic management, particularly in pediatric patients. The sensitivities and specificities of these RSV rapid antigen tests range from 59% to 89% and 93% to 100%, respectively.124, 226
Serologic Testing and Interferon-γ Release Assays
The cause of lower respiratory tract infections can be suggested by detection and quantitation of humoral (e.g., antibody) responses to pathogens. In addition, latent TB infection (LTBI) can be diagnosed by detection of cellular immune responses to M. tuberculosis antigens, such as the interferon-γ release assays (IGRAs). Serologic testing is used commonly to identify infections due to pathogens that are difficult to detect by other conventional methods, to evaluate the course of an infection, and to determine the nature of the infection (primary infection versus reinfection, acute versus chronic infection). Serologic testing and IGRAs are less sensitive in patients with compromised immune systems and therefore cannot be used to rule out infection.227, 228, 229 When possible, microbiologic culture and NATs on respiratory secretions or lung tissue should be performed to detect and confirm the presence of pathogens in immunosuppressed patients who may not be able to mount antibody or cell-mediated immune responses.
The serologic methods commonly used in diagnostic laboratories include enzyme immunoassay, immunoprecipitation, immunodiffusion (ID), complement fixation (CF), immunoblotting (including Western blot), agglutination, hemagglutination inhibition, and indirect immunofluorescence assay. Serologic results are often expressed as a titer, which is the inverse of the greatest dilution, or lowest concentration of a patient's serum that retains measurable specific antibody-antigen reactivity (e.g., dilution of 1 : 16 = titer of 16). A fourfold or greater rise in antibody titer between acute and convalescent sera is usually required for diagnosis. An elevated pathogen-specific IgM antibody titer in a single serum sample suggests recent infection, and a falling titer provides further support for the etiologic significance of this organism. However, false-positive immunoglobulin (Ig) M antibody tests are not rare. Thus serologic testing of pathogen-specific IgG antibody in acute and convalescent sera remains the approach to establish a specific microbial cause of the infection.230
Various commercial assays are available for detection of specific IgM and/or IgG antibodies to respiratory tract pathogens.231 These assays are useful for supporting or confirming the diagnosis of bacterial infections caused by C. pneumoniae, Legionella species, F. tularensis, Y. pestis, C. trachomatis, C. pneumoniae, C. psittaci, and Coxiella burnetii. Although antibody testing is commonly used for the detection of infection with M. pneumoniae, a recent study was unable to distinguish between infection and asymptomatic colonization with this organism.10 The diagnosis of C. pneumoniae infection can also be a problem. In some additional instances, such as infection with M. tuberculosis, commercial serologic tests have been shown to be inconsistent and imprecise, which is the basis of World Health Organization policy statement advising against use of existing serologic tests in TB.232
M. pneumoniae infection is often diagnosed by the presence of specific antibodies in serum.233 Cold agglutinins, detected by agglutination of type O Rh-negative red blood cells at 4° C, are present in the sera of approximately 50% of patients with M. pneumoniae infection, and levels decline to baseline within 6 weeks after acute infection.233 However, cold agglutinins are nonspecific. Antibodies to a chloroform-methanol glycolipid extract of M. pneumoniae are detected by a CF test in more than 85% of culture-positive patients; a single elevated titer of greater than 80 or a greater than fourfold rise in titer between acute and convalescent sera is required to establish a diagnosis. Enzyme immunoassays to detect IgM and IgA antibodies that specifically recognize M. pneumoniae membrane proteins have been developed with improved sensitivity and specificity over the CF assay.233 Specific IgM antibodies appear during the first week of illness and reach peak titers during the third week. However, the IgM antibodies to M. pneumoniae are not consistently produced in adults because of prior sensitization, so that a negative IgM result does not rule out acute M. pneumoniae infection, particularly in older adults. Detection of specific IgA antibodies in the serum has been shown by one group to be a reliable approach for diagnosis, because these antibodies are also produced early in the course of disease and more reliably present in the infected individuals regardless of age.233 Others, however, found them of little value.10
Serologic testing plays an important role in the diagnosis of fungal respiratory tract infections due to C. immitis and H. capsulatum.234 For C. immitis a diagnosis of infection can be based on detection of antibodies to antigens derived from the coccidioidal mycelia or spherules, although there may be cross reactivity with other yeasts and dimorphic fungi. Antibodies to C. immitis can be detected by ID, CF, and enzyme immunoassay. Precipitin-specific IgM antibodies develop in up to 75% of individuals within 2 to 3 weeks after primary C. immitis infection and subsequently disappear except in patients with disseminated infection. Complement-fixing IgG antibodies appear later and persist in relation to the severity of disease, but decline with disease remission. Titers of 32 or higher suggest the possibility of disseminated infection.235, 236 The sensitivity of serologic testing drops 8% to 20% in immunocompromised hosts compared to immunocompetent patients.237
For H. capsulatum, serum antibodies are detected by CF using both yeast and mycelial antigens and an ID assay, which show increased titers in more than 90% of patients with pulmonary histoplasmosis and approximately 80% with disseminated disease.119 The CF test is more sensitive but less specific than the ID test for the diagnosis of subclinical and acute pulmonary histoplasmosis.238 Antibodies become detectable first by CF at 2 to 6 weeks after Histoplasma infection and then by ID 2 to 4 weeks later. However, the ID test remains positive longer than the CF test after resolution of infection, becoming negative 2 to 5 years later. Antibody levels remain high in those with chronic pulmonary infection, progressive disseminated disease, or mediastinal fibrosis. Commercially available serologic tests for blastomycosis exist but suffer from limited accuracy and are of minimal value in patient care.239
Serologic testing is also useful for diagnosis of parasitic infections, especially Paragonimus, T. gondii and S. stercoralis, 240, 241 and for diagnosis of extraintestinal E. histolytica disease. Serologic testing plays an especially important role for screening prospective organ transplant recipients and other patients considered for immunosuppressive therapies.
Serologic testing plays a limited role as an aid to the diagnosis of respiratory viral infections. Although detection of recent respiratory virus infection, for example with influenza A, may be determined via seroconversion or a fourfold or greater rise in antibody titer in a convalescent relative to an acute serum sample, the requirement for two temporally distinct specimens makes serologic results unlikely to factor in clinical decision making.242 In contrast, routine CMV serologic testing in transplant donors and recipients provides valuable information about the risk for subsequent CMV-related sequelae, including the development of respiratory disease.243
IGRAs are in vitro assays used to measure T-cell responses to M. tuberculosis–specific antigens, such as ESAT-6, CFP-10, and TB7.7.244 Two FDA-approved commercial IGRAs are currently available: the QuantiFERON-TB Gold In-Tube assay (QFT-GIT; Qiagen, Carnegie, Australia) and the T-SPOT.TB assay (Oxford Immunotec, Abingdon, United Kingdom). IGRAs were developed as an alternative to the tuberculin skin test (TST) for diagnosis of LTBI.245, 246 Compared to the TST, IGRAs have improved specificity for distinguishing between the responses due to bacillus Calmette-Guérin (BCG) vaccination and latent TB infection, because the antigens used in IGRAs, ESAT-6 and CFP-10, are absent from all strains of BCG. Also, compared to the TST, IGRAs offer logistical advantages because they do not depend on accurate intradermal injection and patients do not have to return to a health facility for the result to be read.244 However, like TSTs, IGRAs cannot distinguish between LTBI and active disease.247 The sensitivity of IGRAs in culture-positive active TB cases has ranged from 65% to 100%,244, 248, 249 and the sensitivity in patients with LTBI who progressed to active TB ranged from 40% to 100%.250 Both IGRAs and the TST have been shown to have similar sensitivity in adults in low- and middle-income countries.251 However, the accuracy of IGRA testing appears to falter at the borderline levels of positivity. Studies conducted in health care workers in low-incidence settings have shown highly variable IGRA results with serial testing. The rate of conversions (negative to positive result using the manufacturer- and CDC-recommended cutoff of ≥0.35 IU/mL) ranged from 2% to 15%, and the rate of reversions (positive to negative result) ranged from 20% to 40%.252, 253 Those with borderline results around the assay cutoff are more likely to revert or convert. Finally, there are limitations to the use of IGRAs. IGRA results have not proven useful for monitoring response to TB treatment. Similarly, IGRAs are no more able than the TST to predict which patients with positive results will go on to develop active TB.254, 255 Currently, therefore, IGRA is most useful as an alternative to the TST, especially in subject populations with a high incidence of BCG vaccination.
Acknowledgment
We thank Stanford Pathology residents Natalia Isaza, Albert Tsai, and Lee Frederick Schroeder for contributing to this chapter.
Key Points.
-
▪
Diagnostic testing should be ordered only if the result will alter treatment decisions.
-
▪
The clinician plays a critical role in preventing accidental laboratory exposure by notifying the microbiology laboratory when highly virulent and transmissible agents are suspected as the cause of disease.
-
▪
Syndromic order sets can improve the accuracy and efficiency of test selection and thus facilitate accurate diagnosis of infectious diseases.
-
▪
Lower respiratory tract secretions collected through the oropharynx are nearly always contaminated with resident microflora of the oral cavity, and therefore microscopy, culture, and nucleic acid test results must be interpreted in the context of other clinical evidence and diagnostic findings.
-
▪
Nucleic acid tests can facilitate rapid and accurate diagnosis of lower respiratory tract infections, especially those caused by viruses and pathogens that are difficult to culture.
Complete reference list available at ExpertConsult.
Key Readings
- Mackowiak PA, Martin RM, Jones SR. Pharyngeal colonization by gram-negative bacilli in aspiration-prone persons. Arch Intern Med. 1978;138:1224–1227. [PubMed] [Google Scholar]
- Mills CC. Occurrence of Mycobacterium other than Mycobacterium tuberculosis in the oral cavity and in sputum. Appl Microbiol. 1972;24:307–310. doi: 10.1128/am.24.3.307-310.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pai M, Elwood K. Interferon-gamma release assays for screening of health care workers in low tuberculosis incidence settings: dynamic patterns and interpretational challenges. Can Respir J. 2012;19:81–83. doi: 10.1155/2012/420392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pauker SG, Kassirer JP. Therapeutic decision making: a cost-benefit analysis. N Engl J Med. 1975;293:229–234. doi: 10.1056/NEJM197507312930505. [DOI] [PubMed] [Google Scholar]
- Rangaka MX, Wilkinson KA, Glynn JR. Predictive value of interferon-gamma release assays for incident active tuberculosis: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12:45–55. doi: 10.1016/S1473-3099(11)70210-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spuesens EB, Fraaij PL, Visser EG. Carriage of Mycoplasma pneumoniae in the upper respiratory tract of symptomatic and asymptomatic children: an observational study. PLoS Med. 2013;10:e1001444. doi: 10.1371/journal.pmed.1001444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tillotson JR, Finland M. Bacterial colonization and clinical superinfection of the respiratory tract complicating antibiotic treatment of pneumonia. J Infect Dis. 1969;119:597–624. doi: 10.1093/infdis/119.6.597. [DOI] [PubMed] [Google Scholar]
- Valenti WM, Trudell RG, Bentley DW. Factors predisposing to oropharyngeal colonization with gram-negative bacilli in the aged. N Engl J Med. 1978;298:1108–1111. doi: 10.1056/NEJM197805182982002. [DOI] [PubMed] [Google Scholar]
- Versalovic J, Carroll KC, Funke G. ed 10. ASM Press; Washington, DC: 2011. Manual of clinical microbiology. [Google Scholar]
- Zwerling A, van den Hof S, Scholten J. Interferon-gamma release assays for tuberculosis screening of healthcare workers: a systematic review. Thorax. 2012;67:62–70. doi: 10.1136/thx.2010.143180. [DOI] [PubMed] [Google Scholar]
References
- 1.Dellit TH, Owens RC, McGowan JE., Jr Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardship. Clin Infect Dis. 2007;44(2):159–177. doi: 10.1086/510393. [DOI] [PubMed] [Google Scholar]
- 2.Tenover FC, Arbeit RD, Goering RV. How to select and interpret molecular strain typing methods for epidemiological studies of bacterial infections: a review for healthcare epidemiologists. Molecular Typing Working Group of the Society for Healthcare Epidemiology of America. Infect Control Hosp Epidemiol. 1997;18(6):426–439. doi: 10.1086/647644. [DOI] [PubMed] [Google Scholar]
- 3.Nguyen DT, Yeh E, Perry S. Real-time PCR testing for mecA reduces vancomycin usage and length of hospitalization for patients infected with methicillin-sensitive staphylococci. J Clin Microbiol. 2010;48(3):785–790. doi: 10.1128/JCM.02150-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Perez K, Olsen R, Musick W. Integrating rapid pathogen identification and antimicrobial stewardship significantly decreases hospital costs. Arch Pathol Lab Med. 2013;137(9):1247–1254. doi: 10.5858/arpa.2012-0651-OA. [DOI] [PubMed] [Google Scholar]
- 5.Versalovic J, Carroll KC, Funke G. ed 10. ASM Press; Washington, DC: 2011. Manual of clinical microbiology. [Google Scholar]
- 6.Bik EM, Long CD, Armitage GC. Bacterial diversity in the oral cavity of 10 healthy individuals. ISME J. 2010;4(8):962–974. doi: 10.1038/ismej.2010.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kuklinska D, Kilian M. Relative proportions of Haemophilus species in the throat of healthy children and adults. Eur J Clin Microbiol. 1984;3(3):249–252. doi: 10.1007/BF02014895. [DOI] [PubMed] [Google Scholar]
- 8.Paisley JW, Lauer BA, McIntosh K. Pathogens associated with acute lower respiratory tract infection in young children. Pediatr Infect Dis. 1984;3(1):14–19. doi: 10.1097/00006454-198401000-00005. [DOI] [PubMed] [Google Scholar]
- 9.Graffelman AW, Knuistingh Neven A, le Cessie S. Pathogens involved in lower respiratory tract infections in general practice. Br J Gen Pract. 2004;54(498):15–19. [PMC free article] [PubMed] [Google Scholar]
- 10.Spuesens EB, Fraaij PL, Visser EG. Carriage of Mycoplasma pneumoniae in the upper respiratory tract of symptomatic and asymptomatic children: an observational study. PLoS Med. 2013;10(5):e1001444. doi: 10.1371/journal.pmed.1001444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mills CC. Occurrence of Mycobacterium other than Mycobacterium tuberculosis in the oral cavity and in sputum. Appl Microbiol. 1972;24(3):307–310. doi: 10.1128/am.24.3.307-310.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Johanson WG, Jr, Pierce AK, Sanford JP. Nosocomial respiratory infections with gram-negative bacilli: the significance of colonization of the respiratory tract. Ann Intern Med. 1972;77(5):701–706. doi: 10.7326/0003-4819-77-5-701. [DOI] [PubMed] [Google Scholar]
- 13.Tillotson JR, Finland M. Bacterial colonization and clinical superinfection of the respiratory tract complicating antibiotic treatment of pneumonia. J Infect Dis. 1969;119(6):597–624. doi: 10.1093/infdis/119.6.597. [DOI] [PubMed] [Google Scholar]
- 14.Mackowiak PA, Martin RM, Jones SR. Pharyngeal colonization by gram-negative bacilli in aspiration-prone persons. Arch Intern Med. 1978;138(8):1224–1227. [PubMed] [Google Scholar]
- 15.Valenti WM, Trudell RG, Bentley DW. Factors predisposing to oropharyngeal colonization with gram-negative bacilli in the aged. N Engl J Med. 1978;298(20):1108–1111. doi: 10.1056/NEJM197805182982002. [DOI] [PubMed] [Google Scholar]
- 16.Fainstein V, Rodriguez V, Turck M. Patterns of oropharyngeal and fecal flora in patients with acute leukemia. J Infect Dis. 1981;144(1):10–18. doi: 10.1093/infdis/144.1.10. [DOI] [PubMed] [Google Scholar]
- 17.Hope WW, Walsh TJ, Denning DW. Laboratory diagnosis of invasive aspergillosis. Lancet Infect Dis. 2005;5(10):609–622. doi: 10.1016/S1473-3099(05)70238-3. [DOI] [PubMed] [Google Scholar]
- 18.Laird AT. A method for increasing the diagnostic value of sputum reports. JAMA. 1909;52:294–296. [Google Scholar]
- 19.Warren JR, Bhattacharya M, De Almeida KN. A minimum 5.0 ml of sputum improves the sensitivity of acid-fast smear for Mycobacterium tuberculosis. Am J Respir Crit Care Med. 2000;161(5):1559–1562. doi: 10.1164/ajrccm.161.5.9908063. [DOI] [PubMed] [Google Scholar]
- 20.Davis JL, Cattamanchi A, Cuevas LE. Diagnostic accuracy of same-day microscopy versus standard microscopy for pulmonary tuberculosis: a systematic review and meta-analysis. Lancet Infect Dis. 2013;13(2):147–154. doi: 10.1016/S1473-3099(12)70232-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.LaRocque RC, Katz JT, Perruzzi P. The utility of sputum induction for diagnosis of Pneumocystis pneumonia in immunocompromised patients without human immunodeficiency virus. Clin Infect Dis. 2003;37(10):1380–1383. doi: 10.1086/379071. [DOI] [PubMed] [Google Scholar]
- 22.Jefferson H, Dalton HP, Escobar MR. Transportation delay and the microbiological quality of clinical specimens. Am J Clin Pathol. 1975;64(5):689–693. doi: 10.1093/ajcp/64.5.689. [DOI] [PubMed] [Google Scholar]
- 23.Penn RL, Silberman R. Effects of overnight refrigeration on the microscopic evaluation of sputum. J Clin Microbiol. 1984;19(2):161–163. doi: 10.1128/jcm.19.2.161-163.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pal N, Sharma B, Malhotra B. Transport and storage of sputum specimen by using cetylpyridinium chloride for isolation of mycobacteria. Indian J Pathol Microbiol. 2009;52(1):59–61. doi: 10.4103/0377-4929.44966. [DOI] [PubMed] [Google Scholar]
- 25.Smithwick RW, Stratigos CB, David HL. Use of cetylpyridinium chloride and sodium chloride for the decontamination of sputum specimens that are transported to the laboratory for the isolation of Mycobacterium tuberculosis. J Clin Microbiol. 1975;1(5):411–413. doi: 10.1128/jcm.1.5.411-413.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tazir M, David HL, Boulahbal F. Evaluation of the chloride and bromide salts of cetylpyridium for the transportation of sputum in tuberculosis bacteriology. Tubercle. 1979;60(1):31–36. doi: 10.1016/0041-3879(79)90053-9. [DOI] [PubMed] [Google Scholar]
- 27.Hariri AR, Hempel HO, Kimberlin CL. Effects of time lapse between sputum collection and culturing on isolation of clinically significant fungi. J Clin Microbiol. 1982;15(3):425–428. doi: 10.1128/jcm.15.3.425-428.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bordelon JY, Jr, Legrand P, Gewin WC. The telescoping plugged catheter in suspected anaerobic infections. A controlled series. Am Rev Respir Dis. 1983;128(3):465–468. doi: 10.1164/arrd.1983.128.3.465. [DOI] [PubMed] [Google Scholar]
- 29.Pollock HM, Hawkins EL, Bonner JR. Diagnosis of bacterial pulmonary infections with quantitative protected catheter cultures obtained during bronchoscopy. J Clin Microbiol. 1983;17(2):255–259. doi: 10.1128/jcm.17.2.255-259.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yeh E, Luo RF, Dyner L. Preferential lower respiratory tract infection in swine-origin 2009 A(H1N1) influenza. Clin Infect Dis. 2010;50(3):391–394. doi: 10.1086/649875. [DOI] [PubMed] [Google Scholar]
- 31.Daley P, Castriciano S, Chernesky M. Comparison of flocked and rayon swabs for collection of respiratory epithelial cells from uninfected volunteers and symptomatic patients. J Clin Microbiol. 2006;44(6):2265–2267. doi: 10.1128/JCM.02055-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hernes SS, Quarsten H, Hagen E. Swabbing for respiratory viral infections in older patients: a comparison of rayon and nylon flocked swabs. Eur J Clin Microbiol Infect Dis. 2011;30(2):159–165. doi: 10.1007/s10096-010-1064-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Abu-Diab A, Azzeh M, Ghneim R. Comparison between pernasal flocked swabs and nasopharyngeal aspirates for detection of common respiratory viruses in samples from children. J Clin Microbiol. 2008;46(7):2414–2417. [Google Scholar]
- 34.Chan KH, Peiris JS, Lim W. Comparison of nasopharyngeal flocked swabs and aspirates for rapid diagnosis of respiratory viruses in children. J Clin Virol. 2008;42(1):65–69. doi: 10.1016/j.jcv.2007.12.003. [DOI] [PubMed] [Google Scholar]
- 35.Debyle C, Bulkow L, Miernyk K. Comparison of nasopharyngeal flocked swabs and nasopharyngeal wash collection methods for respiratory virus detection in hospitalized children using real-time polymerase chain reaction. J Virol Methods. 2012;185(1):89–93. doi: 10.1016/j.jviromet.2012.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Robinson JL, Lee BE, Kothapalli S. Use of throat swab or saliva specimens for detection of respiratory viruses in children. Clin Infect Dis. 2008;46(7):e61–e64. doi: 10.1086/529386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lieberman D, Lieberman D, Shimoni A. Identification of respiratory viruses in adults: nasopharyngeal versus oropharyngeal sampling. J Clin Microbiol. 2009;47(11):3439–3443. doi: 10.1128/JCM.00886-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hammitt LL, Kazungu S, Welch S. Added value of an oropharyngeal swab in detection of viruses in children hospitalized with lower respiratory tract infection. J Clin Microbiol. 2011;49(6):2318–2320. doi: 10.1128/JCM.02605-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kim C, Ahmed JA, Eidex RB. Comparison of nasopharyngeal and oropharyngeal swabs for the diagnosis of eight respiratory viruses by real-time reverse transcription-PCR assays. PLoS One. 2011;6(6):e21610. doi: 10.1371/journal.pone.0021610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Welti M, Jaton K, Altwegg M. Development of a multiplex real-time quantitative PCR assay to detect Chlamydia pneumoniae, Legionella pneumophila and Mycoplasma pneumoniae in respiratory tract secretions. Diagn Microbiol Infect Dis. 2003;45(2):85–95. doi: 10.1016/s0732-8893(02)00484-4. [DOI] [PubMed] [Google Scholar]
- 41.Tondella ML, Talkington DF, Holloway BP. Development and evaluation of real-time PCR-based fluorescence assays for detection of Chlamydia pneumoniae. J Clin Microbiol. 2002;40(2):575–583. doi: 10.1128/JCM.40.2.575-583.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Reischl U, Lehn N, Simnacher U. Rapid and standardized detection of Chlamydia pneumoniae using LightCycler real-time fluorescence PCR. Eur J Clin Microbiol Infect Dis. 2003;22(1):54–57. doi: 10.1007/s10096-002-0858-2. [DOI] [PubMed] [Google Scholar]
- 43.Kuoppa Y, Boman J, Scott L. Quantitative detection of respiratory Chlamydia pneumoniae infection by real-time PCR. J Clin Microbiol. 2002;40(6):2273–2274. doi: 10.1128/JCM.40.6.2273-2274.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ursi D, Dirven K, Loens K. Detection of Mycoplasma pneumoniae in respiratory samples by real-time PCR using an inhibition control. J Microbiol Methods. 2003;55(1):149–153. doi: 10.1016/s0167-7012(03)00131-3. [DOI] [PubMed] [Google Scholar]
- 45.Hardegger D, Nadal D, Bossart W. Rapid detection of Mycoplasma pneumoniae in clinical samples by real-time PCR. J Microbiol Methods. 2000;41(1):45–51. doi: 10.1016/s0167-7012(00)00135-4. [DOI] [PubMed] [Google Scholar]
- 46.Stover DE, Zaman MB, Hajdu SI. Bronchoalveolar lavage in the diagnosis of diffuse pulmonary infiltrates in the immunosuppressed host. Ann Intern Med. 1984;101(1):1–7. doi: 10.7326/0003-4819-101-1-1. [DOI] [PubMed] [Google Scholar]
- 47.van der Eerden MM, Vlaspolder F, de Graaff CS. Value of intensive diagnostic microbiological investigation in low- and high-risk patients with community-acquired pneumonia. Eur J Clin Microbiol Infect Dis. 2005;24(4):241–249. doi: 10.1007/s10096-005-1316-8. [DOI] [PubMed] [Google Scholar]
- 48.Cook DJ, Fitzgerald JM, Guyatt GH. Evaluation of the protected brush catheter and bronchoalveolar lavage in the diagnosis of nosocomial pneumonia. J Intensive Care Med. 1991;6(4):196–205. doi: 10.1177/088506669100600405. [DOI] [PubMed] [Google Scholar]
- 49.Fagon JY. Hospital-acquired pneumonia: diagnostic strategies: lessons from clinical trials. Infect Dis Clin North Am. 2003;17(4):717–726. doi: 10.1016/s0891-5520(03)00071-0. [DOI] [PubMed] [Google Scholar]
- 50.Meduri GU, Baselski V. The role of bronchoalveolar lavage in diagnosing nonopportunistic bacterial pneumonia. Chest. 1991;100(1):179–190. doi: 10.1378/chest.100.1.179. [DOI] [PubMed] [Google Scholar]
- 51.Ramírez P, Valencia M, Torres A. Bronchoalveolar lavage to diagnose respiratory infections. Semin Respir Crit Care Med. 2007;28(5):525–533. doi: 10.1055/s-2007-991524. [DOI] [PubMed] [Google Scholar]
- 52.Canadian Critical Care Trials Group A randomized trial of diagnostic techniques for ventilator-associated pneumonia. N Engl J Med. 2006;355(25):2619–2630. doi: 10.1056/NEJMoa052904. [DOI] [PubMed] [Google Scholar]
- 53.Parashar D, Kabra SK, Lodha R. Does neutralization of gastric aspirates from children with suspected intrathoracic tuberculosis affect mycobacterial yields on MGIT culture? J Clin Microbiol. 2013;51(6):1753–1756. doi: 10.1128/JCM.00202-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zar HJ, Workman L, Isaacs W. Rapid molecular diagnosis of pulmonary tuberculosis in children using nasopharyngeal specimens. Clin Infect Dis. 2012;55(8):1088–1095. doi: 10.1093/cid/cis598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Nicol MP, Spiers K, Workman L. Xpert MTB/RIF testing of stool samples for the diagnosis of pulmonary tuberculosis in children. Clin Infect Dis. 2013;57(3):e18–e21. doi: 10.1093/cid/cit230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Taylor N, Gaur RL, Baron EJ. Can a simple flotation method lower the limit of detection of Mycoplasma pneumoniae in extrapulmonary samples analyzed by the GeneXpert MTB/RIF assay? J Clin Microbiol. 2012;50(7):2272–2276. doi: 10.1128/JCM.01012-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Metersky ML, Ma A, Bratzler DW. Predicting bacteremia in patients with community-acquired pneumonia. Am J Respir Crit Care Med. 2004;169(3):342–347. doi: 10.1164/rccm.200309-1248OC. [DOI] [PubMed] [Google Scholar]
- 58.Luna CM, Videla A, Mattera J. Blood cultures have limited value in predicting severity of illness and as a diagnostic tool in ventilator-associated pneumonia. Chest. 1999;116(4):1075–1084. doi: 10.1378/chest.116.4.1075. [DOI] [PubMed] [Google Scholar]
- 59.Mayer J. Laboratory diagnosis of nosocomial pneumonia. Semin Respir Infect. 2000;15(2):119–131. doi: 10.1053/srin.2000.9594. [DOI] [PubMed] [Google Scholar]
- 60.Abe T, Tokuda Y, Ishimatsu S. Usefulness of initial blood cultures in patients admitted with pneumonia from an emergency department in Japan. J Infect Chemother. 2009;15(3):180–186. doi: 10.1007/s10156-009-0682-z. [DOI] [PubMed] [Google Scholar]
- 61.Shah SS, Dugan MH, Bell LM. Blood cultures in the emergency department evaluation of childhood pneumonia. Pediatr Infect Dis J. 2011;30(6):475–479. doi: 10.1097/INF.0b013e31820a5adb. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cockerill FR, 3rd, Wilson JW, Vetter EA. Optimal testing parameters for blood cultures. Clin Infect Dis. 2004;38(12):1724–1730. doi: 10.1086/421087. [DOI] [PubMed] [Google Scholar]
- 63.Lee A, Mirrett S, Reller LB. Detection of bloodstream infections in adults: How many blood cultures are needed? J Clin Microbiol. 2007;45(11):3546–3548. doi: 10.1128/JCM.01555-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Pohlman JK, Kirkley BA, Easley KA. Controlled clinical comparison of Isolator and BACTEC 9240 Aerobic/F resin bottle for detection of bloodstream infections. J Clin Microbiol. 1995;33(10):2525–2529. doi: 10.1128/jcm.33.10.2525-2529.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Cockerill FR, 3rd, Reed GS, Hughes JG. Clinical comparison of BACTEC 9240 plus aerobic/F resin bottles and the Isolator aerobic culture system for detection of bloodstream infections. J Clin Microbiol. 1997;35(6):1469–1472. doi: 10.1128/jcm.35.6.1469-1472.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hellinger WC, Cawley JJ, Alvarez S. Clinical comparison of the Isolator and BacT/Alert aerobic blood culture systems. J Clin Microbiol. 1995;33(7):1787–1790. doi: 10.1128/jcm.33.7.1787-1790.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Waite RT, Woods GL. Evaluation of BACTEC MYCO/F lytic medium for recovery of mycobacteria and fungi from blood. J Clin Microbiol. 1998;36(5):1176–1179. doi: 10.1128/jcm.36.5.1176-1179.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Pohlman JK, Kirkley BA, Easley KA. Controlled clinical evaluation of BACTEC plus Aerobic/F and BacT/Alert aerobic FAN bottles for detection of bloodstream infections. J Clin Microbiol. 1995;33(11):2856–2858. doi: 10.1128/jcm.33.11.2856-2858.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Mathers G. Bacteriology of the urine in lobar pneumonia. J Infect Dis. 1916;19:416–418. [Google Scholar]
- 70.Roson B, Fernandez-Sabe N, Carratala J. Contribution of a urinary antigen assay (binax NOW) to the early diagnosis of pneumococcal pneumonia. Clin Infect Dis. 2004;38(2):222–226. doi: 10.1086/380639. [DOI] [PubMed] [Google Scholar]
- 71.Kashuba AD, Ballow CH. Legionella urinary antigen testing: potential impact on diagnosis and antibiotic therapy. Diagn Microbiol Infect Dis. 1996;24(3):129–139. doi: 10.1016/0732-8893(96)00010-7. [DOI] [PubMed] [Google Scholar]
- 72.Pappas PG. Blastomycosis. Semin Respir Crit Care Med. 2004;25(2):113–121. doi: 10.1055/s-2004-824896. [DOI] [PubMed] [Google Scholar]
- 73.Kauffman CA. Histoplasmosis: a clinical and laboratory update. Clin Microbiol Rev. 2007;20(1):115–132. doi: 10.1128/CMR.00027-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Morris AJ, Tanner DC, Reller LB. Rejection criteria for endotracheal aspirates from adults. J Clin Microbiol. 1993;31(5):1027–1029. doi: 10.1128/jcm.31.5.1027-1029.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Geckler RW, Gremillion DH, McAllister CK. Microscopic and bacteriological comparison of paired sputa and transtracheal aspirates. J Clin Microbiol. 1977;6(4):396–399. doi: 10.1128/jcm.6.4.396-399.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zaidi AK, Reller LB. Rejection criteria for endotracheal aspirates from pediatric patients. J Clin Microbiol. 1996;34(2):352–354. doi: 10.1128/jcm.34.2.352-354.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Pollock NR, Duong S, Cheng A. Ruling out novel H1N1 influenza virus infection with direct fluorescent antigen testing. Clin Infect Dis. 2009;49(6):e66–e68. doi: 10.1086/644502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Levin DC, Schwarz MI, Matthay RA. Bacteremic Hemophilus influenzae pneumonia in adults. A report of 24 cases and a review of the literature. Am J Med. 1977;62(2):219–224. doi: 10.1016/0002-9343(77)90317-5. [DOI] [PubMed] [Google Scholar]
- 79.Verghese A, Berk SL. Bacterial pneumonia in the elderly. Medicine (Balt) 1983;62(5):271–285. doi: 10.1097/00005792-198309000-00002. [DOI] [PubMed] [Google Scholar]
- 80.Everett ED, Rham AE, Jr, Adaniya R. Haemophilus influenzae pneumonia in adults. JAMA. 1977;238(4):319–321. [PubMed] [Google Scholar]
- 81.Courcol RJ, Damien JM, Ramon P. Presence of alveolar macrophages as a criterion for determining the suitability of sputum specimens for bacterial culture. Eur J Clin Microbiol. 1984;3(2):122–125. [PubMed] [Google Scholar]
- 82.Johnson MA, Drew WL, Roberts M. Branhamella (Neisseria) catarrhalis—a lower respiratory tract pathogen? J Clin Microbiol. 1981;13(6):1066–1069. doi: 10.1128/jcm.13.6.1066-1069.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Putsch RW, Hamilton JD, Wolinsky E. Neisseria meningitidis, a respiratory pathogen? J Infect Dis. 1970;121(1):48–54. doi: 10.1093/infdis/121.1.48. [DOI] [PubMed] [Google Scholar]
- 84.Thornley PE, Aitken J, Drennan CJ. Branhamella catarrhalis infection of the lower respiratory tract: reliable diagnosis by sputum examination. Br Med J (Clin Res Ed) 1982;285(6354):1537–1538. doi: 10.1136/bmj.285.6354.1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rein MF, Gwaltney JM, Jr, O’Brien WM. Accuracy of Gram's stain in identifying pneumococci in sputum. JAMA. 1978;239(25):2671–2673. doi: 10.1001/jama.239.25.2671. [DOI] [PubMed] [Google Scholar]
- 86.Roson B, Carratala J, Verdaguer R. Prospective study of the usefulness of sputum Gram stain in the initial approach to community-acquired pneumonia requiring hospitalization. Clin Infect Dis. 2000;31(4):869–874. doi: 10.1086/318151. [DOI] [PubMed] [Google Scholar]
- 87.Boerner DF, Zwadyk P. The value of the sputum Gram's stain in community-acquired pneumonia. JAMA. 1982;247(5):642–645. [PubMed] [Google Scholar]
- 88.Musher DM, Montoya R, Wanahita A. Diagnostic value of microscopic examination of Gram-stained sputum and sputum cultures in patients with bacteremic pneumococcal pneumonia. Clin Infect Dis. 2004;39(2):165–169. doi: 10.1086/421497. [DOI] [PubMed] [Google Scholar]
- 89.Jones GR, Christodoulides M, Brooks JL. Dynamics of carriage of Neisseria meningitidis in a group of military recruits: Subtype stability and specificity of the immune response following colonization. J Infect Dis. 1998;178(2):451–459. doi: 10.1086/515622. [DOI] [PubMed] [Google Scholar]
- 90.Steingart KR, Henry M, Ng V. Fluorescence versus conventional sputum smear microscopy for tuberculosis: a systematic review. Lancet Infect Dis. 2006;6(9):570–581. doi: 10.1016/S1473-3099(06)70578-3. [DOI] [PubMed] [Google Scholar]
- 91.Parsons LM, Somoskovi A, Gutierrez C. Laboratory diagnosis of tuberculosis in resource-poor countries: challenges and opportunities. Clin Microbiol Rev. 2011;24(2):314–350. doi: 10.1128/CMR.00059-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Yeager H, Jr, Lacy J, Smith LR. Quantitative studies of mycobacterial populations in sputum and saliva. Am Rev Respir Dis. 1967;95(6):998–1004. doi: 10.1164/arrd.1967.95.6.998. [DOI] [PubMed] [Google Scholar]
- 93.Centers for Disease Control and Prevention (CDC) Updated guidelines for the use of nucleic acid amplification tests in the diagnosis of tuberculosis. MMWR Morb Mortal Wkly Rep. 2009;58(1):7–10. [PubMed] [Google Scholar]
- 94.Newton HJ, Ang DK, van Driel IR. Molecular pathogenesis of infections caused by Legionella pneumophila. Clin Microbiol Rev. 2010;23(2):274–298. doi: 10.1128/CMR.00052-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Murdoch DR. Diagnosis of Legionella infection. Clin Infect Dis. 2003;36(1):64–69. doi: 10.1086/345529. [DOI] [PubMed] [Google Scholar]
- 96.Bell TA, Kuo CC, Stamm WE. Direct fluorescent monoclonal antibody stain for rapid detection of infant Chlamydia trachomatis infections. Pediatrics. 1984;74(2):224–228. [PubMed] [Google Scholar]
- 97.Paisley JW, Lauer BA, Melinkovich P. Rapid diagnosis of Chlamydia trachomatis pneumonia in infants by direct immunofluorescence microscopy of nasopharyngeal secretions. J Pediatr. 1986;109(4):653–655. doi: 10.1016/s0022-3476(86)80232-3. [DOI] [PubMed] [Google Scholar]
- 98.Cregan P, Yamamoto A, Lum A. Comparison of four methods for rapid detection of Pneumocystis carinii in respiratory specimens. J Clin Microbiol. 1990;28(11):2432–2436. doi: 10.1128/jcm.28.11.2432-2436.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Fraser JL, Lilly C, Israel E. Diagnostic yield of bronchoalveolar lavage and bronchoscopic lung biopsy for detection of Pneumocystis carinii. Mayo Clin Proc. 1996;71(11):1025–1029. doi: 10.4065/71.11.1025. [DOI] [PubMed] [Google Scholar]
- 100.Aslanzadeh J, Stelmach PS. Detection of Pneumocystis carinii with direct fluorescence antibody and calcofluor white stain. Infection. 1996;24(3):248–250. doi: 10.1007/BF01781104. [DOI] [PubMed] [Google Scholar]
- 101.DiMaio MA, Sahoo MK, Waggoner J. Comparison of Xpert flu rapid nucleic acid testing with rapid antigen testing for the diagnosis of influenza A and B. J Virol Methods. 2012;186(1–2):137–140. doi: 10.1016/j.jviromet.2012.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ginocchio CC, Zhang F, Manji R. Evaluation of multiple test methods for the detection of the novel 2009 influenza A (H1N1) during the New York City outbreak. J Clin Virol. 2009;45(3):191–195. doi: 10.1016/j.jcv.2009.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Monheit JE, Cowan DF, Moore DG. Rapid detection of fungi in tissues using calcofluor white and fluorescence microscopy. Arch Pathol Lab Med. 1984;108(8):616–618. [PubMed] [Google Scholar]
- 104.Kim YK, Parulekar S, Yu PK. Evaluation of calcofluor white stain for detection of Pneumocystis carinii. Diagn Microbiol Infect Dis. 1990;13(4):307–310. doi: 10.1016/0732-8893(90)90022-n. [DOI] [PubMed] [Google Scholar]
- 105.Sangoi AR, Rogers WM, Longacre TA. Challenges and pitfalls of morphologic identification of fungal infections in histologic and cytologic specimens: a ten-year retrospective review at a single institution. Am J Clin Pathol. 2009;131(3):364–375. doi: 10.1309/AJCP99OOOZSNISCZ. [DOI] [PubMed] [Google Scholar]
- 106.Schroeder L, Banaei N. Images in clinical medicine. Strongyloides stercoralis embryonated ova in the lung. N Engl J Med. 2013;368(12):e15. doi: 10.1056/NEJMicm1204579. [DOI] [PubMed] [Google Scholar]
- 107.Scott AL. Lymphatic-dwelling filariae. In: Nutman TB, editor. Lymphatic filariasis. Imperial College Press; London: 2000. pp. 5–39. [Google Scholar]
- 108.Hove MG, Woods GL. Duration of fungal culture incubation in an area endemic for Histoplasma capsulatum. Diagn Microbiol Infect Dis. 1997;28(1):41–43. doi: 10.1016/s0732-8893(97)89158-4. [DOI] [PubMed] [Google Scholar]
- 109.Vickers RM, Rihs JD, Yu VL. Clinical demonstration of isolation of Nocardia asteroides on buffered charcoal-yeast extract media. J Clin Microbiol. 1992;30(1):227–228. doi: 10.1128/jcm.30.1.227-228.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Baselski V, Klutts JS, Baselski V. Quantitative cultures of bronchoscopically obtained specimens should be performed for optimal management of ventilator-associated pneumonia. J Clin Microbiol. 2013;51(3):740–744. doi: 10.1128/JCM.03383-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Craven DE, Hjalmarson KI. Ventilator-associated tracheobronchitis and pneumonia: thinking outside the box. Clin Infect Dis. 2010;51(Suppl 1):S59–S66. doi: 10.1086/653051. [DOI] [PubMed] [Google Scholar]
- 112.Souweine B, Veber B, Bedos JP. Diagnostic accuracy of protected specimen brush and bronchoalveolar lavage in nosocomial pneumonia: impact of previous antimicrobial treatments. Crit Care Med. 1998;26(2):236–244. doi: 10.1097/00003246-199802000-00017. [DOI] [PubMed] [Google Scholar]
- 113.Baselski VS, Wunderink RG. Bronchoscopic diagnosis of pneumonia. Clin Microbiol Rev. 1994;7(4):533–558. doi: 10.1128/cmr.7.4.533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Barrett-Connor E. The nonvalue of sputum culture in the diagnosis of pneumococcal pneumonia. Am Rev Respir Dis. 1971;103(6):845–848. doi: 10.1164/arrd.1971.103.6.845. [DOI] [PubMed] [Google Scholar]
- 115.Bartlett JG, Finegold SM. Anaerobic pleuropulmonary infections. Medicine (Balt) 1972;51(6):413–450. doi: 10.1097/00005792-197211000-00001. [DOI] [PubMed] [Google Scholar]
- 116.Hahn HH, Beaty HN. Transtracheal aspiration in the evaluation of patients with pneumonia. Ann Intern Med. 1970;72(2):183–187. doi: 10.7326/0003-4819-72-2-183. [DOI] [PubMed] [Google Scholar]
- 117.Rello J, Esandi ME, Diaz E. The role of Candida sp isolated from bronchoscopic samples in nonneutropenic patients. Chest. 1998;114(1):146–149. doi: 10.1378/chest.114.1.146. [DOI] [PubMed] [Google Scholar]
- 118.Wheat LJ, Conces D, Allen SD. Pulmonary histoplasmosis syndromes: recognition, diagnosis, and management. Semin Respir Crit Care Med. 2004;25(2):129–144. doi: 10.1055/s-2004-824898. [DOI] [PubMed] [Google Scholar]
- 119.Wheat LJ, Kauffman CA. Histoplasmosis. Infect Dis Clin North Am. 2003;17(1):1–19. doi: 10.1016/s0891-5520(02)00039-9. vii. [DOI] [PubMed] [Google Scholar]
- 120.Koga K, Kasuya S, Khamboonruang C. An evaluation of the agar plate method for the detection of Strongyloides stercoralis in northern Thailand. J Trop Med Hyg. 1990;93(3):183–188. [PubMed] [Google Scholar]
- 121.Koga K, Kasuya S, Ohtomo H. How effective is the agar plate method for Strongyloides stercoralis? J Parasitol. 1992;78(1):155–156. [PubMed] [Google Scholar]
- 122.Intapan PM, Maleewong W, Wongsaroj T. Comparison of the quantitative formalin ethyl acetate concentration technique and agar plate culture for diagnosis of human strongyloidiasis. J Clin Microbiol. 2005;43(4):1932–1933. doi: 10.1128/JCM.43.4.1932-1933.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Arakaki T, Iwanaga M, Kinjo F. Efficacy of agar-plate culture in detection of Strongyloides stercoralis infection. J Parasitol. 1990;76(3):425–428. [PubMed] [Google Scholar]
- 124.Leland DS, Ginocchio CC. Role of cell culture for virus detection in the age of technology. Clin Microbiol Rev. 2007;20(1):49–78. doi: 10.1128/CMR.00002-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Shetty AK, Treynor E, Hill DW. Comparison of conventional viral cultures with direct fluorescent antibody stains for diagnosis of community-acquired respiratory virus infections in hospitalized children. Pediatr Infect Dis J. 2003;22(9):789–794. doi: 10.1097/01.inf.0000083823.43526.97. [DOI] [PubMed] [Google Scholar]
- 126.Fong CK, Lee MK, Griffith BP. Evaluation of R-mix FreshCells in shell vials for detection of respiratory viruses. J Clin Microbiol. 2000;38(12):4660–4662. doi: 10.1128/jcm.38.12.4660-4662.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.LaSala PR, Bufton KK, Ismail N. Prospective comparison of R-mix shell vial system with direct antigen tests and conventional cell culture for respiratory virus detection. J Clin Virol. 2007;38(3):210–216. doi: 10.1016/j.jcv.2006.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kim JS, Kim SH, Bae SY. Enhanced detection of respiratory viruses using cryopreserved R-mix ReadyCells. J Clin Virol. 2008;42(3):264–267. doi: 10.1016/j.jcv.2008.03.015. [DOI] [PubMed] [Google Scholar]
- 129.Paya CV, Wold AD, Smith TF. Detection of cytomegalovirus infections in specimens other than urine by the shell vial assay and conventional tube cell cultures. J Clin Microbiol. 1987;25(5):755–757. doi: 10.1128/jcm.25.5.755-757.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Chemaly RF, Yen-Lieberman B, Chapman J. Clinical utility of cytomegalovirus viral load in bronchoalveolar lavage in lung transplant recipients. Am J Transplant. 2005;5(3):544–548. doi: 10.1111/j.1600-6143.2005.00747.x. [DOI] [PubMed] [Google Scholar]
- 131.Connolly MG, Jr, Baughman RP, Dohn MN. Recovery of viruses other than cytomegalovirus from bronchoalveolar lavage fluid. Chest. 1994;105(6):1775–1781. doi: 10.1378/chest.105.6.1775. [DOI] [PubMed] [Google Scholar]
- 132.Engelmann I, Hesse N, Fegbeutel C. Incidence and impact of herpes simplex and cytomegalovirus detection in the respiratory tract after lung transplantation. Transpl Infect Dis. 2011;13(3):259–265. doi: 10.1111/j.1399-3062.2010.00587.x. [DOI] [PubMed] [Google Scholar]
- 133.Piret J, Boivin G. Resistance of herpes simplex viruses to nucleoside analogues: mechanisms, prevalence, and management. Antimicrob Agents Chemother. 2011;55(2):459–472. doi: 10.1128/AAC.00615-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Pinsky BA, Samson D, Ghafghaichi L. Comparison of real-time PCR and conventional biochemical methods for identification of Staphylococcus lugdunensis. J Clin Microbiol. 2009;47(11):3472–3477. doi: 10.1128/JCM.00342-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Palladino S, Kay ID, Flexman JP. Rapid detection of vanA and vanB genes directly from clinical specimens and enrichment broths by real-time multiplex PCR assay. J Clin Microbiol. 2003;41(6):2483–2486. doi: 10.1128/JCM.41.6.2483-2486.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Boehme CC, Nabeta P, Hillemann D. Rapid molecular detection of tuberculosis and rifampin resistance. N Engl J Med. 2010;363(11):1005–1015. doi: 10.1056/NEJMoa0907847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Niemz A, Ferguson TM, Boyle DS. Point-of-care nucleic acid testing for infectious diseases. Trends Biotechnol. 2011;29(5):240–250. doi: 10.1016/j.tibtech.2011.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Espy MJ, Uhl JR, Sloan LM. Real-time PCR in clinical microbiology: applications for routine laboratory testing. Clin Microbiol Rev. 2006;19(1):165–256. doi: 10.1128/CMR.19.1.165-256.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mahony JB, Petrich A, Smieja M. Molecular diagnosis of respiratory virus infections. Crit Rev Clin Lab Sci. 2011;48(5–6):217–249. doi: 10.3109/10408363.2011.640976. [DOI] [PubMed] [Google Scholar]
- 140.Vassall A, van Kampen S, Sohn H. Rapid diagnosis of tuberculosis with the Xpert MTB/RIF assay in high burden countries: a cost-effectiveness analysis. PLoS Med. 2011;8(11):e1001120. doi: 10.1371/journal.pmed.1001120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Boehme CC, Nicol MP, Nabeta P. Feasibility, diagnostic accuracy, and effectiveness of decentralised use of the Xpert MTB/RIF test for diagnosis of tuberculosis and multidrug resistance: a multicentre implementation study. Lancet. 2011;377(9776):1495–1505. doi: 10.1016/S0140-6736(11)60438-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Ling DI, Zwerling AA, Pai M. GenoType MTBDR assays for the diagnosis of multidrug-resistant tuberculosis: a meta-analysis. Eur Respir J. 2008;32(5):1165–1174. doi: 10.1183/09031936.00061808. [DOI] [PubMed] [Google Scholar]
- 143.Steingart KR, Sohn H, Schiller I. Xpert(R) MTB/RIF assay for pulmonary tuberculosis and rifampicin resistance in adults. Cochrane Database Syst Rev. 2013;(1) doi: 10.1002/14651858.CD009593.pub2. CD009593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Hillemann D, Weizenegger M, Kubica T. Use of the genotype MTBDR assay for rapid detection of rifampin and isoniazid resistance in Mycobacterium tuberculosis complex isolates. J Clin Microbiol. 2005;43(8):3699–3703. doi: 10.1128/JCM.43.8.3699-3703.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Barnard M, Albert H, Coetzee G. Rapid molecular screening for multidrug-resistant tuberculosis in a high-volume public health laboratory in South Africa. Am J Respir Crit Care Med. 2008;177(7):787–792. doi: 10.1164/rccm.200709-1436OC. [DOI] [PubMed] [Google Scholar]
- 146.Binnicker MJ, Buckwalter SP, Eisberner JJ. Detection of Coccidioides species in clinical specimens by real-time PCR. J Clin Microbiol. 2007;45(1):173–178. doi: 10.1128/JCM.01776-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Torelli R, Sanguinetti M, Moody A. Diagnosis of invasive aspergillosis by a commercial real-time PCR assay for Aspergillus DNA in bronchoalveolar lavage fluid samples from high-risk patients compared to a galactomannan enzyme immunoassay. J Clin Microbiol. 2011;49(12):4273–4278. doi: 10.1128/JCM.05026-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Balajee SA, Sigler L, Brandt ME. DNA and the classical way: identification of medically important molds in the 21st century. Med Mycol. 2007;45(6):475–490. doi: 10.1080/13693780701449425. [DOI] [PubMed] [Google Scholar]
- 149.Nguyen MH, Wissel MC, Shields RK. Performance of Candida real-time polymerase chain reaction, beta-D-glucan assay, and blood cultures in the diagnosis of invasive candidiasis. Clin Infect Dis. 2012;54(9):1240–1248. doi: 10.1093/cid/cis200. [DOI] [PubMed] [Google Scholar]
- 150.Mengoli C, Cruciani M, Barnes RA. Use of PCR for diagnosis of invasive aspergillosis: systematic review and meta-analysis. Lancet Infect Dis. 2009;9(2):89–96. doi: 10.1016/S1473-3099(09)70019-2. [DOI] [PubMed] [Google Scholar]
- 151.Durand-Joly I, Chabe M, Soula F. Molecular diagnosis of Pneumocystis pneumonia. FEMS Immunol Med Microbiol. 2005;45(3):405–410. doi: 10.1016/j.femsim.2005.06.006. [DOI] [PubMed] [Google Scholar]
- 152.Samuel CM, Whitelaw A, Corcoran C. Improved detection of Pneumocystis jirovecii in upper and lower respiratory tract specimens from children with suspected Pneumocystis pneumonia using real-time PCR: a prospective study. BMC Infect Dis. 2011;11:329. doi: 10.1186/1471-2334-11-329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Bretagne S, Costa JM, Fleury-Feith J. Quantitative competitive PCR with bronchoalveolar lavage fluid for diagnosis of toxoplasmosis in AIDS patients. J Clin Microbiol. 1995;33(6):1662–1664. doi: 10.1128/jcm.33.6.1662-1664.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Boivin G, Olson CA, Quirk MR. Quantitation of cytomegalovirus DNA and characterization of viral gene expression in bronchoalveolar cells of infected patients with and without pneumonitis. J Infect Dis. 1996;173(6):1304–1312. doi: 10.1093/infdis/173.6.1304. [DOI] [PubMed] [Google Scholar]
- 155.Goto H, Yuasa K, Sakamaki H. Rapid detection cytomegalovirus pneumonia in recipients of bone marrow transplant: evaluation and comparison of five survey methods for bronchoalveolar lavage fluid. Bone Marrow Transplant. 1996;17(5):855–860. [PubMed] [Google Scholar]
- 156.Messmer TO, Skelton SK, Moroney JF. Application of a nested, multiplex PCR to psittacosis outbreaks. J Clin Microbiol. 1997;35(8):2043–2046. doi: 10.1128/jcm.35.8.2043-2046.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Boman J, Allard A, Persson K. Rapid diagnosis of respiratory Chlamydia pneumoniae infection by nested touchdown polymerase chain reaction compared with culture and antigen detection by EIA. J Infect Dis. 1997;175(6):1523–1526. doi: 10.1086/516492. [DOI] [PubMed] [Google Scholar]
- 158.Hauser PM, Bille J, Lass-Florl C. Multicenter, prospective clinical evaluation of respiratory samples from subjects at risk for Pneumocystis jirovecii infection by use of a commercial real-time PCR assay. J Clin Microbiol. 2011;49(5):1872–1878. doi: 10.1128/JCM.02390-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Maskell NA, Waine DJ, Lindley A. Asymptomatic carriage of Pneumocystis jiroveci in subjects undergoing bronchoscopy: A prospective study. Thorax. 2003;58(7):594–597. doi: 10.1136/thorax.58.7.594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Torres J, Goldman M, Wheat LJ. Diagnosis of Pneumocystis carinii pneumonia in human immunodeficiency virus-infected patients with polymerase chain reaction: a blinded comparison to standard methods. Clin Infect Dis. 2000;30(1):141–145. doi: 10.1086/313584. [DOI] [PubMed] [Google Scholar]
- 161.Petti CA. Detection and identification of microorganisms by gene amplification and sequencing. Clin Infect Dis. 2007;44(8):1108–1114. doi: 10.1086/512818. [DOI] [PubMed] [Google Scholar]
- 162.Relman DA, Schmidt TM, MacDermott RP. Identification of the uncultured bacillus of Whipple's disease. N Engl J Med. 1992;327(5):293–301. doi: 10.1056/NEJM199207303270501. [DOI] [PubMed] [Google Scholar]
- 163.Rickerts V, Khot PD, Myerson D. Comparison of quantitative real time PCR with sequencing and ribosomal RNA-FISH for the identification of fungi in formalin fixed, paraffin-embedded tissue specimens. BMC Infect Dis. 2011;11:202. doi: 10.1186/1471-2334-11-202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Lau A, Chen S, Sorrell T. Development and clinical application of a panfungal PCR assay to detect and identify fungal DNA in tissue specimens. J Clin Microbiol. 2007;45(2):380–385. doi: 10.1128/JCM.01862-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Schoch CL, Seifert KA, Huhndorf S. Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for fungi. Proc Natl Acad Sci U S A. 2012;109(16):6241–6246. doi: 10.1073/pnas.1117018109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Sambol AR, Iwen PC, Pieretti M. Validation of the Cepheid Xpert Flu A real time RT-PCR detection panel for emergency use authorization. J Clin Virol. 2010;48(4):234–238. doi: 10.1016/j.jcv.2010.06.001. [DOI] [PubMed] [Google Scholar]
- 167.Miller S, Moayeri M, Wright C. Comparison of GeneXpert FluA PCR to direct fluorescent antibody and respiratory viral panel PCR assays for detection of 2009 novel H1N1 influenza virus. J Clin Microbiol. 2010;48(12):4684–4685. doi: 10.1128/JCM.00707-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Popowitch EB, Rogers E, Miller MB. Retrospective and prospective verification of the Cepheid Xpert influenza virus assay. J Clin Microbiol. 2011;49(9):3368–3369. doi: 10.1128/JCM.01162-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Li M, Brenwald N, Bonigal S. Rapid diagnosis of influenza: an evaluation of two commercially available RT-PCR assays. J Infect. 2012;65(1):60–63. doi: 10.1016/j.jinf.2012.04.003. [DOI] [PubMed] [Google Scholar]
- 170.Novak-Weekley SM, Marlowe EM, Poulter M. Evaluation of the Cepheid Xpert Flu assay for rapid identification and differentiation of influenza A, influenza A 2009 H1N1, and influenza B viruses. J Clin Microbiol. 2012;50(5):1704–1710. doi: 10.1128/JCM.06520-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Salez N, Ninove L, Thirion L. Evaluation of the Xpert Flu test and comparison with in-house real-time RT-PCR assays for detection of influenza virus from 2008 to 2011 in Marseille, France. Clin Microbiol Infect. 2012;18(4):E81–E83. doi: 10.1111/j.1469-0691.2012.03792.x. [DOI] [PubMed] [Google Scholar]
- 172.Alby K, Popowitch EB, Miller MB. Comparative evaluation of the Nanosphere Verigene RV+ assay and the Simplexa Flu A/B & RSV kit for detection of influenza and respiratory syncytial viruses. J Clin Microbiol. 2013;51(1):352–353. doi: 10.1128/JCM.02504-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Krunic N, Merante F, Yaghoubian S. Advances in the diagnosis of respiratory tract infections: role of the Luminex xTAG Respiratory Viral Panel. Ann N Y Acad Sci. 2011;1222:6–13. doi: 10.1111/j.1749-6632.2011.05964.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Pabbaraju K, Tokaryk KL, Wong S. Comparison of the Luminex xTAG Respiratory Viral Panel with in-house nucleic acid amplification tests for diagnosis of respiratory virus infections. J Clin Microbiol. 2008;46(9):3056–3062. doi: 10.1128/JCM.00878-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Gadsby NJ, Hardie A, Claas EC. Comparison of the Luminex Respiratory Virus Panel FAST assay with in-house real-time PCR for respiratory viral infection diagnosis. J Clin Microbiol. 2010;48(6):2213–2216. doi: 10.1128/JCM.02446-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Pabbaraju K, Wong S, Tokaryk KL. Comparison of the Luminex xTAG Respiratory Viral Panel with xTAG Respiratory Viral Panel FAST for diagnosis of respiratory virus infections. J Clin Microbiol. 2011;49(5):1738–1744. doi: 10.1128/JCM.02090-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Rand KH, Rampersaud H, Houck HJ. Comparison of two multiplex methods for detection of respiratory viruses: FilmArray RP and xTAG RVP. J Clin Microbiol. 2011;49(7):2449–2453. doi: 10.1128/JCM.02582-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Raymaekers M, de Rijke B, Pauli I. Timely diagnosis of respiratory tract infections: evaluation of the performance of the Respifinder assay compared to the xTAG respiratory viral panel assay. J Clin Virol. 2011;52(4):314–316. doi: 10.1016/j.jcv.2011.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Babady NE, Mead P, Stiles J. Comparison of the Luminex xTAG RVP FAST assay and the Idaho Technology FilmArray RP assay for detection of respiratory viruses in pediatric patients at a cancer hospital. J Clin Microbiol. 2012;50(7):2282–2288. doi: 10.1128/JCM.06186-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Popowitch EB, O’Neill SS, Miller MB. Comparison of the Biofire FilmArray RP, GenMark eSensor RVP, Luminex xTAG RVPv1, and Luminex xTAG RVP FAST multiplex assays for detection of respiratory viruses. J Clin Microbiol. 2013;51(5):1528–1533. doi: 10.1128/JCM.03368-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Pierce VM, Hodinka RL. Comparison of the GenMark diagnostics eSensor Respiratory Viral Panel to real-time PCR for detection of respiratory viruses in children. J Clin Microbiol. 2012;50(11):3458–3465. doi: 10.1128/JCM.01384-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Poritz MA, Blaschke AJ, Byington CL. FilmArray, an automated nested multiplex PCR system for multi-pathogen detection: development and application to respiratory tract infection. PLoS One. 2011;6(10):e26047. doi: 10.1371/journal.pone.0026047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Loeffelholz MJ, Pong DL, Pyles RB. Comparison of the FilmArray Respiratory Panel and Prodesse real-time PCR assays for detection of respiratory pathogens. J Clin Microbiol. 2011;49(12):4083–4088. doi: 10.1128/JCM.05010-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Hammond SP, Gagne LS, Stock SR. Respiratory virus detection in immunocompromised patients with FilmArray Respiratory Panel compared to conventional methods. J Clin Microbiol. 2012;50(10):3216–3221. doi: 10.1128/JCM.00538-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Hayden RT, Gu Z, Rodriguez A. Comparison of two broadly multiplexed PCR systems for viral detection in clinical respiratory tract specimens from immunocompromised children. J Clin Virol. 2012;53(4):308–313. doi: 10.1016/j.jcv.2011.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Pierce VM, Elkan M, Leet M. Comparison of the Idaho Technology FilmArray system to real-time PCR for detection of respiratory pathogens in children. J Clin Microbiol. 2012;50(2):364–371. doi: 10.1128/JCM.05996-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Renaud C, Crowley J, Jerome KR. Comparison of FilmArray Respiratory Panel and laboratory-developed real-time reverse transcription-polymerase chain reaction assays for respiratory virus detection. Diagn Microbiol Infect Dis. 2012;74(4):379–383. doi: 10.1016/j.diagmicrobio.2012.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Couturier MR, Barney T, Alger G. Evaluation of the FilmArray(R) respiratory panel for clinical use in a large children's hospital. J Clin Lab Anal. 2013;27(2):148–154. doi: 10.1002/jcla.21576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Sinclair A, Xie X, Teltscher M. A urine pneumococcal antigen test for the diagnosis of community acquired Streptococcus pneumoniae pneumonia: systematic review and meta-analysis. J Clin Microbiol. 2013;51(7):2303–2310. doi: 10.1128/JCM.00137-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Dominguez J, Blanco S, Rodrigo C. Usefulness of urinary antigen detection by an immunochromatographic test for diagnosis of pneumococcal pneumonia in children. J Clin Microbiol. 2003;41(5):2161–2163. doi: 10.1128/JCM.41.5.2161-2163.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Rothstein EP, Madore DV, Girone JA. Comparison of antigenuria after immunization with three Haemophilus influenzae type b conjugate vaccines. Pediatr Infect Dis J. 1991;10(4):311–314. doi: 10.1097/00006454-199104000-00009. [DOI] [PubMed] [Google Scholar]
- 192.Shimada T, Noguchi Y, Jackson JL. Systematic review and metaanalysis: urinary antigen tests for legionellosis. Chest. 2009;136(6):1576–1585. doi: 10.1378/chest.08-2602. [DOI] [PubMed] [Google Scholar]
- 193.Kohler RB, Winn WC, Jr, Wheat LJ. Onset and duration of urinary antigen excretion in legionnaires disease. J Clin Microbiol. 1984;20(4):605–607. doi: 10.1128/jcm.20.4.605-607.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Feldmesser M, Harris C, Reichberg S. Serum cryptococcal antigen in patients with AIDS. Clin Infect Dis. 1996;23(4):827–830. doi: 10.1093/clinids/23.4.827. [DOI] [PubMed] [Google Scholar]
- 195.Baughman RP, Rhodes JC, Dohn MN. Detection of cryptococcal antigen in bronchoalveolar lavage fluid: a prospective study of diagnostic utility. Am Rev Respir Dis. 1992;145(5):1226–1229. doi: 10.1164/ajrccm/145.5.1226. [DOI] [PubMed] [Google Scholar]
- 196.Young EJ, Hirsh DD, Fainstein V. Pleural effusions due to Cryptococcus neoformans: a review of the literature and report of two cases with cryptococcal antigen determinations. Am Rev Respir Dis. 1980;121(4):743–747. doi: 10.1164/arrd.1980.121.4.743. [DOI] [PubMed] [Google Scholar]
- 197.Lortholary O, Nunez H, Brauner MW. Pulmonary cryptococcosis. Semin Respir Crit Care Med. 2004;25(2):145–157. doi: 10.1055/s-2004-824899. [DOI] [PubMed] [Google Scholar]
- 198.Cloud JL, Bauman SK, Pelfrey JM. Biased report on the IMMY ALPHA Histoplasma antigen enzyme immunoassay for diagnosis of histoplasmosis. Clin Vaccine Immunol. 2007;14(10):1389–1390. doi: 10.1128/CVI.00280-07. author reply 1390–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Hage CA, Ribes JA, Wengenack NL. A multicenter evaluation of tests for diagnosis of histoplasmosis. Clin Infect Dis. 2011;53(5):448–454. doi: 10.1093/cid/cir435. [DOI] [PubMed] [Google Scholar]
- 200.Connolly PA, Durkin MM, Lemonte AM. Detection of Histoplasma antigen by a quantitative enzyme immunoassay. Clin Vaccine Immunol. 2007;14(12):1587–1591. doi: 10.1128/CVI.00071-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Kuberski T, Myers R, Wheat LJ. Diagnosis of coccidioidomycosis by antigen detection using cross-reaction with a Histoplasma antigen. Clin Infect Dis. 2007;44(5):e50–e54. doi: 10.1086/511684. [DOI] [PubMed] [Google Scholar]
- 202.Connolly P, Hage CA, Bariola JR. Blastomyces dermatitidis antigen detection by quantitative enzyme immunoassay. Clin Vaccine Immunol. 2012;19(1):53–56. doi: 10.1128/CVI.05248-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Pfeiffer CD, Fine JP, Safdar N. Diagnosis of invasive aspergillosis using a galactomannan assay: a meta-analysis. Clin Infect Dis. 2006;42(10):1417–1427. doi: 10.1086/503427. [DOI] [PubMed] [Google Scholar]
- 204.Wheat LJ. Antigen detection, serology, and molecular diagnosis of invasive mycoses in the immunocompromised host. Transpl Infect Dis. 2006;8(3):128–139. doi: 10.1111/j.1399-3062.2006.00165.x. [DOI] [PubMed] [Google Scholar]
- 205.Wheat LJ, Walsh TJ. Diagnosis of invasive aspergillosis by galactomannan antigenemia detection using an enzyme immunoassay. Eur J Clin Microbiol Infect Dis. 2008;27(4):245–251. doi: 10.1007/s10096-007-0437-7. [DOI] [PubMed] [Google Scholar]
- 206.Wheat LJ, Hackett E, Durkin M. Histoplasmosis-associated cross-reactivity in the BioRad Platelia Aspergillus enzyme immunoassay. Clin Vaccine Immunol. 2007;14(5):638–640. doi: 10.1128/CVI.00479-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Xavier MO, Pasqualotto AC, Cardoso IC. Cross-reactivity of Paracoccidioides brasiliensis, Histoplasma capsulatum, and Cryptococcus species in the commercial Platelia Aspergillus enzyme immunoassay. Clin Vaccine Immunol. 2009;16(1):132–133. doi: 10.1128/CVI.00310-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Guarner J, Brandt ME. Histopathologic diagnosis of fungal infections in the 21st century. Clin Microbiol Rev. 2011;24(2):247–280. doi: 10.1128/CMR.00053-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Adam O, Auperin A, Wilquin F. Treatment with piperacillin-tazobactam and false-positive Aspergillus galactomannan antigen test results for patients with hematological malignancies. Clin Infect Dis. 2004;38(6):917–920. doi: 10.1086/383148. [DOI] [PubMed] [Google Scholar]
- 210.Ansorg R, van den Boom R, Rath PM. Detection of Aspergillus galactomannan antigen in foods and antibiotics. Mycoses. 1997;40(9–10):353–357. doi: 10.1111/j.1439-0507.1997.tb00249.x. [DOI] [PubMed] [Google Scholar]
- 211.Viscoli C, Machetti M, Cappellano P. False-positive galactomannan Platelia Aspergillus test results for patients receiving piperacillin-tazobactam. Clin Infect Dis. 2004;38(6):913–916. doi: 10.1086/382224. [DOI] [PubMed] [Google Scholar]
- 212.Petti MC, Prignano G, Mengarelli A. Cross-recognition of Aspergillus galactomannan caused by Listeria monocytogenes infection. Diagn Microbiol Infect Dis. 2013;76(2):250–251. doi: 10.1016/j.diagmicrobio.2013.02.005. [DOI] [PubMed] [Google Scholar]
- 213.Husain S, Clancy CJ, Nguyen MH. Performance characteristics of the Platelia Aspergillus enzyme immunoassay for detection of Aspergillus galactomannan antigen in bronchoalveolar lavage fluid. Clin Vaccine Immunol. 2008;15(12):1760–1763. doi: 10.1128/CVI.00226-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Hage CA, Reynolds JM, Durkin M. Plasmalyte as a cause of false-positive results for Aspergillus galactomannan in bronchoalveolar lavage fluid. J Clin Microbiol. 2007;45(2):676–677. doi: 10.1128/JCM.01940-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Kedzierska A, Kochan P, Pietrzyk A. Current status of fungal cell wall components in the immunodiagnostics of invasive fungal infections in humans: galactomannan, mannan and (1–>3)-beta-D-glucan antigens. Eur J Clin Microbiol Infect Dis. 2007;26(11):755–766. doi: 10.1007/s10096-007-0373-6. [DOI] [PubMed] [Google Scholar]
- 216.Marty FM, Koo S, Bryar J. (1->3)beta-D-glucan assay positivity in patients with Pneumocystis (carinii) jiroveci pneumonia. Ann Intern Med. 2007;147(1):70–72. doi: 10.7326/0003-4819-147-1-200707030-00018. [DOI] [PubMed] [Google Scholar]
- 217.Cuetara MS, Alhambra A, Moragues MD. Detection of (1–>3)-beta-D-glucan as an adjunct to diagnosis in a mixed population with uncommon proven invasive fungal diseases or with an unusual clinical presentation. Clin Vaccine Immunol. 2009;16(3):423–426. doi: 10.1128/CVI.00009-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Karageorgopoulos DE, Vouloumanou EK, Ntziora F. Beta-D-glucan assay for the diagnosis of invasive fungal infections: a meta-analysis. Clin Infect Dis. 2011;52(6):750–770. doi: 10.1093/cid/ciq206. [DOI] [PubMed] [Google Scholar]
- 219.Sax PE, Komarow L, Finkelman MA. Blood (1->3)-beta-D-glucan as a diagnostic test for HIV-related Pneumocystis jirovecii pneumonia. Clin Infect Dis. 2011;53(2):197–202. doi: 10.1093/cid/cir335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Morris A, Norris KA. Colonization by Pneumocystis jirovecii and its role in disease. Clin Microbiol Rev. 2012;25(2):297–317. doi: 10.1128/CMR.00013-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Marty FM, Lowry CM, Lempitski SJ. Reactivity of (1–>3)-beta-D-glucan assay with commonly used intravenous antimicrobials. Antimicrob Agents Chemother. 2006;50(10):3450–3453. doi: 10.1128/AAC.00658-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Clark NM, Lynch JP., 3rd Influenza: epidemiology, clinical features, therapy, and prevention. Semin Respir Crit Care Med. 2011;32(4):373–392. doi: 10.1055/s-0031-1283278. [DOI] [PubMed] [Google Scholar]
- 223.Chartrand C, Pai M. How accurate are rapid influenza diagnostic tests? Expert Rev Anti Infect Ther. 2012;10(6):615–617. doi: 10.1586/eri.12.49. [DOI] [PubMed] [Google Scholar]
- 224.Chartrand C, Leeflang MM, Minion J. Accuracy of rapid influenza diagnostic tests: a meta-analysis. Ann Intern Med. 2012;156(7):500–511. doi: 10.7326/0003-4819-156-7-201204030-00403. [DOI] [PubMed] [Google Scholar]
- 225.Centers for Disease Control and Prevention (CDC): Rapid diagnostic testing for influenza: information for health care professionals, <http://www.cdc.gov/flu/professionals/diagnosis/rapidclin.htm>.
- 226.Ginocchio CC, Swierkosz E, McAdam AJ. Multicenter study of clinical performance of the 3M rapid detection RSV test. J Clin Microbiol. 2010;48(7):2337–2343. doi: 10.1128/JCM.00130-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Fried AJ, Bonilla FA. Pathogenesis, diagnosis, and management of primary antibody deficiencies and infections. Clin Microbiol Rev. 2009;22(3):396–414. doi: 10.1128/CMR.00001-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Picard C, Casanova JL, Puel A. Infectious diseases in patients with IRAK-4, MyD88, NEMO, or IkappaBalpha deficiency. Clin Microbiol Rev. 2011;24(3):490–497. doi: 10.1128/CMR.00001-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Salvana EM, Salata RA. Infectious complications associated with monoclonal antibodies and related small molecules. Clin Microbiol Rev. 2009;22(2):274–290. doi: 10.1128/CMR.00040-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Mandell LA, Wunderink RG, Anzueto A. Infectious Diseases Society of America/American Thoracic Society consensus guidelines on the management of community-acquired pneumonia in adults. Clin Infect Dis. 2007;44(Suppl 2):S27–S72. doi: 10.1086/511159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Hill HR, Matsen JM. Enzyme-linked immunosorbent assay and radioimmunoassay in the serologic diagnosis of infectious diseases. J Infect Dis. 1983;147(2):258–263. doi: 10.1093/infdis/147.2.258. [DOI] [PubMed] [Google Scholar]
- 232.Steingart KR, Flores LL, Dendukuri N. Commercial serological tests for the diagnosis of active pulmonary and extrapulmonary tuberculosis: an updated systematic review and meta-analysis. PLoS Med. 2011;8(8):e1001062. doi: 10.1371/journal.pmed.1001062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Daxboeck F, Krause R, Wenisch C. Laboratory diagnosis of Mycoplasma pneumoniae infection. Clin Microbiol Infect. 2003;9(4):263–273. doi: 10.1046/j.1469-0691.2003.00590.x. [DOI] [PubMed] [Google Scholar]
- 234.Pappagianis D, Zimmer BL. Serology of coccidioidomycosis. Clin Microbiol Rev. 1990;3(3):247–268. doi: 10.1128/cmr.3.3.247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Catanzaro A. Coccidioidomycosis. Semin Respir Crit Care Med. 2004;25(2):123–128. doi: 10.1055/s-2004-824897. [DOI] [PubMed] [Google Scholar]
- 236.Anstead GM, Graybill JR. Coccidioidomycosis. Infect Dis Clin North Am. 2006;20(3):621–643. doi: 10.1016/j.idc.2006.06.005. [DOI] [PubMed] [Google Scholar]
- 237.Blair JE, Coakley B, Santelli AC. Serologic testing for symptomatic coccidioidomycosis in immunocompetent and immunosuppressed hosts. Mycopathologia. 2006;162(5):317–324. doi: 10.1007/s11046-006-0062-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Wheat LJ. Improvements in diagnosis of histoplasmosis. Expert Opin Biol Ther. 2006;6(11):1207–1221. doi: 10.1517/14712598.6.11.1207. [DOI] [PubMed] [Google Scholar]
- 239.McKinnell JA, Pappas PG. Blastomycosis: new insights into diagnosis, prevention, and treatment. Clin Chest Med. 2009;30(2):227–239. doi: 10.1016/j.ccm.2009.02.003. [DOI] [PubMed] [Google Scholar]
- 240.Vijayan VK. How to diagnose and manage common parasitic pneumonias. Curr Opin Pulm Med. 2007;13(3):218–224. doi: 10.1097/MCP.0b013e3280f31b58. [DOI] [PubMed] [Google Scholar]
- 241.Berk SL, Verghese A. Parasitic pneumonia. Semin Respir Infect. 1988;3(2):172–178. [PubMed] [Google Scholar]
- 242.Katz JM, Hancock K, Xu X. Serologic assays for influenza surveillance, diagnosis and vaccine evaluation. Expert Rev Anti Infect Ther. 2011;9(6):669–683. doi: 10.1586/eri.11.51. [DOI] [PubMed] [Google Scholar]
- 243.Snydman DR, Limaye AP, Potena L. Update and review: state-of-the-art management of cytomegalovirus infection and disease following thoracic organ transplantation. Transplant Proc. 2011;43(3 Suppl):S1–S17. doi: 10.1016/j.transproceed.2011.02.069. [DOI] [PubMed] [Google Scholar]
- 244.Menzies D, Pai M, Comstock G. Meta-analysis: new tests for the diagnosis of latent tuberculosis infection: areas of uncertainty and recommendations for research. Ann Intern Med. 2007;146(5):340–354. doi: 10.7326/0003-4819-146-5-200703060-00006. [DOI] [PubMed] [Google Scholar]
- 245.Mazurek GH, Jereb J, Lobue P. Guidelines for using the QuantiFERON-TB gold test for detecting Mycobacterium tuberculosis infection, United States. MMWR Recomm Rep. 2005;54(RR–15):49–55. [PubMed] [Google Scholar]
- 246.Mazurek GH, Jereb J, Vernon A. Updated guidelines for using interferon gamma release assays to detect Mycobacterium tuberculosis infection—United States, 2010. MMWR Recomm Rep. 2010;59(RR–5):1–25. [PubMed] [Google Scholar]
- 247.Barry CE, 3rd, Boshoff HI, Dartois V. The spectrum of latent tuberculosis: rethinking the biology and intervention strategies. Nat Rev Microbiol. 2009;7(12):845–855. doi: 10.1038/nrmicro2236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Diel R, Loddenkemper R, Nienhaus A. Evidence-based comparison of commercial interferon-gamma release assays for detecting active TB: a metaanalysis. Chest. 2010;137(4):952–968. doi: 10.1378/chest.09-2350. [DOI] [PubMed] [Google Scholar]
- 249.Mazurek M, Jereb J, Vernon A. Updated guidelines for using interferon gamma release assays to detect Mycobacterium tuberculosis infection—United States, 2010. MMWR Recomm Rep. 2010;59(RR–5):1–25. [PubMed] [Google Scholar]
- 250.Herrera V, Perry S, Parsonnet J. Clinical application and limitations of interferon-{gamma} release assays for the diagnosis of latent tuberculosis infection. Clin Infect Dis. 2011;52(8):1031–1037. doi: 10.1093/cid/cir068. [DOI] [PubMed] [Google Scholar]
- 251.Metcalfe JZ, Everett CK, Steingart KR. Interferon-gamma release assays for active pulmonary tuberculosis diagnosis in adults in low- and middle-income countries: systematic review and meta-analysis. J Infect Dis. 2011;204(Suppl 4):S1120–S1129. doi: 10.1093/infdis/jir410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Pai M, Elwood K. Interferon-gamma release assays for screening of health care workers in low tuberculosis incidence settings: dynamic patterns and interpretational challenges. Can Respir J. 2012;19(2):81–83. doi: 10.1155/2012/420392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Zwerling A, van den Hof S, Scholten J. Interferon-gamma release assays for tuberculosis screening of healthcare workers: a systematic review. Thorax. 2012;67(1):62–70. doi: 10.1136/thx.2010.143180. [DOI] [PubMed] [Google Scholar]
- 254.Rangaka MX, Wilkinson KA, Glynn JR. Predictive value of interferon-gamma release assays for incident active tuberculosis: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12(1):45–55. doi: 10.1016/S1473-3099(11)70210-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Machingaidze S, Verver S, Mulenga H. Predictive value of recent QuantiFERON conversion for tuberculosis disease in adolescents. Am J Respir Crit Care Med. 2012;186(10):1051–1056. doi: 10.1164/rccm.201206-1134OC. [DOI] [PubMed] [Google Scholar]
- 256.Benitez AJ, Thurman KA, Diaz MH. Comparison of real-time PCR and a microimmunofluorescence serological assay for detection of Chlamydophila pneumoniae infection in an outbreak investigation. J Clin Microbiol. 2012;50(1):151–153. doi: 10.1128/JCM.05357-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Miyashita N, Ouchi K, Kishi F. Rapid and simple diagnosis of Chlamydophila pneumoniae pneumonia by an immunochromatographic test for detection of immunoglobulin M antibodies. Clin Vaccine Immunol. 2008;15(7):1128–1131. [Google Scholar]
- 258.Gaydos CA, Roblin PM, Hammerschlag MR. Diagnostic utility of PCR-enzyme immunoassay, culture, and serology for detection of Chlamydia pneumoniae in symptomatic and asymptomatic patients. J Clin Microbiol. 1994;32(4):903–905. doi: 10.1128/jcm.32.4.903-905.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Tarnvik A, Chu MC. New approaches to diagnosis and therapy of tularemia. Ann N Y Acad Sci. 2007;1105:378–404. doi: 10.1196/annals.1409.017. [DOI] [PubMed] [Google Scholar]
- 260.Sharma N, Hotta A, Yamamoto Y. Detection of Francisella tularensis-specific antibodies in patients with tularemia by a novel competitive enzyme-linked immunosorbent assay. Clin Vaccine Immunol. 2013;20(1):9–16. doi: 10.1128/CVI.00516-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Blazquez RM, Espinosa FJ, Martinez-Toldos CM. Sensitivity of urinary antigen test in relation to clinical severity in a large outbreak of Legionella pneumonia in Spain. Eur J Clin Microbiol Infect Dis. 2005;24(7):488–491. doi: 10.1007/s10096-005-1361-3. [DOI] [PubMed] [Google Scholar]
- 262.Dominguez J, Gali N, Matas L. Evaluation of a rapid immunochromatographic assay for the detection of Legionella antigen in urine samples. Eur J Clin Microbiol Infect Dis. 1999;18(12):896–898. doi: 10.1007/s100960050427. [DOI] [PubMed] [Google Scholar]
- 263.Murdoch DR, Walford EJ, Jennings LC. Use of the polymerase chain reaction to detect Legionella DNA in urine and serum samples from patients with pneumonia. Clin Infect Dis. 1996;23(3):475–480. doi: 10.1093/clinids/23.3.475. [DOI] [PubMed] [Google Scholar]
- 264.Ramirez JA, Ahkee S, Tolentino A. Diagnosis of Legionella pneumophila, Mycoplasma pneumoniae, or Chlamydia pneumoniae lower respiratory infection using the polymerase chain reaction on a single throat swab specimen. Diagn Microbiol Infect Dis. 1996;24(1):7–14. doi: 10.1016/0732-8893(95)00254-5. [DOI] [PubMed] [Google Scholar]
- 265.Beersma MF, Dirven K, van Dam AP. Evaluation of 12 commercial tests and the complement fixation test for Mycoplasma pneumoniae-specific immunoglobulin G (IgG) and IgM antibodies, with PCR used as the “gold standard. J Clin Microbiol. 2005;43(5):2277–2285. doi: 10.1128/JCM.43.5.2277-2285.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Zhang L, Zong ZY, Liu YB. PCR versus serology for diagnosing Mycoplasma pneumoniae infection: a systematic review and meta-analysis. Indian J Med Res. 2011;134:270–280. [PMC free article] [PubMed] [Google Scholar]
- 267.Couble A, Rodriguez-Nava V, de Montclos MP. Direct detection of Nocardia spp. in clinical samples by a rapid molecular method. J Clin Microbiol. 2005;43(4):1921–1924. doi: 10.1128/JCM.43.4.1921-1924.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Murdoch DR, Anderson TP, Beynon KA. Evaluation of a PCR assay for detection of Streptococcus pneumoniae in respiratory and nonrespiratory samples from adults with community-acquired pneumonia. J Clin Microbiol. 2003;41(1):63–66. doi: 10.1128/JCM.41.1.63-66.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Gieseker KE, Mackenzie T, Roe MH. Comparison of two rapid Streptococcus pyogenes diagnostic tests with a rigorous culture standard. Pediatr Infect Dis J. 2002;21(10):922–927. doi: 10.1097/00006454-200210000-00007. [DOI] [PubMed] [Google Scholar]
- 270.Francesconi A, Kasai M, Petraitiene R. Characterization and comparison of galactomannan enzyme immunoassay and quantitative real-time PCR assay for detection of Aspergillus fumigatus in bronchoalveolar lavage fluid from experimental invasive pulmonary aspergillosis. J Clin Microbiol. 2006;44(7):2475–2480. doi: 10.1128/JCM.02693-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Durkin M, Witt J, Lemonte A. Antigen assay with the potential to aid in diagnosis of blastomycosis. J Clin Microbiol. 2004;42(10):4873–4875. doi: 10.1128/JCM.42.10.4873-4875.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Bariola JR, Hage CA, Durkin M. Detection of Blastomyces dermatitidis antigen in patients with newly diagnosed blastomycosis. Diagn Microbiol Infect Dis. 2011;69(2):187–191. doi: 10.1016/j.diagmicrobio.2010.09.015. [DOI] [PubMed] [Google Scholar]
- 273.Klein BS, Vergeront JM, Kaufman L. Serological tests for blastomycosis: assessments during a large point-source outbreak in Wisconsin. J Infect Dis. 1987;155(2):262–268. doi: 10.1093/infdis/155.2.262. [DOI] [PubMed] [Google Scholar]
- 274.Hage CA, Knox KS, Wheat LJ. Endemic mycoses: overlooked causes of community acquired pneumonia. Respir Med. 2012;106(6):769–776. doi: 10.1016/j.rmed.2012.02.004. [DOI] [PubMed] [Google Scholar]
- 275.Binnicker MJ, Buckwalter SP, Eisberner JJ. Detection of Coccidioides species in clinical specimens by real-time PCR. J Clin Microbiol. 2007;45(1):173–178. doi: 10.1128/JCM.01776-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Martins TB, Jaskowski TD, Mouritsen CL. Comparison of commercially available enzyme immunoassay with traditional serological tests for detection of antibodies to Coccidioides immitis. J Clin Microbiol. 1995;33(4):940–943. doi: 10.1128/jcm.33.4.940-943.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.McMullan BJ, Halliday C, Sorrell TC. Clinical utility of the cryptococcal antigen lateral flow assay in a diagnostic mycology laboray. PLoS One. 2012;7(11):e49541. doi: 10.1371/journal.pone.0049541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Tanner DC, Weinstein MP, Fedorciw B. Comparison of commercial kits for detection of cryptococcal antigen. J Clin Microbiol. 1994;32(7):1680–1684. doi: 10.1128/jcm.32.7.1680-1684.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Lindsley MD, Mekha N, Baggett HC. Evaluation of a newly developed lateral flow immunoassay for the diagnosis of cryptococcosis. Clin Infect Dis. 2011;53(4):321–325. doi: 10.1093/cid/cir379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Guimaraes AJ, Nosanchuk JD, Zancope-Oliveira RM. Diagnosis of histoplasmosis. Braz J Microbiol. 2006;37(1):1–13. doi: 10.1590/S1517-83822006000100001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Sekhon AS, Kaufman L, Kobayashi GS. Comparative evaluation of the premier enzyme immunoassay, micro-immunodiffusion and complement fixation tests for the detection of Histoplasma capsulatum var. capsulatum antibodies. Mycoses. 1994;37(9–10):313–316. doi: 10.1111/myc.1994.37.9-10.313. [DOI] [PubMed] [Google Scholar]
- 282.Bastien P. Molecular diagnosis of toxoplasmosis. Trans R Soc Trop Med Hyg. 2002;96(Suppl 1):S205–S215. doi: 10.1016/s0035-9203(02)90078-7. [DOI] [PubMed] [Google Scholar]