Microbiologic Diagnosis of Lung Infection




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. The microbiology laboratory also provides epidemiologic data that assist the hospital epidemiologist in the prevention, detection, investigation, and termination of nosocomial outbreaks. 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.


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).




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

Pathogens That Require Laboratory Notification When Clinically Suspected







ORGANISM



  • Bacillus anthracis



  • Brucella species



  • Clostridium botulinum



  • Coccidioides species



  • Francisella tularensis



  • Hemorrhagic fever viruses



  • Yersinia pestis



  • Variola major



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. Tables 17-2, 17-3, and 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

Community-Acquired Pneumonia Order Set























































































































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 studies for those with defined risks, complications, and/or severity



  • Blood culture for defined risk factors, including ICU admission

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

Hospital-Acquired and Ventilator-Associated Pneumonia Order Set













































































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

Immunocompromised Host Pneumonia Order Set





































































































































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 10 10 to 10 12 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 ), some of which, such as Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, and Neisseria meningitidis, can also be pathogens of the lower respiratory tract. The oropharynx can also contain Mycoplasma pneumoniae and aerobic actinomycetes including Nocardia and nontuberculous mycobacteria in the absence of disease. In addition, aspiration of even minute amounts (0.1 to 1 µL) of oropharyngeal secretions can deliver a bolus of 10 9 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 10 7 CFU of gram-negative bacilli per milliliter in seriously ill patients requiring intensive care, patients treated with antibiotics after hospitalization for acute pulmonary inflammatory disease, chronic alcoholic and diabetic patients, institutionalized older adults and chronically ill patients, and hospitalized patients with acute leukemia. 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. 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

Oropharyngeal Bacteria That Can Be Present without Causing Disease










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. The importance of proper sputum collection was documented by Laird 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. 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. In patients with nonproductive cough or suspected mycobacterial, fungal, or Pneumocystis jirovecii infections, 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. Penn and Silberman 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. 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. 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. 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, protected catheter brushes may be used to obtain samples for culture and identification of organisms causing anaerobic pleuropulmonary disease. 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. 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. In turn, nasopharyngeal aspirates or washes have been shown to be more sensitive than nasopharyngeal flocked swabs. 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. Oropharyngeal swabs may also be used for detecting Chlamydophila pneumoniae, M. pneumoniae, and Legionella species.


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. The use of BAL has also been expanded to include diagnosis of bacterial pneumonia, especially for nosocomial cases. 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, although there is little support for using these procedures to diagnose CAP. Although the results of cultures from protected catheter brushes and BAL specimens are quantitatively similar, Meduri and Baselski 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, (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 . Nasopharyngeal aspirates have also been used for diagnosis of TB, although the sensitivity of culture-confirmed TB is lower compared to induced sputum. Stool samples in children with pulmonary TB may become the specimen of choice if processing methods can be optimized to concentrate the tubercle bacilli.


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 pneumonia but is positive only up to 37% in CAP and in less than 25% in nosocomial pneumonia. 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. 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) or the use of enriched fungal medium bottles.


S. pneumoniae can be recovered from urine cultures in as many as 38% of patients with pneumococcal pneumonia. Urine may be tested for the presence of pneumococcal and Legionella pneumophila serogroup 1 antigens. Fungal antigen tests of urine are also available for diagnosis of histoplasmosis and blastomycosis. 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. 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. Endotracheal aspirates are rejected if the screening Gram-stained smears show no organisms. 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.




Figure 17-1


Gram stain of sputum specimens.

A, This specimen contains numerous polymorphonuclear leukocytes and no visible squamous epithelial cells, indicating that the specimen is acceptable for routine bacteriologic culture. B, This specimen contains numerous squamous epithelial cells and rare polymorphonuclear leukocytes, indicating an inadequate specimen for routine sputum culture.

(From Tille P: Bailey & Scott’s diagnostic microbiology , ed 13, Philadelphia, 2014, Mosby, Fig. 69-4.)




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

Accuracy of Assays Used in Diagnosis of Lower Respiratory Tract Infections Caused by Bacteria, Fungi, and Parasites



















































































































































































































































































































































































































































































































































































































Organism Diagnostic Target Testing Method Sample Type Sensitivity (%) Specificity (%) References
BACTERIA
Chlamydia species (excluding C. trachomatis ) Antibody (IgG) Microimmunofluorescence Serum 65–92 30–51
Antibody (IgM) Microimmunofluorescence Serum 43–75 67–84
Antibody (IgG and IgM) Microimmunofluorescence Serum 87–100 22–40
Antibody (IgM) Enzyme immunoassay Serum 100 92.9
DNA PCR—enzyme immunoassay Nasopharyngeal swab 55–83 91–99
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
Antibody Enzyme immunoassay Serum 93.9 96.1
Antibody Latex microagglutination Serum 81.8 98.0
Legionella species Antibody Direct fluorescent antibody Respiratory samples 25–66 94
Antigen Enzyme immunoassay Urine 37.9–85.7 N/A
Antigen Lateral flow immunoassay Urine 80 97–100
Legionella pneumophila serotype 1 Antigen Immunoassay Urine 74 99.1
DNA PCR Urine/serum 64–73 100
DNA PCR Throat swab 88.2 100
Mycoplasma pneumoniae Antibody Complement fixation Serum 65 97
Antibody (IgG and IgM) Enzyme immunoassay Serum 35–77 49–100
DNA PCR Throat swab 62 96
Nocardia asteroides group DNA PCR Tissue/sputum/BAL 100 100
Streptococcus pneumoniae Antigen Lateral flow immunoassay Urine 67–82 93–99.8
DNA PCR Plasma or sputum 26–100 58–99
Streptococcus pyogenes Antigen Enzyme immunoassay Throat swab 70–90 90–100
MYCOBACTERIA
M. tuberculosis complex DNA NAT Smear negative 33.3–92.9 N/A
DNA NAT Smear positive 85.7–94.6 98
Organism Microscopy Carbolfuchsin 32–94 N/A
Organism Microscopy Fluorochrome (HIV−) 52–97 N/A
Organism Microscopy Fluorochrome (HIV+) 26–100 N/A
INVASIVE FUNGI
Aspergillus species DNA PCR Serum 80 100
Antigen Galactomannan enzyme immunoassay Serum 71 89
Blastomyces dermatitidis Antigen Enzyme immunoassay Urine 80.7–92.9 77–79
Antigen Enzyme immunoassay Serum 81.8 100
Antibody Immunodiffusion Serum 28 100
Antibody Enzyme immunoassay Serum 77–100 86–96
Antibody Complement fixation Serum 9 100
Organism Microscopy Body fluid/tissue 38–97 N/A
Coccidioides species DNA Real-time PCR Respiratory sample 92.9–100 98.1–98.4
Antibody (IgG) Complement fixation Serum 67–75 N/A
Serum (IC patients) 33–100 N/A
Antibody (IgG and IgM) Immunodiffusion Serum 53–73 N/A
Serum (IC patients) 0–75 N/A
Antibody (IgG and IgM) Enzyme immunoassay Serum 75–92.6 84.6–98.3
Serum (IC patients) 25–90 N/A
Cryptococcus neoformans (and Cryptococcus gattii ) Antigen Latex agglutination Serum 83–91.1 92.9–100
Antigen Latex agglutination Urine N/A 100
Antigen Latex agglutination CSF 93–100 93–98
Antigen Lateral flow assay Serum 90.1–100 92.9–100
Antigen Lateral flow assay Urine 70.3–94.4 100
Antigen Enzyme immunoassay Serum 94.1–100 93–100
Antigen Enzyme immunoassay Urine 92% Unknown
Histoplasma capsulatum Antibody (IgG) Complement fixation Serum/urine 72.8–94.3 70–80
Antibody (IgM) Microimmunodiffusion Serum 70–100 100
Antibody (IgG) Enzyme immunoassay Serum 91–100 66–97
Serum (AIDS, disseminated) 69.2 N/A
Serum (other IC patients, disseminated) 84.2 N/A
Serum (non-IC patients, disseminated) 85.7 N/A
Serum (pulmonary subacute infection) 92.3 N/A
Antigen Enzyme immunoassay Urine 30.4–100 Variable
Urine (AIDS, disseminated) 92.1 N/A
Urine (other IC patients, disseminated) 93.5 N/A
Urine (non-IC patients, disseminated) 63.6 N/A
Urine (pulmonary subacute infection) 38.9 N/A
Antibody Latex agglutination Serum 65–97 39
Pneumocystis jirovecii DNA PCR Respiratory samples 93 90
Organism Microscopy with silver stain Induced sputum/BAL 86–92 92–97
Antigen Direct fluorescent antibody Induced sputum/BAL 90–97 85–90
Antigen Indirect fluorescent antibody Induced sputum/BAL 86–97 100
Organism Diff-Quik stain Induced sputum/BAL 81–92 97–100
Antigen β- d -glucan assay Serum/plasma 78–100 70–100
Other (fungi excluding P. jirovecii ) Antigen β- d -glucan assay Serum/plasma 76.8 85.3
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

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. In addition, microscopy lacks sensitivity in specimens with less than 10 4 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. The finding of neutrophils with intracellular organisms is considered indicative of an active infectious process.


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 culture 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 ).


Jul 21, 2019 | Posted by in CARDIOLOGY | Comments Off on Microbiologic Diagnosis of Lung Infection

Full access? Get Clinical Tree

Get Clinical Tree app for offline access