Slot System
Featured Buckets
Featured Buckets Admin
Reverse Chronological Sort

Nontuberculous Mycobacterial Pulmonary Disease

Article Type
Changed
Wed, 02/05/2020 - 12:00
Display Headline
Nontuberculous Mycobacterial Pulmonary Disease

Nontuberculous mycobacterial pulmonary disease is a broad term for a group of pulmonary disorders caused and characterized by exposure to environmental mycobacteria other than those belonging to the Mycobacterium tuberculosis complex and Mycobacterium leprae. Mycobacteria are aerobic, nonmotile organisms that appear positive with acid-fast alcohol stains. Nontuberculous mycobacteria (NTM) are ubiquitous in the environment and have been recovered from domestic and natural water sources, soil, and food products, and from around livestock, cattle, and wildlife.1-3 To date, no evidence exists of human-to-human or animal-to-human transmission of NTM in the general population. Infections in humans are usually acquired from environmental exposures, although the specific source of infection cannot always be identified. Similarly, the mode of infection with NTM has not been established with certainty, but it is highly likely that the organism is implanted, ingested, aspirated, or inhaled. Aerosolization of droplets associated with use of bathroom showerheads and municipal water sources and soil contamination are some of the factors associated with the transmission of infection. Proven routes of transmission include showerheads and potting soil dust.2,3

NTM pulmonary disease occurs in individuals with or without comorbid conditions such as bronchiectasis, chronic obstructive pulmonary disease, pulmonary fibrosis, or structural lung diseases. Slender, middle-aged or elderly white females with marfanoid body habitus, with or without apparent immune or genetic disorders, showing impaired airway and mucus clearance present with this infection as a form of underlying bronchiectasis (Lady Windermere syndrome). It is unclear why NTM infections and escalation to clinical disease occur in certain individuals. Many risk factors, including inherited and acquired defects of host immune response (eg, cystic fibrosis trait and α1 antitrypsin deficiency), have been associated with increased susceptibility to NTM infections.4

NTM infection can lead to chronic symptoms, frequent exacerbations, progressive functional and structural lung destruction, and impaired quality of life, and is associated with an increased risk of hospitalization and higher 5-year all-cause mortality. As such, NTM disease is drawing increasing attention at the clinical, academic, and research levels.5 This case-based review outlines the clinical features of NTM infection, with a focus on the challenges in diagnosis, treatment, and management of NTM pulmonary disease. The cases use Mycobacterium avium complex (MAC), a slow-growing mycobacteria (SGM), and Mycobacterium abscessus, a rapidly growing mycobacteria (RGM), as prototypes in a non–cystic fibrosis, non-HIV clinical setting.

Epidemiology

Of the almost 200 isolated species of NTM, the most prevalent pathogens for respiratory disease in the United States are MAC, Mycobacterium kansasii, and M. abscessus. MAC accounts for more than 80% of cases of NTM respiratory disease in the United States.6 The prevalence of NTM disease is increasing at a rate of about 8% each year, with 75,000 to 105,000 patients diagnosed with NTM lung disease in the United States annually. NTM infections in the United States are increasing among patients aged 65 years and older, a population that is expected to nearly double by 2030.7,8

Isolation and prevalence of many NTM species are higher in certain geographic areas of the United States, especially in the southeast. The US coastal regions have a higher prevalence of NTM pulmonary disease, and account for 70% of NTM cases in the United States each year. Half of patients diagnosed with NTM lung disease reside in 7 states: Florida, New York, Texas, California, Pennsylvania, New Jersey, and Ohio, with 1 in 7 residing in Florida. Three parishes in Louisiana are among the top 10 counties with the highest prevalence in United States. The prevalence of NTM infection–associated hospitalizations is increasing worldwide as well. Co-infection with tuberculosis and multiple NTMs in individual patients has been observed clinically and documented in patients with and without HIV.9,10

It is not clear why the prevalence of NTM pulmonary disease is increasing, but there may be several contributing factors: (1) an increased awareness and identification of NTM infection sources in the environment; (2) an expanding cohort of immunocompromised individuals with exogenous or endogenous immune deficiencies; (3) availability of improved diagnostic techniques, such as use of high-performance liquid chromatography (HPLC), DNA probes, and gene sequencing; and (4) an increased awareness of the morbidity and mortality associated with NTM pulmonary disease. However, it is important to recognize that to best understand the clinical relevance of epidemiologic studies based on laboratory diagnosis and identification, the findings must be evaluated by correlating them with the microbiological and other clinical criteria established by the American Thoracic Society (ATS)/Infectious Diseases Society of America (IDSA) guidelines.11

Continue to: Mycobacterium avium Complex

 

 

Mycobacterium avium Complex

Case Patient 1

A 48-year-old woman who has never smoked and has no past medical problems, except seasonal allergic rhinitis and “colds and flu-like illness” once or twice a year, is evaluated for a chronic lingering cough with occasional sputum production. The patient denies any other chronic symptoms and is otherwise active. Physical examination reveals no specific findings except mild pectus excavatum and mild scoliosis. Body mass index is 22 kg/m2. Chest radiograph shows nonspecific increased markings in the lower zones. Computed tomography (CT) scan of the chest reveals minimal nodular and cylindrical bronchiectasis in both lungs (Figure 1). No previous radiographs are available for comparison. The patient is HIV-negative. Sputum tests reveal normal flora, and both fungus and acid-fast bacilli smear are negative. Culture for mycobacteria shows scanty growth of MAC in 1 specimen.

PDBR01601001_f1.PNG

 

What is the clinical presentation of MAC pulmonary disease?

Among NTM, MAC is the most common cause of pulmonary disease worldwide.6 MAC primarily includes 2 species: M. avium and Mycobacterium intracellulare. M. avium is the more important pathogen in disseminated disease, whereas M. intracellulare is the more common respiratory pathogen.11 These organisms are genetically similar and generally not differentiated in the clinical microbiology laboratory, although there are isolated reports in the literature suggesting differences in prevalence, presentation, and prognosis in M. avium infection versus M. intracellulare infection.12

Three major disease syndromes are produced by MAC in humans: pulmonary disease, usually in adults whose systemic immunity is intact; disseminated disease, usually in patients with advanced HIV infection; and cervical lymphadenitis.13 Pulmonary disease caused by MAC may take on 1 of several clinically different forms, including asymptomatic “colonization” or persistent minimal infection without obvious clinical significance; endobronchial involvement; progressive pulmonary disease with radiographic and clinical deterioration and nodular bronchiectasis or cavitary lung disease; hypersensitivity pneumonitis; or persistent, overwhelming mycobacterial growth with symptomatic manifestations, often in a lung with underlying damage due to either chronic obstructive lung disease or pulmonary fibrosis (Table 1).14

PDBR01601001_t1.PNG

Cavitary Disease

The traditionally recognized presentation of MAC lung disease has been apical cavitary lung disease in men in their late 40s and early 50s who have a history of cigarette smoking, and frequently, excessive alcohol use. If left untreated, or in the case of erratic treatment or macrolide drug resistance, this form of disease is generally progressive within a relatively short time and can result in extensive cavitary lung destruction and progressive respiratory failure.15

Nodular Bronchiectasis

The more common presentation of MAC lung disease, which is outlined in the case described here, is interstitial nodular infiltrates, frequently involving the right middle lobe or lingula and predominantly occurring in postmenopausal, nonsmoking white women. This is sometimes labelled “Lady Windermere syndrome.” These patients with M. avium infection appear to have similar clinical characteristics and body types, including lean build, scoliosis, pectus excavatum, and mitral valve prolapse.16,17 The mechanism by which this body morphotype predisposes to pulmonary mycobacterial infection is not defined, but ineffective mucociliary clearance is a possible explanation. Evidence suggests that some patients may be predisposed to NTM lung disease because of preexisting bronchiectasis. Some potential etiologies of bronchiectasis in this population include chronic sinusitis, gastroesophageal reflux with chronic aspiration, α1 antitrypsin deficiency, and cystic fibrosis genetic traits and mutations.18 Risk factors for increased morbidity and mortality include the development of cavitary disease, age, weight loss, lower body mass index, and other comorbid conditions.

This form of disease, termed nodular bronchiectasis, tends to have a much slower progression than cavitary disease, such that long-term follow-up (months to years) may be necessary to demonstrate clinical or radiographic changes.11 The radiographic term “tree-in-bud” has been used to describe what may reflect inflammatory changes, including bronchiolitis. High-resolution CT scans of the chest are especially helpful for diagnosing this pattern of MAC lung disease, as bronchiectasis and small nodules may not be easily discernible on plain chest radiograph. The nodular/bronchiectasis radiographic pattern can also be seen with other NTM pathogens, including M. abscessus, Mycobacterium simiae, and M. kansasii. Solitary nodules and dense consolidation have also been described. Pleural effusions are uncommon, but reactive pleural thickening is frequently seen. Co-pathogens may be isolated from culture, including Pseudomonas aeruginosa, Staphylococcus aureus, and, occasionally, other NTM such as M. abscessus or Mycobacterium chelonae.19-21

Hypersensitivity Pneumonitis

Hypersensitivity pneumonitis, initially described in patients who were exposed to hot tubs, mimics allergic hypersensitivity pneumonitis, with respiratory symptoms and culture/tissue identification of MAC or sometimes other NTM. It is unclear whether hypersensitivity pneumonitis is an inflammatory process, an infection, or both, and opinion regarding the need for specific antibiotic treatment is divided.11,22 However, avoidance of exposure is prudent and recommended.

Disseminated Disease

Disseminated NTM disease is associated with very low CD4+ lymphocyte counts and is seen in approximately 5% of patients with HIV infection.23-25 Although disseminated NTM disease is rarely seen in immunosuppressed patients without HIV infection, it has been reported in patients who have undergone renal or cardiac transplant, patients on long-term corticosteroid therapy, and those with leukemia or lymphoma. More than 90% of infections are caused by MAC; other potential pathogens include M. kansasii, M. chelonae, M. abscessus, and Mycobacterium haemophilum. Although seen less frequently since the advent of highly active antiretroviral therapy, disseminated infection can develop progressively from an apparently indolent or localized infection or a respiratory or gastrointestinal source. Signs and symptoms of disseminated infection (specifically MAC-associated disease) are nonspecific and include fever, night sweats, weight loss, and abdominal tenderness. Disseminated MAC disease occurs primarily in patients with more advanced HIV disease (CD4+ count typically < 50 cells/μL). Clinically, disseminated MAC manifests as intermittent or persistent fever, constitutional symptoms with organomegaly and organ-specific abnormalities (eg, anemia, neutropenia from bone marrow involvement, adenopathy, hepatosplenomegaly), and elevations of liver enzymes or lung infiltrates from pulmonary involvement.

Continue to: What are the criteria for diagnosing NTM pulmonary disease?

 

 

What are the criteria for diagnosing NTM pulmonary disease?

The diagnosis of NTM disease is based on clinical, radiologic, and mycobacterial correlation with good communication between the experts in this field. The ATS/IDSA criteria for diagnosing NTM lung disease are shown in Figure 2. These criteria best apply to MAC, M. kansasii, and M. abscessus, but are also clinically applied to other NTM respiratory pathogens. The diagnosis of MAC infection is most readily established by culture of blood, bone marrow, respiratory secretions/fluid, or tissue specimens from suspected sites of involvement. Due to erratic shedding of MAC into the respiratory secretions in patients with nodular bronchiectasis, as compared to those with the cavitary form of the disease, sputum may be intermittently positive, with variable colony counts and polyclonal infections.12 Prior to the advent of high-resolution CT, isolation of MAC organisms from the sputum of such patients was frequently dismissed as colonization.

PDBR01601001_f2.PNG

 

Mycobacterial Testing

Because of the nonspecific symptoms and lack of diagnostic specificity of chest imaging, the diagnosis of NTM lung disease requires microbiologic confirmation. Specimens sent to the laboratory for identification of NTM must be handled with care to prevent contamination and false-positive results. Transport media and preservatives should be avoided, and transportation of the specimens should be prompt. These measures will prevent bacterial overgrowth. Furthermore, the yield of NTM may be affected if the patient has used antibiotics, such as macrolides and fluoroquinolones, prior to obtaining the specimen.

NTM should be identified at the species and subspecies level, although this is not practical in community practice settings. The preferred staining procedure in the laboratory is the fluorochrome method. Some species require special growth conditions and/or lower incubation temperatures, and other identification methods may have to be employed, such as DNA probes, polymerase chain reaction genotyping, nucleic acid sequence determination, and high-performance liquid chromatography. As a gold standard, clinical specimens for mycobacterial cultures should be inoculated onto 1 or more solid media (eg, Middlebrook 7H11 media and/or Lowenstein-Jensen media, the former of which is the preferred medium for NTM) and into a liquid medium (eg, BACTEC 12B broth or Mycobacteria growth indicator tube broth). Growth of visible colonies on solid media typically requires 2 to 4 weeks, but liquid media (eg, the radiometric BACTEC system), used as a supplementary and not as an exclusive test, usually produce results within 10 to 14 days. Furthermore, even after initial growth, identification of specific isolates based on the growth characteristics on solid media requires additional time. Use of specific nucleic acid probes for MAC and M. kansasii and HPLC testing of mycolic acid patterns in acid-fast bacilli smear–positive specimens can reduce the turnaround time of specific identification of a primary culture–positive sample. However, HPLC is not sufficient for definitive identification of many NTM species, including the RGM. Other newer techniques, including 16S ribosomal DNA sequencing and polymerase chain reaction-restriction fragment length polymorphism analysis, also allow NTM to be identified and speciated more reliably and rapidly from clinical specimens.

Cost and other practical considerations limit widespread adoption of these techniques. However, the recognition that M. abscessus can be separated into more than 1 subspecies, and that there are important prognostic implications of that separation, lends urgency to the broader adoption of newer molecular techniques in the mycobacteriology laboratory. Susceptibility testing is based on the broth microdilution method; RGM usually grow within 7 days of subculture, and the laboratory time to culture is a helpful hint, although not necessarily specific. Recognizing the morphology of mycobacterial colony growth may also be helpful in identification.

Are skin tests helpful in diagnosing NTM infection?

Tuberculin skin testing remains a nonspecific marker of mycobacterial infection and does not help in further elucidating NTM infection. However, epidemiologic and laboratory studies with well-characterized antigens have shown that dual skin testing with tuberculosis- versus NTM-derived tuberculin can discriminate between prior NTM and prior tuberculosis disease. Species-specific skin test antigens are not commercially available and are not helpful in the diagnosis of NTM disease because of cross-reactivity of M. tuberculosis and some NTM. However, increased prevalence of NTM sensitization based on purified protein derivative testing has been noted in a recent survey, which is consistent with an observed increase in the rates of NTM infections, specifically MAC, in the United States.26,27

Interferon-gamma release assays (IGRAs) are now being used as an alternative to tuberculin skin testing to diagnose M. tuberculosis infection. Certain NTM species also contain gene sequences that encode for ESAT-6 or CFP-10 antigens used in the IGRAs, and hence, yield a positive IGRA test. These include M. marinum, M. szulgai, and M. kansasii.28,29 However, MAC organisms do not produce positive results on assays that use these antigens.

Continue to: What is the approach to management of NTM pulmonary disease?

 

 

What is the approach to management of NTM pulmonary disease?

The correlation of symptoms with radiographic and microbiologic evidence is essential to categorize the disease and determine the need for therapy. Making the diagnosis of NTM lung disease does not necessitate the institution of therapy. The decision to treat should be weighed against potential risks and benefits to the individual patient based on symptomatic, radiographic, and microbiologic criteria, as well as underlying systemic or pulmonary immune status. In the absence of evidence of clinical, radiologic, or mycobacterial progression of disease, pursuing airway clearance therapy and clinical surveillance without initiating specific anti-MAC therapy is a reasonable option.11 Identifying the sustained presence of NTM infection, especially MAC, in a patient with underlying clinical and radiographic evidence of bronchiectasis is of value in determining comprehensive treatment and management strategies. Close observation is indicated if the decision not to treat is made. If treatment is initiated, comprehensive management includes long-term follow-up with periodic bacteriologic surveillance, watching for drug toxicity and drug-drug interactions, ensuring adherence and compliance to treatment, and managing comorbidity.

The Bronchiectasis Severity Index is a useful clinical predictive tool that identifies patients at risk of future mortality, hospitalization, and exacerbations and can be used to evaluate the need for specific treatment.30 The index is based on dyspnea score, lung function tests, colonization of pathogens, and extent of disease.

Case 1 Continued

After approximately 2 months of observation and symptomatic treatment, without specific antibiotic therapy, the patient’s symptoms continue. She now develops intermittent hemoptysis. Repeat sputum studies reveal moderate growth of M. avium. A follow-up CT scan shows progression of disease, with an increase in the tree-in-bud pattern (Figure 3).

PDBR01601001_f3.PNG

What treatment protocols are recommended for MAC pulmonary disease?

As per the ATS/IDSA statement, macrolides are the mainstay of treatment for pulmonary MAC disease.11 Macrolides achieve an increased concentration in the lung, and when used for treatment of pulmonary MAC disease, there is a strong correlation between in vitro susceptibility, in vivo (clinical) response, and the immunomodulating effects of macrolides.31,32 Macrolide-containing regimens have demonstrated efficacy in patients with MAC pulmonary disease33,34; however, macrolide monotherapy should be avoided to prevent the development of resistance.

At the outset, it is critical to establish the objective criteria for determining response and to ensure that the patient understands the goals of the treatment and expectations of the treatment plan. Moreover, experts suggest that due to the possibility of drug intolerance, side effects, and the need for prolonged therapy, a “step ladder” ramping up approach to treatment could be adopted, with gradual introduction of therapy within a short time period; this approach may improve compliance and adherence to treatment.11 If this approach is used, the doses may have to be divided. Patients who are unable to tolerate daily medications, even with dosage adjustment, should be tried on an intermittent treatment regimen. Older female patients frequently require gradual introduction of medications (ie, 1 medication added to the regimen every 1 to 2 weeks) to evaluate tolerance to each medication and medication dose.11 Commonly encountered adverse effects of NTM treatment include intolerance to clarithromycin due to gastrointestinal problems, low body mass index, or age older than 70 years.

After determining that the patient requires therapy, the standard recommended treatment for MAC pulmonary disease includes the following: for most patients with nodular/bronchiectasis disease, a thrice-weekly regimen of clarithromycin (1000 mg) or azithromycin (500 mg), rifampin (600 mg), and ethambutol (25 mg/kg) is recommended. For patients with cavitary MAC pulmonary disease or severe nodular/bronchiectasis disease, the guidelines recommend a daily regimen of clarithromycin (500-1000 mg) or azithromycin (250 mg), rifampin (600 mg) or rifabutin (150–300 mg), and ethambutol (15 mg/kg), with consideration of intravenous (IV) amikacin 3 times/week early in therapy (Table 2).11

PDBR01601001_t2.PNG

The treatment of MAC hypersensitivity-like disease speaks to the controversy of whether this is an inflammatory process, infectious process, or a combination of inflammation and infection. Avoidance of exposure is the mainstay of management. In some cases, steroids are used with or without a short course of anti-MAC therapy (ie, clarithromycin or azithromycin with rifampin and ethambutol).

Prophylaxis for disseminated MAC disease should be given to adults with HIV infection who have a CD4+ count less than 50 cells/μL. Azithromycin 1200 mg/week or clarithromycin 1000 mg/day has proven efficacy, and rifabutin 300 mg/day is also effective but less well tolerated. Rifabutin is more active in vitro against MAC than rifampin, and is used in HIV-positive patients because of drug-drug interaction between antiretroviral drugs and rifampin.

Continue to: Case 1 Continued

 

 

Case 1 Continued

The patient is treated with clarithromycin, rifampin, and ethambutol for 1 year, with sputum conversion after 9 months. In the latter part of her treatment, she experiences decreased visual acuity. Treatment is discontinued prematurely after 1 year due to drug toxicity and continued intolerance to drug therapy. The patient remains asymptomatic for 8 months, and then begins to experience mild to moderate hemoptysis, with increasing cough and sputum production associated with postural changes during exercise. Physical examination overall remains unchanged. Three sputum results reveal heavy growth of MAC, and a CT scan of the chest shows a cavitary lesion in the left upper lobe along with the nodular bronchiectasis (Figure 4).

PDBR01601001_f4.PNG

What are the management options at this stage?

Based on this patient’s continued symptoms, progression of radiologic abnormalities, and current culture growth, she requires re-treatment. With the adverse effects associated with ethambutol during the first round of therapy, the drug regimen needs to be modified. Several considerations are relevant at this stage. Relapse rates range from 20% to 30% after treatment with a macrolide-based therapy.11,34 Obtaining a culture-sensitivity profile is imperative in these cases. Of note, treatment should not be discontinued altogether, but instead the toxic agent should be removed from the treatment regimen. Continuing treatment with a 2-drug regimen of clarithromycin and rifampin may be considered in this patient. Re-infection with multiple genotypes may also occur after successful drug therapy, but this is primarily seen in MAC patients with nodular bronchiectasis.34,35 Patients in whom previous therapy has failed, even those with macrolide-susceptible MAC isolates, are less likely to respond to subsequent therapy. Data suggest that intermittent medication dosing is not effective for patients with severe or cavitary disease or in those in whom previous therapy has failed.36 In this case, treatment should include a daily 3-drug therapy, with an injectable thrice-weekly aminoglycoside. Other agents such as linezolid and clofazimine may have to be tried. Cycloserine, ethionamide, and other agents are sometimes used, but their efficacy is unproven and doubtful. Pyrazinamide and isoniazid have no activity against MAC.

Treatment Failure and Drug Resistance

Treatment failure is considered to have occurred if patients have not had a response (microbiologic, clinical, or radiographic) after 6 months of appropriate therapy or had not achieved conversion of sputum to culture-negative after 12 months of appropriate therapy.11 This occurs in about 40% of patients. Multiple factors can interfere with the successful treatment of MAC pulmonary disease, including medication nonadherence, medication side effects or intolerance, lack of response to a medication regimen, or the emergence of a macrolide-resistant or multidrug-resistant strain. Inducible macrolide resistance remains a potential factor.34-36 A number of characteristics of NTM contribute to the poor response to currently used antibiotics: the organisms have a lipid outer membrane and prefer to adhere to surfaces and form biofilms, which makes them relatively impermeable to antibiotics.37 Also, NTM replicate in phagocytic cells, allowing them to subvert normal cellular defense mechanisms. Furthermore, NTM can display colony variants, whereby single colony isolates switch between antibiotic-susceptible and -resistant variants. These factors have also impeded in development of new antibiotics for NTM infection.37

Recent limited approval of amikacin liposomal inhalation suspension (ALIS) for treatment failure and refractory MAC infection in combination with guideline-based antimicrobial therapy (GBT) is a promising addition to the available treatment armamentarium. In a multinational trial, the addition of ALIS to GBT for treatment-refractory MAC lung disease achieved significantly greater culture conversion rates by month 6 than GBT alone, with comparable rates of serious adverse events.38

Is therapeutic drug monitoring recommended during treatment of MAC pulmonary disease?

Treatment failure may also be drug-related, including poor drug penetration into the damaged lung tissue or drug-drug interactions leading to suboptimal drug levels. Peak serum concentrations have been found to be below target ranges in approximately 50% of patients using a macrolide and ethambutol. Concurrent use of rifampin decreases the peak serum concentration of macrolides and quinolones, with acceptable target levels seen in only 18% to 57% of cases. Whether this alters patient outcomes is not clear.39-42 Factors identified as contributing to the poor response to therapy include poor compliance, cavitary disease, previous treatment for MAC pulmonary disease, and a history of chronic obstructive lung disease. Studies by Koh and colleagues40 and van Ingen and colleagues41 with pharmacokinetic and pharmacodynamics data showed that, in patients on MAC treatment with both clarithromycin and rifampicin, plasma levels of clarithromycin were lower than the recommended minimal inhibitory concentrations (MIC) against MAC for that drug. The studies also showed that rifampicin lowered clarithromycin concentrations more than did rifabutin, with the AUC/MIC ratio being suboptimal in nearly half the cases. However, low plasma clarithromycin concentrations did not have any correlation with treatment outcomes, as the peak plasma drug concentrations and the peak plasma drug concentration/MIC ratios did not differ between patients with unfavorable treatment outcomes and those with favorable outcomes. This is further compounded by the fact that macrolides achieve higher levels in lung tissue than in plasma, and hence the significance of low plasma levels is unclear; however, it is postulated that achieving higher drug levels could, in fact, lead to better clinical outcomes. Pending specific well-designed, prospective randomized controlled trials, routine therapeutic drug monitoring is not currently recommended, although some referral centers do this as their practice pattern.

Is surgery an option in this case?

The overall 5-year mortality for MAC pulmonary disease was approximately 28% in a retrospective analysis, with patients with cavitary disease at increased risk for death at 5 years.42 As such, surgery is an option in selected cases as part of adjunctive therapy along with anti-MAC therapy based on mycobacterial sensitivity. Surgery is used as either a curative approach or a “debulking” measure.11 When present, clearly localized disease, especially in the upper lobe, lends itself best to surgical intervention. Surgical resection of a solitary pulmonary nodule due to MAC, in addition to concomitant medical treatment, is recommended. Surgical intervention should be considered early in the course of the disease because it may provide a cure without prolonged treatment and its associated problems, and this approach may lead to early sputum conversion. Surgery should also be considered in patients with macrolide-resistant or multidrug-resistant MAC infection or in those who cannot tolerate the side effects of therapy, provided that the disease is focal and limited. Patients with poor preoperative lung function have poorer outcomes than those with good lung function, and postoperative complications arising from treatment, especially with a right-sided pneumonectomy, tend to occur more frequently in these patients. Thoracic surgery for NTM pulmonary disease must be considered cautiously, as this is associated with significant morbidity and mortality and is best performed at specialized centers that have expertise and experience in this field.43

Continue to: Mycobacterium abscessus Complex

 

 

Mycobacterium abscessus Complex

Case Patient 2

A 64-year-old man who is an ex-smoker presents with chronic cough, mild shortness of breath on exertion, low-grade fever, and unintentional weight loss of 10 lb. Physical exam is unremarkable. The patient was diagnosed with immunoglobulin deficiency (low IgM and low IgG4) in 2002, and has been on replacement therapy since then. He also has had multiple episodes of NTM infection, with MAC and M. kansasii infections diagnosed in 2012-2014, which required 18 months of multi-drug antibiotic treatment that resulted in sputum conversion. Pulmonary function testing done on this visit in 2017 shows mild obstructive impairment.

    Chest radiograph and CT scan show bilateral bronchiectasis (Figure 5 and Figure 6).

    PDBR01601001_f5.PNG

    The results of serial sputum microbiology testing performed over the course of 6 months are outlined below:

    • 5/2017 (bronchoalveolar lavage): 2+; M. abscessus
    • 9/2017 × 2: smear (–); group IV RGM
    • 11/2017: smear (–); M. abscessus (> 50 CFU)
    • 12/2017: smear (–); M. abscessus (> 50 CFU)

     

    PDBR01601001_f6.PNG

    What are the clinical considerations in this patient with multiple NTM infections?

    M. abscessus complex was originally described in soft tissue abscesses and skin infections possibly resulting from soil or water contamination. Subspeciation of M. abscessus complex during laboratory testing is critical to facilitate selection of a specific therapeutic approach; treatment decisions are impacted by the presence of an active erm gene and in vitro macrolide sensitivity, which differ between subspecies. The most acceptable classification outlines 3 species in the M. abscessus complex: Mycobacterium abscessus subsp abscessus, Mycobacterium abscessus subsp bolletii (both with an active erm gene responsible for macrolide resistance), and Mycobacterium abscessus subsp massiliense (with an inactive erm gene and therefore susceptible to macrolides).44

    RGM typically manifest in skin, soft tissue, and bone, and can cause soft tissue, surgical wound, and catheter-related infections. Although the role of RGM as pulmonary pathogens is unclear, underlying diseases associated with RGM include previously treated mycobacterial disease, coexistent pulmonary diseases with or without MAC, cystic fibrosis, malignancies, and gastroesophageal disorders. M. abscessus is the third most commonly identified respiratory NTM and accounts for the majority (80%) of RGM respiratory isolates. Other NTM reported to cause both lung disease and skin, bone, and joint infections include Mycobacterium simiae, Mycobacterium xenopi, and Mycobacterium malmoense. Ocular granulomatous diseases, such as chorioretinitis and keratitis, have been reported with both RGM and Runyon group III SGM, such as MAC or M. szulgai, following trauma or refractive surgery. These can mimic fungal, herpetic, or amebic keratitis. The pulmonary syndromes associated with multiple culture positivity are seen in elderly women with bronchiectasis or cavitary lung disease and/or associated with gastrointestinal symptoms of acid reflux, with or without achalasia and concomitant lipoid interstitial pneumonia.45

    Generally, pulmonary disease progresses slowly, but lung disease attributed to RGM can result in respiratory failure. Thus, RGM should be recognized as a possible cause of chronic mycobacterial lung disease, especially in immunocompromised patients, and respiratory isolates should be assessed carefully. Identification and drug susceptibility testing are essential before initiation of treatment for RGM.

    What is the approach to management of M. abscessus pulmonary disease in a patient without cystic fibrosis?

    The management of M. abscessus pulmonary infection as a subset of RGM requires a considered step-wise approach. The criteria for diagnosis and threshold for starting treatment are the same as those used in the management of MAC pulmonary disease,11 but the treatment of M. abscessus pulmonary infection is more complex and has lower rates of success and cure. Also, antibiotic treatment presents challenges related to rapid identification of the causative organism, nomenclature, resistance patterns, and tolerance of treatment and side effects. If a source such as catheter, access port, or any surgical site is identified, prompt removal and clearance of the infected site are strongly advised

    In the absence of any controlled clinical trials, treatment of RGM is based on in vitro susceptibility testing and expert opinion. As in MAC pulmonary disease, macrolides are the mainstay of treatment, with an induction phase of intravenous antibiotics. Treatment may include a combination of injectable aminoglycosides, imipenem, or cefoxitin and oral drugs such as a macrolide (eg, clarithromycin, azithromycin), doxycycline, fluoroquinolones, trimethoprim/sulfamethoxazole, or linezolid. While antibiotic treatment of M. abscessus pulmonary disease is based on in vitro sensitivity pattern to a greater degree than is treatment of MAC pulmonary disease, this approach has significant practical limitations and hence variable applicability. The final choice of antibiotics is best based on the extended susceptibility results, if available. The presence of an active erm gene on a prolonged growth specimen in M. abscessus subsp abscessus and M. abscessus subsp bolletii precludes the use of a macrolide. In such cases, amikacin, especially in an intravenous form, is the mainstay of treatment based on MIC. Recently, there has been a resurgence in interest in the use of clofazimine in combination with amikacin when treatment is not successful in patients with M. abscessus subsp abscessus or M. bolletii with an active erm gene.45,46 When localized abscess formation is noted, surgery may be the best option, with emphasis on removal of implants and catheters if implicated in RGM infection.

    Attention must also be given to confounding pulmonary and associated comorbidities. This includes management of bronchiectasis with appropriately aggressive airway clearance techniques; anti-reflux measures for prevention of micro-aspiration; and management of other comorbid pulmonary conditions, such as chronic obstructive pulmonary disease, pulmonary fibrosis, and sarcoidosis, if applicable. These interventions play a critical role in clearing the M. abscessus infection, preventing progression of disease, and reducing morbidity. The role of immunomodulatory therapy needs to be considered on a regular, ongoing basis. Identification of genetic factors and correction of immune deficiencies may help in managing the infection.

    Case Patient 2 Conclusion

    The treatment regimen adopted in this case includes a 3-month course of daily intravenous amikacin and imipenem with oral azithromycin, followed by a continuation phase of azithromycin with clofazimine and linezolid. Airway clearance techniques such as Vest/Acapella/CPT are intensified and monthly intravenous immunoglobulin therapy is continued. The patient responds to treatment, with resolution of his clinical symptoms and reduction in the colony count of M. abscessus in the sputum.

    Summary

    NTM are ubiquitous in the environment, and NTM infection has variable manifestations, especially in patients with no recognizable immune impairments. Underlying comorbid conditions with bronchiectasis complicate its management. Treatment strategies must be individualized based on degree of involvement, associated comorbidities, immune deficiencies, goals of therapy, outcome-based risk-benefit ratio assessment, and patient engagement and expectations. In diffuse pulmonary disease, drug treatment remains difficult due to poor match of in vitro and in vivo culture sensitivity, side effects of medications, and high failure rates. When a localized resectable foci of infection is identified, especially in RGM disease, surgical treatment may be the best approach in selected patients, but it must be performed in centers with expertise and experience in this field. 

    References

    1. Johnson MM, Odell JA. Nontuberculous mycobacterial pulmonary infections. J Thorac Dis. 2014;6:210-220.

    2. Falkinham JO III. Environmental sources of NTM. Clin Chest Med. 2015;36:35-41.

    3. Falkinham JO III, Current epidemiological trends in NTM. Curr Environ Health Rep. 2016;3:161-167.

    4. Honda JR, Knight V, Chan ED. Pathogenesis and risk factors for nontuberculous mycobacterial lung disease. Clin Chest Med. 2015;36:1-11.

    5. Marras TK, Mirsaeidi M, Chou E, et al. Health care utilization and expenditures following diagnosis of nontuberculous mycobacterial lung disease in the United States. Manag Care Spec Pharm. 2018;24:964-974.

    6. Prevots DR, Shaw PA, Strickland D, et al. Nontuberculous mycobacterial lung disease prevalence at four integrated healthcare delivery systems. Am J Respir Crit Care Med. 2010;182:970-976.

    7. Winthrop KL, McNelley E, Kendall B, et al. Pulmonary nontuberculous mycobacterial disease prevalence and clinical features: an emerging public health disease. Am J Respir Crit Care Med. 2010;182:977-982.

    8. Adjemian, Olivier KN, Seitz AE, J et al. Prevalence of nontuberculous mycobacterial lung disease in US Medicare beneficiaries. Am J Respir Crit Care Med. 2012;185;881-886.

    9. Ringshausen FC, Apel RM, Bange FC, et al. Burden and trends of hospitalizations associated with pulmonary nontuberculous mycobacterial infections in Germany, 2005-2011. BMC Infect Dis. 2013;13:231.

    10. Aliyu G, El-Kamary SS, Abimiku A, et al. Prevalence of non-tuberculous mycobacterial infections among tuberculosis suspects in Nigeria. PLoS One. 2013;8:e63170.

    11. Griffith DE, Aksamit T, Brown-Elliott, et al; American Thoracic Society; Infectious Diseases Society of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367-415.

    12. Wallace RJ Jr, Zhang Y, Brown BA, et al. Polyclonal Mycobacterium avium complex infections in patients with nodular bronchiectasis. Am J Respir Crit Care Med. 1998;158:1235-1244.

    13. Gordin FM, Horsburgh CR Jr. Mycobacterium avium complex. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Elsevier; 2015.

    14. Chitty S, Ali J. Mycobacterium avium complex pulmonary disease in immune competent patients. South Med J. 2005;98:646-52.

    15. Ramirez J, Mason C, Ali J, Lopez FA. MAC pulmonary disease: management options in HIV-negative patients. J La State Med Soc. 2008;160:248-254.

    16. Iseman MD, Buschman DL, Ackerson LM. Pectus excavatum and scoliosis. Thoracic anomalies associated with pulmonary disease caused by Mycobacterium avium complex. Am Rev Respir Dis. 1991;144:914-916.

    17. Kim RD, Greenburg DE, Ehrmantraut ME, et al. Pulmonary nontuberculous mycobacterial disease: prospective study of a distinct preexisting syndrome. Am J Respir Crit Care Med. 2008;178:1066-1074.

    18. Ziedalski TM, Kao PN, Henig NR, et al. Prospective analysis of cystic fibrosis transmembrane regulator mutations in adults with bronchiectasis or pulmonary nontuberculous mycobacterial infection. Chest. 2006;130:995-1002.

    19. Koh WJ, Lee KS, Kwon OJ, et al. Bilateral bronchiectasis and bronchiolitis at thin-section CT: diagnostic implications in nontuberculous mycobacterial pulmonary infection. Radiology. 2005;235:282-288.

    20. Swensen SJ, Hartman TE, Williams DE. Computed tomographic diagnosis of Mycobacterium avium-intracellulare complex in patients with bronchiectasis. Chest. 1994;105:49-52.

    21. Huang JH, Kao PN, Adi V, Ruoss SJ. Mycobacterium avium intracellulare pulmonary infection in HIV-negative patients without preexisting lung disease: diagnostic and management limitations. Chest. 1999;115:1033-1040.

    22. Cappelluti E, Fraire AE, Schaefer OP. A case of “hot tub lung” due to Mycobacterium avium complex in an immunocompetent host. Arch Intern Med. 2003;163:845-848.

    23. Nightingale SD, Byrd LT, Southern PM, et al. Incidence of Mycobacterium avium-intracellulare complex bacteremia in human immunodeficiency virus-positive patients. J Infect Dis. 1992;165:1082-1085.

    24. Horsburgh CR Jr, Selik RM. The epidemiology of disseminated tuberculous mycobacterial infection in the acquired immunodeficiency syndrome (AIDS). Am Rev Respir Dis. 1989;139:4-7.

    25. Chin DP, Hopewell PC, Yajko DM, et al. Mycobacterium avium complex in the respiratory or gastrointestinal tract and the risk of M. avium complex bacteremia in patients with human immunodeficiency virus infection. J Infect Dis. 1994;169:289-295.

    26. Khan K, Wang J, Marras TK. Nontuberculous mycobacterial sensitization in the United States: national trends over three decades. Am J Respir Crit Care Med. 2007;176:306-313.

    27. Lillo M, Orengo S, Cernoch P, Harris RL. Pulmonary and disseminated infection due to Mycobacterium kansasii: a decade of experience. Rev Infect Dis. 1990;12:760-767.

    28. Andersen P, Munk ME, Pollock JM, Doherty TM. Specific immune-based diagnosis of tuberculosis. Lancet. 2000;356:1099-1104.

    29. Arend SM, van Meijgaarden KE, de Boer K, et al. Tuberculin skin testing and in vitro T cell responses to ESAT-6 and culture filtrate protein 10 after infection with Mycobacterium marinum or M. kansasii. J Infect Dis. 2002;186:1797-1807.

    30. James D, Chalmers JD, Goeminne P, et al. The Bronchiectasis Severity Index: an international derivation and validation study. Am J Respir Crit Care Med. 2014;189:576-585.

    31. Heifets L. MIC as a quantitative measurement of the susceptibility of Mycobacterium avium strains to seven antituberculosis drugs. Antimicrob Agents Chemother. 1988;32:1131-1136.

    32. Horsburgh CR Jr, Mason UG 3rd, Heifits LB, et al. Response to therapy of pulmonary Mycobacterium avium intracellulare infection correlates with results of in vitro susceptibility testing. Am Rev Respir Dis. 1987;135:418-421.

    33. Rubin BK, Henke MO. Immunomodulatory activity and effectiveness of macrolides in chronic airway disease. Chest. 2004;125(2 Suppl):70S-78S.

    34. Wallace RJ Jr, Brown BA, Griffith DE, et al. Clarithromycin regimens for pulmonary Mycobacterium avium complex. The first 50 patients. Am J Respir Crit Care Med. 1996;153:1766-1772.

    35. Griffith DE, Brown-Elliott BA, Langsjoen B, et al. Clinical and molecular analysis of macrolide resistance in Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;174:928-934.

    36. Lam PK, Griffith DE, Aksamit TR, et al. Factors related to response to intermittent treatment of Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;173:1283-1289.

    37. Falkinham J III. Challenges of NTM drug development. Front Microbiol. 2018;9:1613.

    38. Griffith DE, Eagle G, Thomson R, et al. Amikacin liposome inhalation suspension for treatment-refractory lung disease caused by Mycobacterium avium complex (CONVERT). A prospective, open-label, randomized study. Am J Respir Crit Care Med. 2018;198:1559-1569.

    39. Schluger NW. Treatment of pulmonary Mycobacterium avium complex infections: do drug levels matter? Am J Respir Crit Care Med. 2012;186:710-711.

    40. Van Ingen J, Egelund EF, Levin A, et al. The pharmacokinetics and pharmacodynamics of pulmonary Mycobacterium avium complex disease treatment. Am J Respir Crit Care Med. 2012;186:559-565.

    41. Koh WJ, Jeong BH, Jeon K, et al. Therapeutic drug monitoring in the treatment of Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2012;186:797-802.

    42. Ito Y, Hirai T, Maekawa K, et al. Predictors of 5-year mortality in pulmonary MAC disease. Int J Tuberc Lung Dis. 2012;16:408-414.

    43. Yuji S, Yutsuki N, Keiichiso T, et al. Surgery for Mycobacterium avium lung disease in the clarithromycin era. Eur J Cardiothor Surg. 2002;21:314-318.

    44. Tortoli E, Kohl TA, Brown-Elliott BA, et al. Emended description of Mycobacterium abscessus, Mycobacterium abscessus subsp. abscessus and Mycobacterium abscessus subsp. bolletii and designation of Mycobacterium abscessus subsp. massiliense comb. Int J Syst Evol Microbiol. 2016; 66:4471-4479.

    45. Griffith DE, Girard WM, Wallace RJ Jr. Clinical features of pulmonary disease caused by rapidly growing mycobacteria. An analysis of 154 patients. Am Rev Respir Dis. 1993;147:1271-1278.

    46. Koh WJ, Jeong BH, Kim SY, et al. Mycobacterial characteristics and treatment outcomes in Mycobacterium abscessus lung disease. Clin Infect Dis. 2017;64:309-316.

    Author and Disclosure Information

    Juzar Ali, MD, FRCP(C), FCCP LSU
    Alumni Klein Professor of Medicine, Section of Pulmonary/Critical Care, Louisiana State University Health Sciences Center New Orleans; Director, Wetmore Mycobacterial Disease & Bronchiectasis Program, New Orleans, LA; Adjunct Professor, Department of Tropical Medicine, Tulane University Health Sciences Center, New Orleans, LA.

    Publications
    Topics
    Sections
    Author and Disclosure Information

    Juzar Ali, MD, FRCP(C), FCCP LSU
    Alumni Klein Professor of Medicine, Section of Pulmonary/Critical Care, Louisiana State University Health Sciences Center New Orleans; Director, Wetmore Mycobacterial Disease & Bronchiectasis Program, New Orleans, LA; Adjunct Professor, Department of Tropical Medicine, Tulane University Health Sciences Center, New Orleans, LA.

    Author and Disclosure Information

    Juzar Ali, MD, FRCP(C), FCCP LSU
    Alumni Klein Professor of Medicine, Section of Pulmonary/Critical Care, Louisiana State University Health Sciences Center New Orleans; Director, Wetmore Mycobacterial Disease & Bronchiectasis Program, New Orleans, LA; Adjunct Professor, Department of Tropical Medicine, Tulane University Health Sciences Center, New Orleans, LA.

    Nontuberculous mycobacterial pulmonary disease is a broad term for a group of pulmonary disorders caused and characterized by exposure to environmental mycobacteria other than those belonging to the Mycobacterium tuberculosis complex and Mycobacterium leprae. Mycobacteria are aerobic, nonmotile organisms that appear positive with acid-fast alcohol stains. Nontuberculous mycobacteria (NTM) are ubiquitous in the environment and have been recovered from domestic and natural water sources, soil, and food products, and from around livestock, cattle, and wildlife.1-3 To date, no evidence exists of human-to-human or animal-to-human transmission of NTM in the general population. Infections in humans are usually acquired from environmental exposures, although the specific source of infection cannot always be identified. Similarly, the mode of infection with NTM has not been established with certainty, but it is highly likely that the organism is implanted, ingested, aspirated, or inhaled. Aerosolization of droplets associated with use of bathroom showerheads and municipal water sources and soil contamination are some of the factors associated with the transmission of infection. Proven routes of transmission include showerheads and potting soil dust.2,3

    NTM pulmonary disease occurs in individuals with or without comorbid conditions such as bronchiectasis, chronic obstructive pulmonary disease, pulmonary fibrosis, or structural lung diseases. Slender, middle-aged or elderly white females with marfanoid body habitus, with or without apparent immune or genetic disorders, showing impaired airway and mucus clearance present with this infection as a form of underlying bronchiectasis (Lady Windermere syndrome). It is unclear why NTM infections and escalation to clinical disease occur in certain individuals. Many risk factors, including inherited and acquired defects of host immune response (eg, cystic fibrosis trait and α1 antitrypsin deficiency), have been associated with increased susceptibility to NTM infections.4

    NTM infection can lead to chronic symptoms, frequent exacerbations, progressive functional and structural lung destruction, and impaired quality of life, and is associated with an increased risk of hospitalization and higher 5-year all-cause mortality. As such, NTM disease is drawing increasing attention at the clinical, academic, and research levels.5 This case-based review outlines the clinical features of NTM infection, with a focus on the challenges in diagnosis, treatment, and management of NTM pulmonary disease. The cases use Mycobacterium avium complex (MAC), a slow-growing mycobacteria (SGM), and Mycobacterium abscessus, a rapidly growing mycobacteria (RGM), as prototypes in a non–cystic fibrosis, non-HIV clinical setting.

    Epidemiology

    Of the almost 200 isolated species of NTM, the most prevalent pathogens for respiratory disease in the United States are MAC, Mycobacterium kansasii, and M. abscessus. MAC accounts for more than 80% of cases of NTM respiratory disease in the United States.6 The prevalence of NTM disease is increasing at a rate of about 8% each year, with 75,000 to 105,000 patients diagnosed with NTM lung disease in the United States annually. NTM infections in the United States are increasing among patients aged 65 years and older, a population that is expected to nearly double by 2030.7,8

    Isolation and prevalence of many NTM species are higher in certain geographic areas of the United States, especially in the southeast. The US coastal regions have a higher prevalence of NTM pulmonary disease, and account for 70% of NTM cases in the United States each year. Half of patients diagnosed with NTM lung disease reside in 7 states: Florida, New York, Texas, California, Pennsylvania, New Jersey, and Ohio, with 1 in 7 residing in Florida. Three parishes in Louisiana are among the top 10 counties with the highest prevalence in United States. The prevalence of NTM infection–associated hospitalizations is increasing worldwide as well. Co-infection with tuberculosis and multiple NTMs in individual patients has been observed clinically and documented in patients with and without HIV.9,10

    It is not clear why the prevalence of NTM pulmonary disease is increasing, but there may be several contributing factors: (1) an increased awareness and identification of NTM infection sources in the environment; (2) an expanding cohort of immunocompromised individuals with exogenous or endogenous immune deficiencies; (3) availability of improved diagnostic techniques, such as use of high-performance liquid chromatography (HPLC), DNA probes, and gene sequencing; and (4) an increased awareness of the morbidity and mortality associated with NTM pulmonary disease. However, it is important to recognize that to best understand the clinical relevance of epidemiologic studies based on laboratory diagnosis and identification, the findings must be evaluated by correlating them with the microbiological and other clinical criteria established by the American Thoracic Society (ATS)/Infectious Diseases Society of America (IDSA) guidelines.11

    Continue to: Mycobacterium avium Complex

     

     

    Mycobacterium avium Complex

    Case Patient 1

    A 48-year-old woman who has never smoked and has no past medical problems, except seasonal allergic rhinitis and “colds and flu-like illness” once or twice a year, is evaluated for a chronic lingering cough with occasional sputum production. The patient denies any other chronic symptoms and is otherwise active. Physical examination reveals no specific findings except mild pectus excavatum and mild scoliosis. Body mass index is 22 kg/m2. Chest radiograph shows nonspecific increased markings in the lower zones. Computed tomography (CT) scan of the chest reveals minimal nodular and cylindrical bronchiectasis in both lungs (Figure 1). No previous radiographs are available for comparison. The patient is HIV-negative. Sputum tests reveal normal flora, and both fungus and acid-fast bacilli smear are negative. Culture for mycobacteria shows scanty growth of MAC in 1 specimen.

    PDBR01601001_f1.PNG

     

    What is the clinical presentation of MAC pulmonary disease?

    Among NTM, MAC is the most common cause of pulmonary disease worldwide.6 MAC primarily includes 2 species: M. avium and Mycobacterium intracellulare. M. avium is the more important pathogen in disseminated disease, whereas M. intracellulare is the more common respiratory pathogen.11 These organisms are genetically similar and generally not differentiated in the clinical microbiology laboratory, although there are isolated reports in the literature suggesting differences in prevalence, presentation, and prognosis in M. avium infection versus M. intracellulare infection.12

    Three major disease syndromes are produced by MAC in humans: pulmonary disease, usually in adults whose systemic immunity is intact; disseminated disease, usually in patients with advanced HIV infection; and cervical lymphadenitis.13 Pulmonary disease caused by MAC may take on 1 of several clinically different forms, including asymptomatic “colonization” or persistent minimal infection without obvious clinical significance; endobronchial involvement; progressive pulmonary disease with radiographic and clinical deterioration and nodular bronchiectasis or cavitary lung disease; hypersensitivity pneumonitis; or persistent, overwhelming mycobacterial growth with symptomatic manifestations, often in a lung with underlying damage due to either chronic obstructive lung disease or pulmonary fibrosis (Table 1).14

    PDBR01601001_t1.PNG

    Cavitary Disease

    The traditionally recognized presentation of MAC lung disease has been apical cavitary lung disease in men in their late 40s and early 50s who have a history of cigarette smoking, and frequently, excessive alcohol use. If left untreated, or in the case of erratic treatment or macrolide drug resistance, this form of disease is generally progressive within a relatively short time and can result in extensive cavitary lung destruction and progressive respiratory failure.15

    Nodular Bronchiectasis

    The more common presentation of MAC lung disease, which is outlined in the case described here, is interstitial nodular infiltrates, frequently involving the right middle lobe or lingula and predominantly occurring in postmenopausal, nonsmoking white women. This is sometimes labelled “Lady Windermere syndrome.” These patients with M. avium infection appear to have similar clinical characteristics and body types, including lean build, scoliosis, pectus excavatum, and mitral valve prolapse.16,17 The mechanism by which this body morphotype predisposes to pulmonary mycobacterial infection is not defined, but ineffective mucociliary clearance is a possible explanation. Evidence suggests that some patients may be predisposed to NTM lung disease because of preexisting bronchiectasis. Some potential etiologies of bronchiectasis in this population include chronic sinusitis, gastroesophageal reflux with chronic aspiration, α1 antitrypsin deficiency, and cystic fibrosis genetic traits and mutations.18 Risk factors for increased morbidity and mortality include the development of cavitary disease, age, weight loss, lower body mass index, and other comorbid conditions.

    This form of disease, termed nodular bronchiectasis, tends to have a much slower progression than cavitary disease, such that long-term follow-up (months to years) may be necessary to demonstrate clinical or radiographic changes.11 The radiographic term “tree-in-bud” has been used to describe what may reflect inflammatory changes, including bronchiolitis. High-resolution CT scans of the chest are especially helpful for diagnosing this pattern of MAC lung disease, as bronchiectasis and small nodules may not be easily discernible on plain chest radiograph. The nodular/bronchiectasis radiographic pattern can also be seen with other NTM pathogens, including M. abscessus, Mycobacterium simiae, and M. kansasii. Solitary nodules and dense consolidation have also been described. Pleural effusions are uncommon, but reactive pleural thickening is frequently seen. Co-pathogens may be isolated from culture, including Pseudomonas aeruginosa, Staphylococcus aureus, and, occasionally, other NTM such as M. abscessus or Mycobacterium chelonae.19-21

    Hypersensitivity Pneumonitis

    Hypersensitivity pneumonitis, initially described in patients who were exposed to hot tubs, mimics allergic hypersensitivity pneumonitis, with respiratory symptoms and culture/tissue identification of MAC or sometimes other NTM. It is unclear whether hypersensitivity pneumonitis is an inflammatory process, an infection, or both, and opinion regarding the need for specific antibiotic treatment is divided.11,22 However, avoidance of exposure is prudent and recommended.

    Disseminated Disease

    Disseminated NTM disease is associated with very low CD4+ lymphocyte counts and is seen in approximately 5% of patients with HIV infection.23-25 Although disseminated NTM disease is rarely seen in immunosuppressed patients without HIV infection, it has been reported in patients who have undergone renal or cardiac transplant, patients on long-term corticosteroid therapy, and those with leukemia or lymphoma. More than 90% of infections are caused by MAC; other potential pathogens include M. kansasii, M. chelonae, M. abscessus, and Mycobacterium haemophilum. Although seen less frequently since the advent of highly active antiretroviral therapy, disseminated infection can develop progressively from an apparently indolent or localized infection or a respiratory or gastrointestinal source. Signs and symptoms of disseminated infection (specifically MAC-associated disease) are nonspecific and include fever, night sweats, weight loss, and abdominal tenderness. Disseminated MAC disease occurs primarily in patients with more advanced HIV disease (CD4+ count typically < 50 cells/μL). Clinically, disseminated MAC manifests as intermittent or persistent fever, constitutional symptoms with organomegaly and organ-specific abnormalities (eg, anemia, neutropenia from bone marrow involvement, adenopathy, hepatosplenomegaly), and elevations of liver enzymes or lung infiltrates from pulmonary involvement.

    Continue to: What are the criteria for diagnosing NTM pulmonary disease?

     

     

    What are the criteria for diagnosing NTM pulmonary disease?

    The diagnosis of NTM disease is based on clinical, radiologic, and mycobacterial correlation with good communication between the experts in this field. The ATS/IDSA criteria for diagnosing NTM lung disease are shown in Figure 2. These criteria best apply to MAC, M. kansasii, and M. abscessus, but are also clinically applied to other NTM respiratory pathogens. The diagnosis of MAC infection is most readily established by culture of blood, bone marrow, respiratory secretions/fluid, or tissue specimens from suspected sites of involvement. Due to erratic shedding of MAC into the respiratory secretions in patients with nodular bronchiectasis, as compared to those with the cavitary form of the disease, sputum may be intermittently positive, with variable colony counts and polyclonal infections.12 Prior to the advent of high-resolution CT, isolation of MAC organisms from the sputum of such patients was frequently dismissed as colonization.

    PDBR01601001_f2.PNG

     

    Mycobacterial Testing

    Because of the nonspecific symptoms and lack of diagnostic specificity of chest imaging, the diagnosis of NTM lung disease requires microbiologic confirmation. Specimens sent to the laboratory for identification of NTM must be handled with care to prevent contamination and false-positive results. Transport media and preservatives should be avoided, and transportation of the specimens should be prompt. These measures will prevent bacterial overgrowth. Furthermore, the yield of NTM may be affected if the patient has used antibiotics, such as macrolides and fluoroquinolones, prior to obtaining the specimen.

    NTM should be identified at the species and subspecies level, although this is not practical in community practice settings. The preferred staining procedure in the laboratory is the fluorochrome method. Some species require special growth conditions and/or lower incubation temperatures, and other identification methods may have to be employed, such as DNA probes, polymerase chain reaction genotyping, nucleic acid sequence determination, and high-performance liquid chromatography. As a gold standard, clinical specimens for mycobacterial cultures should be inoculated onto 1 or more solid media (eg, Middlebrook 7H11 media and/or Lowenstein-Jensen media, the former of which is the preferred medium for NTM) and into a liquid medium (eg, BACTEC 12B broth or Mycobacteria growth indicator tube broth). Growth of visible colonies on solid media typically requires 2 to 4 weeks, but liquid media (eg, the radiometric BACTEC system), used as a supplementary and not as an exclusive test, usually produce results within 10 to 14 days. Furthermore, even after initial growth, identification of specific isolates based on the growth characteristics on solid media requires additional time. Use of specific nucleic acid probes for MAC and M. kansasii and HPLC testing of mycolic acid patterns in acid-fast bacilli smear–positive specimens can reduce the turnaround time of specific identification of a primary culture–positive sample. However, HPLC is not sufficient for definitive identification of many NTM species, including the RGM. Other newer techniques, including 16S ribosomal DNA sequencing and polymerase chain reaction-restriction fragment length polymorphism analysis, also allow NTM to be identified and speciated more reliably and rapidly from clinical specimens.

    Cost and other practical considerations limit widespread adoption of these techniques. However, the recognition that M. abscessus can be separated into more than 1 subspecies, and that there are important prognostic implications of that separation, lends urgency to the broader adoption of newer molecular techniques in the mycobacteriology laboratory. Susceptibility testing is based on the broth microdilution method; RGM usually grow within 7 days of subculture, and the laboratory time to culture is a helpful hint, although not necessarily specific. Recognizing the morphology of mycobacterial colony growth may also be helpful in identification.

    Are skin tests helpful in diagnosing NTM infection?

    Tuberculin skin testing remains a nonspecific marker of mycobacterial infection and does not help in further elucidating NTM infection. However, epidemiologic and laboratory studies with well-characterized antigens have shown that dual skin testing with tuberculosis- versus NTM-derived tuberculin can discriminate between prior NTM and prior tuberculosis disease. Species-specific skin test antigens are not commercially available and are not helpful in the diagnosis of NTM disease because of cross-reactivity of M. tuberculosis and some NTM. However, increased prevalence of NTM sensitization based on purified protein derivative testing has been noted in a recent survey, which is consistent with an observed increase in the rates of NTM infections, specifically MAC, in the United States.26,27

    Interferon-gamma release assays (IGRAs) are now being used as an alternative to tuberculin skin testing to diagnose M. tuberculosis infection. Certain NTM species also contain gene sequences that encode for ESAT-6 or CFP-10 antigens used in the IGRAs, and hence, yield a positive IGRA test. These include M. marinum, M. szulgai, and M. kansasii.28,29 However, MAC organisms do not produce positive results on assays that use these antigens.

    Continue to: What is the approach to management of NTM pulmonary disease?

     

     

    What is the approach to management of NTM pulmonary disease?

    The correlation of symptoms with radiographic and microbiologic evidence is essential to categorize the disease and determine the need for therapy. Making the diagnosis of NTM lung disease does not necessitate the institution of therapy. The decision to treat should be weighed against potential risks and benefits to the individual patient based on symptomatic, radiographic, and microbiologic criteria, as well as underlying systemic or pulmonary immune status. In the absence of evidence of clinical, radiologic, or mycobacterial progression of disease, pursuing airway clearance therapy and clinical surveillance without initiating specific anti-MAC therapy is a reasonable option.11 Identifying the sustained presence of NTM infection, especially MAC, in a patient with underlying clinical and radiographic evidence of bronchiectasis is of value in determining comprehensive treatment and management strategies. Close observation is indicated if the decision not to treat is made. If treatment is initiated, comprehensive management includes long-term follow-up with periodic bacteriologic surveillance, watching for drug toxicity and drug-drug interactions, ensuring adherence and compliance to treatment, and managing comorbidity.

    The Bronchiectasis Severity Index is a useful clinical predictive tool that identifies patients at risk of future mortality, hospitalization, and exacerbations and can be used to evaluate the need for specific treatment.30 The index is based on dyspnea score, lung function tests, colonization of pathogens, and extent of disease.

    Case 1 Continued

    After approximately 2 months of observation and symptomatic treatment, without specific antibiotic therapy, the patient’s symptoms continue. She now develops intermittent hemoptysis. Repeat sputum studies reveal moderate growth of M. avium. A follow-up CT scan shows progression of disease, with an increase in the tree-in-bud pattern (Figure 3).

    PDBR01601001_f3.PNG

    What treatment protocols are recommended for MAC pulmonary disease?

    As per the ATS/IDSA statement, macrolides are the mainstay of treatment for pulmonary MAC disease.11 Macrolides achieve an increased concentration in the lung, and when used for treatment of pulmonary MAC disease, there is a strong correlation between in vitro susceptibility, in vivo (clinical) response, and the immunomodulating effects of macrolides.31,32 Macrolide-containing regimens have demonstrated efficacy in patients with MAC pulmonary disease33,34; however, macrolide monotherapy should be avoided to prevent the development of resistance.

    At the outset, it is critical to establish the objective criteria for determining response and to ensure that the patient understands the goals of the treatment and expectations of the treatment plan. Moreover, experts suggest that due to the possibility of drug intolerance, side effects, and the need for prolonged therapy, a “step ladder” ramping up approach to treatment could be adopted, with gradual introduction of therapy within a short time period; this approach may improve compliance and adherence to treatment.11 If this approach is used, the doses may have to be divided. Patients who are unable to tolerate daily medications, even with dosage adjustment, should be tried on an intermittent treatment regimen. Older female patients frequently require gradual introduction of medications (ie, 1 medication added to the regimen every 1 to 2 weeks) to evaluate tolerance to each medication and medication dose.11 Commonly encountered adverse effects of NTM treatment include intolerance to clarithromycin due to gastrointestinal problems, low body mass index, or age older than 70 years.

    After determining that the patient requires therapy, the standard recommended treatment for MAC pulmonary disease includes the following: for most patients with nodular/bronchiectasis disease, a thrice-weekly regimen of clarithromycin (1000 mg) or azithromycin (500 mg), rifampin (600 mg), and ethambutol (25 mg/kg) is recommended. For patients with cavitary MAC pulmonary disease or severe nodular/bronchiectasis disease, the guidelines recommend a daily regimen of clarithromycin (500-1000 mg) or azithromycin (250 mg), rifampin (600 mg) or rifabutin (150–300 mg), and ethambutol (15 mg/kg), with consideration of intravenous (IV) amikacin 3 times/week early in therapy (Table 2).11

    PDBR01601001_t2.PNG

    The treatment of MAC hypersensitivity-like disease speaks to the controversy of whether this is an inflammatory process, infectious process, or a combination of inflammation and infection. Avoidance of exposure is the mainstay of management. In some cases, steroids are used with or without a short course of anti-MAC therapy (ie, clarithromycin or azithromycin with rifampin and ethambutol).

    Prophylaxis for disseminated MAC disease should be given to adults with HIV infection who have a CD4+ count less than 50 cells/μL. Azithromycin 1200 mg/week or clarithromycin 1000 mg/day has proven efficacy, and rifabutin 300 mg/day is also effective but less well tolerated. Rifabutin is more active in vitro against MAC than rifampin, and is used in HIV-positive patients because of drug-drug interaction between antiretroviral drugs and rifampin.

    Continue to: Case 1 Continued

     

     

    Case 1 Continued

    The patient is treated with clarithromycin, rifampin, and ethambutol for 1 year, with sputum conversion after 9 months. In the latter part of her treatment, she experiences decreased visual acuity. Treatment is discontinued prematurely after 1 year due to drug toxicity and continued intolerance to drug therapy. The patient remains asymptomatic for 8 months, and then begins to experience mild to moderate hemoptysis, with increasing cough and sputum production associated with postural changes during exercise. Physical examination overall remains unchanged. Three sputum results reveal heavy growth of MAC, and a CT scan of the chest shows a cavitary lesion in the left upper lobe along with the nodular bronchiectasis (Figure 4).

    PDBR01601001_f4.PNG

    What are the management options at this stage?

    Based on this patient’s continued symptoms, progression of radiologic abnormalities, and current culture growth, she requires re-treatment. With the adverse effects associated with ethambutol during the first round of therapy, the drug regimen needs to be modified. Several considerations are relevant at this stage. Relapse rates range from 20% to 30% after treatment with a macrolide-based therapy.11,34 Obtaining a culture-sensitivity profile is imperative in these cases. Of note, treatment should not be discontinued altogether, but instead the toxic agent should be removed from the treatment regimen. Continuing treatment with a 2-drug regimen of clarithromycin and rifampin may be considered in this patient. Re-infection with multiple genotypes may also occur after successful drug therapy, but this is primarily seen in MAC patients with nodular bronchiectasis.34,35 Patients in whom previous therapy has failed, even those with macrolide-susceptible MAC isolates, are less likely to respond to subsequent therapy. Data suggest that intermittent medication dosing is not effective for patients with severe or cavitary disease or in those in whom previous therapy has failed.36 In this case, treatment should include a daily 3-drug therapy, with an injectable thrice-weekly aminoglycoside. Other agents such as linezolid and clofazimine may have to be tried. Cycloserine, ethionamide, and other agents are sometimes used, but their efficacy is unproven and doubtful. Pyrazinamide and isoniazid have no activity against MAC.

    Treatment Failure and Drug Resistance

    Treatment failure is considered to have occurred if patients have not had a response (microbiologic, clinical, or radiographic) after 6 months of appropriate therapy or had not achieved conversion of sputum to culture-negative after 12 months of appropriate therapy.11 This occurs in about 40% of patients. Multiple factors can interfere with the successful treatment of MAC pulmonary disease, including medication nonadherence, medication side effects or intolerance, lack of response to a medication regimen, or the emergence of a macrolide-resistant or multidrug-resistant strain. Inducible macrolide resistance remains a potential factor.34-36 A number of characteristics of NTM contribute to the poor response to currently used antibiotics: the organisms have a lipid outer membrane and prefer to adhere to surfaces and form biofilms, which makes them relatively impermeable to antibiotics.37 Also, NTM replicate in phagocytic cells, allowing them to subvert normal cellular defense mechanisms. Furthermore, NTM can display colony variants, whereby single colony isolates switch between antibiotic-susceptible and -resistant variants. These factors have also impeded in development of new antibiotics for NTM infection.37

    Recent limited approval of amikacin liposomal inhalation suspension (ALIS) for treatment failure and refractory MAC infection in combination with guideline-based antimicrobial therapy (GBT) is a promising addition to the available treatment armamentarium. In a multinational trial, the addition of ALIS to GBT for treatment-refractory MAC lung disease achieved significantly greater culture conversion rates by month 6 than GBT alone, with comparable rates of serious adverse events.38

    Is therapeutic drug monitoring recommended during treatment of MAC pulmonary disease?

    Treatment failure may also be drug-related, including poor drug penetration into the damaged lung tissue or drug-drug interactions leading to suboptimal drug levels. Peak serum concentrations have been found to be below target ranges in approximately 50% of patients using a macrolide and ethambutol. Concurrent use of rifampin decreases the peak serum concentration of macrolides and quinolones, with acceptable target levels seen in only 18% to 57% of cases. Whether this alters patient outcomes is not clear.39-42 Factors identified as contributing to the poor response to therapy include poor compliance, cavitary disease, previous treatment for MAC pulmonary disease, and a history of chronic obstructive lung disease. Studies by Koh and colleagues40 and van Ingen and colleagues41 with pharmacokinetic and pharmacodynamics data showed that, in patients on MAC treatment with both clarithromycin and rifampicin, plasma levels of clarithromycin were lower than the recommended minimal inhibitory concentrations (MIC) against MAC for that drug. The studies also showed that rifampicin lowered clarithromycin concentrations more than did rifabutin, with the AUC/MIC ratio being suboptimal in nearly half the cases. However, low plasma clarithromycin concentrations did not have any correlation with treatment outcomes, as the peak plasma drug concentrations and the peak plasma drug concentration/MIC ratios did not differ between patients with unfavorable treatment outcomes and those with favorable outcomes. This is further compounded by the fact that macrolides achieve higher levels in lung tissue than in plasma, and hence the significance of low plasma levels is unclear; however, it is postulated that achieving higher drug levels could, in fact, lead to better clinical outcomes. Pending specific well-designed, prospective randomized controlled trials, routine therapeutic drug monitoring is not currently recommended, although some referral centers do this as their practice pattern.

    Is surgery an option in this case?

    The overall 5-year mortality for MAC pulmonary disease was approximately 28% in a retrospective analysis, with patients with cavitary disease at increased risk for death at 5 years.42 As such, surgery is an option in selected cases as part of adjunctive therapy along with anti-MAC therapy based on mycobacterial sensitivity. Surgery is used as either a curative approach or a “debulking” measure.11 When present, clearly localized disease, especially in the upper lobe, lends itself best to surgical intervention. Surgical resection of a solitary pulmonary nodule due to MAC, in addition to concomitant medical treatment, is recommended. Surgical intervention should be considered early in the course of the disease because it may provide a cure without prolonged treatment and its associated problems, and this approach may lead to early sputum conversion. Surgery should also be considered in patients with macrolide-resistant or multidrug-resistant MAC infection or in those who cannot tolerate the side effects of therapy, provided that the disease is focal and limited. Patients with poor preoperative lung function have poorer outcomes than those with good lung function, and postoperative complications arising from treatment, especially with a right-sided pneumonectomy, tend to occur more frequently in these patients. Thoracic surgery for NTM pulmonary disease must be considered cautiously, as this is associated with significant morbidity and mortality and is best performed at specialized centers that have expertise and experience in this field.43

    Continue to: Mycobacterium abscessus Complex

     

     

    Mycobacterium abscessus Complex

    Case Patient 2

    A 64-year-old man who is an ex-smoker presents with chronic cough, mild shortness of breath on exertion, low-grade fever, and unintentional weight loss of 10 lb. Physical exam is unremarkable. The patient was diagnosed with immunoglobulin deficiency (low IgM and low IgG4) in 2002, and has been on replacement therapy since then. He also has had multiple episodes of NTM infection, with MAC and M. kansasii infections diagnosed in 2012-2014, which required 18 months of multi-drug antibiotic treatment that resulted in sputum conversion. Pulmonary function testing done on this visit in 2017 shows mild obstructive impairment.

      Chest radiograph and CT scan show bilateral bronchiectasis (Figure 5 and Figure 6).

      PDBR01601001_f5.PNG

      The results of serial sputum microbiology testing performed over the course of 6 months are outlined below:

      • 5/2017 (bronchoalveolar lavage): 2+; M. abscessus
      • 9/2017 × 2: smear (–); group IV RGM
      • 11/2017: smear (–); M. abscessus (> 50 CFU)
      • 12/2017: smear (–); M. abscessus (> 50 CFU)

       

      PDBR01601001_f6.PNG

      What are the clinical considerations in this patient with multiple NTM infections?

      M. abscessus complex was originally described in soft tissue abscesses and skin infections possibly resulting from soil or water contamination. Subspeciation of M. abscessus complex during laboratory testing is critical to facilitate selection of a specific therapeutic approach; treatment decisions are impacted by the presence of an active erm gene and in vitro macrolide sensitivity, which differ between subspecies. The most acceptable classification outlines 3 species in the M. abscessus complex: Mycobacterium abscessus subsp abscessus, Mycobacterium abscessus subsp bolletii (both with an active erm gene responsible for macrolide resistance), and Mycobacterium abscessus subsp massiliense (with an inactive erm gene and therefore susceptible to macrolides).44

      RGM typically manifest in skin, soft tissue, and bone, and can cause soft tissue, surgical wound, and catheter-related infections. Although the role of RGM as pulmonary pathogens is unclear, underlying diseases associated with RGM include previously treated mycobacterial disease, coexistent pulmonary diseases with or without MAC, cystic fibrosis, malignancies, and gastroesophageal disorders. M. abscessus is the third most commonly identified respiratory NTM and accounts for the majority (80%) of RGM respiratory isolates. Other NTM reported to cause both lung disease and skin, bone, and joint infections include Mycobacterium simiae, Mycobacterium xenopi, and Mycobacterium malmoense. Ocular granulomatous diseases, such as chorioretinitis and keratitis, have been reported with both RGM and Runyon group III SGM, such as MAC or M. szulgai, following trauma or refractive surgery. These can mimic fungal, herpetic, or amebic keratitis. The pulmonary syndromes associated with multiple culture positivity are seen in elderly women with bronchiectasis or cavitary lung disease and/or associated with gastrointestinal symptoms of acid reflux, with or without achalasia and concomitant lipoid interstitial pneumonia.45

      Generally, pulmonary disease progresses slowly, but lung disease attributed to RGM can result in respiratory failure. Thus, RGM should be recognized as a possible cause of chronic mycobacterial lung disease, especially in immunocompromised patients, and respiratory isolates should be assessed carefully. Identification and drug susceptibility testing are essential before initiation of treatment for RGM.

      What is the approach to management of M. abscessus pulmonary disease in a patient without cystic fibrosis?

      The management of M. abscessus pulmonary infection as a subset of RGM requires a considered step-wise approach. The criteria for diagnosis and threshold for starting treatment are the same as those used in the management of MAC pulmonary disease,11 but the treatment of M. abscessus pulmonary infection is more complex and has lower rates of success and cure. Also, antibiotic treatment presents challenges related to rapid identification of the causative organism, nomenclature, resistance patterns, and tolerance of treatment and side effects. If a source such as catheter, access port, or any surgical site is identified, prompt removal and clearance of the infected site are strongly advised

      In the absence of any controlled clinical trials, treatment of RGM is based on in vitro susceptibility testing and expert opinion. As in MAC pulmonary disease, macrolides are the mainstay of treatment, with an induction phase of intravenous antibiotics. Treatment may include a combination of injectable aminoglycosides, imipenem, or cefoxitin and oral drugs such as a macrolide (eg, clarithromycin, azithromycin), doxycycline, fluoroquinolones, trimethoprim/sulfamethoxazole, or linezolid. While antibiotic treatment of M. abscessus pulmonary disease is based on in vitro sensitivity pattern to a greater degree than is treatment of MAC pulmonary disease, this approach has significant practical limitations and hence variable applicability. The final choice of antibiotics is best based on the extended susceptibility results, if available. The presence of an active erm gene on a prolonged growth specimen in M. abscessus subsp abscessus and M. abscessus subsp bolletii precludes the use of a macrolide. In such cases, amikacin, especially in an intravenous form, is the mainstay of treatment based on MIC. Recently, there has been a resurgence in interest in the use of clofazimine in combination with amikacin when treatment is not successful in patients with M. abscessus subsp abscessus or M. bolletii with an active erm gene.45,46 When localized abscess formation is noted, surgery may be the best option, with emphasis on removal of implants and catheters if implicated in RGM infection.

      Attention must also be given to confounding pulmonary and associated comorbidities. This includes management of bronchiectasis with appropriately aggressive airway clearance techniques; anti-reflux measures for prevention of micro-aspiration; and management of other comorbid pulmonary conditions, such as chronic obstructive pulmonary disease, pulmonary fibrosis, and sarcoidosis, if applicable. These interventions play a critical role in clearing the M. abscessus infection, preventing progression of disease, and reducing morbidity. The role of immunomodulatory therapy needs to be considered on a regular, ongoing basis. Identification of genetic factors and correction of immune deficiencies may help in managing the infection.

      Case Patient 2 Conclusion

      The treatment regimen adopted in this case includes a 3-month course of daily intravenous amikacin and imipenem with oral azithromycin, followed by a continuation phase of azithromycin with clofazimine and linezolid. Airway clearance techniques such as Vest/Acapella/CPT are intensified and monthly intravenous immunoglobulin therapy is continued. The patient responds to treatment, with resolution of his clinical symptoms and reduction in the colony count of M. abscessus in the sputum.

      Summary

      NTM are ubiquitous in the environment, and NTM infection has variable manifestations, especially in patients with no recognizable immune impairments. Underlying comorbid conditions with bronchiectasis complicate its management. Treatment strategies must be individualized based on degree of involvement, associated comorbidities, immune deficiencies, goals of therapy, outcome-based risk-benefit ratio assessment, and patient engagement and expectations. In diffuse pulmonary disease, drug treatment remains difficult due to poor match of in vitro and in vivo culture sensitivity, side effects of medications, and high failure rates. When a localized resectable foci of infection is identified, especially in RGM disease, surgical treatment may be the best approach in selected patients, but it must be performed in centers with expertise and experience in this field. 

      Nontuberculous mycobacterial pulmonary disease is a broad term for a group of pulmonary disorders caused and characterized by exposure to environmental mycobacteria other than those belonging to the Mycobacterium tuberculosis complex and Mycobacterium leprae. Mycobacteria are aerobic, nonmotile organisms that appear positive with acid-fast alcohol stains. Nontuberculous mycobacteria (NTM) are ubiquitous in the environment and have been recovered from domestic and natural water sources, soil, and food products, and from around livestock, cattle, and wildlife.1-3 To date, no evidence exists of human-to-human or animal-to-human transmission of NTM in the general population. Infections in humans are usually acquired from environmental exposures, although the specific source of infection cannot always be identified. Similarly, the mode of infection with NTM has not been established with certainty, but it is highly likely that the organism is implanted, ingested, aspirated, or inhaled. Aerosolization of droplets associated with use of bathroom showerheads and municipal water sources and soil contamination are some of the factors associated with the transmission of infection. Proven routes of transmission include showerheads and potting soil dust.2,3

      NTM pulmonary disease occurs in individuals with or without comorbid conditions such as bronchiectasis, chronic obstructive pulmonary disease, pulmonary fibrosis, or structural lung diseases. Slender, middle-aged or elderly white females with marfanoid body habitus, with or without apparent immune or genetic disorders, showing impaired airway and mucus clearance present with this infection as a form of underlying bronchiectasis (Lady Windermere syndrome). It is unclear why NTM infections and escalation to clinical disease occur in certain individuals. Many risk factors, including inherited and acquired defects of host immune response (eg, cystic fibrosis trait and α1 antitrypsin deficiency), have been associated with increased susceptibility to NTM infections.4

      NTM infection can lead to chronic symptoms, frequent exacerbations, progressive functional and structural lung destruction, and impaired quality of life, and is associated with an increased risk of hospitalization and higher 5-year all-cause mortality. As such, NTM disease is drawing increasing attention at the clinical, academic, and research levels.5 This case-based review outlines the clinical features of NTM infection, with a focus on the challenges in diagnosis, treatment, and management of NTM pulmonary disease. The cases use Mycobacterium avium complex (MAC), a slow-growing mycobacteria (SGM), and Mycobacterium abscessus, a rapidly growing mycobacteria (RGM), as prototypes in a non–cystic fibrosis, non-HIV clinical setting.

      Epidemiology

      Of the almost 200 isolated species of NTM, the most prevalent pathogens for respiratory disease in the United States are MAC, Mycobacterium kansasii, and M. abscessus. MAC accounts for more than 80% of cases of NTM respiratory disease in the United States.6 The prevalence of NTM disease is increasing at a rate of about 8% each year, with 75,000 to 105,000 patients diagnosed with NTM lung disease in the United States annually. NTM infections in the United States are increasing among patients aged 65 years and older, a population that is expected to nearly double by 2030.7,8

      Isolation and prevalence of many NTM species are higher in certain geographic areas of the United States, especially in the southeast. The US coastal regions have a higher prevalence of NTM pulmonary disease, and account for 70% of NTM cases in the United States each year. Half of patients diagnosed with NTM lung disease reside in 7 states: Florida, New York, Texas, California, Pennsylvania, New Jersey, and Ohio, with 1 in 7 residing in Florida. Three parishes in Louisiana are among the top 10 counties with the highest prevalence in United States. The prevalence of NTM infection–associated hospitalizations is increasing worldwide as well. Co-infection with tuberculosis and multiple NTMs in individual patients has been observed clinically and documented in patients with and without HIV.9,10

      It is not clear why the prevalence of NTM pulmonary disease is increasing, but there may be several contributing factors: (1) an increased awareness and identification of NTM infection sources in the environment; (2) an expanding cohort of immunocompromised individuals with exogenous or endogenous immune deficiencies; (3) availability of improved diagnostic techniques, such as use of high-performance liquid chromatography (HPLC), DNA probes, and gene sequencing; and (4) an increased awareness of the morbidity and mortality associated with NTM pulmonary disease. However, it is important to recognize that to best understand the clinical relevance of epidemiologic studies based on laboratory diagnosis and identification, the findings must be evaluated by correlating them with the microbiological and other clinical criteria established by the American Thoracic Society (ATS)/Infectious Diseases Society of America (IDSA) guidelines.11

      Continue to: Mycobacterium avium Complex

       

       

      Mycobacterium avium Complex

      Case Patient 1

      A 48-year-old woman who has never smoked and has no past medical problems, except seasonal allergic rhinitis and “colds and flu-like illness” once or twice a year, is evaluated for a chronic lingering cough with occasional sputum production. The patient denies any other chronic symptoms and is otherwise active. Physical examination reveals no specific findings except mild pectus excavatum and mild scoliosis. Body mass index is 22 kg/m2. Chest radiograph shows nonspecific increased markings in the lower zones. Computed tomography (CT) scan of the chest reveals minimal nodular and cylindrical bronchiectasis in both lungs (Figure 1). No previous radiographs are available for comparison. The patient is HIV-negative. Sputum tests reveal normal flora, and both fungus and acid-fast bacilli smear are negative. Culture for mycobacteria shows scanty growth of MAC in 1 specimen.

      PDBR01601001_f1.PNG

       

      What is the clinical presentation of MAC pulmonary disease?

      Among NTM, MAC is the most common cause of pulmonary disease worldwide.6 MAC primarily includes 2 species: M. avium and Mycobacterium intracellulare. M. avium is the more important pathogen in disseminated disease, whereas M. intracellulare is the more common respiratory pathogen.11 These organisms are genetically similar and generally not differentiated in the clinical microbiology laboratory, although there are isolated reports in the literature suggesting differences in prevalence, presentation, and prognosis in M. avium infection versus M. intracellulare infection.12

      Three major disease syndromes are produced by MAC in humans: pulmonary disease, usually in adults whose systemic immunity is intact; disseminated disease, usually in patients with advanced HIV infection; and cervical lymphadenitis.13 Pulmonary disease caused by MAC may take on 1 of several clinically different forms, including asymptomatic “colonization” or persistent minimal infection without obvious clinical significance; endobronchial involvement; progressive pulmonary disease with radiographic and clinical deterioration and nodular bronchiectasis or cavitary lung disease; hypersensitivity pneumonitis; or persistent, overwhelming mycobacterial growth with symptomatic manifestations, often in a lung with underlying damage due to either chronic obstructive lung disease or pulmonary fibrosis (Table 1).14

      PDBR01601001_t1.PNG

      Cavitary Disease

      The traditionally recognized presentation of MAC lung disease has been apical cavitary lung disease in men in their late 40s and early 50s who have a history of cigarette smoking, and frequently, excessive alcohol use. If left untreated, or in the case of erratic treatment or macrolide drug resistance, this form of disease is generally progressive within a relatively short time and can result in extensive cavitary lung destruction and progressive respiratory failure.15

      Nodular Bronchiectasis

      The more common presentation of MAC lung disease, which is outlined in the case described here, is interstitial nodular infiltrates, frequently involving the right middle lobe or lingula and predominantly occurring in postmenopausal, nonsmoking white women. This is sometimes labelled “Lady Windermere syndrome.” These patients with M. avium infection appear to have similar clinical characteristics and body types, including lean build, scoliosis, pectus excavatum, and mitral valve prolapse.16,17 The mechanism by which this body morphotype predisposes to pulmonary mycobacterial infection is not defined, but ineffective mucociliary clearance is a possible explanation. Evidence suggests that some patients may be predisposed to NTM lung disease because of preexisting bronchiectasis. Some potential etiologies of bronchiectasis in this population include chronic sinusitis, gastroesophageal reflux with chronic aspiration, α1 antitrypsin deficiency, and cystic fibrosis genetic traits and mutations.18 Risk factors for increased morbidity and mortality include the development of cavitary disease, age, weight loss, lower body mass index, and other comorbid conditions.

      This form of disease, termed nodular bronchiectasis, tends to have a much slower progression than cavitary disease, such that long-term follow-up (months to years) may be necessary to demonstrate clinical or radiographic changes.11 The radiographic term “tree-in-bud” has been used to describe what may reflect inflammatory changes, including bronchiolitis. High-resolution CT scans of the chest are especially helpful for diagnosing this pattern of MAC lung disease, as bronchiectasis and small nodules may not be easily discernible on plain chest radiograph. The nodular/bronchiectasis radiographic pattern can also be seen with other NTM pathogens, including M. abscessus, Mycobacterium simiae, and M. kansasii. Solitary nodules and dense consolidation have also been described. Pleural effusions are uncommon, but reactive pleural thickening is frequently seen. Co-pathogens may be isolated from culture, including Pseudomonas aeruginosa, Staphylococcus aureus, and, occasionally, other NTM such as M. abscessus or Mycobacterium chelonae.19-21

      Hypersensitivity Pneumonitis

      Hypersensitivity pneumonitis, initially described in patients who were exposed to hot tubs, mimics allergic hypersensitivity pneumonitis, with respiratory symptoms and culture/tissue identification of MAC or sometimes other NTM. It is unclear whether hypersensitivity pneumonitis is an inflammatory process, an infection, or both, and opinion regarding the need for specific antibiotic treatment is divided.11,22 However, avoidance of exposure is prudent and recommended.

      Disseminated Disease

      Disseminated NTM disease is associated with very low CD4+ lymphocyte counts and is seen in approximately 5% of patients with HIV infection.23-25 Although disseminated NTM disease is rarely seen in immunosuppressed patients without HIV infection, it has been reported in patients who have undergone renal or cardiac transplant, patients on long-term corticosteroid therapy, and those with leukemia or lymphoma. More than 90% of infections are caused by MAC; other potential pathogens include M. kansasii, M. chelonae, M. abscessus, and Mycobacterium haemophilum. Although seen less frequently since the advent of highly active antiretroviral therapy, disseminated infection can develop progressively from an apparently indolent or localized infection or a respiratory or gastrointestinal source. Signs and symptoms of disseminated infection (specifically MAC-associated disease) are nonspecific and include fever, night sweats, weight loss, and abdominal tenderness. Disseminated MAC disease occurs primarily in patients with more advanced HIV disease (CD4+ count typically < 50 cells/μL). Clinically, disseminated MAC manifests as intermittent or persistent fever, constitutional symptoms with organomegaly and organ-specific abnormalities (eg, anemia, neutropenia from bone marrow involvement, adenopathy, hepatosplenomegaly), and elevations of liver enzymes or lung infiltrates from pulmonary involvement.

      Continue to: What are the criteria for diagnosing NTM pulmonary disease?

       

       

      What are the criteria for diagnosing NTM pulmonary disease?

      The diagnosis of NTM disease is based on clinical, radiologic, and mycobacterial correlation with good communication between the experts in this field. The ATS/IDSA criteria for diagnosing NTM lung disease are shown in Figure 2. These criteria best apply to MAC, M. kansasii, and M. abscessus, but are also clinically applied to other NTM respiratory pathogens. The diagnosis of MAC infection is most readily established by culture of blood, bone marrow, respiratory secretions/fluid, or tissue specimens from suspected sites of involvement. Due to erratic shedding of MAC into the respiratory secretions in patients with nodular bronchiectasis, as compared to those with the cavitary form of the disease, sputum may be intermittently positive, with variable colony counts and polyclonal infections.12 Prior to the advent of high-resolution CT, isolation of MAC organisms from the sputum of such patients was frequently dismissed as colonization.

      PDBR01601001_f2.PNG

       

      Mycobacterial Testing

      Because of the nonspecific symptoms and lack of diagnostic specificity of chest imaging, the diagnosis of NTM lung disease requires microbiologic confirmation. Specimens sent to the laboratory for identification of NTM must be handled with care to prevent contamination and false-positive results. Transport media and preservatives should be avoided, and transportation of the specimens should be prompt. These measures will prevent bacterial overgrowth. Furthermore, the yield of NTM may be affected if the patient has used antibiotics, such as macrolides and fluoroquinolones, prior to obtaining the specimen.

      NTM should be identified at the species and subspecies level, although this is not practical in community practice settings. The preferred staining procedure in the laboratory is the fluorochrome method. Some species require special growth conditions and/or lower incubation temperatures, and other identification methods may have to be employed, such as DNA probes, polymerase chain reaction genotyping, nucleic acid sequence determination, and high-performance liquid chromatography. As a gold standard, clinical specimens for mycobacterial cultures should be inoculated onto 1 or more solid media (eg, Middlebrook 7H11 media and/or Lowenstein-Jensen media, the former of which is the preferred medium for NTM) and into a liquid medium (eg, BACTEC 12B broth or Mycobacteria growth indicator tube broth). Growth of visible colonies on solid media typically requires 2 to 4 weeks, but liquid media (eg, the radiometric BACTEC system), used as a supplementary and not as an exclusive test, usually produce results within 10 to 14 days. Furthermore, even after initial growth, identification of specific isolates based on the growth characteristics on solid media requires additional time. Use of specific nucleic acid probes for MAC and M. kansasii and HPLC testing of mycolic acid patterns in acid-fast bacilli smear–positive specimens can reduce the turnaround time of specific identification of a primary culture–positive sample. However, HPLC is not sufficient for definitive identification of many NTM species, including the RGM. Other newer techniques, including 16S ribosomal DNA sequencing and polymerase chain reaction-restriction fragment length polymorphism analysis, also allow NTM to be identified and speciated more reliably and rapidly from clinical specimens.

      Cost and other practical considerations limit widespread adoption of these techniques. However, the recognition that M. abscessus can be separated into more than 1 subspecies, and that there are important prognostic implications of that separation, lends urgency to the broader adoption of newer molecular techniques in the mycobacteriology laboratory. Susceptibility testing is based on the broth microdilution method; RGM usually grow within 7 days of subculture, and the laboratory time to culture is a helpful hint, although not necessarily specific. Recognizing the morphology of mycobacterial colony growth may also be helpful in identification.

      Are skin tests helpful in diagnosing NTM infection?

      Tuberculin skin testing remains a nonspecific marker of mycobacterial infection and does not help in further elucidating NTM infection. However, epidemiologic and laboratory studies with well-characterized antigens have shown that dual skin testing with tuberculosis- versus NTM-derived tuberculin can discriminate between prior NTM and prior tuberculosis disease. Species-specific skin test antigens are not commercially available and are not helpful in the diagnosis of NTM disease because of cross-reactivity of M. tuberculosis and some NTM. However, increased prevalence of NTM sensitization based on purified protein derivative testing has been noted in a recent survey, which is consistent with an observed increase in the rates of NTM infections, specifically MAC, in the United States.26,27

      Interferon-gamma release assays (IGRAs) are now being used as an alternative to tuberculin skin testing to diagnose M. tuberculosis infection. Certain NTM species also contain gene sequences that encode for ESAT-6 or CFP-10 antigens used in the IGRAs, and hence, yield a positive IGRA test. These include M. marinum, M. szulgai, and M. kansasii.28,29 However, MAC organisms do not produce positive results on assays that use these antigens.

      Continue to: What is the approach to management of NTM pulmonary disease?

       

       

      What is the approach to management of NTM pulmonary disease?

      The correlation of symptoms with radiographic and microbiologic evidence is essential to categorize the disease and determine the need for therapy. Making the diagnosis of NTM lung disease does not necessitate the institution of therapy. The decision to treat should be weighed against potential risks and benefits to the individual patient based on symptomatic, radiographic, and microbiologic criteria, as well as underlying systemic or pulmonary immune status. In the absence of evidence of clinical, radiologic, or mycobacterial progression of disease, pursuing airway clearance therapy and clinical surveillance without initiating specific anti-MAC therapy is a reasonable option.11 Identifying the sustained presence of NTM infection, especially MAC, in a patient with underlying clinical and radiographic evidence of bronchiectasis is of value in determining comprehensive treatment and management strategies. Close observation is indicated if the decision not to treat is made. If treatment is initiated, comprehensive management includes long-term follow-up with periodic bacteriologic surveillance, watching for drug toxicity and drug-drug interactions, ensuring adherence and compliance to treatment, and managing comorbidity.

      The Bronchiectasis Severity Index is a useful clinical predictive tool that identifies patients at risk of future mortality, hospitalization, and exacerbations and can be used to evaluate the need for specific treatment.30 The index is based on dyspnea score, lung function tests, colonization of pathogens, and extent of disease.

      Case 1 Continued

      After approximately 2 months of observation and symptomatic treatment, without specific antibiotic therapy, the patient’s symptoms continue. She now develops intermittent hemoptysis. Repeat sputum studies reveal moderate growth of M. avium. A follow-up CT scan shows progression of disease, with an increase in the tree-in-bud pattern (Figure 3).

      PDBR01601001_f3.PNG

      What treatment protocols are recommended for MAC pulmonary disease?

      As per the ATS/IDSA statement, macrolides are the mainstay of treatment for pulmonary MAC disease.11 Macrolides achieve an increased concentration in the lung, and when used for treatment of pulmonary MAC disease, there is a strong correlation between in vitro susceptibility, in vivo (clinical) response, and the immunomodulating effects of macrolides.31,32 Macrolide-containing regimens have demonstrated efficacy in patients with MAC pulmonary disease33,34; however, macrolide monotherapy should be avoided to prevent the development of resistance.

      At the outset, it is critical to establish the objective criteria for determining response and to ensure that the patient understands the goals of the treatment and expectations of the treatment plan. Moreover, experts suggest that due to the possibility of drug intolerance, side effects, and the need for prolonged therapy, a “step ladder” ramping up approach to treatment could be adopted, with gradual introduction of therapy within a short time period; this approach may improve compliance and adherence to treatment.11 If this approach is used, the doses may have to be divided. Patients who are unable to tolerate daily medications, even with dosage adjustment, should be tried on an intermittent treatment regimen. Older female patients frequently require gradual introduction of medications (ie, 1 medication added to the regimen every 1 to 2 weeks) to evaluate tolerance to each medication and medication dose.11 Commonly encountered adverse effects of NTM treatment include intolerance to clarithromycin due to gastrointestinal problems, low body mass index, or age older than 70 years.

      After determining that the patient requires therapy, the standard recommended treatment for MAC pulmonary disease includes the following: for most patients with nodular/bronchiectasis disease, a thrice-weekly regimen of clarithromycin (1000 mg) or azithromycin (500 mg), rifampin (600 mg), and ethambutol (25 mg/kg) is recommended. For patients with cavitary MAC pulmonary disease or severe nodular/bronchiectasis disease, the guidelines recommend a daily regimen of clarithromycin (500-1000 mg) or azithromycin (250 mg), rifampin (600 mg) or rifabutin (150–300 mg), and ethambutol (15 mg/kg), with consideration of intravenous (IV) amikacin 3 times/week early in therapy (Table 2).11

      PDBR01601001_t2.PNG

      The treatment of MAC hypersensitivity-like disease speaks to the controversy of whether this is an inflammatory process, infectious process, or a combination of inflammation and infection. Avoidance of exposure is the mainstay of management. In some cases, steroids are used with or without a short course of anti-MAC therapy (ie, clarithromycin or azithromycin with rifampin and ethambutol).

      Prophylaxis for disseminated MAC disease should be given to adults with HIV infection who have a CD4+ count less than 50 cells/μL. Azithromycin 1200 mg/week or clarithromycin 1000 mg/day has proven efficacy, and rifabutin 300 mg/day is also effective but less well tolerated. Rifabutin is more active in vitro against MAC than rifampin, and is used in HIV-positive patients because of drug-drug interaction between antiretroviral drugs and rifampin.

      Continue to: Case 1 Continued

       

       

      Case 1 Continued

      The patient is treated with clarithromycin, rifampin, and ethambutol for 1 year, with sputum conversion after 9 months. In the latter part of her treatment, she experiences decreased visual acuity. Treatment is discontinued prematurely after 1 year due to drug toxicity and continued intolerance to drug therapy. The patient remains asymptomatic for 8 months, and then begins to experience mild to moderate hemoptysis, with increasing cough and sputum production associated with postural changes during exercise. Physical examination overall remains unchanged. Three sputum results reveal heavy growth of MAC, and a CT scan of the chest shows a cavitary lesion in the left upper lobe along with the nodular bronchiectasis (Figure 4).

      PDBR01601001_f4.PNG

      What are the management options at this stage?

      Based on this patient’s continued symptoms, progression of radiologic abnormalities, and current culture growth, she requires re-treatment. With the adverse effects associated with ethambutol during the first round of therapy, the drug regimen needs to be modified. Several considerations are relevant at this stage. Relapse rates range from 20% to 30% after treatment with a macrolide-based therapy.11,34 Obtaining a culture-sensitivity profile is imperative in these cases. Of note, treatment should not be discontinued altogether, but instead the toxic agent should be removed from the treatment regimen. Continuing treatment with a 2-drug regimen of clarithromycin and rifampin may be considered in this patient. Re-infection with multiple genotypes may also occur after successful drug therapy, but this is primarily seen in MAC patients with nodular bronchiectasis.34,35 Patients in whom previous therapy has failed, even those with macrolide-susceptible MAC isolates, are less likely to respond to subsequent therapy. Data suggest that intermittent medication dosing is not effective for patients with severe or cavitary disease or in those in whom previous therapy has failed.36 In this case, treatment should include a daily 3-drug therapy, with an injectable thrice-weekly aminoglycoside. Other agents such as linezolid and clofazimine may have to be tried. Cycloserine, ethionamide, and other agents are sometimes used, but their efficacy is unproven and doubtful. Pyrazinamide and isoniazid have no activity against MAC.

      Treatment Failure and Drug Resistance

      Treatment failure is considered to have occurred if patients have not had a response (microbiologic, clinical, or radiographic) after 6 months of appropriate therapy or had not achieved conversion of sputum to culture-negative after 12 months of appropriate therapy.11 This occurs in about 40% of patients. Multiple factors can interfere with the successful treatment of MAC pulmonary disease, including medication nonadherence, medication side effects or intolerance, lack of response to a medication regimen, or the emergence of a macrolide-resistant or multidrug-resistant strain. Inducible macrolide resistance remains a potential factor.34-36 A number of characteristics of NTM contribute to the poor response to currently used antibiotics: the organisms have a lipid outer membrane and prefer to adhere to surfaces and form biofilms, which makes them relatively impermeable to antibiotics.37 Also, NTM replicate in phagocytic cells, allowing them to subvert normal cellular defense mechanisms. Furthermore, NTM can display colony variants, whereby single colony isolates switch between antibiotic-susceptible and -resistant variants. These factors have also impeded in development of new antibiotics for NTM infection.37

      Recent limited approval of amikacin liposomal inhalation suspension (ALIS) for treatment failure and refractory MAC infection in combination with guideline-based antimicrobial therapy (GBT) is a promising addition to the available treatment armamentarium. In a multinational trial, the addition of ALIS to GBT for treatment-refractory MAC lung disease achieved significantly greater culture conversion rates by month 6 than GBT alone, with comparable rates of serious adverse events.38

      Is therapeutic drug monitoring recommended during treatment of MAC pulmonary disease?

      Treatment failure may also be drug-related, including poor drug penetration into the damaged lung tissue or drug-drug interactions leading to suboptimal drug levels. Peak serum concentrations have been found to be below target ranges in approximately 50% of patients using a macrolide and ethambutol. Concurrent use of rifampin decreases the peak serum concentration of macrolides and quinolones, with acceptable target levels seen in only 18% to 57% of cases. Whether this alters patient outcomes is not clear.39-42 Factors identified as contributing to the poor response to therapy include poor compliance, cavitary disease, previous treatment for MAC pulmonary disease, and a history of chronic obstructive lung disease. Studies by Koh and colleagues40 and van Ingen and colleagues41 with pharmacokinetic and pharmacodynamics data showed that, in patients on MAC treatment with both clarithromycin and rifampicin, plasma levels of clarithromycin were lower than the recommended minimal inhibitory concentrations (MIC) against MAC for that drug. The studies also showed that rifampicin lowered clarithromycin concentrations more than did rifabutin, with the AUC/MIC ratio being suboptimal in nearly half the cases. However, low plasma clarithromycin concentrations did not have any correlation with treatment outcomes, as the peak plasma drug concentrations and the peak plasma drug concentration/MIC ratios did not differ between patients with unfavorable treatment outcomes and those with favorable outcomes. This is further compounded by the fact that macrolides achieve higher levels in lung tissue than in plasma, and hence the significance of low plasma levels is unclear; however, it is postulated that achieving higher drug levels could, in fact, lead to better clinical outcomes. Pending specific well-designed, prospective randomized controlled trials, routine therapeutic drug monitoring is not currently recommended, although some referral centers do this as their practice pattern.

      Is surgery an option in this case?

      The overall 5-year mortality for MAC pulmonary disease was approximately 28% in a retrospective analysis, with patients with cavitary disease at increased risk for death at 5 years.42 As such, surgery is an option in selected cases as part of adjunctive therapy along with anti-MAC therapy based on mycobacterial sensitivity. Surgery is used as either a curative approach or a “debulking” measure.11 When present, clearly localized disease, especially in the upper lobe, lends itself best to surgical intervention. Surgical resection of a solitary pulmonary nodule due to MAC, in addition to concomitant medical treatment, is recommended. Surgical intervention should be considered early in the course of the disease because it may provide a cure without prolonged treatment and its associated problems, and this approach may lead to early sputum conversion. Surgery should also be considered in patients with macrolide-resistant or multidrug-resistant MAC infection or in those who cannot tolerate the side effects of therapy, provided that the disease is focal and limited. Patients with poor preoperative lung function have poorer outcomes than those with good lung function, and postoperative complications arising from treatment, especially with a right-sided pneumonectomy, tend to occur more frequently in these patients. Thoracic surgery for NTM pulmonary disease must be considered cautiously, as this is associated with significant morbidity and mortality and is best performed at specialized centers that have expertise and experience in this field.43

      Continue to: Mycobacterium abscessus Complex

       

       

      Mycobacterium abscessus Complex

      Case Patient 2

      A 64-year-old man who is an ex-smoker presents with chronic cough, mild shortness of breath on exertion, low-grade fever, and unintentional weight loss of 10 lb. Physical exam is unremarkable. The patient was diagnosed with immunoglobulin deficiency (low IgM and low IgG4) in 2002, and has been on replacement therapy since then. He also has had multiple episodes of NTM infection, with MAC and M. kansasii infections diagnosed in 2012-2014, which required 18 months of multi-drug antibiotic treatment that resulted in sputum conversion. Pulmonary function testing done on this visit in 2017 shows mild obstructive impairment.

        Chest radiograph and CT scan show bilateral bronchiectasis (Figure 5 and Figure 6).

        PDBR01601001_f5.PNG

        The results of serial sputum microbiology testing performed over the course of 6 months are outlined below:

        • 5/2017 (bronchoalveolar lavage): 2+; M. abscessus
        • 9/2017 × 2: smear (–); group IV RGM
        • 11/2017: smear (–); M. abscessus (> 50 CFU)
        • 12/2017: smear (–); M. abscessus (> 50 CFU)

         

        PDBR01601001_f6.PNG

        What are the clinical considerations in this patient with multiple NTM infections?

        M. abscessus complex was originally described in soft tissue abscesses and skin infections possibly resulting from soil or water contamination. Subspeciation of M. abscessus complex during laboratory testing is critical to facilitate selection of a specific therapeutic approach; treatment decisions are impacted by the presence of an active erm gene and in vitro macrolide sensitivity, which differ between subspecies. The most acceptable classification outlines 3 species in the M. abscessus complex: Mycobacterium abscessus subsp abscessus, Mycobacterium abscessus subsp bolletii (both with an active erm gene responsible for macrolide resistance), and Mycobacterium abscessus subsp massiliense (with an inactive erm gene and therefore susceptible to macrolides).44

        RGM typically manifest in skin, soft tissue, and bone, and can cause soft tissue, surgical wound, and catheter-related infections. Although the role of RGM as pulmonary pathogens is unclear, underlying diseases associated with RGM include previously treated mycobacterial disease, coexistent pulmonary diseases with or without MAC, cystic fibrosis, malignancies, and gastroesophageal disorders. M. abscessus is the third most commonly identified respiratory NTM and accounts for the majority (80%) of RGM respiratory isolates. Other NTM reported to cause both lung disease and skin, bone, and joint infections include Mycobacterium simiae, Mycobacterium xenopi, and Mycobacterium malmoense. Ocular granulomatous diseases, such as chorioretinitis and keratitis, have been reported with both RGM and Runyon group III SGM, such as MAC or M. szulgai, following trauma or refractive surgery. These can mimic fungal, herpetic, or amebic keratitis. The pulmonary syndromes associated with multiple culture positivity are seen in elderly women with bronchiectasis or cavitary lung disease and/or associated with gastrointestinal symptoms of acid reflux, with or without achalasia and concomitant lipoid interstitial pneumonia.45

        Generally, pulmonary disease progresses slowly, but lung disease attributed to RGM can result in respiratory failure. Thus, RGM should be recognized as a possible cause of chronic mycobacterial lung disease, especially in immunocompromised patients, and respiratory isolates should be assessed carefully. Identification and drug susceptibility testing are essential before initiation of treatment for RGM.

        What is the approach to management of M. abscessus pulmonary disease in a patient without cystic fibrosis?

        The management of M. abscessus pulmonary infection as a subset of RGM requires a considered step-wise approach. The criteria for diagnosis and threshold for starting treatment are the same as those used in the management of MAC pulmonary disease,11 but the treatment of M. abscessus pulmonary infection is more complex and has lower rates of success and cure. Also, antibiotic treatment presents challenges related to rapid identification of the causative organism, nomenclature, resistance patterns, and tolerance of treatment and side effects. If a source such as catheter, access port, or any surgical site is identified, prompt removal and clearance of the infected site are strongly advised

        In the absence of any controlled clinical trials, treatment of RGM is based on in vitro susceptibility testing and expert opinion. As in MAC pulmonary disease, macrolides are the mainstay of treatment, with an induction phase of intravenous antibiotics. Treatment may include a combination of injectable aminoglycosides, imipenem, or cefoxitin and oral drugs such as a macrolide (eg, clarithromycin, azithromycin), doxycycline, fluoroquinolones, trimethoprim/sulfamethoxazole, or linezolid. While antibiotic treatment of M. abscessus pulmonary disease is based on in vitro sensitivity pattern to a greater degree than is treatment of MAC pulmonary disease, this approach has significant practical limitations and hence variable applicability. The final choice of antibiotics is best based on the extended susceptibility results, if available. The presence of an active erm gene on a prolonged growth specimen in M. abscessus subsp abscessus and M. abscessus subsp bolletii precludes the use of a macrolide. In such cases, amikacin, especially in an intravenous form, is the mainstay of treatment based on MIC. Recently, there has been a resurgence in interest in the use of clofazimine in combination with amikacin when treatment is not successful in patients with M. abscessus subsp abscessus or M. bolletii with an active erm gene.45,46 When localized abscess formation is noted, surgery may be the best option, with emphasis on removal of implants and catheters if implicated in RGM infection.

        Attention must also be given to confounding pulmonary and associated comorbidities. This includes management of bronchiectasis with appropriately aggressive airway clearance techniques; anti-reflux measures for prevention of micro-aspiration; and management of other comorbid pulmonary conditions, such as chronic obstructive pulmonary disease, pulmonary fibrosis, and sarcoidosis, if applicable. These interventions play a critical role in clearing the M. abscessus infection, preventing progression of disease, and reducing morbidity. The role of immunomodulatory therapy needs to be considered on a regular, ongoing basis. Identification of genetic factors and correction of immune deficiencies may help in managing the infection.

        Case Patient 2 Conclusion

        The treatment regimen adopted in this case includes a 3-month course of daily intravenous amikacin and imipenem with oral azithromycin, followed by a continuation phase of azithromycin with clofazimine and linezolid. Airway clearance techniques such as Vest/Acapella/CPT are intensified and monthly intravenous immunoglobulin therapy is continued. The patient responds to treatment, with resolution of his clinical symptoms and reduction in the colony count of M. abscessus in the sputum.

        Summary

        NTM are ubiquitous in the environment, and NTM infection has variable manifestations, especially in patients with no recognizable immune impairments. Underlying comorbid conditions with bronchiectasis complicate its management. Treatment strategies must be individualized based on degree of involvement, associated comorbidities, immune deficiencies, goals of therapy, outcome-based risk-benefit ratio assessment, and patient engagement and expectations. In diffuse pulmonary disease, drug treatment remains difficult due to poor match of in vitro and in vivo culture sensitivity, side effects of medications, and high failure rates. When a localized resectable foci of infection is identified, especially in RGM disease, surgical treatment may be the best approach in selected patients, but it must be performed in centers with expertise and experience in this field. 

        References

        1. Johnson MM, Odell JA. Nontuberculous mycobacterial pulmonary infections. J Thorac Dis. 2014;6:210-220.

        2. Falkinham JO III. Environmental sources of NTM. Clin Chest Med. 2015;36:35-41.

        3. Falkinham JO III, Current epidemiological trends in NTM. Curr Environ Health Rep. 2016;3:161-167.

        4. Honda JR, Knight V, Chan ED. Pathogenesis and risk factors for nontuberculous mycobacterial lung disease. Clin Chest Med. 2015;36:1-11.

        5. Marras TK, Mirsaeidi M, Chou E, et al. Health care utilization and expenditures following diagnosis of nontuberculous mycobacterial lung disease in the United States. Manag Care Spec Pharm. 2018;24:964-974.

        6. Prevots DR, Shaw PA, Strickland D, et al. Nontuberculous mycobacterial lung disease prevalence at four integrated healthcare delivery systems. Am J Respir Crit Care Med. 2010;182:970-976.

        7. Winthrop KL, McNelley E, Kendall B, et al. Pulmonary nontuberculous mycobacterial disease prevalence and clinical features: an emerging public health disease. Am J Respir Crit Care Med. 2010;182:977-982.

        8. Adjemian, Olivier KN, Seitz AE, J et al. Prevalence of nontuberculous mycobacterial lung disease in US Medicare beneficiaries. Am J Respir Crit Care Med. 2012;185;881-886.

        9. Ringshausen FC, Apel RM, Bange FC, et al. Burden and trends of hospitalizations associated with pulmonary nontuberculous mycobacterial infections in Germany, 2005-2011. BMC Infect Dis. 2013;13:231.

        10. Aliyu G, El-Kamary SS, Abimiku A, et al. Prevalence of non-tuberculous mycobacterial infections among tuberculosis suspects in Nigeria. PLoS One. 2013;8:e63170.

        11. Griffith DE, Aksamit T, Brown-Elliott, et al; American Thoracic Society; Infectious Diseases Society of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367-415.

        12. Wallace RJ Jr, Zhang Y, Brown BA, et al. Polyclonal Mycobacterium avium complex infections in patients with nodular bronchiectasis. Am J Respir Crit Care Med. 1998;158:1235-1244.

        13. Gordin FM, Horsburgh CR Jr. Mycobacterium avium complex. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Elsevier; 2015.

        14. Chitty S, Ali J. Mycobacterium avium complex pulmonary disease in immune competent patients. South Med J. 2005;98:646-52.

        15. Ramirez J, Mason C, Ali J, Lopez FA. MAC pulmonary disease: management options in HIV-negative patients. J La State Med Soc. 2008;160:248-254.

        16. Iseman MD, Buschman DL, Ackerson LM. Pectus excavatum and scoliosis. Thoracic anomalies associated with pulmonary disease caused by Mycobacterium avium complex. Am Rev Respir Dis. 1991;144:914-916.

        17. Kim RD, Greenburg DE, Ehrmantraut ME, et al. Pulmonary nontuberculous mycobacterial disease: prospective study of a distinct preexisting syndrome. Am J Respir Crit Care Med. 2008;178:1066-1074.

        18. Ziedalski TM, Kao PN, Henig NR, et al. Prospective analysis of cystic fibrosis transmembrane regulator mutations in adults with bronchiectasis or pulmonary nontuberculous mycobacterial infection. Chest. 2006;130:995-1002.

        19. Koh WJ, Lee KS, Kwon OJ, et al. Bilateral bronchiectasis and bronchiolitis at thin-section CT: diagnostic implications in nontuberculous mycobacterial pulmonary infection. Radiology. 2005;235:282-288.

        20. Swensen SJ, Hartman TE, Williams DE. Computed tomographic diagnosis of Mycobacterium avium-intracellulare complex in patients with bronchiectasis. Chest. 1994;105:49-52.

        21. Huang JH, Kao PN, Adi V, Ruoss SJ. Mycobacterium avium intracellulare pulmonary infection in HIV-negative patients without preexisting lung disease: diagnostic and management limitations. Chest. 1999;115:1033-1040.

        22. Cappelluti E, Fraire AE, Schaefer OP. A case of “hot tub lung” due to Mycobacterium avium complex in an immunocompetent host. Arch Intern Med. 2003;163:845-848.

        23. Nightingale SD, Byrd LT, Southern PM, et al. Incidence of Mycobacterium avium-intracellulare complex bacteremia in human immunodeficiency virus-positive patients. J Infect Dis. 1992;165:1082-1085.

        24. Horsburgh CR Jr, Selik RM. The epidemiology of disseminated tuberculous mycobacterial infection in the acquired immunodeficiency syndrome (AIDS). Am Rev Respir Dis. 1989;139:4-7.

        25. Chin DP, Hopewell PC, Yajko DM, et al. Mycobacterium avium complex in the respiratory or gastrointestinal tract and the risk of M. avium complex bacteremia in patients with human immunodeficiency virus infection. J Infect Dis. 1994;169:289-295.

        26. Khan K, Wang J, Marras TK. Nontuberculous mycobacterial sensitization in the United States: national trends over three decades. Am J Respir Crit Care Med. 2007;176:306-313.

        27. Lillo M, Orengo S, Cernoch P, Harris RL. Pulmonary and disseminated infection due to Mycobacterium kansasii: a decade of experience. Rev Infect Dis. 1990;12:760-767.

        28. Andersen P, Munk ME, Pollock JM, Doherty TM. Specific immune-based diagnosis of tuberculosis. Lancet. 2000;356:1099-1104.

        29. Arend SM, van Meijgaarden KE, de Boer K, et al. Tuberculin skin testing and in vitro T cell responses to ESAT-6 and culture filtrate protein 10 after infection with Mycobacterium marinum or M. kansasii. J Infect Dis. 2002;186:1797-1807.

        30. James D, Chalmers JD, Goeminne P, et al. The Bronchiectasis Severity Index: an international derivation and validation study. Am J Respir Crit Care Med. 2014;189:576-585.

        31. Heifets L. MIC as a quantitative measurement of the susceptibility of Mycobacterium avium strains to seven antituberculosis drugs. Antimicrob Agents Chemother. 1988;32:1131-1136.

        32. Horsburgh CR Jr, Mason UG 3rd, Heifits LB, et al. Response to therapy of pulmonary Mycobacterium avium intracellulare infection correlates with results of in vitro susceptibility testing. Am Rev Respir Dis. 1987;135:418-421.

        33. Rubin BK, Henke MO. Immunomodulatory activity and effectiveness of macrolides in chronic airway disease. Chest. 2004;125(2 Suppl):70S-78S.

        34. Wallace RJ Jr, Brown BA, Griffith DE, et al. Clarithromycin regimens for pulmonary Mycobacterium avium complex. The first 50 patients. Am J Respir Crit Care Med. 1996;153:1766-1772.

        35. Griffith DE, Brown-Elliott BA, Langsjoen B, et al. Clinical and molecular analysis of macrolide resistance in Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;174:928-934.

        36. Lam PK, Griffith DE, Aksamit TR, et al. Factors related to response to intermittent treatment of Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;173:1283-1289.

        37. Falkinham J III. Challenges of NTM drug development. Front Microbiol. 2018;9:1613.

        38. Griffith DE, Eagle G, Thomson R, et al. Amikacin liposome inhalation suspension for treatment-refractory lung disease caused by Mycobacterium avium complex (CONVERT). A prospective, open-label, randomized study. Am J Respir Crit Care Med. 2018;198:1559-1569.

        39. Schluger NW. Treatment of pulmonary Mycobacterium avium complex infections: do drug levels matter? Am J Respir Crit Care Med. 2012;186:710-711.

        40. Van Ingen J, Egelund EF, Levin A, et al. The pharmacokinetics and pharmacodynamics of pulmonary Mycobacterium avium complex disease treatment. Am J Respir Crit Care Med. 2012;186:559-565.

        41. Koh WJ, Jeong BH, Jeon K, et al. Therapeutic drug monitoring in the treatment of Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2012;186:797-802.

        42. Ito Y, Hirai T, Maekawa K, et al. Predictors of 5-year mortality in pulmonary MAC disease. Int J Tuberc Lung Dis. 2012;16:408-414.

        43. Yuji S, Yutsuki N, Keiichiso T, et al. Surgery for Mycobacterium avium lung disease in the clarithromycin era. Eur J Cardiothor Surg. 2002;21:314-318.

        44. Tortoli E, Kohl TA, Brown-Elliott BA, et al. Emended description of Mycobacterium abscessus, Mycobacterium abscessus subsp. abscessus and Mycobacterium abscessus subsp. bolletii and designation of Mycobacterium abscessus subsp. massiliense comb. Int J Syst Evol Microbiol. 2016; 66:4471-4479.

        45. Griffith DE, Girard WM, Wallace RJ Jr. Clinical features of pulmonary disease caused by rapidly growing mycobacteria. An analysis of 154 patients. Am Rev Respir Dis. 1993;147:1271-1278.

        46. Koh WJ, Jeong BH, Kim SY, et al. Mycobacterial characteristics and treatment outcomes in Mycobacterium abscessus lung disease. Clin Infect Dis. 2017;64:309-316.

        References

        1. Johnson MM, Odell JA. Nontuberculous mycobacterial pulmonary infections. J Thorac Dis. 2014;6:210-220.

        2. Falkinham JO III. Environmental sources of NTM. Clin Chest Med. 2015;36:35-41.

        3. Falkinham JO III, Current epidemiological trends in NTM. Curr Environ Health Rep. 2016;3:161-167.

        4. Honda JR, Knight V, Chan ED. Pathogenesis and risk factors for nontuberculous mycobacterial lung disease. Clin Chest Med. 2015;36:1-11.

        5. Marras TK, Mirsaeidi M, Chou E, et al. Health care utilization and expenditures following diagnosis of nontuberculous mycobacterial lung disease in the United States. Manag Care Spec Pharm. 2018;24:964-974.

        6. Prevots DR, Shaw PA, Strickland D, et al. Nontuberculous mycobacterial lung disease prevalence at four integrated healthcare delivery systems. Am J Respir Crit Care Med. 2010;182:970-976.

        7. Winthrop KL, McNelley E, Kendall B, et al. Pulmonary nontuberculous mycobacterial disease prevalence and clinical features: an emerging public health disease. Am J Respir Crit Care Med. 2010;182:977-982.

        8. Adjemian, Olivier KN, Seitz AE, J et al. Prevalence of nontuberculous mycobacterial lung disease in US Medicare beneficiaries. Am J Respir Crit Care Med. 2012;185;881-886.

        9. Ringshausen FC, Apel RM, Bange FC, et al. Burden and trends of hospitalizations associated with pulmonary nontuberculous mycobacterial infections in Germany, 2005-2011. BMC Infect Dis. 2013;13:231.

        10. Aliyu G, El-Kamary SS, Abimiku A, et al. Prevalence of non-tuberculous mycobacterial infections among tuberculosis suspects in Nigeria. PLoS One. 2013;8:e63170.

        11. Griffith DE, Aksamit T, Brown-Elliott, et al; American Thoracic Society; Infectious Diseases Society of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175:367-415.

        12. Wallace RJ Jr, Zhang Y, Brown BA, et al. Polyclonal Mycobacterium avium complex infections in patients with nodular bronchiectasis. Am J Respir Crit Care Med. 1998;158:1235-1244.

        13. Gordin FM, Horsburgh CR Jr. Mycobacterium avium complex. In: Mandell GL, Bennett JE, Dolin R, eds. Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Elsevier; 2015.

        14. Chitty S, Ali J. Mycobacterium avium complex pulmonary disease in immune competent patients. South Med J. 2005;98:646-52.

        15. Ramirez J, Mason C, Ali J, Lopez FA. MAC pulmonary disease: management options in HIV-negative patients. J La State Med Soc. 2008;160:248-254.

        16. Iseman MD, Buschman DL, Ackerson LM. Pectus excavatum and scoliosis. Thoracic anomalies associated with pulmonary disease caused by Mycobacterium avium complex. Am Rev Respir Dis. 1991;144:914-916.

        17. Kim RD, Greenburg DE, Ehrmantraut ME, et al. Pulmonary nontuberculous mycobacterial disease: prospective study of a distinct preexisting syndrome. Am J Respir Crit Care Med. 2008;178:1066-1074.

        18. Ziedalski TM, Kao PN, Henig NR, et al. Prospective analysis of cystic fibrosis transmembrane regulator mutations in adults with bronchiectasis or pulmonary nontuberculous mycobacterial infection. Chest. 2006;130:995-1002.

        19. Koh WJ, Lee KS, Kwon OJ, et al. Bilateral bronchiectasis and bronchiolitis at thin-section CT: diagnostic implications in nontuberculous mycobacterial pulmonary infection. Radiology. 2005;235:282-288.

        20. Swensen SJ, Hartman TE, Williams DE. Computed tomographic diagnosis of Mycobacterium avium-intracellulare complex in patients with bronchiectasis. Chest. 1994;105:49-52.

        21. Huang JH, Kao PN, Adi V, Ruoss SJ. Mycobacterium avium intracellulare pulmonary infection in HIV-negative patients without preexisting lung disease: diagnostic and management limitations. Chest. 1999;115:1033-1040.

        22. Cappelluti E, Fraire AE, Schaefer OP. A case of “hot tub lung” due to Mycobacterium avium complex in an immunocompetent host. Arch Intern Med. 2003;163:845-848.

        23. Nightingale SD, Byrd LT, Southern PM, et al. Incidence of Mycobacterium avium-intracellulare complex bacteremia in human immunodeficiency virus-positive patients. J Infect Dis. 1992;165:1082-1085.

        24. Horsburgh CR Jr, Selik RM. The epidemiology of disseminated tuberculous mycobacterial infection in the acquired immunodeficiency syndrome (AIDS). Am Rev Respir Dis. 1989;139:4-7.

        25. Chin DP, Hopewell PC, Yajko DM, et al. Mycobacterium avium complex in the respiratory or gastrointestinal tract and the risk of M. avium complex bacteremia in patients with human immunodeficiency virus infection. J Infect Dis. 1994;169:289-295.

        26. Khan K, Wang J, Marras TK. Nontuberculous mycobacterial sensitization in the United States: national trends over three decades. Am J Respir Crit Care Med. 2007;176:306-313.

        27. Lillo M, Orengo S, Cernoch P, Harris RL. Pulmonary and disseminated infection due to Mycobacterium kansasii: a decade of experience. Rev Infect Dis. 1990;12:760-767.

        28. Andersen P, Munk ME, Pollock JM, Doherty TM. Specific immune-based diagnosis of tuberculosis. Lancet. 2000;356:1099-1104.

        29. Arend SM, van Meijgaarden KE, de Boer K, et al. Tuberculin skin testing and in vitro T cell responses to ESAT-6 and culture filtrate protein 10 after infection with Mycobacterium marinum or M. kansasii. J Infect Dis. 2002;186:1797-1807.

        30. James D, Chalmers JD, Goeminne P, et al. The Bronchiectasis Severity Index: an international derivation and validation study. Am J Respir Crit Care Med. 2014;189:576-585.

        31. Heifets L. MIC as a quantitative measurement of the susceptibility of Mycobacterium avium strains to seven antituberculosis drugs. Antimicrob Agents Chemother. 1988;32:1131-1136.

        32. Horsburgh CR Jr, Mason UG 3rd, Heifits LB, et al. Response to therapy of pulmonary Mycobacterium avium intracellulare infection correlates with results of in vitro susceptibility testing. Am Rev Respir Dis. 1987;135:418-421.

        33. Rubin BK, Henke MO. Immunomodulatory activity and effectiveness of macrolides in chronic airway disease. Chest. 2004;125(2 Suppl):70S-78S.

        34. Wallace RJ Jr, Brown BA, Griffith DE, et al. Clarithromycin regimens for pulmonary Mycobacterium avium complex. The first 50 patients. Am J Respir Crit Care Med. 1996;153:1766-1772.

        35. Griffith DE, Brown-Elliott BA, Langsjoen B, et al. Clinical and molecular analysis of macrolide resistance in Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;174:928-934.

        36. Lam PK, Griffith DE, Aksamit TR, et al. Factors related to response to intermittent treatment of Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;173:1283-1289.

        37. Falkinham J III. Challenges of NTM drug development. Front Microbiol. 2018;9:1613.

        38. Griffith DE, Eagle G, Thomson R, et al. Amikacin liposome inhalation suspension for treatment-refractory lung disease caused by Mycobacterium avium complex (CONVERT). A prospective, open-label, randomized study. Am J Respir Crit Care Med. 2018;198:1559-1569.

        39. Schluger NW. Treatment of pulmonary Mycobacterium avium complex infections: do drug levels matter? Am J Respir Crit Care Med. 2012;186:710-711.

        40. Van Ingen J, Egelund EF, Levin A, et al. The pharmacokinetics and pharmacodynamics of pulmonary Mycobacterium avium complex disease treatment. Am J Respir Crit Care Med. 2012;186:559-565.

        41. Koh WJ, Jeong BH, Jeon K, et al. Therapeutic drug monitoring in the treatment of Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2012;186:797-802.

        42. Ito Y, Hirai T, Maekawa K, et al. Predictors of 5-year mortality in pulmonary MAC disease. Int J Tuberc Lung Dis. 2012;16:408-414.

        43. Yuji S, Yutsuki N, Keiichiso T, et al. Surgery for Mycobacterium avium lung disease in the clarithromycin era. Eur J Cardiothor Surg. 2002;21:314-318.

        44. Tortoli E, Kohl TA, Brown-Elliott BA, et al. Emended description of Mycobacterium abscessus, Mycobacterium abscessus subsp. abscessus and Mycobacterium abscessus subsp. bolletii and designation of Mycobacterium abscessus subsp. massiliense comb. Int J Syst Evol Microbiol. 2016; 66:4471-4479.

        45. Griffith DE, Girard WM, Wallace RJ Jr. Clinical features of pulmonary disease caused by rapidly growing mycobacteria. An analysis of 154 patients. Am Rev Respir Dis. 1993;147:1271-1278.

        46. Koh WJ, Jeong BH, Kim SY, et al. Mycobacterial characteristics and treatment outcomes in Mycobacterium abscessus lung disease. Clin Infect Dis. 2017;64:309-316.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Nontuberculous Mycobacterial Pulmonary Disease
        Display Headline
        Nontuberculous Mycobacterial Pulmonary Disease
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2020 February;16(5)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Thu, 01/30/2020 - 13:00
        Un-Gate On Date
        Thu, 01/30/2020 - 13:00
        Use ProPublica
        CFC Schedule Remove Status
        Thu, 01/30/2020 - 13:00
        Hide sidebar & use full width
        render the right sidebar.

        Portopulmonary Hypertension: Treatment

        Article Type
        Changed
        Wed, 12/11/2019 - 11:37
        Display Headline
        Portopulmonary Hypertension: Treatment

        Portopulmonary hypertension (POPH) is a form of group 1 pulmonary arterial hypertension. When treating patients with POPH, baseline assessment is necessary so that response to therapy can be measured as the change from baseline. Patients should undergo echocardiography and right heart catheterization, and their exercise capacity and NYHA functional class should be determined. Patients with POPH should be considered for treatment if they are NYHA functional class II or above and/or their mean pulmonary artery pressure (MPAP) is greater than 35 mm Hg in transplant candidates. The goal in the treatment and management of POPH is to improve pulmonary hemodynamics by reducing the obstruction to pulmonary arterial flow and to preserve right ventricular function (Table). This article, the second in a 2-part review of POPH in patients with liver disease, reviews the role of medical therapy and liver transplantation in treatment. Evaluation and diagnosis of POPH are discussed in a separate article.

        pdbr01612002_t1.png

        Medical Therapy

        Prostanoids

        Although prostacyclin and prostaglandin analogs entered routine clinical practice for POPH in the 1990s, reports of investigational use date back to the 1980s. Prostanoids are potent vasodilators with antiplatelet aggregation and antiproliferative properties. Prostacyclin synthase is reduced in patients with PAH, resulting in decreased concentration of prostacyclin with vasoconstriction and proliferative changes in the pulmonary vasculature.1

        Epoprostenol

        Epoprostenol is also known as synthetic prostaglandin I2 or prostacyclin. It was the first therapy approved for the treatment of PAH in 1995 by the US Food and Drug Administration (FDA) as a continuous intravenous infusion.2,3 It also inhibits platelet aggregation and may help modulate pulmonary vascular remodeling.4,5 Epoprostenol is derived from the metabolism of arachidonic acid and is a potent pulmonary and systemic vasodilator. One study reported an immediate 11.8% decrease in MPAP, 24% decrease in pulmonary vascular resistance (PVR) and 28% drop in systemic vascular resistance (SVR) during an epoprostenol infusion.6 The authors reported that epoprostenol was a more potent vasodilator than nitric oxide and may have a role in predicting the reversibility of POPH. In a case series of 33 patients with secondary pulmonary hypertension (including 7 patients with POPH) treated with continuous intravenous prostacyclin for approximately 1 year, exercise tolerance, NYHA functional class, and pulmonary hemodynamics improved in each patient compared to baseline.7 Krowka et al studied 14 patients with moderate to severe POPH treated with intravenous epoprostenol.8 No significant side effects were noted and treatment resulted in significant improvements in PVR, MPAP, and cardiac output. In 2007, Fix et al published a large retrospective cohort of patients with moderate to severe POPH.9 Nineteen patients treated with epoprostenol were compared to 17 patients with no treatment. After a median treatment period of 15.4 months, the epoprostenol group showed significant improvement in MPAP, PVR and cardiac output, but survival did not differ between the 2 groups.

        Epoprostenol has often been considered a bridge to transplant in patients with POPH. Sussman et al described 8 consecutive patients with POPH who were treated with intravenous epoprostenol (2 to 8 ng/kg/min dose).10 Liver transplant was considered in 7 of the 8 patients when MPAP decreased to less than 35 mm Hg. Six patients were eventually listed for liver transplant, but 2 died waiting on the list. Long-term outcomes in the group of transplanted recipients were excellent. They remained alive and well at least 9 to 18 months post-transplant, and half did not require long-term vasodilator therapy post-orthotopic liver transplant. Similarly, Ashfaq et al published their data on 16 patients with moderate-to-severe POPH who were treated with vasodilator therapy.11 MPAP decreased to acceptable levels in 75% of the treated patients, and 11 went on to liver transplantation. Rates of 1- and 5-year survival in the transplanted patients were 91% and 67% respectively. None of the patients who failed vasodilator therapy survived.

        Epoprostenol has a short half-life (3 to 5 minutes) and requires continuous infusion through central access via an infusion pump. Aseptic technique must be maintained to avoid blood stream infections. Pump failure or loss of vascular access can result in rebound pulmonary vasoconstriction that can be life-threatening and requires immediate attention. Side effects associated with epoprostenol include flushing, headache, nausea/vomiting, bradycardia, chest pain, jaw pain, diarrhea, and musculoskeletal pain.

        Patients on epoprostenol should be monitored for prostanoid overdose. In the case of patients with chronic liver disease, epoprostenol increases systemic vasodilation in patients with already low systemic vascular tone. As a result, cardiac output may increase to the point of high cardiac output failure. MPAP will remain elevated secondary to high cardiac output rather than high PVR. In these patients, right heart catheterization will show an elevated MPAP in the setting of normal to low PVR/transpulmonary gradient (TPG) values. Lowering the epoprostenol dose will successfully reduce both cardiac output and MPAP.

        Treprostinil

        Treprostinil is a prostacyclin analog that is available in intravenous, inhalational, and subcutaneous form, although subcutaneous dosing may be limited by pain. Sakai et al published a small case series of 3 patients with PAH and end-stage liver disease treated with intravenous treprostinil.12 Pulmonary hemodynamics improved in all patients, and 2 patients went on to an uneventful liver transplantation. More than 10 years later, data were published on 255 patients with PAH on therapy with bosentan or sildenafil randomized to additional inhaled treprostinil.13 Treprostinil proved to be safe and well tolerated, with improvement in quality of life measures but no improvement in other secondary endpoints.

         

         

        Iloprost

        Inhaled iloprost is another prostacyclin that has a short therapeutic half-life of 20 to 30 minutes and requires frequent administration (6 to 9 times daily). In study in which patients with severe POPH were treated for up to 3 years with inhaled iloprost,14 survival rates at 1, 2, and 3 years were 77%, 62%, and 46%, respectively. A second study published in 2010 was designed to assess the acute effects of inhaled iloprost on pulmonary hemodynamics and evaluate the clinical outcome after 12 months of treatment.15 Iloprost was found to rapidly reduce pulmonary arterial pressure and PVR. In the long-term evaluation, inhaled iloprost increased the 6-minute walk distance (6MWD) and functional class, but no change was noted in the systolic pulmonary artery pressure. The authors concluded that iloprost might provide symptomatic improvement and improvement in exercise capacity.

        Selexipag

        Selexipag is an oral selective IP prostacyclin receptor agonist that is structurally distinct from other prostacyclins.16 In a phase 3 randomized double blind clinical trial, PAH patients treated with selexipag had lower composite of death or complication of PAH to the end of the study period.17 This effect was consistent across all dose ranges, but POPH patients were excluded from this study. Safety and efficacy of selexipag has not been evaluated in POPH patients.

        Endothelin Receptor Antagonists

        Endothelin receptor antagonists block the production of endothelin-1 (ET-1), a potent vasoconstrictor and smooth muscle mitogen that may contribute to the development of PAH. Three different receptors have been described: endothelin A, endothelin B, and endothelin B2. Elevated ET-1 levels have been reported in patients with chronic liver disease and may originate from hepatosplanchnic circulation.18

        Bosentan

        Bosentan is an oral, nonspecific, ET-1A and ET-1B receptor antagonist. Initial use of bosentan in patients with POPH was limited because of concern for hepatotoxicity. Approximately 10% of patients on bosentan were reported to have mild hepatic side effects in the form of elevated aminotransferases, but severe injury has been reported.19 One of the first clinical experiences of bosentan in patients with POPH was published in 2005. Hoeper et al followed 11 patients with Child A cirrhosis and severe POPH.20 All patients included were in NYHA functional class III or IV and were treated with bosentan for over 1 year. Exercise capacity and symptoms improved in all treated patients. The medication was tolerated well and there was no evidence of drug-induced liver injury. A single case report showed the effectiveness of bosentan in a 43-year-old man with alcohol-related liver disease (Child-Pugh A) and right ventricular enlargement and dysfunction secondary to POPH.21 Pulmonary arterial pressure decreased, exercise capacity increased, and improvement was maintained over 2 years.

        In a group of 31 patients with Child A or B cirrhosis and severe POPH, bosentan had significantly better effects than inhaled iloprost on exercise capacity, hemodynamics, and survival.14 One, 2, and 3-year survival rates in the bosentan group were 94%, 89%, and 89% (compared to 77%, 62%, and 46% in the iloprost group). Both drugs were considered safe with no reported hepatotoxicity. In 2013, Savale et al published data on 34 patients with POPH, Child-Pugh A and/or B who were treated with bosentan for a median of 43 months.22 The authors reported significant improvements in hemodynamics, NYHA functional class, and 6WMD. Event-free survival rates at 1, 2, and 3 years were 82%, 63%, and 47%, respectively.

         

         

        Ambrisentan

        Ambrisentan is a highly selective ET-1A receptor antagonist with once daily dosing and a lower risk of hepatotoxicity compared to bosentan. Fourteen patients with moderate to severe POPH treated with ambrisentan in 4 German hospitals were retrospectively analyzed.23 Median follow-up was 16 months, and the study demonstrated significant improvement in exercise capacity and clinical symptoms without significant change in liver function tests. Cartin-Ceba et al published their experience of 13 patients with moderate to severe POPH treated with ambrisentan monotherapy.24 Patients were followed for a median of 613 days and on treatment for a median time of 390 days. Significant improvements were shown in pulmonary arterial pressure and PVR without adverse effect on hepatic function. Over 270 patients with PAH (6% with POPH) received ambrisentan from March 2009 through June 2013 at a large United Kingdom portal hypertension referral center.25 Discontinuation due to side effects was higher than previously reported. Discontinuation due to abnormal transaminases was uncommon.

        Macitentan

        Macitentan is a dual endothelin-receptor antagonist developed by modifying the structure of bosentan to increase efficacy and safety. The SERAPHIN trial compared oral macitentan to placebo in 250 patients with moderate to severe PAH, some of whom were also on a stable dose of oral or inhaled therapy for PAH.26 Over a 2-year period, patients treated with macitentan were less likely to have progression of their disease or die on therapy (38% and 31% versus 46%), regardless of if they were receiving additional oral therapy and more likely to have improvement of their exercise capacity and WHO functional class. Nasopharyngitis and significant anemia were more common in the macitentan group, but there was no difference in the rate of liver function test abnormalities compared to placebo. Trials with macitentan are currently ongoing in patients with POPH.

        Phosphodiesterase-5 Inhibitors

        Cyclic guanosine monophosphate (cGMP) is the mediator of nitric oxide–induced vasodilation. Phosphodiesterase-5 (PDE-5) inhibitors prolong the vasodilatory effects of cyclic guanosine monophosphate by preventing its hydrolysis, thereby reducing the pulmonary arterial pressure.

        Sildenafil

        Sildenafil is the most widely accepted PDE-5 inhibitor for POPH. Fourteen patients with moderate to severe POPH were treated with sildenafil (50 mg 3 times per day) in an observational study published by Reichenberger et al in 2006.27 Eight patients were newly started on sildenafil, whereas sildenafil was added to inhaled prostanoids in the remaining 6x patients. Sildenafil significantly decreased 66MWD, MPAP, PVR, and cardiac index alone or in combination with inhaled prostanoids.

        Sildenafil has also been used as a bridge to transplant in liver transplant candidates with POPH. Ten patients with POPH treated with sildenafil monotherapy were followed for a 21±16 months.28 Patients improved symptomatically and increased their 6MWD at 1 year by 30 meters or more. Three patients became transplant eligible and another 3 patients were stable, without progression of their liver disease or POPH. Four patients were not considered transplant candidates, 2 because of refractory POPH and 2 for other comorbidities. The authors concluded that sildenafil monotherapy could stabilize or improve pulmonary hemodynamics in patients with POPH and eventually lead to liver transplantation. Gough et al took a similar look at 9 patients with POPH treated with sildenafil.29 All patients had initial and follow-up right heart catheterizations within a period of 3 years. Mean PVR improved in all patients, decreasing from 575 to 375 dynes/s/cm–5. MPAP decreased to ≤ 35 mmHg in 4 patients, 1 of whom went on to receive a liver transplant. Overall sildenafil improved pulmonary hemodynamics in this small cohort of POPH patients.

         

         

        Tadalafil

        Tadalafil is another oral PDE-5 inhibitor but with a longer half-life than sildenafil. Unlike sildenafil, which requires 3 times daily dosing, tadalafil requires once daily administration. A few case reports have demonstrated tadalafil’s effectiveness for POPH in combination with other medical therapy (eg, sildenafil, ambrisentan).30,31

        Guanylate Cyclase Stimulator

        Riociguat

        Riociguat is a first-in-class activator of soluble form of guanylate cyclase that increases levels of cyclic GMP. Two randomized clinical trials, PATENT, a study in PAH patients, and CHEST, a study in patients with chronic thromboembolic pulmonary hypertension showed improvement in 6MWD at 12 weeks (PATENT) or 16 weeks (CHEST), with improvement in secondary endpoints such as PVR, N-terminal pro b-type natriuretic peptide and WHO functional class.32,33 Riociguat may have potential advantages in patients with POPH given that it has a favorable liver safety profile. A subgroup analysis of patients enrolled in the PATENT study showed that 13 had POPH and 11 were randomized to receive riociguat 2.5 mg 3 times daily dose and 2 received placebo.34 Riociguat was well tolerated and improved 6MWD that was maintained over 2 years in the open label extension.

        Medications to Avoid

        Nonselective beta-blockers are commonly recommended in patients with portal hypertension to help prevent variceal hemorrhage. However, in patients with POPH, beta-blockers have been shown to decrease exercise capacity and worsen pulmonary hemodynamics. A study of 10 patients with moderate to severe POPH who were receiving beta-blockers for variceal bleeding prophylaxis showed that 6MWD improved in almost all of the patients, cardiac output increased by 28%, and PVR decreased by 19% when beta-blockers were discontinued.35 The authors concluded that the use of beta-blockers should be avoided in this patient population.

        Calcium channel blockers should not be used in patients with POPH because they can cause significant hypotension due to systemic vasodilatation and decreased right ventricular filling. Patients with portal hypertension and chronic liver disease commonly have low systemic vascular resistance and are particularly susceptible to the deleterious effects of calcium channel blockers.

        Transplantation

        Liver transplantation is a potential cure for POPH and its role in POPH has evolved over the past 2 decades. In 1997, Ramsay et al published their review of 1205 consecutive liver transplants at Baylor University Medical Center (BUMC) in Texas.36 The incidence of POPH in this group was 8.5%, with the majority of patients having mild POPH. Liver transplant outcomes were not affected by mild and moderate pulmonary hypertension. However, patients with severe POPH (n = 7, systolic pulmonary artery pressure > 60 mm Hg) had a mortality rate of 42% at 9 months post-transplantation and 71% at 36 months post-transplant. The surviving patients continued to deteriorate with progressive right heart failure and no improvement in POPH.

         

         

        To understand the effect of liver transplantation on POPH, one must understand the hemodynamic changes that occur with POPH and during liver transplant. The right ventricle is able to manage the same volume as the left ventricle under normal circumstances, but is unable to pump against a significant pressure gradient.37 In the setting of POPH, right ventricular hypertrophy occurs and RV output remains stable for some time. With time, pulmonary artery pressure increases secondary to pulmonary arteriolar vasoconstriction, intimal thickening, and progressive occlusion of the pulmonary vascular bed. Right ventricular failure may occur as a result. Cardiac output increases significantly at the time of reperfusion during liver transplant (up to 3-fold in 15 minutes),38 and in the setting of a noncompliant vascular bed, the patient is at risk for right heart failure. This is the likely explanation to such high perioperative mortality rates in patients with uncontrolled POPH. Failure to decrease MPAP to less than 50 mm Hg is considered a complete contraindication to liver transplant at most institutions. Many transplant centers will list patients for liver transplant if MPAP can be decreased to less than 35 mm Hg and PVR < 400 dynes/s/cm–5. These parameters are thought to represent an adequate right ventricular reserve and a compliant pulmonary vascular bed.37 However, even with good pressure control, the anesthesiology and critical care teams must be prepared to deal with acute right heart failure peri-operatively. Intraoperative transesophageal echocardiography has been recommended to closely follow right ventricular function.38 Inhaled or intravenous dilators are the most effective agents in the event of a pulmonary hypertensive crisis.

        Review of Outcomes

        A retrospective review evaluated 43 patients with untreated POPH who underwent attempted liver transplantation.39 Data were collected from 18 peer-reviewed studies and 7 patients at the authors’ institution. Overall mortality was 35% (15 patients), with almost all of the deaths secondary to cardiac dysfunction. Two deaths occurred intraoperatively and 8 deaths occurred during the transplant hospitalization. The transplant could not be successfully completed in 4 of the patients. MPAP > 50 mm Hg was associated with 100% mortality, whereas patients with MPAP between 35 mm Hg and 50 mm Hg had a 50% mortality. No mortality was noted in patients with MPAP < 35 mm Hg.

        Liver transplantation has been shown to be successful in patients with controlled POPH. Sussman et al published their data on 8 patients with severe POPH in 2006. In this prospective study, all patients were treated with sequential epoprostenol infusions and 7 of the 8 patients experienced a significant reduction in MPAP and PVR. Six patients were listed for liver transplant, 4 of who were transplanted successfully and alive up to 5 years later.

        The Baylor University Medical Center published their data on POPH patients who received liver transplants in 2007.11 POPH was confirmed by right heart catheterization in 30 patients evaluated for liver transplant. Sixteen patients were considered to be suitable candidates for transplant and MPAP was decreased to less than 35 mmHg in 12 patients with vasodilator therapy. Eleven patients eventually underwent liver transplant and 1- and 5-year survival rates were 91% and 67%.

        Compared to medical therapy or liver transplant alone, patients who receive medical therapy followed by liver transplantation have the best survival. The Mayo Clinic retrospectively reviewed 74 POPH patients identified between 1994 and 2007.40 Patients were categorized in 1 of 3 categories: no medical therapy, medical therapy alone for POPH, or medical therapy for POPH followed by liver transplantation. Patients who received no medical therapy for POPH and no liver transplant had the worst outcomes, with a dismal 5-year survival of only 14% with over 50% deceased at 1 year of diagnosis. Five-year survival was 45% in patients who received medical therapy only. Patients who received medical therapy with prostacyclin followed by liver transplantation had the best outcomes, with a 5-year survival of 67% versus 25% in those who were transplanted without prior prostacyclin therapy.

         

         

        We reported the longest follow-up study for patients undergoing liver transplantation with POPH in 2014.41 Seven patients with moderate to severe POPH received a liver transplant at our institution between June 2004 and January 2011. Mean pulmonary artery pressure was reduced to < 35 mm Hg, with appropriate POPH therapy in all of the patients. Both the graft and patient survival rates were 85.7% after a median follow-up of 7.8 years. The 1 patient who did not survive died from complications related to recurrent hepatitis C and cirrhosis, not from POPH-related issues. Four of the remaining 6 patients continue to require oral vasodilator therapy post-transplant, suggesting irreversible remodeling of the pulmonary vasculature. Two patients (4.4 and 8.5 years post-transplant) have no evidence of pulmonary hypertension post-transplant and therefore do not require medical treatment for pulmonary hypertension. We concluded that POPH responsive to vasodilator therapy is an appropriate indication for liver transplant, with excellent long-term survival.

        Hollatz et al published their data on 11 patients with moderate to severe POPH who were successfully treated (mostly with oral sildenafil and subcutaneous treprostinil) as a bridge to liver transplant.42 The mortality rate was 0, with a follow-up duration of 7 to 60 months. Interestingly, 7 of the 11 patients (64%) were off all pulmonary vasodilators post-transplant. Ashfaq et al reported similar results.11 Nine of 11 patients with treated moderate to severe POPH who received liver transplants stopped vasodilator therapy at a median period of 9.2 months post-transplant. Raevens et al described a group of 3 patients with POPH who went on to liver transplant after their pulmonary pressures were decreased with combined oral vasodilator therapy: 1 required continued long-term vasodilator therapy, another was weaned off medications after transplant, and the third patient died during the liver transplant from perioperative complications that induced uncontrolled pulmonary hypertension.43

        Patient Selection

        In 2006, the United Network for Organ Sharing (UNOS) initiated a policy whereby a higher priority for liver transplantation was granted for highly selected patients in the United States.44 UNOS policy 3.6.4.5.6 upgraded POPH patients to a MELD score of 22, with an increase in MELD every 3 months as long as MPAP remained < 35 mm Hg and PVR remained < 400 dynes/s/cm–5. One hundred fifty-five patients were granted MELD exception points for POPH between 2002 and 2010 and went on to receive liver transplants.45 Goldberg et al collected data from the Organ Procurement and Transplantation Network (OPTN) and compared outcomes of patients with approved POPH MELD exception points versus waitlist candidates with no exception points.46 One hundred fifty-five waitlisted patients received POPH MELD exception points, with only 43.1% meeting OPTN exception requirements. One-third did not fulfill hemodynamic criteria consistent with POPH or had missing data, and 80% went on to receive a liver transplant. Waitlist candidates receiving POPH MELD exception points also had increased waitlist mortality and several early post-transplant deaths. The authors felt these data highlighted the need for OPTN/UNOS to revise their policy for POPH MELD exceptions points, revise how points are rewarded, and continue research to help risk stratify these patients to minimize perioperative complications.

        Conclusion

        Several effective medical treatment regimens are available, including prostanoids, endothelin receptor antagonists, and PDE-5 inhibitors. Liver transplantation is a potential cure but is only recommended if MPAP can be decreased to ≤ 35 mmHg. Long-term follow-up studies have shown these patients do well several years post-transplant but may continue to require oral therapy for their POPH.

        References

        1. Tuder RM, Cool CD, Geraci MW, et al. Prostacyclin synthase expression is decreased in lungs from patients with severe pulmonary hypertension. Am J Respir Crit Care Med. 1999;159:1925-1932.

        2. Chin K, Rubin L. Pulmonary arterial hypertension. Am Coll Cardiol. 2008;51:1527-1538.

        3. Doran A, Harris S, Goetz B. Advances in prostanoid infusion therapy for pulmonary arterial hypertension. J Infus Nurs. 2008;31:336-345.

        4. Chin KM, Channick RN, De Lemos JA, ET AL. Hemodynamics and epoprostenol use are associated with thrombocytopenia in pulmonary arterial hypertension. Chest. 2009;135:130-136.

        5. Hoshikawa Y, Voelkel NF, Gesell TL, et al. Prostacyclin receptor-dependent modulation of pulmonary vascular remodeling. Am J Respir Crit Care Med. 2001;164:314-318.

        6. Ricci GL, Melgosa MT, Burgos F, et al. Assessment of acute pulmonary vascular reactivity in portopulmonary hypertension. Liver Transplant. 2007;13:1506-1514.

        7. McLaughlin V V, Genthner DE, Panella MM, et al. Compassionate use of continuous prostacyclin in the management of secondary pulmonary hypertension: a case series. Ann Intern Med. 1999;130:740-743.

        8. Krowka MJ, Frantz RP, McGoon MD, et al. Improvement in pulmonary hemodynamics during intravenous epoprostenol (prostacyclin): A study of 15 patients with moderate to severe portopulmonary hypertension. Hepatology. 1999;30:641-648.

        9. Fix OK, Bass NM, De Morco T, Merriman RB. Long-term follow-up of portopulmonary hypertension: Effect of treatment with epoprostenol. Liver Transplant. 2007;13:875-885.

        10. Sussman N, Kaza V, Barshes N, et al. Successful liver transplantation following medical management of portopulmonary hypertension: a single-center series. Am J Transplant. 2006;6:2177-2182.

        11. Ashfaq M, Chinnakotla S, Rogers L, et al. The impact of treatment of portopulmonary hypertension on survival following liver transplantation. Am J Transplant. 2007;7:1258-1264.

        12. Sakai T, Planinsic RM, Mathier MA, et al. initial experience using continuous intravenous treprostinil to manage pulmonary arterial hypertension in patients with end-stage liver disease. Transpl Int. 2009;22:554-561.

        13. McLaughlin VV, Benza RL, Rubin LJ, et al. Addition of inhaled treprostinil to oral therapy for pulmonary arterial hypertension: A randomized controlled clinical trial. J Am Coll Cardiol. 2010;55:1915-1922.

        14. Hoeper MM, Seyfarth HJ, Hoeffken G, et al. Experience with inhaled iloprost and bosentan in portopulmonary hypertension. Eur Respir J. 2007;30:1096-1102.

        15. Melgosa MT, Ricci GL, Garcia-Pagan JC et al. Acute and long-term effects of inhaled iloprost in portopulmonary hypertension. Liver Transplant. 2010;16:348-356.

        16. Simonneau G, Torbicki A, Hoeper MM, et al. Selexipag: an oral, selective prostacyclin receptor agonist for the treatment of pulmonary arterial hypertension. Eur Respir J. 2012;40:874-880

        17. Sitbon O, Channick R, Chin, KM, et al. Selexipag for the treatment of pulmonary arterial hypertension. N Engl J Med. 2015;373:2522-2533.

        18. Moller S, Gulberg V, Henriksen JH, Gerbes AL. Endothelin-1 and endothelin-3 in cirrhosis: Relations to systemic and splanchnic haemodynamics. J Hepatol. 1995;23:135-144.

        19. Eriksson C, Gustavsson A, Kronvall T, Tysk C. Hepatotoxicity by bosentan in a patient with portopulmonary hypertension : a case-report and review of the literature. J Gastrointestin Liver Dis. 2011;20:77-80.

        20. Hoeper MM, Halank M, Marx C, et al. Bosentan therapy for portopulmonary hypertension. Eur Respir J. 2005;25:502-508.

        21. Stähler G, Von Hunnius P. Successful treatment of portopulmonary hypertension with bosentan: Case report.: Eur J Clin Investig. 2006;36:62-66.

        22. Savale L, Magnier R, Le Pavec J, et al. Efficacy, safety and pharmacokinetics of bosentan in portopulmonary hypertension. Eur. 2013;41:96-103.

        23. Halank M, Knudsen L, Seyfarth H, et al. Ambrisentan improves exercise capacity and symptoms in patients with portopulmonary hypertension. Z Gastroenterol. 2011;49:1258-1262.

        24. Cartin-Ceba R, Swanson K, Iyer V, et al. Safety and efficacy of ambrisentan for the treatment of portopulmonary hypertension. Chest. 2011;139:109-114.

        25. Condliffe R, Elliot C, Hurdman J, et al. Ambrisentan therapy in pulmonary hypertension: clinical use and tolerability in a referral centre. Ther Adv Respir Dis. 2014;8:71-77.

        26. Pulido T, Adzerikho I, Channick RN, et al. Macitentan and morbidity and mortality in pulmonary arterial hypertension. N Engl J Med. 2013;369:809-818.

        27. Reichenberger F, Voswinckel R, Steveling E, et al. Sildenafil treatment for portopulmonary hypertension. Eur Respir J. 2006;28:563-567.

        28. Hemnes AR RI. Sildenafil monotherapy in portopulmonary hypertension can facilitate liver transplantation. Liver Transplant. 2009;15:15-19.

        29. Gough WR. Sildenafil therapy is associated with improved hemodynamics in liver transplantation candidates with pulmonary arterial hypertension. Liver Transplant. 2009;15:30-36.

        30. Yamashita Y. Hemodynamic effects of ambrisentan-tadalafil combination therapy on progressive portopulmonary hypertension. World J Hepatol. 2014;6:825.

        31. Bremer HC, Kreisel W, Roecker K, et al. Phosphodiesterase 5 inhibitors lower both portal and pulmonary pressure in portopulmonary hypertension: a case report. J Med Case Rep. 2007;1:46.

        32. Ghofrani HA, Galie N, Grimminger F, et al. Riociguat for the treatment of pulmonary arterial hypertension. N Engl J Med. 2013:369;330-340.

        33. Ghofrani HA, Galie N, Grimminger F, et al. Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N Engl J Med. 2013:369;319-329

        34. Cartin-Ceba R, Halank M, Ghofrani HA, et al. Riociguat treatment for portopulmonary hypertension: a subgroup analysis from the PATENT-1/-2 studies. Pulm Circ. 2018: 8:2045894018769305.

        35. Provencher S, Herve P, Jais X, et al. Deleterious effects of beta-blockers on exercise capacity and hemodynamics in patients with portopulmonary hypertension. 2006. Gastroenterology. 2006;130:120-126.

        36. Ramsay M a, Simpson BR, Nguyen T, et al. Severe pulmonary hypertension in liver transplant candidates. Liver Transpl Surg. 1997;3:494-500.

        37. Safdar Z, Bartolome S, Sussman N. Portopulmonary hypertension : an update. Liver Tranpl. 2012;18:881-891.

        38. Ramsay M. Portopulmonary hypertension and right heart failure in patients with cirrhosis. Curr Opin Anaesthesiol. 2010;23:145-150.

        39. Krowka MJ, Plevak DJ, Findlay JY, et al. Pulmonary hemodynamics and perioperative cardiopulmonary-related mortality in patients with portopulmonary hypertension undergoing liver transplantation. Liver Transpl. 2000;6:443-450.

        40. Swanson KL, Wiesner RH, Nyberg SL, et al. Survival in portopulmonary hypertension: Mayo Clinic experience categorized by treatment subgroups. Am J Transplant. 2008;8:2445-2453.

        41. Khaderi S, Khan R, Safdar Z, et al. Long-term follow-up of portopulmonary hypertension patients after liver transplantation. Liver Transplant. 2014;20:724-727.

        42. Hollatz TJ, Musat A, Westphal S, et al. Treatment with sildenafil and treprostinil allows successful liver transplantation of patients with moderate to severe portopulmonary hypertension. Liver Transpl. 2012:686-695.

        43. Raevens S, De Pauw M, Reyntjens K, et al. Oral vasodilator therapy in patients with moderate to severe portopulmonary hypertension as a bridge to liver transplantation. Eur J Gastroenterol Hepatol. 2012:1-8.

        44. Krowka M, Fallon M, Mulligan D. Model for end-stage liver disease (MELD) exception for portopulmonary hypertension. Liver Transplant. 2006;12:S114-S116.

        45. Krowka M, Wiesner R, Rosen C. Portopulmonary hypertension outcomes in the era of MELD exception. Liver Transplant. 2012;18:S259.

        46. Goldberg DS, Batra S, Sahay S, et al. MELD Exceptions for portopulmonary hypertension: current policy and future implementation. Am J Transplant. 2014;14:2081-2087.

        Author and Disclosure Information

        Saira Aijaz Khaderi, MD
        Abdominal Transplant & Liver Disease Center, Baylor College of Medicine, Houston, TX

        Zeenat Safdar, MD, MS
        Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston, TX

        Publications
        Topics
        Sections
        Author and Disclosure Information

        Saira Aijaz Khaderi, MD
        Abdominal Transplant & Liver Disease Center, Baylor College of Medicine, Houston, TX

        Zeenat Safdar, MD, MS
        Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston, TX

        Author and Disclosure Information

        Saira Aijaz Khaderi, MD
        Abdominal Transplant & Liver Disease Center, Baylor College of Medicine, Houston, TX

        Zeenat Safdar, MD, MS
        Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston, TX

        Portopulmonary hypertension (POPH) is a form of group 1 pulmonary arterial hypertension. When treating patients with POPH, baseline assessment is necessary so that response to therapy can be measured as the change from baseline. Patients should undergo echocardiography and right heart catheterization, and their exercise capacity and NYHA functional class should be determined. Patients with POPH should be considered for treatment if they are NYHA functional class II or above and/or their mean pulmonary artery pressure (MPAP) is greater than 35 mm Hg in transplant candidates. The goal in the treatment and management of POPH is to improve pulmonary hemodynamics by reducing the obstruction to pulmonary arterial flow and to preserve right ventricular function (Table). This article, the second in a 2-part review of POPH in patients with liver disease, reviews the role of medical therapy and liver transplantation in treatment. Evaluation and diagnosis of POPH are discussed in a separate article.

        pdbr01612002_t1.png

        Medical Therapy

        Prostanoids

        Although prostacyclin and prostaglandin analogs entered routine clinical practice for POPH in the 1990s, reports of investigational use date back to the 1980s. Prostanoids are potent vasodilators with antiplatelet aggregation and antiproliferative properties. Prostacyclin synthase is reduced in patients with PAH, resulting in decreased concentration of prostacyclin with vasoconstriction and proliferative changes in the pulmonary vasculature.1

        Epoprostenol

        Epoprostenol is also known as synthetic prostaglandin I2 or prostacyclin. It was the first therapy approved for the treatment of PAH in 1995 by the US Food and Drug Administration (FDA) as a continuous intravenous infusion.2,3 It also inhibits platelet aggregation and may help modulate pulmonary vascular remodeling.4,5 Epoprostenol is derived from the metabolism of arachidonic acid and is a potent pulmonary and systemic vasodilator. One study reported an immediate 11.8% decrease in MPAP, 24% decrease in pulmonary vascular resistance (PVR) and 28% drop in systemic vascular resistance (SVR) during an epoprostenol infusion.6 The authors reported that epoprostenol was a more potent vasodilator than nitric oxide and may have a role in predicting the reversibility of POPH. In a case series of 33 patients with secondary pulmonary hypertension (including 7 patients with POPH) treated with continuous intravenous prostacyclin for approximately 1 year, exercise tolerance, NYHA functional class, and pulmonary hemodynamics improved in each patient compared to baseline.7 Krowka et al studied 14 patients with moderate to severe POPH treated with intravenous epoprostenol.8 No significant side effects were noted and treatment resulted in significant improvements in PVR, MPAP, and cardiac output. In 2007, Fix et al published a large retrospective cohort of patients with moderate to severe POPH.9 Nineteen patients treated with epoprostenol were compared to 17 patients with no treatment. After a median treatment period of 15.4 months, the epoprostenol group showed significant improvement in MPAP, PVR and cardiac output, but survival did not differ between the 2 groups.

        Epoprostenol has often been considered a bridge to transplant in patients with POPH. Sussman et al described 8 consecutive patients with POPH who were treated with intravenous epoprostenol (2 to 8 ng/kg/min dose).10 Liver transplant was considered in 7 of the 8 patients when MPAP decreased to less than 35 mm Hg. Six patients were eventually listed for liver transplant, but 2 died waiting on the list. Long-term outcomes in the group of transplanted recipients were excellent. They remained alive and well at least 9 to 18 months post-transplant, and half did not require long-term vasodilator therapy post-orthotopic liver transplant. Similarly, Ashfaq et al published their data on 16 patients with moderate-to-severe POPH who were treated with vasodilator therapy.11 MPAP decreased to acceptable levels in 75% of the treated patients, and 11 went on to liver transplantation. Rates of 1- and 5-year survival in the transplanted patients were 91% and 67% respectively. None of the patients who failed vasodilator therapy survived.

        Epoprostenol has a short half-life (3 to 5 minutes) and requires continuous infusion through central access via an infusion pump. Aseptic technique must be maintained to avoid blood stream infections. Pump failure or loss of vascular access can result in rebound pulmonary vasoconstriction that can be life-threatening and requires immediate attention. Side effects associated with epoprostenol include flushing, headache, nausea/vomiting, bradycardia, chest pain, jaw pain, diarrhea, and musculoskeletal pain.

        Patients on epoprostenol should be monitored for prostanoid overdose. In the case of patients with chronic liver disease, epoprostenol increases systemic vasodilation in patients with already low systemic vascular tone. As a result, cardiac output may increase to the point of high cardiac output failure. MPAP will remain elevated secondary to high cardiac output rather than high PVR. In these patients, right heart catheterization will show an elevated MPAP in the setting of normal to low PVR/transpulmonary gradient (TPG) values. Lowering the epoprostenol dose will successfully reduce both cardiac output and MPAP.

        Treprostinil

        Treprostinil is a prostacyclin analog that is available in intravenous, inhalational, and subcutaneous form, although subcutaneous dosing may be limited by pain. Sakai et al published a small case series of 3 patients with PAH and end-stage liver disease treated with intravenous treprostinil.12 Pulmonary hemodynamics improved in all patients, and 2 patients went on to an uneventful liver transplantation. More than 10 years later, data were published on 255 patients with PAH on therapy with bosentan or sildenafil randomized to additional inhaled treprostinil.13 Treprostinil proved to be safe and well tolerated, with improvement in quality of life measures but no improvement in other secondary endpoints.

         

         

        Iloprost

        Inhaled iloprost is another prostacyclin that has a short therapeutic half-life of 20 to 30 minutes and requires frequent administration (6 to 9 times daily). In study in which patients with severe POPH were treated for up to 3 years with inhaled iloprost,14 survival rates at 1, 2, and 3 years were 77%, 62%, and 46%, respectively. A second study published in 2010 was designed to assess the acute effects of inhaled iloprost on pulmonary hemodynamics and evaluate the clinical outcome after 12 months of treatment.15 Iloprost was found to rapidly reduce pulmonary arterial pressure and PVR. In the long-term evaluation, inhaled iloprost increased the 6-minute walk distance (6MWD) and functional class, but no change was noted in the systolic pulmonary artery pressure. The authors concluded that iloprost might provide symptomatic improvement and improvement in exercise capacity.

        Selexipag

        Selexipag is an oral selective IP prostacyclin receptor agonist that is structurally distinct from other prostacyclins.16 In a phase 3 randomized double blind clinical trial, PAH patients treated with selexipag had lower composite of death or complication of PAH to the end of the study period.17 This effect was consistent across all dose ranges, but POPH patients were excluded from this study. Safety and efficacy of selexipag has not been evaluated in POPH patients.

        Endothelin Receptor Antagonists

        Endothelin receptor antagonists block the production of endothelin-1 (ET-1), a potent vasoconstrictor and smooth muscle mitogen that may contribute to the development of PAH. Three different receptors have been described: endothelin A, endothelin B, and endothelin B2. Elevated ET-1 levels have been reported in patients with chronic liver disease and may originate from hepatosplanchnic circulation.18

        Bosentan

        Bosentan is an oral, nonspecific, ET-1A and ET-1B receptor antagonist. Initial use of bosentan in patients with POPH was limited because of concern for hepatotoxicity. Approximately 10% of patients on bosentan were reported to have mild hepatic side effects in the form of elevated aminotransferases, but severe injury has been reported.19 One of the first clinical experiences of bosentan in patients with POPH was published in 2005. Hoeper et al followed 11 patients with Child A cirrhosis and severe POPH.20 All patients included were in NYHA functional class III or IV and were treated with bosentan for over 1 year. Exercise capacity and symptoms improved in all treated patients. The medication was tolerated well and there was no evidence of drug-induced liver injury. A single case report showed the effectiveness of bosentan in a 43-year-old man with alcohol-related liver disease (Child-Pugh A) and right ventricular enlargement and dysfunction secondary to POPH.21 Pulmonary arterial pressure decreased, exercise capacity increased, and improvement was maintained over 2 years.

        In a group of 31 patients with Child A or B cirrhosis and severe POPH, bosentan had significantly better effects than inhaled iloprost on exercise capacity, hemodynamics, and survival.14 One, 2, and 3-year survival rates in the bosentan group were 94%, 89%, and 89% (compared to 77%, 62%, and 46% in the iloprost group). Both drugs were considered safe with no reported hepatotoxicity. In 2013, Savale et al published data on 34 patients with POPH, Child-Pugh A and/or B who were treated with bosentan for a median of 43 months.22 The authors reported significant improvements in hemodynamics, NYHA functional class, and 6WMD. Event-free survival rates at 1, 2, and 3 years were 82%, 63%, and 47%, respectively.

         

         

        Ambrisentan

        Ambrisentan is a highly selective ET-1A receptor antagonist with once daily dosing and a lower risk of hepatotoxicity compared to bosentan. Fourteen patients with moderate to severe POPH treated with ambrisentan in 4 German hospitals were retrospectively analyzed.23 Median follow-up was 16 months, and the study demonstrated significant improvement in exercise capacity and clinical symptoms without significant change in liver function tests. Cartin-Ceba et al published their experience of 13 patients with moderate to severe POPH treated with ambrisentan monotherapy.24 Patients were followed for a median of 613 days and on treatment for a median time of 390 days. Significant improvements were shown in pulmonary arterial pressure and PVR without adverse effect on hepatic function. Over 270 patients with PAH (6% with POPH) received ambrisentan from March 2009 through June 2013 at a large United Kingdom portal hypertension referral center.25 Discontinuation due to side effects was higher than previously reported. Discontinuation due to abnormal transaminases was uncommon.

        Macitentan

        Macitentan is a dual endothelin-receptor antagonist developed by modifying the structure of bosentan to increase efficacy and safety. The SERAPHIN trial compared oral macitentan to placebo in 250 patients with moderate to severe PAH, some of whom were also on a stable dose of oral or inhaled therapy for PAH.26 Over a 2-year period, patients treated with macitentan were less likely to have progression of their disease or die on therapy (38% and 31% versus 46%), regardless of if they were receiving additional oral therapy and more likely to have improvement of their exercise capacity and WHO functional class. Nasopharyngitis and significant anemia were more common in the macitentan group, but there was no difference in the rate of liver function test abnormalities compared to placebo. Trials with macitentan are currently ongoing in patients with POPH.

        Phosphodiesterase-5 Inhibitors

        Cyclic guanosine monophosphate (cGMP) is the mediator of nitric oxide–induced vasodilation. Phosphodiesterase-5 (PDE-5) inhibitors prolong the vasodilatory effects of cyclic guanosine monophosphate by preventing its hydrolysis, thereby reducing the pulmonary arterial pressure.

        Sildenafil

        Sildenafil is the most widely accepted PDE-5 inhibitor for POPH. Fourteen patients with moderate to severe POPH were treated with sildenafil (50 mg 3 times per day) in an observational study published by Reichenberger et al in 2006.27 Eight patients were newly started on sildenafil, whereas sildenafil was added to inhaled prostanoids in the remaining 6x patients. Sildenafil significantly decreased 66MWD, MPAP, PVR, and cardiac index alone or in combination with inhaled prostanoids.

        Sildenafil has also been used as a bridge to transplant in liver transplant candidates with POPH. Ten patients with POPH treated with sildenafil monotherapy were followed for a 21±16 months.28 Patients improved symptomatically and increased their 6MWD at 1 year by 30 meters or more. Three patients became transplant eligible and another 3 patients were stable, without progression of their liver disease or POPH. Four patients were not considered transplant candidates, 2 because of refractory POPH and 2 for other comorbidities. The authors concluded that sildenafil monotherapy could stabilize or improve pulmonary hemodynamics in patients with POPH and eventually lead to liver transplantation. Gough et al took a similar look at 9 patients with POPH treated with sildenafil.29 All patients had initial and follow-up right heart catheterizations within a period of 3 years. Mean PVR improved in all patients, decreasing from 575 to 375 dynes/s/cm–5. MPAP decreased to ≤ 35 mmHg in 4 patients, 1 of whom went on to receive a liver transplant. Overall sildenafil improved pulmonary hemodynamics in this small cohort of POPH patients.

         

         

        Tadalafil

        Tadalafil is another oral PDE-5 inhibitor but with a longer half-life than sildenafil. Unlike sildenafil, which requires 3 times daily dosing, tadalafil requires once daily administration. A few case reports have demonstrated tadalafil’s effectiveness for POPH in combination with other medical therapy (eg, sildenafil, ambrisentan).30,31

        Guanylate Cyclase Stimulator

        Riociguat

        Riociguat is a first-in-class activator of soluble form of guanylate cyclase that increases levels of cyclic GMP. Two randomized clinical trials, PATENT, a study in PAH patients, and CHEST, a study in patients with chronic thromboembolic pulmonary hypertension showed improvement in 6MWD at 12 weeks (PATENT) or 16 weeks (CHEST), with improvement in secondary endpoints such as PVR, N-terminal pro b-type natriuretic peptide and WHO functional class.32,33 Riociguat may have potential advantages in patients with POPH given that it has a favorable liver safety profile. A subgroup analysis of patients enrolled in the PATENT study showed that 13 had POPH and 11 were randomized to receive riociguat 2.5 mg 3 times daily dose and 2 received placebo.34 Riociguat was well tolerated and improved 6MWD that was maintained over 2 years in the open label extension.

        Medications to Avoid

        Nonselective beta-blockers are commonly recommended in patients with portal hypertension to help prevent variceal hemorrhage. However, in patients with POPH, beta-blockers have been shown to decrease exercise capacity and worsen pulmonary hemodynamics. A study of 10 patients with moderate to severe POPH who were receiving beta-blockers for variceal bleeding prophylaxis showed that 6MWD improved in almost all of the patients, cardiac output increased by 28%, and PVR decreased by 19% when beta-blockers were discontinued.35 The authors concluded that the use of beta-blockers should be avoided in this patient population.

        Calcium channel blockers should not be used in patients with POPH because they can cause significant hypotension due to systemic vasodilatation and decreased right ventricular filling. Patients with portal hypertension and chronic liver disease commonly have low systemic vascular resistance and are particularly susceptible to the deleterious effects of calcium channel blockers.

        Transplantation

        Liver transplantation is a potential cure for POPH and its role in POPH has evolved over the past 2 decades. In 1997, Ramsay et al published their review of 1205 consecutive liver transplants at Baylor University Medical Center (BUMC) in Texas.36 The incidence of POPH in this group was 8.5%, with the majority of patients having mild POPH. Liver transplant outcomes were not affected by mild and moderate pulmonary hypertension. However, patients with severe POPH (n = 7, systolic pulmonary artery pressure > 60 mm Hg) had a mortality rate of 42% at 9 months post-transplantation and 71% at 36 months post-transplant. The surviving patients continued to deteriorate with progressive right heart failure and no improvement in POPH.

         

         

        To understand the effect of liver transplantation on POPH, one must understand the hemodynamic changes that occur with POPH and during liver transplant. The right ventricle is able to manage the same volume as the left ventricle under normal circumstances, but is unable to pump against a significant pressure gradient.37 In the setting of POPH, right ventricular hypertrophy occurs and RV output remains stable for some time. With time, pulmonary artery pressure increases secondary to pulmonary arteriolar vasoconstriction, intimal thickening, and progressive occlusion of the pulmonary vascular bed. Right ventricular failure may occur as a result. Cardiac output increases significantly at the time of reperfusion during liver transplant (up to 3-fold in 15 minutes),38 and in the setting of a noncompliant vascular bed, the patient is at risk for right heart failure. This is the likely explanation to such high perioperative mortality rates in patients with uncontrolled POPH. Failure to decrease MPAP to less than 50 mm Hg is considered a complete contraindication to liver transplant at most institutions. Many transplant centers will list patients for liver transplant if MPAP can be decreased to less than 35 mm Hg and PVR < 400 dynes/s/cm–5. These parameters are thought to represent an adequate right ventricular reserve and a compliant pulmonary vascular bed.37 However, even with good pressure control, the anesthesiology and critical care teams must be prepared to deal with acute right heart failure peri-operatively. Intraoperative transesophageal echocardiography has been recommended to closely follow right ventricular function.38 Inhaled or intravenous dilators are the most effective agents in the event of a pulmonary hypertensive crisis.

        Review of Outcomes

        A retrospective review evaluated 43 patients with untreated POPH who underwent attempted liver transplantation.39 Data were collected from 18 peer-reviewed studies and 7 patients at the authors’ institution. Overall mortality was 35% (15 patients), with almost all of the deaths secondary to cardiac dysfunction. Two deaths occurred intraoperatively and 8 deaths occurred during the transplant hospitalization. The transplant could not be successfully completed in 4 of the patients. MPAP > 50 mm Hg was associated with 100% mortality, whereas patients with MPAP between 35 mm Hg and 50 mm Hg had a 50% mortality. No mortality was noted in patients with MPAP < 35 mm Hg.

        Liver transplantation has been shown to be successful in patients with controlled POPH. Sussman et al published their data on 8 patients with severe POPH in 2006. In this prospective study, all patients were treated with sequential epoprostenol infusions and 7 of the 8 patients experienced a significant reduction in MPAP and PVR. Six patients were listed for liver transplant, 4 of who were transplanted successfully and alive up to 5 years later.

        The Baylor University Medical Center published their data on POPH patients who received liver transplants in 2007.11 POPH was confirmed by right heart catheterization in 30 patients evaluated for liver transplant. Sixteen patients were considered to be suitable candidates for transplant and MPAP was decreased to less than 35 mmHg in 12 patients with vasodilator therapy. Eleven patients eventually underwent liver transplant and 1- and 5-year survival rates were 91% and 67%.

        Compared to medical therapy or liver transplant alone, patients who receive medical therapy followed by liver transplantation have the best survival. The Mayo Clinic retrospectively reviewed 74 POPH patients identified between 1994 and 2007.40 Patients were categorized in 1 of 3 categories: no medical therapy, medical therapy alone for POPH, or medical therapy for POPH followed by liver transplantation. Patients who received no medical therapy for POPH and no liver transplant had the worst outcomes, with a dismal 5-year survival of only 14% with over 50% deceased at 1 year of diagnosis. Five-year survival was 45% in patients who received medical therapy only. Patients who received medical therapy with prostacyclin followed by liver transplantation had the best outcomes, with a 5-year survival of 67% versus 25% in those who were transplanted without prior prostacyclin therapy.

         

         

        We reported the longest follow-up study for patients undergoing liver transplantation with POPH in 2014.41 Seven patients with moderate to severe POPH received a liver transplant at our institution between June 2004 and January 2011. Mean pulmonary artery pressure was reduced to < 35 mm Hg, with appropriate POPH therapy in all of the patients. Both the graft and patient survival rates were 85.7% after a median follow-up of 7.8 years. The 1 patient who did not survive died from complications related to recurrent hepatitis C and cirrhosis, not from POPH-related issues. Four of the remaining 6 patients continue to require oral vasodilator therapy post-transplant, suggesting irreversible remodeling of the pulmonary vasculature. Two patients (4.4 and 8.5 years post-transplant) have no evidence of pulmonary hypertension post-transplant and therefore do not require medical treatment for pulmonary hypertension. We concluded that POPH responsive to vasodilator therapy is an appropriate indication for liver transplant, with excellent long-term survival.

        Hollatz et al published their data on 11 patients with moderate to severe POPH who were successfully treated (mostly with oral sildenafil and subcutaneous treprostinil) as a bridge to liver transplant.42 The mortality rate was 0, with a follow-up duration of 7 to 60 months. Interestingly, 7 of the 11 patients (64%) were off all pulmonary vasodilators post-transplant. Ashfaq et al reported similar results.11 Nine of 11 patients with treated moderate to severe POPH who received liver transplants stopped vasodilator therapy at a median period of 9.2 months post-transplant. Raevens et al described a group of 3 patients with POPH who went on to liver transplant after their pulmonary pressures were decreased with combined oral vasodilator therapy: 1 required continued long-term vasodilator therapy, another was weaned off medications after transplant, and the third patient died during the liver transplant from perioperative complications that induced uncontrolled pulmonary hypertension.43

        Patient Selection

        In 2006, the United Network for Organ Sharing (UNOS) initiated a policy whereby a higher priority for liver transplantation was granted for highly selected patients in the United States.44 UNOS policy 3.6.4.5.6 upgraded POPH patients to a MELD score of 22, with an increase in MELD every 3 months as long as MPAP remained < 35 mm Hg and PVR remained < 400 dynes/s/cm–5. One hundred fifty-five patients were granted MELD exception points for POPH between 2002 and 2010 and went on to receive liver transplants.45 Goldberg et al collected data from the Organ Procurement and Transplantation Network (OPTN) and compared outcomes of patients with approved POPH MELD exception points versus waitlist candidates with no exception points.46 One hundred fifty-five waitlisted patients received POPH MELD exception points, with only 43.1% meeting OPTN exception requirements. One-third did not fulfill hemodynamic criteria consistent with POPH or had missing data, and 80% went on to receive a liver transplant. Waitlist candidates receiving POPH MELD exception points also had increased waitlist mortality and several early post-transplant deaths. The authors felt these data highlighted the need for OPTN/UNOS to revise their policy for POPH MELD exceptions points, revise how points are rewarded, and continue research to help risk stratify these patients to minimize perioperative complications.

        Conclusion

        Several effective medical treatment regimens are available, including prostanoids, endothelin receptor antagonists, and PDE-5 inhibitors. Liver transplantation is a potential cure but is only recommended if MPAP can be decreased to ≤ 35 mmHg. Long-term follow-up studies have shown these patients do well several years post-transplant but may continue to require oral therapy for their POPH.

        Portopulmonary hypertension (POPH) is a form of group 1 pulmonary arterial hypertension. When treating patients with POPH, baseline assessment is necessary so that response to therapy can be measured as the change from baseline. Patients should undergo echocardiography and right heart catheterization, and their exercise capacity and NYHA functional class should be determined. Patients with POPH should be considered for treatment if they are NYHA functional class II or above and/or their mean pulmonary artery pressure (MPAP) is greater than 35 mm Hg in transplant candidates. The goal in the treatment and management of POPH is to improve pulmonary hemodynamics by reducing the obstruction to pulmonary arterial flow and to preserve right ventricular function (Table). This article, the second in a 2-part review of POPH in patients with liver disease, reviews the role of medical therapy and liver transplantation in treatment. Evaluation and diagnosis of POPH are discussed in a separate article.

        pdbr01612002_t1.png

        Medical Therapy

        Prostanoids

        Although prostacyclin and prostaglandin analogs entered routine clinical practice for POPH in the 1990s, reports of investigational use date back to the 1980s. Prostanoids are potent vasodilators with antiplatelet aggregation and antiproliferative properties. Prostacyclin synthase is reduced in patients with PAH, resulting in decreased concentration of prostacyclin with vasoconstriction and proliferative changes in the pulmonary vasculature.1

        Epoprostenol

        Epoprostenol is also known as synthetic prostaglandin I2 or prostacyclin. It was the first therapy approved for the treatment of PAH in 1995 by the US Food and Drug Administration (FDA) as a continuous intravenous infusion.2,3 It also inhibits platelet aggregation and may help modulate pulmonary vascular remodeling.4,5 Epoprostenol is derived from the metabolism of arachidonic acid and is a potent pulmonary and systemic vasodilator. One study reported an immediate 11.8% decrease in MPAP, 24% decrease in pulmonary vascular resistance (PVR) and 28% drop in systemic vascular resistance (SVR) during an epoprostenol infusion.6 The authors reported that epoprostenol was a more potent vasodilator than nitric oxide and may have a role in predicting the reversibility of POPH. In a case series of 33 patients with secondary pulmonary hypertension (including 7 patients with POPH) treated with continuous intravenous prostacyclin for approximately 1 year, exercise tolerance, NYHA functional class, and pulmonary hemodynamics improved in each patient compared to baseline.7 Krowka et al studied 14 patients with moderate to severe POPH treated with intravenous epoprostenol.8 No significant side effects were noted and treatment resulted in significant improvements in PVR, MPAP, and cardiac output. In 2007, Fix et al published a large retrospective cohort of patients with moderate to severe POPH.9 Nineteen patients treated with epoprostenol were compared to 17 patients with no treatment. After a median treatment period of 15.4 months, the epoprostenol group showed significant improvement in MPAP, PVR and cardiac output, but survival did not differ between the 2 groups.

        Epoprostenol has often been considered a bridge to transplant in patients with POPH. Sussman et al described 8 consecutive patients with POPH who were treated with intravenous epoprostenol (2 to 8 ng/kg/min dose).10 Liver transplant was considered in 7 of the 8 patients when MPAP decreased to less than 35 mm Hg. Six patients were eventually listed for liver transplant, but 2 died waiting on the list. Long-term outcomes in the group of transplanted recipients were excellent. They remained alive and well at least 9 to 18 months post-transplant, and half did not require long-term vasodilator therapy post-orthotopic liver transplant. Similarly, Ashfaq et al published their data on 16 patients with moderate-to-severe POPH who were treated with vasodilator therapy.11 MPAP decreased to acceptable levels in 75% of the treated patients, and 11 went on to liver transplantation. Rates of 1- and 5-year survival in the transplanted patients were 91% and 67% respectively. None of the patients who failed vasodilator therapy survived.

        Epoprostenol has a short half-life (3 to 5 minutes) and requires continuous infusion through central access via an infusion pump. Aseptic technique must be maintained to avoid blood stream infections. Pump failure or loss of vascular access can result in rebound pulmonary vasoconstriction that can be life-threatening and requires immediate attention. Side effects associated with epoprostenol include flushing, headache, nausea/vomiting, bradycardia, chest pain, jaw pain, diarrhea, and musculoskeletal pain.

        Patients on epoprostenol should be monitored for prostanoid overdose. In the case of patients with chronic liver disease, epoprostenol increases systemic vasodilation in patients with already low systemic vascular tone. As a result, cardiac output may increase to the point of high cardiac output failure. MPAP will remain elevated secondary to high cardiac output rather than high PVR. In these patients, right heart catheterization will show an elevated MPAP in the setting of normal to low PVR/transpulmonary gradient (TPG) values. Lowering the epoprostenol dose will successfully reduce both cardiac output and MPAP.

        Treprostinil

        Treprostinil is a prostacyclin analog that is available in intravenous, inhalational, and subcutaneous form, although subcutaneous dosing may be limited by pain. Sakai et al published a small case series of 3 patients with PAH and end-stage liver disease treated with intravenous treprostinil.12 Pulmonary hemodynamics improved in all patients, and 2 patients went on to an uneventful liver transplantation. More than 10 years later, data were published on 255 patients with PAH on therapy with bosentan or sildenafil randomized to additional inhaled treprostinil.13 Treprostinil proved to be safe and well tolerated, with improvement in quality of life measures but no improvement in other secondary endpoints.

         

         

        Iloprost

        Inhaled iloprost is another prostacyclin that has a short therapeutic half-life of 20 to 30 minutes and requires frequent administration (6 to 9 times daily). In study in which patients with severe POPH were treated for up to 3 years with inhaled iloprost,14 survival rates at 1, 2, and 3 years were 77%, 62%, and 46%, respectively. A second study published in 2010 was designed to assess the acute effects of inhaled iloprost on pulmonary hemodynamics and evaluate the clinical outcome after 12 months of treatment.15 Iloprost was found to rapidly reduce pulmonary arterial pressure and PVR. In the long-term evaluation, inhaled iloprost increased the 6-minute walk distance (6MWD) and functional class, but no change was noted in the systolic pulmonary artery pressure. The authors concluded that iloprost might provide symptomatic improvement and improvement in exercise capacity.

        Selexipag

        Selexipag is an oral selective IP prostacyclin receptor agonist that is structurally distinct from other prostacyclins.16 In a phase 3 randomized double blind clinical trial, PAH patients treated with selexipag had lower composite of death or complication of PAH to the end of the study period.17 This effect was consistent across all dose ranges, but POPH patients were excluded from this study. Safety and efficacy of selexipag has not been evaluated in POPH patients.

        Endothelin Receptor Antagonists

        Endothelin receptor antagonists block the production of endothelin-1 (ET-1), a potent vasoconstrictor and smooth muscle mitogen that may contribute to the development of PAH. Three different receptors have been described: endothelin A, endothelin B, and endothelin B2. Elevated ET-1 levels have been reported in patients with chronic liver disease and may originate from hepatosplanchnic circulation.18

        Bosentan

        Bosentan is an oral, nonspecific, ET-1A and ET-1B receptor antagonist. Initial use of bosentan in patients with POPH was limited because of concern for hepatotoxicity. Approximately 10% of patients on bosentan were reported to have mild hepatic side effects in the form of elevated aminotransferases, but severe injury has been reported.19 One of the first clinical experiences of bosentan in patients with POPH was published in 2005. Hoeper et al followed 11 patients with Child A cirrhosis and severe POPH.20 All patients included were in NYHA functional class III or IV and were treated with bosentan for over 1 year. Exercise capacity and symptoms improved in all treated patients. The medication was tolerated well and there was no evidence of drug-induced liver injury. A single case report showed the effectiveness of bosentan in a 43-year-old man with alcohol-related liver disease (Child-Pugh A) and right ventricular enlargement and dysfunction secondary to POPH.21 Pulmonary arterial pressure decreased, exercise capacity increased, and improvement was maintained over 2 years.

        In a group of 31 patients with Child A or B cirrhosis and severe POPH, bosentan had significantly better effects than inhaled iloprost on exercise capacity, hemodynamics, and survival.14 One, 2, and 3-year survival rates in the bosentan group were 94%, 89%, and 89% (compared to 77%, 62%, and 46% in the iloprost group). Both drugs were considered safe with no reported hepatotoxicity. In 2013, Savale et al published data on 34 patients with POPH, Child-Pugh A and/or B who were treated with bosentan for a median of 43 months.22 The authors reported significant improvements in hemodynamics, NYHA functional class, and 6WMD. Event-free survival rates at 1, 2, and 3 years were 82%, 63%, and 47%, respectively.

         

         

        Ambrisentan

        Ambrisentan is a highly selective ET-1A receptor antagonist with once daily dosing and a lower risk of hepatotoxicity compared to bosentan. Fourteen patients with moderate to severe POPH treated with ambrisentan in 4 German hospitals were retrospectively analyzed.23 Median follow-up was 16 months, and the study demonstrated significant improvement in exercise capacity and clinical symptoms without significant change in liver function tests. Cartin-Ceba et al published their experience of 13 patients with moderate to severe POPH treated with ambrisentan monotherapy.24 Patients were followed for a median of 613 days and on treatment for a median time of 390 days. Significant improvements were shown in pulmonary arterial pressure and PVR without adverse effect on hepatic function. Over 270 patients with PAH (6% with POPH) received ambrisentan from March 2009 through June 2013 at a large United Kingdom portal hypertension referral center.25 Discontinuation due to side effects was higher than previously reported. Discontinuation due to abnormal transaminases was uncommon.

        Macitentan

        Macitentan is a dual endothelin-receptor antagonist developed by modifying the structure of bosentan to increase efficacy and safety. The SERAPHIN trial compared oral macitentan to placebo in 250 patients with moderate to severe PAH, some of whom were also on a stable dose of oral or inhaled therapy for PAH.26 Over a 2-year period, patients treated with macitentan were less likely to have progression of their disease or die on therapy (38% and 31% versus 46%), regardless of if they were receiving additional oral therapy and more likely to have improvement of their exercise capacity and WHO functional class. Nasopharyngitis and significant anemia were more common in the macitentan group, but there was no difference in the rate of liver function test abnormalities compared to placebo. Trials with macitentan are currently ongoing in patients with POPH.

        Phosphodiesterase-5 Inhibitors

        Cyclic guanosine monophosphate (cGMP) is the mediator of nitric oxide–induced vasodilation. Phosphodiesterase-5 (PDE-5) inhibitors prolong the vasodilatory effects of cyclic guanosine monophosphate by preventing its hydrolysis, thereby reducing the pulmonary arterial pressure.

        Sildenafil

        Sildenafil is the most widely accepted PDE-5 inhibitor for POPH. Fourteen patients with moderate to severe POPH were treated with sildenafil (50 mg 3 times per day) in an observational study published by Reichenberger et al in 2006.27 Eight patients were newly started on sildenafil, whereas sildenafil was added to inhaled prostanoids in the remaining 6x patients. Sildenafil significantly decreased 66MWD, MPAP, PVR, and cardiac index alone or in combination with inhaled prostanoids.

        Sildenafil has also been used as a bridge to transplant in liver transplant candidates with POPH. Ten patients with POPH treated with sildenafil monotherapy were followed for a 21±16 months.28 Patients improved symptomatically and increased their 6MWD at 1 year by 30 meters or more. Three patients became transplant eligible and another 3 patients were stable, without progression of their liver disease or POPH. Four patients were not considered transplant candidates, 2 because of refractory POPH and 2 for other comorbidities. The authors concluded that sildenafil monotherapy could stabilize or improve pulmonary hemodynamics in patients with POPH and eventually lead to liver transplantation. Gough et al took a similar look at 9 patients with POPH treated with sildenafil.29 All patients had initial and follow-up right heart catheterizations within a period of 3 years. Mean PVR improved in all patients, decreasing from 575 to 375 dynes/s/cm–5. MPAP decreased to ≤ 35 mmHg in 4 patients, 1 of whom went on to receive a liver transplant. Overall sildenafil improved pulmonary hemodynamics in this small cohort of POPH patients.

         

         

        Tadalafil

        Tadalafil is another oral PDE-5 inhibitor but with a longer half-life than sildenafil. Unlike sildenafil, which requires 3 times daily dosing, tadalafil requires once daily administration. A few case reports have demonstrated tadalafil’s effectiveness for POPH in combination with other medical therapy (eg, sildenafil, ambrisentan).30,31

        Guanylate Cyclase Stimulator

        Riociguat

        Riociguat is a first-in-class activator of soluble form of guanylate cyclase that increases levels of cyclic GMP. Two randomized clinical trials, PATENT, a study in PAH patients, and CHEST, a study in patients with chronic thromboembolic pulmonary hypertension showed improvement in 6MWD at 12 weeks (PATENT) or 16 weeks (CHEST), with improvement in secondary endpoints such as PVR, N-terminal pro b-type natriuretic peptide and WHO functional class.32,33 Riociguat may have potential advantages in patients with POPH given that it has a favorable liver safety profile. A subgroup analysis of patients enrolled in the PATENT study showed that 13 had POPH and 11 were randomized to receive riociguat 2.5 mg 3 times daily dose and 2 received placebo.34 Riociguat was well tolerated and improved 6MWD that was maintained over 2 years in the open label extension.

        Medications to Avoid

        Nonselective beta-blockers are commonly recommended in patients with portal hypertension to help prevent variceal hemorrhage. However, in patients with POPH, beta-blockers have been shown to decrease exercise capacity and worsen pulmonary hemodynamics. A study of 10 patients with moderate to severe POPH who were receiving beta-blockers for variceal bleeding prophylaxis showed that 6MWD improved in almost all of the patients, cardiac output increased by 28%, and PVR decreased by 19% when beta-blockers were discontinued.35 The authors concluded that the use of beta-blockers should be avoided in this patient population.

        Calcium channel blockers should not be used in patients with POPH because they can cause significant hypotension due to systemic vasodilatation and decreased right ventricular filling. Patients with portal hypertension and chronic liver disease commonly have low systemic vascular resistance and are particularly susceptible to the deleterious effects of calcium channel blockers.

        Transplantation

        Liver transplantation is a potential cure for POPH and its role in POPH has evolved over the past 2 decades. In 1997, Ramsay et al published their review of 1205 consecutive liver transplants at Baylor University Medical Center (BUMC) in Texas.36 The incidence of POPH in this group was 8.5%, with the majority of patients having mild POPH. Liver transplant outcomes were not affected by mild and moderate pulmonary hypertension. However, patients with severe POPH (n = 7, systolic pulmonary artery pressure > 60 mm Hg) had a mortality rate of 42% at 9 months post-transplantation and 71% at 36 months post-transplant. The surviving patients continued to deteriorate with progressive right heart failure and no improvement in POPH.

         

         

        To understand the effect of liver transplantation on POPH, one must understand the hemodynamic changes that occur with POPH and during liver transplant. The right ventricle is able to manage the same volume as the left ventricle under normal circumstances, but is unable to pump against a significant pressure gradient.37 In the setting of POPH, right ventricular hypertrophy occurs and RV output remains stable for some time. With time, pulmonary artery pressure increases secondary to pulmonary arteriolar vasoconstriction, intimal thickening, and progressive occlusion of the pulmonary vascular bed. Right ventricular failure may occur as a result. Cardiac output increases significantly at the time of reperfusion during liver transplant (up to 3-fold in 15 minutes),38 and in the setting of a noncompliant vascular bed, the patient is at risk for right heart failure. This is the likely explanation to such high perioperative mortality rates in patients with uncontrolled POPH. Failure to decrease MPAP to less than 50 mm Hg is considered a complete contraindication to liver transplant at most institutions. Many transplant centers will list patients for liver transplant if MPAP can be decreased to less than 35 mm Hg and PVR < 400 dynes/s/cm–5. These parameters are thought to represent an adequate right ventricular reserve and a compliant pulmonary vascular bed.37 However, even with good pressure control, the anesthesiology and critical care teams must be prepared to deal with acute right heart failure peri-operatively. Intraoperative transesophageal echocardiography has been recommended to closely follow right ventricular function.38 Inhaled or intravenous dilators are the most effective agents in the event of a pulmonary hypertensive crisis.

        Review of Outcomes

        A retrospective review evaluated 43 patients with untreated POPH who underwent attempted liver transplantation.39 Data were collected from 18 peer-reviewed studies and 7 patients at the authors’ institution. Overall mortality was 35% (15 patients), with almost all of the deaths secondary to cardiac dysfunction. Two deaths occurred intraoperatively and 8 deaths occurred during the transplant hospitalization. The transplant could not be successfully completed in 4 of the patients. MPAP > 50 mm Hg was associated with 100% mortality, whereas patients with MPAP between 35 mm Hg and 50 mm Hg had a 50% mortality. No mortality was noted in patients with MPAP < 35 mm Hg.

        Liver transplantation has been shown to be successful in patients with controlled POPH. Sussman et al published their data on 8 patients with severe POPH in 2006. In this prospective study, all patients were treated with sequential epoprostenol infusions and 7 of the 8 patients experienced a significant reduction in MPAP and PVR. Six patients were listed for liver transplant, 4 of who were transplanted successfully and alive up to 5 years later.

        The Baylor University Medical Center published their data on POPH patients who received liver transplants in 2007.11 POPH was confirmed by right heart catheterization in 30 patients evaluated for liver transplant. Sixteen patients were considered to be suitable candidates for transplant and MPAP was decreased to less than 35 mmHg in 12 patients with vasodilator therapy. Eleven patients eventually underwent liver transplant and 1- and 5-year survival rates were 91% and 67%.

        Compared to medical therapy or liver transplant alone, patients who receive medical therapy followed by liver transplantation have the best survival. The Mayo Clinic retrospectively reviewed 74 POPH patients identified between 1994 and 2007.40 Patients were categorized in 1 of 3 categories: no medical therapy, medical therapy alone for POPH, or medical therapy for POPH followed by liver transplantation. Patients who received no medical therapy for POPH and no liver transplant had the worst outcomes, with a dismal 5-year survival of only 14% with over 50% deceased at 1 year of diagnosis. Five-year survival was 45% in patients who received medical therapy only. Patients who received medical therapy with prostacyclin followed by liver transplantation had the best outcomes, with a 5-year survival of 67% versus 25% in those who were transplanted without prior prostacyclin therapy.

         

         

        We reported the longest follow-up study for patients undergoing liver transplantation with POPH in 2014.41 Seven patients with moderate to severe POPH received a liver transplant at our institution between June 2004 and January 2011. Mean pulmonary artery pressure was reduced to < 35 mm Hg, with appropriate POPH therapy in all of the patients. Both the graft and patient survival rates were 85.7% after a median follow-up of 7.8 years. The 1 patient who did not survive died from complications related to recurrent hepatitis C and cirrhosis, not from POPH-related issues. Four of the remaining 6 patients continue to require oral vasodilator therapy post-transplant, suggesting irreversible remodeling of the pulmonary vasculature. Two patients (4.4 and 8.5 years post-transplant) have no evidence of pulmonary hypertension post-transplant and therefore do not require medical treatment for pulmonary hypertension. We concluded that POPH responsive to vasodilator therapy is an appropriate indication for liver transplant, with excellent long-term survival.

        Hollatz et al published their data on 11 patients with moderate to severe POPH who were successfully treated (mostly with oral sildenafil and subcutaneous treprostinil) as a bridge to liver transplant.42 The mortality rate was 0, with a follow-up duration of 7 to 60 months. Interestingly, 7 of the 11 patients (64%) were off all pulmonary vasodilators post-transplant. Ashfaq et al reported similar results.11 Nine of 11 patients with treated moderate to severe POPH who received liver transplants stopped vasodilator therapy at a median period of 9.2 months post-transplant. Raevens et al described a group of 3 patients with POPH who went on to liver transplant after their pulmonary pressures were decreased with combined oral vasodilator therapy: 1 required continued long-term vasodilator therapy, another was weaned off medications after transplant, and the third patient died during the liver transplant from perioperative complications that induced uncontrolled pulmonary hypertension.43

        Patient Selection

        In 2006, the United Network for Organ Sharing (UNOS) initiated a policy whereby a higher priority for liver transplantation was granted for highly selected patients in the United States.44 UNOS policy 3.6.4.5.6 upgraded POPH patients to a MELD score of 22, with an increase in MELD every 3 months as long as MPAP remained < 35 mm Hg and PVR remained < 400 dynes/s/cm–5. One hundred fifty-five patients were granted MELD exception points for POPH between 2002 and 2010 and went on to receive liver transplants.45 Goldberg et al collected data from the Organ Procurement and Transplantation Network (OPTN) and compared outcomes of patients with approved POPH MELD exception points versus waitlist candidates with no exception points.46 One hundred fifty-five waitlisted patients received POPH MELD exception points, with only 43.1% meeting OPTN exception requirements. One-third did not fulfill hemodynamic criteria consistent with POPH or had missing data, and 80% went on to receive a liver transplant. Waitlist candidates receiving POPH MELD exception points also had increased waitlist mortality and several early post-transplant deaths. The authors felt these data highlighted the need for OPTN/UNOS to revise their policy for POPH MELD exceptions points, revise how points are rewarded, and continue research to help risk stratify these patients to minimize perioperative complications.

        Conclusion

        Several effective medical treatment regimens are available, including prostanoids, endothelin receptor antagonists, and PDE-5 inhibitors. Liver transplantation is a potential cure but is only recommended if MPAP can be decreased to ≤ 35 mmHg. Long-term follow-up studies have shown these patients do well several years post-transplant but may continue to require oral therapy for their POPH.

        References

        1. Tuder RM, Cool CD, Geraci MW, et al. Prostacyclin synthase expression is decreased in lungs from patients with severe pulmonary hypertension. Am J Respir Crit Care Med. 1999;159:1925-1932.

        2. Chin K, Rubin L. Pulmonary arterial hypertension. Am Coll Cardiol. 2008;51:1527-1538.

        3. Doran A, Harris S, Goetz B. Advances in prostanoid infusion therapy for pulmonary arterial hypertension. J Infus Nurs. 2008;31:336-345.

        4. Chin KM, Channick RN, De Lemos JA, ET AL. Hemodynamics and epoprostenol use are associated with thrombocytopenia in pulmonary arterial hypertension. Chest. 2009;135:130-136.

        5. Hoshikawa Y, Voelkel NF, Gesell TL, et al. Prostacyclin receptor-dependent modulation of pulmonary vascular remodeling. Am J Respir Crit Care Med. 2001;164:314-318.

        6. Ricci GL, Melgosa MT, Burgos F, et al. Assessment of acute pulmonary vascular reactivity in portopulmonary hypertension. Liver Transplant. 2007;13:1506-1514.

        7. McLaughlin V V, Genthner DE, Panella MM, et al. Compassionate use of continuous prostacyclin in the management of secondary pulmonary hypertension: a case series. Ann Intern Med. 1999;130:740-743.

        8. Krowka MJ, Frantz RP, McGoon MD, et al. Improvement in pulmonary hemodynamics during intravenous epoprostenol (prostacyclin): A study of 15 patients with moderate to severe portopulmonary hypertension. Hepatology. 1999;30:641-648.

        9. Fix OK, Bass NM, De Morco T, Merriman RB. Long-term follow-up of portopulmonary hypertension: Effect of treatment with epoprostenol. Liver Transplant. 2007;13:875-885.

        10. Sussman N, Kaza V, Barshes N, et al. Successful liver transplantation following medical management of portopulmonary hypertension: a single-center series. Am J Transplant. 2006;6:2177-2182.

        11. Ashfaq M, Chinnakotla S, Rogers L, et al. The impact of treatment of portopulmonary hypertension on survival following liver transplantation. Am J Transplant. 2007;7:1258-1264.

        12. Sakai T, Planinsic RM, Mathier MA, et al. initial experience using continuous intravenous treprostinil to manage pulmonary arterial hypertension in patients with end-stage liver disease. Transpl Int. 2009;22:554-561.

        13. McLaughlin VV, Benza RL, Rubin LJ, et al. Addition of inhaled treprostinil to oral therapy for pulmonary arterial hypertension: A randomized controlled clinical trial. J Am Coll Cardiol. 2010;55:1915-1922.

        14. Hoeper MM, Seyfarth HJ, Hoeffken G, et al. Experience with inhaled iloprost and bosentan in portopulmonary hypertension. Eur Respir J. 2007;30:1096-1102.

        15. Melgosa MT, Ricci GL, Garcia-Pagan JC et al. Acute and long-term effects of inhaled iloprost in portopulmonary hypertension. Liver Transplant. 2010;16:348-356.

        16. Simonneau G, Torbicki A, Hoeper MM, et al. Selexipag: an oral, selective prostacyclin receptor agonist for the treatment of pulmonary arterial hypertension. Eur Respir J. 2012;40:874-880

        17. Sitbon O, Channick R, Chin, KM, et al. Selexipag for the treatment of pulmonary arterial hypertension. N Engl J Med. 2015;373:2522-2533.

        18. Moller S, Gulberg V, Henriksen JH, Gerbes AL. Endothelin-1 and endothelin-3 in cirrhosis: Relations to systemic and splanchnic haemodynamics. J Hepatol. 1995;23:135-144.

        19. Eriksson C, Gustavsson A, Kronvall T, Tysk C. Hepatotoxicity by bosentan in a patient with portopulmonary hypertension : a case-report and review of the literature. J Gastrointestin Liver Dis. 2011;20:77-80.

        20. Hoeper MM, Halank M, Marx C, et al. Bosentan therapy for portopulmonary hypertension. Eur Respir J. 2005;25:502-508.

        21. Stähler G, Von Hunnius P. Successful treatment of portopulmonary hypertension with bosentan: Case report.: Eur J Clin Investig. 2006;36:62-66.

        22. Savale L, Magnier R, Le Pavec J, et al. Efficacy, safety and pharmacokinetics of bosentan in portopulmonary hypertension. Eur. 2013;41:96-103.

        23. Halank M, Knudsen L, Seyfarth H, et al. Ambrisentan improves exercise capacity and symptoms in patients with portopulmonary hypertension. Z Gastroenterol. 2011;49:1258-1262.

        24. Cartin-Ceba R, Swanson K, Iyer V, et al. Safety and efficacy of ambrisentan for the treatment of portopulmonary hypertension. Chest. 2011;139:109-114.

        25. Condliffe R, Elliot C, Hurdman J, et al. Ambrisentan therapy in pulmonary hypertension: clinical use and tolerability in a referral centre. Ther Adv Respir Dis. 2014;8:71-77.

        26. Pulido T, Adzerikho I, Channick RN, et al. Macitentan and morbidity and mortality in pulmonary arterial hypertension. N Engl J Med. 2013;369:809-818.

        27. Reichenberger F, Voswinckel R, Steveling E, et al. Sildenafil treatment for portopulmonary hypertension. Eur Respir J. 2006;28:563-567.

        28. Hemnes AR RI. Sildenafil monotherapy in portopulmonary hypertension can facilitate liver transplantation. Liver Transplant. 2009;15:15-19.

        29. Gough WR. Sildenafil therapy is associated with improved hemodynamics in liver transplantation candidates with pulmonary arterial hypertension. Liver Transplant. 2009;15:30-36.

        30. Yamashita Y. Hemodynamic effects of ambrisentan-tadalafil combination therapy on progressive portopulmonary hypertension. World J Hepatol. 2014;6:825.

        31. Bremer HC, Kreisel W, Roecker K, et al. Phosphodiesterase 5 inhibitors lower both portal and pulmonary pressure in portopulmonary hypertension: a case report. J Med Case Rep. 2007;1:46.

        32. Ghofrani HA, Galie N, Grimminger F, et al. Riociguat for the treatment of pulmonary arterial hypertension. N Engl J Med. 2013:369;330-340.

        33. Ghofrani HA, Galie N, Grimminger F, et al. Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N Engl J Med. 2013:369;319-329

        34. Cartin-Ceba R, Halank M, Ghofrani HA, et al. Riociguat treatment for portopulmonary hypertension: a subgroup analysis from the PATENT-1/-2 studies. Pulm Circ. 2018: 8:2045894018769305.

        35. Provencher S, Herve P, Jais X, et al. Deleterious effects of beta-blockers on exercise capacity and hemodynamics in patients with portopulmonary hypertension. 2006. Gastroenterology. 2006;130:120-126.

        36. Ramsay M a, Simpson BR, Nguyen T, et al. Severe pulmonary hypertension in liver transplant candidates. Liver Transpl Surg. 1997;3:494-500.

        37. Safdar Z, Bartolome S, Sussman N. Portopulmonary hypertension : an update. Liver Tranpl. 2012;18:881-891.

        38. Ramsay M. Portopulmonary hypertension and right heart failure in patients with cirrhosis. Curr Opin Anaesthesiol. 2010;23:145-150.

        39. Krowka MJ, Plevak DJ, Findlay JY, et al. Pulmonary hemodynamics and perioperative cardiopulmonary-related mortality in patients with portopulmonary hypertension undergoing liver transplantation. Liver Transpl. 2000;6:443-450.

        40. Swanson KL, Wiesner RH, Nyberg SL, et al. Survival in portopulmonary hypertension: Mayo Clinic experience categorized by treatment subgroups. Am J Transplant. 2008;8:2445-2453.

        41. Khaderi S, Khan R, Safdar Z, et al. Long-term follow-up of portopulmonary hypertension patients after liver transplantation. Liver Transplant. 2014;20:724-727.

        42. Hollatz TJ, Musat A, Westphal S, et al. Treatment with sildenafil and treprostinil allows successful liver transplantation of patients with moderate to severe portopulmonary hypertension. Liver Transpl. 2012:686-695.

        43. Raevens S, De Pauw M, Reyntjens K, et al. Oral vasodilator therapy in patients with moderate to severe portopulmonary hypertension as a bridge to liver transplantation. Eur J Gastroenterol Hepatol. 2012:1-8.

        44. Krowka M, Fallon M, Mulligan D. Model for end-stage liver disease (MELD) exception for portopulmonary hypertension. Liver Transplant. 2006;12:S114-S116.

        45. Krowka M, Wiesner R, Rosen C. Portopulmonary hypertension outcomes in the era of MELD exception. Liver Transplant. 2012;18:S259.

        46. Goldberg DS, Batra S, Sahay S, et al. MELD Exceptions for portopulmonary hypertension: current policy and future implementation. Am J Transplant. 2014;14:2081-2087.

        References

        1. Tuder RM, Cool CD, Geraci MW, et al. Prostacyclin synthase expression is decreased in lungs from patients with severe pulmonary hypertension. Am J Respir Crit Care Med. 1999;159:1925-1932.

        2. Chin K, Rubin L. Pulmonary arterial hypertension. Am Coll Cardiol. 2008;51:1527-1538.

        3. Doran A, Harris S, Goetz B. Advances in prostanoid infusion therapy for pulmonary arterial hypertension. J Infus Nurs. 2008;31:336-345.

        4. Chin KM, Channick RN, De Lemos JA, ET AL. Hemodynamics and epoprostenol use are associated with thrombocytopenia in pulmonary arterial hypertension. Chest. 2009;135:130-136.

        5. Hoshikawa Y, Voelkel NF, Gesell TL, et al. Prostacyclin receptor-dependent modulation of pulmonary vascular remodeling. Am J Respir Crit Care Med. 2001;164:314-318.

        6. Ricci GL, Melgosa MT, Burgos F, et al. Assessment of acute pulmonary vascular reactivity in portopulmonary hypertension. Liver Transplant. 2007;13:1506-1514.

        7. McLaughlin V V, Genthner DE, Panella MM, et al. Compassionate use of continuous prostacyclin in the management of secondary pulmonary hypertension: a case series. Ann Intern Med. 1999;130:740-743.

        8. Krowka MJ, Frantz RP, McGoon MD, et al. Improvement in pulmonary hemodynamics during intravenous epoprostenol (prostacyclin): A study of 15 patients with moderate to severe portopulmonary hypertension. Hepatology. 1999;30:641-648.

        9. Fix OK, Bass NM, De Morco T, Merriman RB. Long-term follow-up of portopulmonary hypertension: Effect of treatment with epoprostenol. Liver Transplant. 2007;13:875-885.

        10. Sussman N, Kaza V, Barshes N, et al. Successful liver transplantation following medical management of portopulmonary hypertension: a single-center series. Am J Transplant. 2006;6:2177-2182.

        11. Ashfaq M, Chinnakotla S, Rogers L, et al. The impact of treatment of portopulmonary hypertension on survival following liver transplantation. Am J Transplant. 2007;7:1258-1264.

        12. Sakai T, Planinsic RM, Mathier MA, et al. initial experience using continuous intravenous treprostinil to manage pulmonary arterial hypertension in patients with end-stage liver disease. Transpl Int. 2009;22:554-561.

        13. McLaughlin VV, Benza RL, Rubin LJ, et al. Addition of inhaled treprostinil to oral therapy for pulmonary arterial hypertension: A randomized controlled clinical trial. J Am Coll Cardiol. 2010;55:1915-1922.

        14. Hoeper MM, Seyfarth HJ, Hoeffken G, et al. Experience with inhaled iloprost and bosentan in portopulmonary hypertension. Eur Respir J. 2007;30:1096-1102.

        15. Melgosa MT, Ricci GL, Garcia-Pagan JC et al. Acute and long-term effects of inhaled iloprost in portopulmonary hypertension. Liver Transplant. 2010;16:348-356.

        16. Simonneau G, Torbicki A, Hoeper MM, et al. Selexipag: an oral, selective prostacyclin receptor agonist for the treatment of pulmonary arterial hypertension. Eur Respir J. 2012;40:874-880

        17. Sitbon O, Channick R, Chin, KM, et al. Selexipag for the treatment of pulmonary arterial hypertension. N Engl J Med. 2015;373:2522-2533.

        18. Moller S, Gulberg V, Henriksen JH, Gerbes AL. Endothelin-1 and endothelin-3 in cirrhosis: Relations to systemic and splanchnic haemodynamics. J Hepatol. 1995;23:135-144.

        19. Eriksson C, Gustavsson A, Kronvall T, Tysk C. Hepatotoxicity by bosentan in a patient with portopulmonary hypertension : a case-report and review of the literature. J Gastrointestin Liver Dis. 2011;20:77-80.

        20. Hoeper MM, Halank M, Marx C, et al. Bosentan therapy for portopulmonary hypertension. Eur Respir J. 2005;25:502-508.

        21. Stähler G, Von Hunnius P. Successful treatment of portopulmonary hypertension with bosentan: Case report.: Eur J Clin Investig. 2006;36:62-66.

        22. Savale L, Magnier R, Le Pavec J, et al. Efficacy, safety and pharmacokinetics of bosentan in portopulmonary hypertension. Eur. 2013;41:96-103.

        23. Halank M, Knudsen L, Seyfarth H, et al. Ambrisentan improves exercise capacity and symptoms in patients with portopulmonary hypertension. Z Gastroenterol. 2011;49:1258-1262.

        24. Cartin-Ceba R, Swanson K, Iyer V, et al. Safety and efficacy of ambrisentan for the treatment of portopulmonary hypertension. Chest. 2011;139:109-114.

        25. Condliffe R, Elliot C, Hurdman J, et al. Ambrisentan therapy in pulmonary hypertension: clinical use and tolerability in a referral centre. Ther Adv Respir Dis. 2014;8:71-77.

        26. Pulido T, Adzerikho I, Channick RN, et al. Macitentan and morbidity and mortality in pulmonary arterial hypertension. N Engl J Med. 2013;369:809-818.

        27. Reichenberger F, Voswinckel R, Steveling E, et al. Sildenafil treatment for portopulmonary hypertension. Eur Respir J. 2006;28:563-567.

        28. Hemnes AR RI. Sildenafil monotherapy in portopulmonary hypertension can facilitate liver transplantation. Liver Transplant. 2009;15:15-19.

        29. Gough WR. Sildenafil therapy is associated with improved hemodynamics in liver transplantation candidates with pulmonary arterial hypertension. Liver Transplant. 2009;15:30-36.

        30. Yamashita Y. Hemodynamic effects of ambrisentan-tadalafil combination therapy on progressive portopulmonary hypertension. World J Hepatol. 2014;6:825.

        31. Bremer HC, Kreisel W, Roecker K, et al. Phosphodiesterase 5 inhibitors lower both portal and pulmonary pressure in portopulmonary hypertension: a case report. J Med Case Rep. 2007;1:46.

        32. Ghofrani HA, Galie N, Grimminger F, et al. Riociguat for the treatment of pulmonary arterial hypertension. N Engl J Med. 2013:369;330-340.

        33. Ghofrani HA, Galie N, Grimminger F, et al. Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N Engl J Med. 2013:369;319-329

        34. Cartin-Ceba R, Halank M, Ghofrani HA, et al. Riociguat treatment for portopulmonary hypertension: a subgroup analysis from the PATENT-1/-2 studies. Pulm Circ. 2018: 8:2045894018769305.

        35. Provencher S, Herve P, Jais X, et al. Deleterious effects of beta-blockers on exercise capacity and hemodynamics in patients with portopulmonary hypertension. 2006. Gastroenterology. 2006;130:120-126.

        36. Ramsay M a, Simpson BR, Nguyen T, et al. Severe pulmonary hypertension in liver transplant candidates. Liver Transpl Surg. 1997;3:494-500.

        37. Safdar Z, Bartolome S, Sussman N. Portopulmonary hypertension : an update. Liver Tranpl. 2012;18:881-891.

        38. Ramsay M. Portopulmonary hypertension and right heart failure in patients with cirrhosis. Curr Opin Anaesthesiol. 2010;23:145-150.

        39. Krowka MJ, Plevak DJ, Findlay JY, et al. Pulmonary hemodynamics and perioperative cardiopulmonary-related mortality in patients with portopulmonary hypertension undergoing liver transplantation. Liver Transpl. 2000;6:443-450.

        40. Swanson KL, Wiesner RH, Nyberg SL, et al. Survival in portopulmonary hypertension: Mayo Clinic experience categorized by treatment subgroups. Am J Transplant. 2008;8:2445-2453.

        41. Khaderi S, Khan R, Safdar Z, et al. Long-term follow-up of portopulmonary hypertension patients after liver transplantation. Liver Transplant. 2014;20:724-727.

        42. Hollatz TJ, Musat A, Westphal S, et al. Treatment with sildenafil and treprostinil allows successful liver transplantation of patients with moderate to severe portopulmonary hypertension. Liver Transpl. 2012:686-695.

        43. Raevens S, De Pauw M, Reyntjens K, et al. Oral vasodilator therapy in patients with moderate to severe portopulmonary hypertension as a bridge to liver transplantation. Eur J Gastroenterol Hepatol. 2012:1-8.

        44. Krowka M, Fallon M, Mulligan D. Model for end-stage liver disease (MELD) exception for portopulmonary hypertension. Liver Transplant. 2006;12:S114-S116.

        45. Krowka M, Wiesner R, Rosen C. Portopulmonary hypertension outcomes in the era of MELD exception. Liver Transplant. 2012;18:S259.

        46. Goldberg DS, Batra S, Sahay S, et al. MELD Exceptions for portopulmonary hypertension: current policy and future implementation. Am J Transplant. 2014;14:2081-2087.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Portopulmonary Hypertension: Treatment
        Display Headline
        Portopulmonary Hypertension: Treatment
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 December;16(4)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Wed, 12/04/2019 - 11:30
        Un-Gate On Date
        Wed, 12/04/2019 - 11:30
        Use ProPublica
        CFC Schedule Remove Status
        Wed, 12/04/2019 - 11:30
        Hide sidebar & use full width
        render the right sidebar.

        Portopulmonary Hypertension: Evaluation and Diagnosis

        Article Type
        Changed
        Wed, 12/11/2019 - 11:37
        Display Headline
        Portopulmonary Hypertension: Evaluation and Diagnosis

        Pulmonary arterial hypertension (PAH) is a rare disease that is associated with high mortality and is characterized by pulmonary vascular remodeling. Portopulmonary hypertension (POPH) is a form of PAH that occurs in patients with portal hypertension where no alternative cause of PAH can be identified. POPH is documented in approximately 4.5% to 8.5% of liver transplant candidates,1,2 but there is no relationship between the existence or severity of POPH and the severity of liver dysfunction.3 Mantz and Craig described the first case of POPH in a 53-year-old woman with enlarged pulmonary arteries that exhibited forceful pulsations more characteristic of the aorta than a low-pressure pulmonary trunk.4 Autopsy revealed findings of chronic liver disease including a stenotic portal vein, portocaval shunt, and esophageal varices. In both PAH and POPH, pre-capillary pulmonary arteries have characteristic lesions, such as intimal thickening, endothelial proliferation, and thrombotic changes. This 2-part article reviews the diagnosis and treatment of patients with POPH. Here, we review the epidemiology, prognosis, pathogenesis, and diagnosis of POPH; current treatment options for POPH are reviewed in a separate article.

        Definition

        The term POPH was first used by Yoshida et al in 1993 to describe the first successful liver transplant in a patient with POPH, a 39-year-old man with chronic hepatitis.5 The World Health Organization (WHO) classifies POPH as a form of Group 1 PAH.6 The criteria that must be met to make a diagnosis of POPH are shown in the Table 1.7

        pdbr01612001_t1.png

        Moderate POPH is defined as a mean pulmonary artery pressure (MPAP) between 35 mm Hg and < 45 mm Hg, whereas severe POPH is MPAP ≥ 45 mm Hg. Moderate and severe POPH are considered contraindications to liver transplant because of high perioperative and postoperative mortality rates.8 In 2000, the Mayo Clinic retrospectively reviewed 43 patients with POPH who underwent attempted liver transplantation.9 The cardiopulmonary-related mortality rate in patients with a MPAP of 35 to < 50 mm Hg was 50% and 100% for those with MPAP > 50 mm Hg. No mortality was noted in patients with a pre-liver transplant MPAP of < 35 mm Hg and transpulmonary gradient (TPG) < 15 mm Hg.

        Epidemiology

        In 1983, a series of 17,901 autopsied patients showed a primary pulmonary hypertension prevalence of 0.13% and a prevalence of 0.73% in patients with cirrhosis.10 In 1987, Rich et al published data from the National Institutes of Health’s national registry of primary pulmonary hypertension.11 The registry included data from 187 patients from 32 centers. Further analyses by Groves et al concluded that 8.3% of the patients likely had POPH.12 Humbert et al published data on the French pulmonary hypertension registry experience in 2006.13 The French registry included 674 patients from 17 university hospitals; 10.4% of these patients had POPH. The largest prospective study was published by Hadengue et al in 1991.14 In this study, 507 patients hospitalized with portal hypertension but without known pulmonary hypertension underwent cardiac catheterization; 10 patients (2%) had pulmonary hypertension and more than half were clinically asymptomatic. Finally, the Registry to Evaluate Early And Long-term pulmonary arterial hypertension disease management (REVEAL registry) documented a 5.3% frequency of POPH (174 of 3525) in the United States.15

        Prognosis

        Individuals with POPH have worse outcomes compared to other forms of PAH. Median survival prior to the introduction of vasodilator therapy was a dismal 6 months and mean survival was 15 months.16 The cause of death in patients with POPH is equally distributed between right heart failure from POPH and direct complications of chronic liver disease.1 Le Pavec et al retrospectively analyzed all patients referred to the French Referral Center with POPH between 1984 and 2004 (154 patients).1 Approximately 50% of the patients were Child-Turcotte-Pugh class B or C, and 60% were classified as New York Health Association (NYHA) class III or IV. In these patients, 1-, 3-, and 5-year survival rates were 88%, 75%, and 68%, respectively. Major independent prognostic risk factors were presence and severity of cirrhosis and preservation of right ventricular function. Interestingly, NYHA functional class was not related to survival in this study, although it has clearly been identified as a strong prognostic factor in idiopathic PAH.

        Krowka et al evaluated 174 patients with POPH enrolled in the REVEAL Registry,15 a multicenter, observational, US-based study comprised of more than 3500 patients with PAH. Despite having better hemodynamic parameters at diagnosis, patients with POPH had significantly poorer survival and all-cause hospitalization compared with patients with idiopathic PAH (IPAH) or hereditary PAH (HPAH). Two-year survival from enrollment was 67% in POPH versus 85% in those with IPAH/HPAH (P < 0.001). Five-year survival from time of diagnosis was 40% versus 64% (P < 0.001). Additionally, patients with POPH were less likely to be on PAH-specific therapy at enrollment, with only 25% on treatment at the time of entry. These findings were replicated in 2005 when Kawut et al retrospectively compared 13 patients with POPH with 33 patients with IPAH.17 Despite having a higher cardiac index and lower pulmonary vascular resistance than patients with IPAH, patients with POPH had a higher risk of death (hazard ratio, 2.8, P = 0.04), likely reflecting the combination of 2 serious diseases.

        In 2008 the Mayo Clinic published their retrospective analysis of patients with POPH to determine the natural history of POPH.18 Patients were categorized into 3 groups: (1) no medical therapy for POPH and no liver transplant; (2) medical therapy for POPH alone; (3) medical therapy for POPH followed by liver transplant. The study included 74 patients between 1994 through 2007; 19 patients who did not receive treatment for POPH or liver transplant truly represented the natural history of POPH. Their 5-year survival was only 14%, and over half were deceased 1 year after diagnosis. The largest group consisted of patients who received therapy for POPH but no liver transplant. This group did remarkably better than those who received no therapy at all, with a 5-year survival of 45%. However, the patients with the overall best survival were those who received a combination of treatment for POPH followed by liver transplant. Their 5-year survival was 67%. Survival at 5 years was only 25% for the small group of patients who received transplant without PAH therapy. Once again, mortality did not correlate with the severity of hepatic dysfunction or baseline hemodynamic data.

        Pathogenesis

        The pathogenesis of POPH is unclear. Multiple studies have shown that there is minimal, if any, association with pulmonary hypertension and the severity of liver disease or portal hypertension.19,20 Portal hypertension is the result of an increase in intrahepatic resistance and an increase in blood flow into the portal circulation. Collateral vessels develop and blood from the splanchnic circulation is allowed directly into the systemic venous circulation, bypassing the liver. One of the most widely accepted theories is that a humoral substance, that would otherwise be metabolized by the liver, is able to reach the pulmonary circulation through collaterals, resulting in POPH.21 Pelicelli et al evaluated the possible role of endothelin-1, interleukin-6, interleukin 1β, and tumor necrosis factor in the pathogenesis of POPH.22 Plasma concentrations of these cytokines were compared between patients with POPH and patients with cirrhosis but no POPH. Patients with POPH had higher concentrations of endothelin-1 and interleukin-6, suggesting antagonists for these cytokines may have a role in the treatment of POPH. The role of endothelin-1 was further supported by Kamath et al in 200023 when they determined the pulmonary vascular bed is exposed to increased levels of circulating endothelin-1a in the setting of cirrhosis. Endothelin-1 is a potent vasoconstrictor and facilitator of smooth muscle proliferation.

        In addition to collateral circulation allowing mediators to reach the pulmonary arterial bed in portal hypertension, high flow may trigger a vasoproliferative process in the pulmonary vascular bed. Patients with advanced liver disease have a low systemic vascular resistance, with a compensatory increase in cardiac output. An increase in cardiac output can lead to shear stress of the pulmonary vascular endothelial layer. Although the resistance of the pulmonary vasculature may decrease rapidly to help normalize pulmonary pressures, persistent circulatory overload could result in irreversible vascular changes. Autopsy and lung explant studies show that POPH is characterized by obstructive and remodeling changes in the pulmonary arterial bed.24 Initially, medial hypertrophy with smooth muscle proliferation is present. As the disease advances, platelet aggregates, in situ thrombosis, and intimal fibrosis develop. Finally, web-like lesions involving the entire pulmonary wall develop with recanalization for the passage of pulmonary arterial flow. These changes are identical to the changes observed in patients with other forms of PAH.

        Not all patients with portal hypertension develop POPH, suggesting that genetic predisposition may play a role in POPH development. The Pulmonary Vascular Complications of Liver Study Group published a multicenter case-control study that attempted to identify genetic risk factors for POPH in patients with advanced liver disease.25 More than 1000 common single nucleotide polymorphisms (SNPs) in 93 candidate genes were genotyped in each patient. When compared to controls, multiple SNPs in the genes coding for estrogen receptor 1, aromatase, phosphodiesterase 5, angiopoietin 1, and calcium binding protein A4 were associated with an increased risk of POPH. One year earlier, the same study group concluded that female sex (adjusted odds ratio [OR], 2.90) and autoimmune hepatitis (adjusted OR, 4.02) were associated with a higher risk for POPH, whereas hepatitis C was associated with a decreased risk.20

        Clinical Presentation

        Clinical presentation is variable in POPH. Patients referred to a pulmonologist will usually present with symptoms similar to patients with other forms of PAH. In a retrospective analysis of patients referred to the French Referral Center for Pulmonary Hypertension, 60% of the patients belonged to NYHA functional class III or IV.1 In a series of 78 patients with POPH, the most common presenting pulmonary symptom was dyspnea on exertion (81%), followed by syncope, chest pain, and fatigue (< 33%).16 Symptoms such as syncope and chest pain are usually markers of severe POPH.3 Stigmata of portal hypertension, such as ascites, spider angiomata, and palmar erythema, may be present on exam. An accentuated pulmonary component of the second heart sound can be seen in 82% of patients and a systolic murmur caused by tricuspid regurgitation in 69% of patients.16 Patients with severe POPH may have jugular vein distention, peripheral edema, and a third heart sound.

        Diagnostic Evaluation

        Chest x-rays may show prominent pulmonary arteries and cardiomegaly in patients with POPH, whereas electrocardiogram can suggest right ventricular hypertrophy and right axis deviation. The best screening test for POPH in patients with portal hypertension is echocardiography. Routine screening for POPH is recommended during liver transplant evaluation in the practice guidelines from the American Association for the Study of Liver Disease.26 Right-sided cardiac chamber enlargement and right ventricular pressure or volume overload can be assessed on echocardiography. Colle et al followed 165 patients evaluated for liver transplantation who underwent transthoracic Doppler echocardiography and right heart catheterization.27 Seventeen patients met the criteria for POPH on echocardiography (presence of tricuspid regurgitation and calculated systolic pulmonary artery pressure over 30 mm Hg) and right heart catheterization confirmed the diagnosis in 10 patients. Right ventricular systolic pressure (RVSP) estimate of ≤ 30 mm Hg on 2-dimensional echo had a 100% sensitivity and negative predictive value. Positive predictive value was poor at 59%, reiterating the need for right heart catheterization in the diagnosis of POPH. When Kim et al used a RVSP threshold of 50 mm Hg, 72% had at least moderate pulmonary hypertension, including 30% with severe pulmonary hypertension.28 Raevens et al analyzed data from 152 patients who underwent pretransplant echocardiography and catheterization.2 Their data show a RVSP threshold of greater than 38 mm Hg by echocardiography had a specificity of 82% and sensitivity and negative predictive value of 100%. The European Respiratory Society recommendations state that PAH should be considered unlikely if echocardiography estimates a RVSP ≤36 mm Hg and likely if the RVSP is estimated at > 50 mm Hg.29 We recommend repeating echocardiography every 6 to 12 months in patients listed for liver transplantation, as pulmonary hemodynamics may change over time.

        Computed tomography (CT) may have a complementary role in the future for the noninvasive detection of POPH. In a study published in 2014, 49 patients referred for liver transplantation were retrospectively reviewed.30 Measured CT signs included the main pulmonary artery/ascending aorta diameter ratio, the mean left and right main pulmonary artery diameter, and the enlargement of the pulmonary artery compared to the ascending aorta. Compared to the transthoracic echocardiography alone, an algorithm incorporating CT and echocardiography improved the detection of POPH (area under curve = 0.8, P < 0.0001).

        A diagnosis of POPH can only be confirmed when PAH exists in a patient with portal hypertension, as determined by right heart catheterization, and no other cause of PAH can be identified. MPAP should be 25 mm Hg or greater, PVR of 240 dynes/s/cm–5, wedge pressure of 15 mm Hg or less, and TPG greater than 12 mm Hg. Krowka et al showed the value of right heart catheterization in their 10-year prospective, echocardiography-catheterization algorithm study.19 Of 1235 liver transplant candidates who underwent echocardiography, 104 patients had a RVSP exceeding 50 mm Hg. Almost all of these patients had a right heart catheterization. All cause pulmonary hypertension (MPAP > 25 mm Hg) was confirmed in 90% of the patients, and 35% had a PVR < 240 dynes/s/cm–5 and pulmonary capillary wedge pressure (PCWP) > 15 mm Hg, suggesting fluid overload. Forty-one patients had significant POPH, with a PVR > 400 dynes/s/cm–5, and 24% also had an elevated PCWP. TPG was > 12 mm Hg in all of these patients, confirming POPH. As demonstrated by this study, right heart catheterization is required to confirm the diagnosis of POPH because high flow and fluid overload can lead to elevated pulmonary artery pressures.

        Patients with POPH have a unique clinical profile with characteristics common to patients with primary pulmonary hypertension and chronic liver disease. In a retrospective review that compared 30 patients with PAH, 30 patients with chronic liver disease only, and 30 patients with catheterization-proved POPH,31 patients with POPH had elevated MPAP similar to those with primary PAH, but they also had reduced SVR and elevated cardiac index similar to those with chronic liver disease alone.

        Besides POPH, 2 other common causes can lead to increased pulmonary arterial blood flow in patients with portal hypertension. First is a high-flow condition caused by increased cardiac output but with a normal PVR and PCWP. Fluid overload can also lead to pulmonary venous hypertension with increased PCWP, normal cardiac output, and normal PVR. Up to 25% of patients with POPH may present with marked excess volume caused by fluid retention.3 There can be an increase in both PCWP and PVR depending on the presence and the degree of fluid retention. TPG (MPAP – PCWP) > 12 mm Hg was introduced to make such patients less confusing and to help correct for increased PCWP secondary to fluid overload. Obstruction to pulmonary arterial flow is manifest by an increased TPG (Table 2).

        pdbr01612001_t2.png

        POPH should be distinguished from hepatopulmonary syndrome (HPS), which is another pulmonary vascular consequence of liver disease. Unlike POPH, HPS is characterized by a defect in arterial oxygenation induced by pulmonary vascular dilation.32 Similar to other patients with liver disease, patients with HPS have a normal PVR and increased cardiac output secondary to a high-flow state. HPS is diagnosed by confirmation of an intrapulmonary shunt by echocardiogram. Injection of agitated saline results in saline bubbles being visualized in the left atrium 3 or more cardiac cycles after they appear in the right atrium. Currently, there is no effective medical treatment for HPS and liver transplantation is the only successful treatment.

        Conclusion

        POPH is an uncommon complication of chronic liver disease. It is defined as PAH in a patient with portal hypertension excluding other causes of PAH. The following criteria must be met to make a diagnosis of POPH: (1) evidence of portal hypertension; (2) MPAP ≥ 35 mm Hg; (3) PVR ≥ 240 dynes/s/cm5; (4) pulmonary capillary wedge pressure ≤ 15 mm Hg; and (5) TPG > 12 mm Hg. Individuals with POPH have worse outcomes compared to other forms of PAH, with a median survival of 6 months without medical therapy. The pathogenesis of POPH is unclear but may be related to a genetic predisposition since not all patients with portal hypertension develop POPH. Echocardiography is an excellent screening test for POPH, but a right heart catheterization must be performed to confirm the diagnosis.

        References

        1. Le Pavec J, Souza R, Herve P, et al. Portopulmonary hypertension: survival and prognostic factors. Am J Respir Crit Care Med. 2008;178:637-643.

        2. Raevens S, Colle I, Reyntjens K, et al. Echocardiography for the detection of portopulmonary hypertension in liver transplant candidates: An analysis of cutoff values. Liver Transplant. 2013;19:602-610.

        3. Krowka MJ. Portopulmonary hypertension. Semin Respir Crit Care Med. 2012;33:17-25.

        4. Mantz F. Portal axis thrombosis with spontaneous portocaval shunt and resultant cor pulmonale. AMA Arch Pathol. 1951;52:91-97.

        5. Yoshida EM, Erb SR, Pflugfelder PW, et al. Single-lung versus liver transplantation for the treatment of portopulmonary hypertension--a comparison of two patients. Transplantation. 1993;55:688-690.

        6. Badesch DB, Champion HC, Gomez Sanchez MA, et al. Diagnosis and assessment of pulmonary arterial hypertension. J Am Coll Cardiol. 2009;54 54(1 Suppl):S55-66.

        7. Cartin-Ceba R, Krowka MJ. Portopulmonary hypertension. Clin Liver Dis. 2014;18:421-438.

        8. Ramsay M, Simpson BR, Nguyen T, et al. Severe pulmonary hypertension in liver transplant candidates. Liver Transpl Surg. 1997;3:494-500.

        9. Krowka MJ, Plevak DJ, Findlay JY, et al. Pulmonary hemodynamics and perioperative cardiopulmonary-related mortality in patients with portopulmonary hypertension undergoing liver transplantation. Liver Transpl. 2000;6:443-450.

        10. McDonnell P, Toye P, Hutchins G. Primary pulmonary hypertension and cirrhosis: are they related? Am Rev Respir Dis. 1983;127:437-441.

        11. Rich S, Dantzker D, Ayres S, et al. Primary pulmonary hypertension. A national prospective study. Ann Intern Med. 1987;107:216-223.

        12. Groves B. Pulmonary Hypertension Associated with Cirrhosis. Philadelphia: University of Pennsylvania Press; 1990.

        13. Habib G, Gressin V, Yaici A, et al. Pulmonary arterial hypertension in France results from a national registry. Am J Respir Crit Care Med. 2006;173:1023-1030.

        14. Hadengue A, Benhayoun M, Lebrec D, Benhamou J. Pulmonary hypertension complicating portal hypertension: prevalence and relation to splanchnic hemodynamics. Gastroenterology. 1991;100:520-528.

        15. Krowka MJ, Miller DP, Barst RJ, et al. Portopulmonary hypertension: a report from the US-based REVEAL Registry. Chest. 2012;141:906-915.

        16. Robalino BD, Moodie DS. Association between primary pulmonary hypertension and portal hypertension: analysis of its pathophysiology and clinical, laboratory and hemodynamic manifestations. J Am Coll Cardiol. 1991;17:492-498.

        17. Kawut SM, Taichman DB, Ahya VN, et al. Hemodynamics and survival of patients with portopulmonary hypertension. Liver Transpl. 2005;11:1107-1111.

        18. Swanson KL, Wiesner RH, Nyberg SL, et al. Survival in portopulmonary hypertension: Mayo Clinic experience categorized by treatment subgroups. Am J Transplant. 2008;8:2445-2453.

        19. Krowka MJ, Swanson KL, Frantz RP, et al. Portopulmonary hypertension: Results from a 10-year screening algorithm. Hepatology. 2006;44:1502-1510.

        20. Kawut SM, Krowka MJ, Trotter JF, et al. Clinical risk factors for portopulmonary hypertension. Hepatology. 2008;48:196-203.

        21. Lebrec D, Capron JP, Dhumeaux D, Benhamou JP. pulmonary hypertension complicating portal hypertension. Am J Rev Resp Dis. 1979;120:849-856.

        22. Pellicelli AM, Barbaro G, Puoti C, et al. Plasma cytokines and portopulmonary hypertension in patients with cirrhosis waiting for orthotopic liver transplantation. Angiology. 2010;61:802-806.

        23. Kamath PS, Carpenter HA, Lloyd RV, et al. Hepatic localization of endothelin-1 in patients with idiopathic portal hypertension and cirrhosis of the liver. Liver Transpl. 2000;6:596-602.

        24. Krowka MJ, Edwards WD. A spectrum of pulmonary vascular pathology in portopulmonary hypertension. Liver Transpl. 2000;6:241-242.

        25. Roberts KE, Fallon MB, Krowka MJ, et al. Genetic risk factors for portopulmonary hypertension in patients with advanced liver disease. Am J Respir Crit Care Med. 2009;179:835-842.

        26. Murray KF, Carithers RL. AASLD practice guidelines: Evaluation of the patient for liver transplantation. Hepatology. 2005;41:1407-1432.

        27. Colle IO, Moreau R, Godinho E, et al. Diagnosis of portopulmonary hypertension in candidates for liver transplantation: a prospective study. Hepatology. 2003;37:401-409.

        28. Kim WR, Krowka MJ, Plevak DJ, et al. Accuracy of Doppler echocardiography in the assessment of pulmonary hypertension in liver transplant candidates. Liver Transpl. 2000;6:453-458.

        29. Galiè N, Hoeper MM, Humbert M, et al. Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2009;34:1219-1263.

        30. Devaraj A, Loveridge R, Bosanac D, et al. Portopulmonary hypertension: Improved detection using CT and echocardiography in combination. Eur Radiol. 2014;24:2385-2393.

        31. Kuo P, Plotkin J, Johnson L, et al. Distinctive clinical features of portopulmonary hypertension. Chest. 1997;112:980-986.

        32. Rodríguez-Roisin R, Krowka MJ. Hepatopulmonary syndrome--a liver-induced lung vascular disorder. N Engl J Med. 2008;358:2378-2387.

        Author and Disclosure Information

        Saira Aijaz Khaderi, MD
        Abdominal Transplant & Liver Disease Center, Baylor College of Medicine, Houston, TX

        Zeenat Safdar, MD, MS
        Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston, TX

        Publications
        Topics
        Sections
        Author and Disclosure Information

        Saira Aijaz Khaderi, MD
        Abdominal Transplant & Liver Disease Center, Baylor College of Medicine, Houston, TX

        Zeenat Safdar, MD, MS
        Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston, TX

        Author and Disclosure Information

        Saira Aijaz Khaderi, MD
        Abdominal Transplant & Liver Disease Center, Baylor College of Medicine, Houston, TX

        Zeenat Safdar, MD, MS
        Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston, TX

        Pulmonary arterial hypertension (PAH) is a rare disease that is associated with high mortality and is characterized by pulmonary vascular remodeling. Portopulmonary hypertension (POPH) is a form of PAH that occurs in patients with portal hypertension where no alternative cause of PAH can be identified. POPH is documented in approximately 4.5% to 8.5% of liver transplant candidates,1,2 but there is no relationship between the existence or severity of POPH and the severity of liver dysfunction.3 Mantz and Craig described the first case of POPH in a 53-year-old woman with enlarged pulmonary arteries that exhibited forceful pulsations more characteristic of the aorta than a low-pressure pulmonary trunk.4 Autopsy revealed findings of chronic liver disease including a stenotic portal vein, portocaval shunt, and esophageal varices. In both PAH and POPH, pre-capillary pulmonary arteries have characteristic lesions, such as intimal thickening, endothelial proliferation, and thrombotic changes. This 2-part article reviews the diagnosis and treatment of patients with POPH. Here, we review the epidemiology, prognosis, pathogenesis, and diagnosis of POPH; current treatment options for POPH are reviewed in a separate article.

        Definition

        The term POPH was first used by Yoshida et al in 1993 to describe the first successful liver transplant in a patient with POPH, a 39-year-old man with chronic hepatitis.5 The World Health Organization (WHO) classifies POPH as a form of Group 1 PAH.6 The criteria that must be met to make a diagnosis of POPH are shown in the Table 1.7

        pdbr01612001_t1.png

        Moderate POPH is defined as a mean pulmonary artery pressure (MPAP) between 35 mm Hg and < 45 mm Hg, whereas severe POPH is MPAP ≥ 45 mm Hg. Moderate and severe POPH are considered contraindications to liver transplant because of high perioperative and postoperative mortality rates.8 In 2000, the Mayo Clinic retrospectively reviewed 43 patients with POPH who underwent attempted liver transplantation.9 The cardiopulmonary-related mortality rate in patients with a MPAP of 35 to < 50 mm Hg was 50% and 100% for those with MPAP > 50 mm Hg. No mortality was noted in patients with a pre-liver transplant MPAP of < 35 mm Hg and transpulmonary gradient (TPG) < 15 mm Hg.

        Epidemiology

        In 1983, a series of 17,901 autopsied patients showed a primary pulmonary hypertension prevalence of 0.13% and a prevalence of 0.73% in patients with cirrhosis.10 In 1987, Rich et al published data from the National Institutes of Health’s national registry of primary pulmonary hypertension.11 The registry included data from 187 patients from 32 centers. Further analyses by Groves et al concluded that 8.3% of the patients likely had POPH.12 Humbert et al published data on the French pulmonary hypertension registry experience in 2006.13 The French registry included 674 patients from 17 university hospitals; 10.4% of these patients had POPH. The largest prospective study was published by Hadengue et al in 1991.14 In this study, 507 patients hospitalized with portal hypertension but without known pulmonary hypertension underwent cardiac catheterization; 10 patients (2%) had pulmonary hypertension and more than half were clinically asymptomatic. Finally, the Registry to Evaluate Early And Long-term pulmonary arterial hypertension disease management (REVEAL registry) documented a 5.3% frequency of POPH (174 of 3525) in the United States.15

        Prognosis

        Individuals with POPH have worse outcomes compared to other forms of PAH. Median survival prior to the introduction of vasodilator therapy was a dismal 6 months and mean survival was 15 months.16 The cause of death in patients with POPH is equally distributed between right heart failure from POPH and direct complications of chronic liver disease.1 Le Pavec et al retrospectively analyzed all patients referred to the French Referral Center with POPH between 1984 and 2004 (154 patients).1 Approximately 50% of the patients were Child-Turcotte-Pugh class B or C, and 60% were classified as New York Health Association (NYHA) class III or IV. In these patients, 1-, 3-, and 5-year survival rates were 88%, 75%, and 68%, respectively. Major independent prognostic risk factors were presence and severity of cirrhosis and preservation of right ventricular function. Interestingly, NYHA functional class was not related to survival in this study, although it has clearly been identified as a strong prognostic factor in idiopathic PAH.

        Krowka et al evaluated 174 patients with POPH enrolled in the REVEAL Registry,15 a multicenter, observational, US-based study comprised of more than 3500 patients with PAH. Despite having better hemodynamic parameters at diagnosis, patients with POPH had significantly poorer survival and all-cause hospitalization compared with patients with idiopathic PAH (IPAH) or hereditary PAH (HPAH). Two-year survival from enrollment was 67% in POPH versus 85% in those with IPAH/HPAH (P < 0.001). Five-year survival from time of diagnosis was 40% versus 64% (P < 0.001). Additionally, patients with POPH were less likely to be on PAH-specific therapy at enrollment, with only 25% on treatment at the time of entry. These findings were replicated in 2005 when Kawut et al retrospectively compared 13 patients with POPH with 33 patients with IPAH.17 Despite having a higher cardiac index and lower pulmonary vascular resistance than patients with IPAH, patients with POPH had a higher risk of death (hazard ratio, 2.8, P = 0.04), likely reflecting the combination of 2 serious diseases.

        In 2008 the Mayo Clinic published their retrospective analysis of patients with POPH to determine the natural history of POPH.18 Patients were categorized into 3 groups: (1) no medical therapy for POPH and no liver transplant; (2) medical therapy for POPH alone; (3) medical therapy for POPH followed by liver transplant. The study included 74 patients between 1994 through 2007; 19 patients who did not receive treatment for POPH or liver transplant truly represented the natural history of POPH. Their 5-year survival was only 14%, and over half were deceased 1 year after diagnosis. The largest group consisted of patients who received therapy for POPH but no liver transplant. This group did remarkably better than those who received no therapy at all, with a 5-year survival of 45%. However, the patients with the overall best survival were those who received a combination of treatment for POPH followed by liver transplant. Their 5-year survival was 67%. Survival at 5 years was only 25% for the small group of patients who received transplant without PAH therapy. Once again, mortality did not correlate with the severity of hepatic dysfunction or baseline hemodynamic data.

        Pathogenesis

        The pathogenesis of POPH is unclear. Multiple studies have shown that there is minimal, if any, association with pulmonary hypertension and the severity of liver disease or portal hypertension.19,20 Portal hypertension is the result of an increase in intrahepatic resistance and an increase in blood flow into the portal circulation. Collateral vessels develop and blood from the splanchnic circulation is allowed directly into the systemic venous circulation, bypassing the liver. One of the most widely accepted theories is that a humoral substance, that would otherwise be metabolized by the liver, is able to reach the pulmonary circulation through collaterals, resulting in POPH.21 Pelicelli et al evaluated the possible role of endothelin-1, interleukin-6, interleukin 1β, and tumor necrosis factor in the pathogenesis of POPH.22 Plasma concentrations of these cytokines were compared between patients with POPH and patients with cirrhosis but no POPH. Patients with POPH had higher concentrations of endothelin-1 and interleukin-6, suggesting antagonists for these cytokines may have a role in the treatment of POPH. The role of endothelin-1 was further supported by Kamath et al in 200023 when they determined the pulmonary vascular bed is exposed to increased levels of circulating endothelin-1a in the setting of cirrhosis. Endothelin-1 is a potent vasoconstrictor and facilitator of smooth muscle proliferation.

        In addition to collateral circulation allowing mediators to reach the pulmonary arterial bed in portal hypertension, high flow may trigger a vasoproliferative process in the pulmonary vascular bed. Patients with advanced liver disease have a low systemic vascular resistance, with a compensatory increase in cardiac output. An increase in cardiac output can lead to shear stress of the pulmonary vascular endothelial layer. Although the resistance of the pulmonary vasculature may decrease rapidly to help normalize pulmonary pressures, persistent circulatory overload could result in irreversible vascular changes. Autopsy and lung explant studies show that POPH is characterized by obstructive and remodeling changes in the pulmonary arterial bed.24 Initially, medial hypertrophy with smooth muscle proliferation is present. As the disease advances, platelet aggregates, in situ thrombosis, and intimal fibrosis develop. Finally, web-like lesions involving the entire pulmonary wall develop with recanalization for the passage of pulmonary arterial flow. These changes are identical to the changes observed in patients with other forms of PAH.

        Not all patients with portal hypertension develop POPH, suggesting that genetic predisposition may play a role in POPH development. The Pulmonary Vascular Complications of Liver Study Group published a multicenter case-control study that attempted to identify genetic risk factors for POPH in patients with advanced liver disease.25 More than 1000 common single nucleotide polymorphisms (SNPs) in 93 candidate genes were genotyped in each patient. When compared to controls, multiple SNPs in the genes coding for estrogen receptor 1, aromatase, phosphodiesterase 5, angiopoietin 1, and calcium binding protein A4 were associated with an increased risk of POPH. One year earlier, the same study group concluded that female sex (adjusted odds ratio [OR], 2.90) and autoimmune hepatitis (adjusted OR, 4.02) were associated with a higher risk for POPH, whereas hepatitis C was associated with a decreased risk.20

        Clinical Presentation

        Clinical presentation is variable in POPH. Patients referred to a pulmonologist will usually present with symptoms similar to patients with other forms of PAH. In a retrospective analysis of patients referred to the French Referral Center for Pulmonary Hypertension, 60% of the patients belonged to NYHA functional class III or IV.1 In a series of 78 patients with POPH, the most common presenting pulmonary symptom was dyspnea on exertion (81%), followed by syncope, chest pain, and fatigue (< 33%).16 Symptoms such as syncope and chest pain are usually markers of severe POPH.3 Stigmata of portal hypertension, such as ascites, spider angiomata, and palmar erythema, may be present on exam. An accentuated pulmonary component of the second heart sound can be seen in 82% of patients and a systolic murmur caused by tricuspid regurgitation in 69% of patients.16 Patients with severe POPH may have jugular vein distention, peripheral edema, and a third heart sound.

        Diagnostic Evaluation

        Chest x-rays may show prominent pulmonary arteries and cardiomegaly in patients with POPH, whereas electrocardiogram can suggest right ventricular hypertrophy and right axis deviation. The best screening test for POPH in patients with portal hypertension is echocardiography. Routine screening for POPH is recommended during liver transplant evaluation in the practice guidelines from the American Association for the Study of Liver Disease.26 Right-sided cardiac chamber enlargement and right ventricular pressure or volume overload can be assessed on echocardiography. Colle et al followed 165 patients evaluated for liver transplantation who underwent transthoracic Doppler echocardiography and right heart catheterization.27 Seventeen patients met the criteria for POPH on echocardiography (presence of tricuspid regurgitation and calculated systolic pulmonary artery pressure over 30 mm Hg) and right heart catheterization confirmed the diagnosis in 10 patients. Right ventricular systolic pressure (RVSP) estimate of ≤ 30 mm Hg on 2-dimensional echo had a 100% sensitivity and negative predictive value. Positive predictive value was poor at 59%, reiterating the need for right heart catheterization in the diagnosis of POPH. When Kim et al used a RVSP threshold of 50 mm Hg, 72% had at least moderate pulmonary hypertension, including 30% with severe pulmonary hypertension.28 Raevens et al analyzed data from 152 patients who underwent pretransplant echocardiography and catheterization.2 Their data show a RVSP threshold of greater than 38 mm Hg by echocardiography had a specificity of 82% and sensitivity and negative predictive value of 100%. The European Respiratory Society recommendations state that PAH should be considered unlikely if echocardiography estimates a RVSP ≤36 mm Hg and likely if the RVSP is estimated at > 50 mm Hg.29 We recommend repeating echocardiography every 6 to 12 months in patients listed for liver transplantation, as pulmonary hemodynamics may change over time.

        Computed tomography (CT) may have a complementary role in the future for the noninvasive detection of POPH. In a study published in 2014, 49 patients referred for liver transplantation were retrospectively reviewed.30 Measured CT signs included the main pulmonary artery/ascending aorta diameter ratio, the mean left and right main pulmonary artery diameter, and the enlargement of the pulmonary artery compared to the ascending aorta. Compared to the transthoracic echocardiography alone, an algorithm incorporating CT and echocardiography improved the detection of POPH (area under curve = 0.8, P < 0.0001).

        A diagnosis of POPH can only be confirmed when PAH exists in a patient with portal hypertension, as determined by right heart catheterization, and no other cause of PAH can be identified. MPAP should be 25 mm Hg or greater, PVR of 240 dynes/s/cm–5, wedge pressure of 15 mm Hg or less, and TPG greater than 12 mm Hg. Krowka et al showed the value of right heart catheterization in their 10-year prospective, echocardiography-catheterization algorithm study.19 Of 1235 liver transplant candidates who underwent echocardiography, 104 patients had a RVSP exceeding 50 mm Hg. Almost all of these patients had a right heart catheterization. All cause pulmonary hypertension (MPAP > 25 mm Hg) was confirmed in 90% of the patients, and 35% had a PVR < 240 dynes/s/cm–5 and pulmonary capillary wedge pressure (PCWP) > 15 mm Hg, suggesting fluid overload. Forty-one patients had significant POPH, with a PVR > 400 dynes/s/cm–5, and 24% also had an elevated PCWP. TPG was > 12 mm Hg in all of these patients, confirming POPH. As demonstrated by this study, right heart catheterization is required to confirm the diagnosis of POPH because high flow and fluid overload can lead to elevated pulmonary artery pressures.

        Patients with POPH have a unique clinical profile with characteristics common to patients with primary pulmonary hypertension and chronic liver disease. In a retrospective review that compared 30 patients with PAH, 30 patients with chronic liver disease only, and 30 patients with catheterization-proved POPH,31 patients with POPH had elevated MPAP similar to those with primary PAH, but they also had reduced SVR and elevated cardiac index similar to those with chronic liver disease alone.

        Besides POPH, 2 other common causes can lead to increased pulmonary arterial blood flow in patients with portal hypertension. First is a high-flow condition caused by increased cardiac output but with a normal PVR and PCWP. Fluid overload can also lead to pulmonary venous hypertension with increased PCWP, normal cardiac output, and normal PVR. Up to 25% of patients with POPH may present with marked excess volume caused by fluid retention.3 There can be an increase in both PCWP and PVR depending on the presence and the degree of fluid retention. TPG (MPAP – PCWP) > 12 mm Hg was introduced to make such patients less confusing and to help correct for increased PCWP secondary to fluid overload. Obstruction to pulmonary arterial flow is manifest by an increased TPG (Table 2).

        pdbr01612001_t2.png

        POPH should be distinguished from hepatopulmonary syndrome (HPS), which is another pulmonary vascular consequence of liver disease. Unlike POPH, HPS is characterized by a defect in arterial oxygenation induced by pulmonary vascular dilation.32 Similar to other patients with liver disease, patients with HPS have a normal PVR and increased cardiac output secondary to a high-flow state. HPS is diagnosed by confirmation of an intrapulmonary shunt by echocardiogram. Injection of agitated saline results in saline bubbles being visualized in the left atrium 3 or more cardiac cycles after they appear in the right atrium. Currently, there is no effective medical treatment for HPS and liver transplantation is the only successful treatment.

        Conclusion

        POPH is an uncommon complication of chronic liver disease. It is defined as PAH in a patient with portal hypertension excluding other causes of PAH. The following criteria must be met to make a diagnosis of POPH: (1) evidence of portal hypertension; (2) MPAP ≥ 35 mm Hg; (3) PVR ≥ 240 dynes/s/cm5; (4) pulmonary capillary wedge pressure ≤ 15 mm Hg; and (5) TPG > 12 mm Hg. Individuals with POPH have worse outcomes compared to other forms of PAH, with a median survival of 6 months without medical therapy. The pathogenesis of POPH is unclear but may be related to a genetic predisposition since not all patients with portal hypertension develop POPH. Echocardiography is an excellent screening test for POPH, but a right heart catheterization must be performed to confirm the diagnosis.

        Pulmonary arterial hypertension (PAH) is a rare disease that is associated with high mortality and is characterized by pulmonary vascular remodeling. Portopulmonary hypertension (POPH) is a form of PAH that occurs in patients with portal hypertension where no alternative cause of PAH can be identified. POPH is documented in approximately 4.5% to 8.5% of liver transplant candidates,1,2 but there is no relationship between the existence or severity of POPH and the severity of liver dysfunction.3 Mantz and Craig described the first case of POPH in a 53-year-old woman with enlarged pulmonary arteries that exhibited forceful pulsations more characteristic of the aorta than a low-pressure pulmonary trunk.4 Autopsy revealed findings of chronic liver disease including a stenotic portal vein, portocaval shunt, and esophageal varices. In both PAH and POPH, pre-capillary pulmonary arteries have characteristic lesions, such as intimal thickening, endothelial proliferation, and thrombotic changes. This 2-part article reviews the diagnosis and treatment of patients with POPH. Here, we review the epidemiology, prognosis, pathogenesis, and diagnosis of POPH; current treatment options for POPH are reviewed in a separate article.

        Definition

        The term POPH was first used by Yoshida et al in 1993 to describe the first successful liver transplant in a patient with POPH, a 39-year-old man with chronic hepatitis.5 The World Health Organization (WHO) classifies POPH as a form of Group 1 PAH.6 The criteria that must be met to make a diagnosis of POPH are shown in the Table 1.7

        pdbr01612001_t1.png

        Moderate POPH is defined as a mean pulmonary artery pressure (MPAP) between 35 mm Hg and < 45 mm Hg, whereas severe POPH is MPAP ≥ 45 mm Hg. Moderate and severe POPH are considered contraindications to liver transplant because of high perioperative and postoperative mortality rates.8 In 2000, the Mayo Clinic retrospectively reviewed 43 patients with POPH who underwent attempted liver transplantation.9 The cardiopulmonary-related mortality rate in patients with a MPAP of 35 to < 50 mm Hg was 50% and 100% for those with MPAP > 50 mm Hg. No mortality was noted in patients with a pre-liver transplant MPAP of < 35 mm Hg and transpulmonary gradient (TPG) < 15 mm Hg.

        Epidemiology

        In 1983, a series of 17,901 autopsied patients showed a primary pulmonary hypertension prevalence of 0.13% and a prevalence of 0.73% in patients with cirrhosis.10 In 1987, Rich et al published data from the National Institutes of Health’s national registry of primary pulmonary hypertension.11 The registry included data from 187 patients from 32 centers. Further analyses by Groves et al concluded that 8.3% of the patients likely had POPH.12 Humbert et al published data on the French pulmonary hypertension registry experience in 2006.13 The French registry included 674 patients from 17 university hospitals; 10.4% of these patients had POPH. The largest prospective study was published by Hadengue et al in 1991.14 In this study, 507 patients hospitalized with portal hypertension but without known pulmonary hypertension underwent cardiac catheterization; 10 patients (2%) had pulmonary hypertension and more than half were clinically asymptomatic. Finally, the Registry to Evaluate Early And Long-term pulmonary arterial hypertension disease management (REVEAL registry) documented a 5.3% frequency of POPH (174 of 3525) in the United States.15

        Prognosis

        Individuals with POPH have worse outcomes compared to other forms of PAH. Median survival prior to the introduction of vasodilator therapy was a dismal 6 months and mean survival was 15 months.16 The cause of death in patients with POPH is equally distributed between right heart failure from POPH and direct complications of chronic liver disease.1 Le Pavec et al retrospectively analyzed all patients referred to the French Referral Center with POPH between 1984 and 2004 (154 patients).1 Approximately 50% of the patients were Child-Turcotte-Pugh class B or C, and 60% were classified as New York Health Association (NYHA) class III or IV. In these patients, 1-, 3-, and 5-year survival rates were 88%, 75%, and 68%, respectively. Major independent prognostic risk factors were presence and severity of cirrhosis and preservation of right ventricular function. Interestingly, NYHA functional class was not related to survival in this study, although it has clearly been identified as a strong prognostic factor in idiopathic PAH.

        Krowka et al evaluated 174 patients with POPH enrolled in the REVEAL Registry,15 a multicenter, observational, US-based study comprised of more than 3500 patients with PAH. Despite having better hemodynamic parameters at diagnosis, patients with POPH had significantly poorer survival and all-cause hospitalization compared with patients with idiopathic PAH (IPAH) or hereditary PAH (HPAH). Two-year survival from enrollment was 67% in POPH versus 85% in those with IPAH/HPAH (P < 0.001). Five-year survival from time of diagnosis was 40% versus 64% (P < 0.001). Additionally, patients with POPH were less likely to be on PAH-specific therapy at enrollment, with only 25% on treatment at the time of entry. These findings were replicated in 2005 when Kawut et al retrospectively compared 13 patients with POPH with 33 patients with IPAH.17 Despite having a higher cardiac index and lower pulmonary vascular resistance than patients with IPAH, patients with POPH had a higher risk of death (hazard ratio, 2.8, P = 0.04), likely reflecting the combination of 2 serious diseases.

        In 2008 the Mayo Clinic published their retrospective analysis of patients with POPH to determine the natural history of POPH.18 Patients were categorized into 3 groups: (1) no medical therapy for POPH and no liver transplant; (2) medical therapy for POPH alone; (3) medical therapy for POPH followed by liver transplant. The study included 74 patients between 1994 through 2007; 19 patients who did not receive treatment for POPH or liver transplant truly represented the natural history of POPH. Their 5-year survival was only 14%, and over half were deceased 1 year after diagnosis. The largest group consisted of patients who received therapy for POPH but no liver transplant. This group did remarkably better than those who received no therapy at all, with a 5-year survival of 45%. However, the patients with the overall best survival were those who received a combination of treatment for POPH followed by liver transplant. Their 5-year survival was 67%. Survival at 5 years was only 25% for the small group of patients who received transplant without PAH therapy. Once again, mortality did not correlate with the severity of hepatic dysfunction or baseline hemodynamic data.

        Pathogenesis

        The pathogenesis of POPH is unclear. Multiple studies have shown that there is minimal, if any, association with pulmonary hypertension and the severity of liver disease or portal hypertension.19,20 Portal hypertension is the result of an increase in intrahepatic resistance and an increase in blood flow into the portal circulation. Collateral vessels develop and blood from the splanchnic circulation is allowed directly into the systemic venous circulation, bypassing the liver. One of the most widely accepted theories is that a humoral substance, that would otherwise be metabolized by the liver, is able to reach the pulmonary circulation through collaterals, resulting in POPH.21 Pelicelli et al evaluated the possible role of endothelin-1, interleukin-6, interleukin 1β, and tumor necrosis factor in the pathogenesis of POPH.22 Plasma concentrations of these cytokines were compared between patients with POPH and patients with cirrhosis but no POPH. Patients with POPH had higher concentrations of endothelin-1 and interleukin-6, suggesting antagonists for these cytokines may have a role in the treatment of POPH. The role of endothelin-1 was further supported by Kamath et al in 200023 when they determined the pulmonary vascular bed is exposed to increased levels of circulating endothelin-1a in the setting of cirrhosis. Endothelin-1 is a potent vasoconstrictor and facilitator of smooth muscle proliferation.

        In addition to collateral circulation allowing mediators to reach the pulmonary arterial bed in portal hypertension, high flow may trigger a vasoproliferative process in the pulmonary vascular bed. Patients with advanced liver disease have a low systemic vascular resistance, with a compensatory increase in cardiac output. An increase in cardiac output can lead to shear stress of the pulmonary vascular endothelial layer. Although the resistance of the pulmonary vasculature may decrease rapidly to help normalize pulmonary pressures, persistent circulatory overload could result in irreversible vascular changes. Autopsy and lung explant studies show that POPH is characterized by obstructive and remodeling changes in the pulmonary arterial bed.24 Initially, medial hypertrophy with smooth muscle proliferation is present. As the disease advances, platelet aggregates, in situ thrombosis, and intimal fibrosis develop. Finally, web-like lesions involving the entire pulmonary wall develop with recanalization for the passage of pulmonary arterial flow. These changes are identical to the changes observed in patients with other forms of PAH.

        Not all patients with portal hypertension develop POPH, suggesting that genetic predisposition may play a role in POPH development. The Pulmonary Vascular Complications of Liver Study Group published a multicenter case-control study that attempted to identify genetic risk factors for POPH in patients with advanced liver disease.25 More than 1000 common single nucleotide polymorphisms (SNPs) in 93 candidate genes were genotyped in each patient. When compared to controls, multiple SNPs in the genes coding for estrogen receptor 1, aromatase, phosphodiesterase 5, angiopoietin 1, and calcium binding protein A4 were associated with an increased risk of POPH. One year earlier, the same study group concluded that female sex (adjusted odds ratio [OR], 2.90) and autoimmune hepatitis (adjusted OR, 4.02) were associated with a higher risk for POPH, whereas hepatitis C was associated with a decreased risk.20

        Clinical Presentation

        Clinical presentation is variable in POPH. Patients referred to a pulmonologist will usually present with symptoms similar to patients with other forms of PAH. In a retrospective analysis of patients referred to the French Referral Center for Pulmonary Hypertension, 60% of the patients belonged to NYHA functional class III or IV.1 In a series of 78 patients with POPH, the most common presenting pulmonary symptom was dyspnea on exertion (81%), followed by syncope, chest pain, and fatigue (< 33%).16 Symptoms such as syncope and chest pain are usually markers of severe POPH.3 Stigmata of portal hypertension, such as ascites, spider angiomata, and palmar erythema, may be present on exam. An accentuated pulmonary component of the second heart sound can be seen in 82% of patients and a systolic murmur caused by tricuspid regurgitation in 69% of patients.16 Patients with severe POPH may have jugular vein distention, peripheral edema, and a third heart sound.

        Diagnostic Evaluation

        Chest x-rays may show prominent pulmonary arteries and cardiomegaly in patients with POPH, whereas electrocardiogram can suggest right ventricular hypertrophy and right axis deviation. The best screening test for POPH in patients with portal hypertension is echocardiography. Routine screening for POPH is recommended during liver transplant evaluation in the practice guidelines from the American Association for the Study of Liver Disease.26 Right-sided cardiac chamber enlargement and right ventricular pressure or volume overload can be assessed on echocardiography. Colle et al followed 165 patients evaluated for liver transplantation who underwent transthoracic Doppler echocardiography and right heart catheterization.27 Seventeen patients met the criteria for POPH on echocardiography (presence of tricuspid regurgitation and calculated systolic pulmonary artery pressure over 30 mm Hg) and right heart catheterization confirmed the diagnosis in 10 patients. Right ventricular systolic pressure (RVSP) estimate of ≤ 30 mm Hg on 2-dimensional echo had a 100% sensitivity and negative predictive value. Positive predictive value was poor at 59%, reiterating the need for right heart catheterization in the diagnosis of POPH. When Kim et al used a RVSP threshold of 50 mm Hg, 72% had at least moderate pulmonary hypertension, including 30% with severe pulmonary hypertension.28 Raevens et al analyzed data from 152 patients who underwent pretransplant echocardiography and catheterization.2 Their data show a RVSP threshold of greater than 38 mm Hg by echocardiography had a specificity of 82% and sensitivity and negative predictive value of 100%. The European Respiratory Society recommendations state that PAH should be considered unlikely if echocardiography estimates a RVSP ≤36 mm Hg and likely if the RVSP is estimated at > 50 mm Hg.29 We recommend repeating echocardiography every 6 to 12 months in patients listed for liver transplantation, as pulmonary hemodynamics may change over time.

        Computed tomography (CT) may have a complementary role in the future for the noninvasive detection of POPH. In a study published in 2014, 49 patients referred for liver transplantation were retrospectively reviewed.30 Measured CT signs included the main pulmonary artery/ascending aorta diameter ratio, the mean left and right main pulmonary artery diameter, and the enlargement of the pulmonary artery compared to the ascending aorta. Compared to the transthoracic echocardiography alone, an algorithm incorporating CT and echocardiography improved the detection of POPH (area under curve = 0.8, P < 0.0001).

        A diagnosis of POPH can only be confirmed when PAH exists in a patient with portal hypertension, as determined by right heart catheterization, and no other cause of PAH can be identified. MPAP should be 25 mm Hg or greater, PVR of 240 dynes/s/cm–5, wedge pressure of 15 mm Hg or less, and TPG greater than 12 mm Hg. Krowka et al showed the value of right heart catheterization in their 10-year prospective, echocardiography-catheterization algorithm study.19 Of 1235 liver transplant candidates who underwent echocardiography, 104 patients had a RVSP exceeding 50 mm Hg. Almost all of these patients had a right heart catheterization. All cause pulmonary hypertension (MPAP > 25 mm Hg) was confirmed in 90% of the patients, and 35% had a PVR < 240 dynes/s/cm–5 and pulmonary capillary wedge pressure (PCWP) > 15 mm Hg, suggesting fluid overload. Forty-one patients had significant POPH, with a PVR > 400 dynes/s/cm–5, and 24% also had an elevated PCWP. TPG was > 12 mm Hg in all of these patients, confirming POPH. As demonstrated by this study, right heart catheterization is required to confirm the diagnosis of POPH because high flow and fluid overload can lead to elevated pulmonary artery pressures.

        Patients with POPH have a unique clinical profile with characteristics common to patients with primary pulmonary hypertension and chronic liver disease. In a retrospective review that compared 30 patients with PAH, 30 patients with chronic liver disease only, and 30 patients with catheterization-proved POPH,31 patients with POPH had elevated MPAP similar to those with primary PAH, but they also had reduced SVR and elevated cardiac index similar to those with chronic liver disease alone.

        Besides POPH, 2 other common causes can lead to increased pulmonary arterial blood flow in patients with portal hypertension. First is a high-flow condition caused by increased cardiac output but with a normal PVR and PCWP. Fluid overload can also lead to pulmonary venous hypertension with increased PCWP, normal cardiac output, and normal PVR. Up to 25% of patients with POPH may present with marked excess volume caused by fluid retention.3 There can be an increase in both PCWP and PVR depending on the presence and the degree of fluid retention. TPG (MPAP – PCWP) > 12 mm Hg was introduced to make such patients less confusing and to help correct for increased PCWP secondary to fluid overload. Obstruction to pulmonary arterial flow is manifest by an increased TPG (Table 2).

        pdbr01612001_t2.png

        POPH should be distinguished from hepatopulmonary syndrome (HPS), which is another pulmonary vascular consequence of liver disease. Unlike POPH, HPS is characterized by a defect in arterial oxygenation induced by pulmonary vascular dilation.32 Similar to other patients with liver disease, patients with HPS have a normal PVR and increased cardiac output secondary to a high-flow state. HPS is diagnosed by confirmation of an intrapulmonary shunt by echocardiogram. Injection of agitated saline results in saline bubbles being visualized in the left atrium 3 or more cardiac cycles after they appear in the right atrium. Currently, there is no effective medical treatment for HPS and liver transplantation is the only successful treatment.

        Conclusion

        POPH is an uncommon complication of chronic liver disease. It is defined as PAH in a patient with portal hypertension excluding other causes of PAH. The following criteria must be met to make a diagnosis of POPH: (1) evidence of portal hypertension; (2) MPAP ≥ 35 mm Hg; (3) PVR ≥ 240 dynes/s/cm5; (4) pulmonary capillary wedge pressure ≤ 15 mm Hg; and (5) TPG > 12 mm Hg. Individuals with POPH have worse outcomes compared to other forms of PAH, with a median survival of 6 months without medical therapy. The pathogenesis of POPH is unclear but may be related to a genetic predisposition since not all patients with portal hypertension develop POPH. Echocardiography is an excellent screening test for POPH, but a right heart catheterization must be performed to confirm the diagnosis.

        References

        1. Le Pavec J, Souza R, Herve P, et al. Portopulmonary hypertension: survival and prognostic factors. Am J Respir Crit Care Med. 2008;178:637-643.

        2. Raevens S, Colle I, Reyntjens K, et al. Echocardiography for the detection of portopulmonary hypertension in liver transplant candidates: An analysis of cutoff values. Liver Transplant. 2013;19:602-610.

        3. Krowka MJ. Portopulmonary hypertension. Semin Respir Crit Care Med. 2012;33:17-25.

        4. Mantz F. Portal axis thrombosis with spontaneous portocaval shunt and resultant cor pulmonale. AMA Arch Pathol. 1951;52:91-97.

        5. Yoshida EM, Erb SR, Pflugfelder PW, et al. Single-lung versus liver transplantation for the treatment of portopulmonary hypertension--a comparison of two patients. Transplantation. 1993;55:688-690.

        6. Badesch DB, Champion HC, Gomez Sanchez MA, et al. Diagnosis and assessment of pulmonary arterial hypertension. J Am Coll Cardiol. 2009;54 54(1 Suppl):S55-66.

        7. Cartin-Ceba R, Krowka MJ. Portopulmonary hypertension. Clin Liver Dis. 2014;18:421-438.

        8. Ramsay M, Simpson BR, Nguyen T, et al. Severe pulmonary hypertension in liver transplant candidates. Liver Transpl Surg. 1997;3:494-500.

        9. Krowka MJ, Plevak DJ, Findlay JY, et al. Pulmonary hemodynamics and perioperative cardiopulmonary-related mortality in patients with portopulmonary hypertension undergoing liver transplantation. Liver Transpl. 2000;6:443-450.

        10. McDonnell P, Toye P, Hutchins G. Primary pulmonary hypertension and cirrhosis: are they related? Am Rev Respir Dis. 1983;127:437-441.

        11. Rich S, Dantzker D, Ayres S, et al. Primary pulmonary hypertension. A national prospective study. Ann Intern Med. 1987;107:216-223.

        12. Groves B. Pulmonary Hypertension Associated with Cirrhosis. Philadelphia: University of Pennsylvania Press; 1990.

        13. Habib G, Gressin V, Yaici A, et al. Pulmonary arterial hypertension in France results from a national registry. Am J Respir Crit Care Med. 2006;173:1023-1030.

        14. Hadengue A, Benhayoun M, Lebrec D, Benhamou J. Pulmonary hypertension complicating portal hypertension: prevalence and relation to splanchnic hemodynamics. Gastroenterology. 1991;100:520-528.

        15. Krowka MJ, Miller DP, Barst RJ, et al. Portopulmonary hypertension: a report from the US-based REVEAL Registry. Chest. 2012;141:906-915.

        16. Robalino BD, Moodie DS. Association between primary pulmonary hypertension and portal hypertension: analysis of its pathophysiology and clinical, laboratory and hemodynamic manifestations. J Am Coll Cardiol. 1991;17:492-498.

        17. Kawut SM, Taichman DB, Ahya VN, et al. Hemodynamics and survival of patients with portopulmonary hypertension. Liver Transpl. 2005;11:1107-1111.

        18. Swanson KL, Wiesner RH, Nyberg SL, et al. Survival in portopulmonary hypertension: Mayo Clinic experience categorized by treatment subgroups. Am J Transplant. 2008;8:2445-2453.

        19. Krowka MJ, Swanson KL, Frantz RP, et al. Portopulmonary hypertension: Results from a 10-year screening algorithm. Hepatology. 2006;44:1502-1510.

        20. Kawut SM, Krowka MJ, Trotter JF, et al. Clinical risk factors for portopulmonary hypertension. Hepatology. 2008;48:196-203.

        21. Lebrec D, Capron JP, Dhumeaux D, Benhamou JP. pulmonary hypertension complicating portal hypertension. Am J Rev Resp Dis. 1979;120:849-856.

        22. Pellicelli AM, Barbaro G, Puoti C, et al. Plasma cytokines and portopulmonary hypertension in patients with cirrhosis waiting for orthotopic liver transplantation. Angiology. 2010;61:802-806.

        23. Kamath PS, Carpenter HA, Lloyd RV, et al. Hepatic localization of endothelin-1 in patients with idiopathic portal hypertension and cirrhosis of the liver. Liver Transpl. 2000;6:596-602.

        24. Krowka MJ, Edwards WD. A spectrum of pulmonary vascular pathology in portopulmonary hypertension. Liver Transpl. 2000;6:241-242.

        25. Roberts KE, Fallon MB, Krowka MJ, et al. Genetic risk factors for portopulmonary hypertension in patients with advanced liver disease. Am J Respir Crit Care Med. 2009;179:835-842.

        26. Murray KF, Carithers RL. AASLD practice guidelines: Evaluation of the patient for liver transplantation. Hepatology. 2005;41:1407-1432.

        27. Colle IO, Moreau R, Godinho E, et al. Diagnosis of portopulmonary hypertension in candidates for liver transplantation: a prospective study. Hepatology. 2003;37:401-409.

        28. Kim WR, Krowka MJ, Plevak DJ, et al. Accuracy of Doppler echocardiography in the assessment of pulmonary hypertension in liver transplant candidates. Liver Transpl. 2000;6:453-458.

        29. Galiè N, Hoeper MM, Humbert M, et al. Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2009;34:1219-1263.

        30. Devaraj A, Loveridge R, Bosanac D, et al. Portopulmonary hypertension: Improved detection using CT and echocardiography in combination. Eur Radiol. 2014;24:2385-2393.

        31. Kuo P, Plotkin J, Johnson L, et al. Distinctive clinical features of portopulmonary hypertension. Chest. 1997;112:980-986.

        32. Rodríguez-Roisin R, Krowka MJ. Hepatopulmonary syndrome--a liver-induced lung vascular disorder. N Engl J Med. 2008;358:2378-2387.

        References

        1. Le Pavec J, Souza R, Herve P, et al. Portopulmonary hypertension: survival and prognostic factors. Am J Respir Crit Care Med. 2008;178:637-643.

        2. Raevens S, Colle I, Reyntjens K, et al. Echocardiography for the detection of portopulmonary hypertension in liver transplant candidates: An analysis of cutoff values. Liver Transplant. 2013;19:602-610.

        3. Krowka MJ. Portopulmonary hypertension. Semin Respir Crit Care Med. 2012;33:17-25.

        4. Mantz F. Portal axis thrombosis with spontaneous portocaval shunt and resultant cor pulmonale. AMA Arch Pathol. 1951;52:91-97.

        5. Yoshida EM, Erb SR, Pflugfelder PW, et al. Single-lung versus liver transplantation for the treatment of portopulmonary hypertension--a comparison of two patients. Transplantation. 1993;55:688-690.

        6. Badesch DB, Champion HC, Gomez Sanchez MA, et al. Diagnosis and assessment of pulmonary arterial hypertension. J Am Coll Cardiol. 2009;54 54(1 Suppl):S55-66.

        7. Cartin-Ceba R, Krowka MJ. Portopulmonary hypertension. Clin Liver Dis. 2014;18:421-438.

        8. Ramsay M, Simpson BR, Nguyen T, et al. Severe pulmonary hypertension in liver transplant candidates. Liver Transpl Surg. 1997;3:494-500.

        9. Krowka MJ, Plevak DJ, Findlay JY, et al. Pulmonary hemodynamics and perioperative cardiopulmonary-related mortality in patients with portopulmonary hypertension undergoing liver transplantation. Liver Transpl. 2000;6:443-450.

        10. McDonnell P, Toye P, Hutchins G. Primary pulmonary hypertension and cirrhosis: are they related? Am Rev Respir Dis. 1983;127:437-441.

        11. Rich S, Dantzker D, Ayres S, et al. Primary pulmonary hypertension. A national prospective study. Ann Intern Med. 1987;107:216-223.

        12. Groves B. Pulmonary Hypertension Associated with Cirrhosis. Philadelphia: University of Pennsylvania Press; 1990.

        13. Habib G, Gressin V, Yaici A, et al. Pulmonary arterial hypertension in France results from a national registry. Am J Respir Crit Care Med. 2006;173:1023-1030.

        14. Hadengue A, Benhayoun M, Lebrec D, Benhamou J. Pulmonary hypertension complicating portal hypertension: prevalence and relation to splanchnic hemodynamics. Gastroenterology. 1991;100:520-528.

        15. Krowka MJ, Miller DP, Barst RJ, et al. Portopulmonary hypertension: a report from the US-based REVEAL Registry. Chest. 2012;141:906-915.

        16. Robalino BD, Moodie DS. Association between primary pulmonary hypertension and portal hypertension: analysis of its pathophysiology and clinical, laboratory and hemodynamic manifestations. J Am Coll Cardiol. 1991;17:492-498.

        17. Kawut SM, Taichman DB, Ahya VN, et al. Hemodynamics and survival of patients with portopulmonary hypertension. Liver Transpl. 2005;11:1107-1111.

        18. Swanson KL, Wiesner RH, Nyberg SL, et al. Survival in portopulmonary hypertension: Mayo Clinic experience categorized by treatment subgroups. Am J Transplant. 2008;8:2445-2453.

        19. Krowka MJ, Swanson KL, Frantz RP, et al. Portopulmonary hypertension: Results from a 10-year screening algorithm. Hepatology. 2006;44:1502-1510.

        20. Kawut SM, Krowka MJ, Trotter JF, et al. Clinical risk factors for portopulmonary hypertension. Hepatology. 2008;48:196-203.

        21. Lebrec D, Capron JP, Dhumeaux D, Benhamou JP. pulmonary hypertension complicating portal hypertension. Am J Rev Resp Dis. 1979;120:849-856.

        22. Pellicelli AM, Barbaro G, Puoti C, et al. Plasma cytokines and portopulmonary hypertension in patients with cirrhosis waiting for orthotopic liver transplantation. Angiology. 2010;61:802-806.

        23. Kamath PS, Carpenter HA, Lloyd RV, et al. Hepatic localization of endothelin-1 in patients with idiopathic portal hypertension and cirrhosis of the liver. Liver Transpl. 2000;6:596-602.

        24. Krowka MJ, Edwards WD. A spectrum of pulmonary vascular pathology in portopulmonary hypertension. Liver Transpl. 2000;6:241-242.

        25. Roberts KE, Fallon MB, Krowka MJ, et al. Genetic risk factors for portopulmonary hypertension in patients with advanced liver disease. Am J Respir Crit Care Med. 2009;179:835-842.

        26. Murray KF, Carithers RL. AASLD practice guidelines: Evaluation of the patient for liver transplantation. Hepatology. 2005;41:1407-1432.

        27. Colle IO, Moreau R, Godinho E, et al. Diagnosis of portopulmonary hypertension in candidates for liver transplantation: a prospective study. Hepatology. 2003;37:401-409.

        28. Kim WR, Krowka MJ, Plevak DJ, et al. Accuracy of Doppler echocardiography in the assessment of pulmonary hypertension in liver transplant candidates. Liver Transpl. 2000;6:453-458.

        29. Galiè N, Hoeper MM, Humbert M, et al. Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2009;34:1219-1263.

        30. Devaraj A, Loveridge R, Bosanac D, et al. Portopulmonary hypertension: Improved detection using CT and echocardiography in combination. Eur Radiol. 2014;24:2385-2393.

        31. Kuo P, Plotkin J, Johnson L, et al. Distinctive clinical features of portopulmonary hypertension. Chest. 1997;112:980-986.

        32. Rodríguez-Roisin R, Krowka MJ. Hepatopulmonary syndrome--a liver-induced lung vascular disorder. N Engl J Med. 2008;358:2378-2387.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Portopulmonary Hypertension: Evaluation and Diagnosis
        Display Headline
        Portopulmonary Hypertension: Evaluation and Diagnosis
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 December;16(4)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Wed, 12/04/2019 - 11:00
        Un-Gate On Date
        Wed, 12/04/2019 - 11:00
        Use ProPublica
        CFC Schedule Remove Status
        Wed, 12/04/2019 - 11:00
        Hide sidebar & use full width
        render the right sidebar.

        Community-Acquired Pneumonia: Treatment

        Article Type
        Changed
        Mon, 10/07/2019 - 09:49
        Display Headline
        Community-Acquired Pneumonia: Treatment

        Initial management decisions for patients with community-acquired pneumonia (CAP) will depend on severity of infection, with need for hospitalization being one of the first decisions. Because empiric antibiotics are the mainstay of treatment and the causative organisms are seldom identified, underlying medical conditions and epidemiologic risk factors are considered when selecting an empiric regimen. As with other infections, duration of therapy is not standardized, but rather is guided by clinical improvement. Prevention of pneumonia centers around vaccination and smoking cessation. This article, the second in a 2-part review of CAP in adults, focuses on site of care decision, empiric and directed therapies, length of treatment, and prevention strategies. Evaluation and diagnosis of CAP are discussed in a separate article.

        Site of Care Decision

        For patients diagnosed with CAP, the clinician must decide whether treatment will be done in an outpatient or inpatient setting, and for those in the inpatient setting, whether they can safely be treated on the general medical ward or in the intensive care unit (ICU). Two common scoring systems that can be used to aid the clinician in determining severity of the infection and guide site-of-care decisions are the Pneumonia Severity Index (PSI) and CURB-65 scores.

        The PSI score uses 20 different parameters, including comorbidities, laboratory parameters, and radiographic findings, to stratify patients into 5 mortality risk classes.1 On the basis of associated mortality rates, it has been suggested that risk class I and II patients should be treated as outpatients, risk class III patients should be treated in an observation unit or with a short hospitalization, and risk class IV and V patients should be treated as inpatients.1

        The CURB-65 method of risk stratification is based on 5 clinical parameters: confusion, urea level, respiratory rate, systolic blood pressure, and age ≥ 65 years (Table 1).2,3 A modification to the CURB-65 algorithm tool was CRB-65, which excludes urea nitrogen, making it optimal for making determinations in a clinic-based setting. It should be emphasized that these tools do not take into account other factors that should be used in determining location of treatment, such as stable home, mental illness, or concerns about compliance with medications. In many instances, it is these factors that preclude low-risk patients from being treated as outpatients.4,5 Similarly, these scoring systems have not been validated for immunocompromised patients or those who would qualify as having health care–associated pneumonia.

        PDBR01609002_t1.PNG

        Patients with CURB-65 scores of 4 or 5 are considered to have severe pneumonia, and admission to the ICU should be considered for these patients. Aside from the CURB-65 score, anyone requiring vasopressor support or mechanical ventilation merits admission to the ICU.6 American Thoracic Society (ATS) and Infectious Diseases Society of America (IDSA) guidelines also recommend the use of “minor criteria” for making ICU admission decisions; these include respiratory rate ≥ 30 breaths/minute, PaO2 fraction ≤ 250 mm Hg, multilobar infiltrates, confusion, blood urea nitrogen ≥ 20 mg/dL, leukopenia, thrombocytopenia, hypothermia, and hypotension.6 These factors are associated with increased mortality due to CAP, and ICU admission is indicated if 3 of the minor criteria for severe CAP are present.

        Another clinical calculator that can be used for assessing severity of CAP is SMART-COP (systolic blood pressure, multilobar chest radiography involvement, albumin level, respiratory rate, tachycardia, confusion, oxygenation and arterial pH).7 This scoring system uses 8 weighted criteria to predict which patients will require intensive respiratory or vasopressor support. SMART-COP has a sensitivity of 79% and a specificity of 64% in predicting ICU admission, whereas CURB-65 has a pooled sensitivity of 57.2% and specificity of 77.2%.8

         

         

        Antibiotic Therapy

        Antibiotics are the mainstay of treatment for CAP, with the majority of patients with CAP treated empirically taking into account the site of care, likely pathogen, and antimicrobial resistance issues. Patients with pneumonia who are treated as outpatients usually respond well to empiric antibiotic treatment, and a causative pathogen is not usually sought. Patients who are hospitalized for treatment of CAP usually receive empiric antibiotic on admission. Once the etiology has been determined by microbiologic or serologic means, antimicrobial therapy should be adjusted accordingly. A CDC study found that the burden of viral etiologies was higher than previously thought, with rhinovirus and influenza accounting for 15% of cases and Streptococcus pneumoniae for only 5%.9 This study highlighted the fact that despite advances in molecular techniques, no pathogen is identified for most patients with pneumonia.9 Given the lack of discernable pathogens in the majority of cases, patients should continue to be treated with antibiotics unless a nonbacterial etiology is found.

        Outpatients without comorbidities or risk factors for drug-resistant S. pneumoniae (Table 2)10 can be treated with monotherapy. Hospitalized patients are usually treated with combination intravenous therapy, although non-ICU patients who receive a respiratory fluoroquinolone can be treated orally.

        PDBR01609002_t2.PNG

        As previously mentioned, antibiotic therapy is typically empiric, since neither clinical features nor radiographic features are sufficient to include or exclude infectious etiologies. Epidemiologic risk factors should be considered and, in certain cases, antimicrobial coverage should be expanded to include those entities; for example, treatment of anaerobes in the setting of lung abscess and antipseudomonal antibiotics for patients with bronchiectasis.

        Of concern in the treatment of CAP is the increased prevalence of antimicrobial resistance among S. pneumoniae. The IDSA guidelines report that drug-resistant S. pneumoniae is more common in persons aged < 2 or > 65 years, and those with β-lactam therapy within the previous 3 months, alcoholism, medical comorbidities, immunosuppressive illness or therapy, or exposure to a child who attends a day care center.6

        Staphylococcus aureus should be considered during influenza outbreaks, with either vancomycin or linezolid being the recommended agents in the setting of methicillin-resistant S. aureus (MRSA). In a study comparing vancomycin versus linezolid for nosocomial pneumonia, the all-cause 60-day mortality was similar for both agents.11 Daptomycin, another agent used against MRSA, is not indicated in the setting of pneumonia because daptomycin binds to surfactant, yielding it ineffective in the treatment of pneumonia.12 Ceftaroline is a newer cephalosporin with activity against MRSA; its role in treatment of community-acquired MRSA pneumonia has not been fully elucidated, but it appears to be a useful agent for this indication.13,14 Similarly, other agents known to have antibacterial properties against MRSA, such as trimethoprim/sulfamethoxazole and doxycycline, have not been studied for this indication. Clindamycin has been used to treat MRSA in children, and IDSA guidelines on the treatment of MRSA list clindamycin as an alternative15 if MRSA is known to be sensitive.

        A summary of recommended empiric antibiotic therapy is presented in Table 3.16

        PDBR01609002_t3.PNG

        Three antibiotics were approved by the US Food and Drug Administration (FDA) for the treatment of CAP after the release of the IDSA/ATS guidelines in 2007. Ceftaroline fosamil is a fifth-generation cephalosporin that has coverage for MRSA and was approved in November 2010.17 It can only be administered intravenously and needs dose adjustment for renal function. Omadacycline is a new tetracycline that was approved by the FDA in October 2018.18 It is available in both intravenous injectable and oral forms. No dose adjustment is needed for renal function. Lefamulin is a first-in-class novel pleuromutilin antibiotic which was FDA-approved in August 2019. It can be administered intravenously or orally, with no dosage adjustment necessary in patients with renal impairment.19

         

         

        Antibiotic Therapy for Selected Pathogens

        Streptococcus pneumoniae

        Patients with pneumococcal pneumonia who have penicillin-susceptible strains can be treated with intravenous penicillin (2 or 3 million units every 4 hours) or ceftriaxone. Once a patient meets criteria of stability, they can then be transitioned to oral penicillin, amoxicillin, or clarithromycin. Those with strains with reduced susceptibility can still be treated with penicillin, but at a higher dose (4 million units intravenously [IV] every 4 hours), or a third-generation cephalosporin. Those whose pneumococcal pneumonia is complicated by bacteremia will benefit from dual therapy if severely ill, requiring ICU monitoring. Those not severely ill can be treated with monotherapy.20

        Staphylococcus aureus

        Staphylococcus aureus is more commonly associated with hospital-acquired pneumonia, but it may also be seen during the influenza season and in those with severe necrotizing CAP. Both linezolid and vancomycin can be used to treat MRSA CAP. As noted, ceftaroline has activity against MRSA and is approved for treatment of CAP, but is not approved by the FDA for MRSA CAP treatment. Similarly, tigecycline is approved for CAP and has activity against MRSA, but is not approved for MRSA CAP. Moreover, the FDA has warned of increased risk of death with tigecycline and has a black box warning to that effect.21

        Legionella

        Legionellosis can be treated with tetra¬cyclines, macrolides, or fluoroquinolones. For non-immunocompromised patients with mild pneumonia, any of the listed antibiotics is considered appropriate. However, patients with severe infection or those with immunosuppression should be treated with either levofloxacin or azithromycin for 7 to 10 days.22

        Chlamydophila pneumoniae

        As with other atypical organisms, Chlamydophila pneumoniae can be treated with doxycycline, a macrolide, or respiratory fluoroquinolones. However, length of therapy varies by regimen used; treating with doxycycline 100 mg twice daily generally requires 14 to 21 days, whereas moxifloxacin 400 mg daily requires 10 days.23

        Mycoplasma pneumoniae

        As with C. pneumoniae, length of therapy of Mycoplasma pneumoniae varies by which antimicrobial regimen is used. Shortest courses are seen with the use of macrolides for 5 days, whereas 14 days is considered standard for doxycycline or a respiratory fluoroquinolone.24 It should be noted that there has been increasingly documented resistance to macrolides, with known resistance of 8.2% in the United States.25

         

         

        Duration of Treatment

        Most patients with CAP respond to appropriate therapy within 72 hours. IDSA/ATS guidelines recommend that patients with routine cases of CAP be treated for a minimum of 5 days. Despite this, many patients are treated for an excessive amount of time, with over 70% of patients reported to have received antibiotics for more than 10 days for uncomplicated CAP.26 There are instances that require longer courses of antibiotics, including cases caused by Pseudomonas aeruginosa, S. aureus, and Legionella species and patients with lung abscesses or necrotizing infections, among others.27

        Hospitalized patients do not need to be monitored for an additional day once they have reached clinical stability (Table 4), are able to maintain oral intake, and have normal mentation, provided that other comorbidities are stable and social needs have been met.6 C-reactive protein (CRP) level has been postulated as an additional measure of stability, specifically monitoring for a greater than 50% reduction in CRP; however, this was validated only for those with complicated pneumonia.28 Patients discharged from the hospital with instability have higher risk of readmission or death.29

        PDBR01609002_t4.PNG

        Transition to Oral Therapy

        IDSA/ATS guidelines6 recommend that patients should be transitioned from intravenous to oral antibiotics when they are improving clinically, have stable vital signs, and are able to ingest food/fluids and medications.

        Management of Nonresponders

        Although the majority of patients respond to antibiotics within 72 hours, treatment failure occurs in up to 15% of patients.15 Nonresponding pneumonia is generally seen in 2 patterns: worsening of clinical status despite empiric antibiotics or delay in achieving clinical stability, as defined in Table 4, after 72 hours of treatment.30 Risk factors associated with nonresponding pneumonia31 are:

        • Radiographic: multilobar infiltrates, pleural effusion, cavitation
        • Bacteriologic: MRSA, gram-negative or Legionella pneumonia
        • Severity index: PSI > 90
        • Pharmacologic: incorrect antibiotic choice based on susceptibility

        Patients with acute deterioration of clinical status require prompt transfer to a higher level of care and may require mechanical ventilator support. In those with delay in achieving clinical stability, a question centers on whether the same antibiotics can be continued while doing further radiographic/microbiologic work-up and/or changing antibiotics. History should be reviewed, with particular attention to exposures, travel history, and microbiologic and radiographic data. Clinicians should recall that viruses account for up to 20% of pneumonias and that there are also noninfectious causes that can mimic pyogenic infections.32 If adequate initial cultures were not obtained, they should be obtained; however, care must be taken in reviewing new sets of cultures while on antibiotics, as they may reveal colonization selected out by antibiotics and not a true pathogen. If repeat evaluation is unrevealing, then further evaluation with computed tomography (CT) scan and bronchoscopy with bronchoalveolar lavage and biopsy is warranted. CT scans can show pleural effusions, bronchial obstructions, or a pattern suggestive of cryptogenic pneumonia. A bronchoscopy might yield a microbiologic diagnosis and, when combined with biopsy, can also evaluate for noninfectious causes.

        As with other infections, if escalation of antibiotics is undertaken, clinicians should try to determine the reason for nonresponse. To simply broaden antimicrobial therapy without attempts at establishing a microbiologic or radiographic cause for nonresponse may lead to inappropriate treatment and recurrence of infection. Aside from patients who have bacteremic pneumococcal pneumonia in an ICU setting, there are no published reports pointing to superiority of combination antibiotics.20

         

         

        Other Treatment

        Several agents have been evaluated as adjunctive treatment of pneumonia to decrease the inflammatory response associated with pneumonia; namely, steroids, macrolide antibiotics, and statins. To date, only the use of steroids (methylprednisolone 0.5 mg/kg every 12 hours for 5 days) in those with severe CAP and high initial anti-inflammatory response (CRP > 150) has been shown to decrease treatment failure, decrease risk of acute respiratory distress syndrome, and possibly reduce length of stay and duration of intravenous antibiotics, without effect on mortality or adverse side effects.33,34 However, a recent double-blind randomized study conducted in Australia in which patients admitted with CAP were prescribed prednisolone acetate (50 mg/day for 7 days) and de-escalated from parenteral to oral antibiotics according to standardized criteria revealed no difference in mortality, length of stay, or readmission rates between the corticosteroids group and the control group at 90-day follow-up.35 At this point, corticosteroid as an adjunctive treatment for CAP is still controversial and the new 2019 ATS/IDSA guidelines recommend not routinely using corticosteroids in all patients with CAP.36 Other adjunctive methods have not been found to have significant impact.6

        Prevention of Pneumonia

        Prevention of pneumococcal pneumonia involves vaccinations to prevent infection caused by S. pneumoniae and influenza viruses. As influenza is a risk factor for bacterial infection, specifically with S. pneumoniae, influenza vaccination can help prevent bacterial pneumonia.37 In their most recent recommendations, the CDC continues to recommend routine influenza vaccination for all persons older than age 6 months, unless otherwise contraindicated.38

        There are 2 vaccines for prevention of pneumococcal disease: the pneumococcal polysaccharide vaccine (PPSV23) and a conjugate vaccine (PCV13). Following vaccination with PPSV23, 80% of adults develop antibodies against at least 18 of the 23 serotypes.39 PPSV23 is reported to be protective against invasive pneumococcal infection, although there is no consensus regarding whether PPSV23 leads to decreased rates of pneumonia.40 On the other hand, PCV13 vaccination was associated with prevention of both invasive disease and CAP in adults aged 65 years or older.41 The CDC recommends that all children aged 2 years or younger receive PCV13, and those aged 65 or older receive PCV13 followed by a dose of PPSV23.42,43 The dose of PPSV23 should be given at least 1 year after the dose of PCV13 is administered.44 Persons younger than 65 years with immunocompromising and certain other conditions should also receive vaccination (Table 5).44 Full recommendations, many scenarios, and details on timing of vaccinations can be found at the CDC’s website.

        PDBR01609002_t5.PNG

        Cigarette smoking increases the risk of respiratory infections, as evidenced by smokers accounting for almost half of all patients with invasive pneumococcal disease.11 As this is a modifiable risk factor, smoking cessation should be part of a comprehensive approach toward prevention of pneumonia.

         

         

        Summary

        Most patients with CAP are treated empirically with antibiotics, with therapy selection based on the site of care, likely pathogen, and antimicrobial resistance issues. Those treated as outpatients usually respond well to empiric antibiotic treatment, and a causative pathogen is not usually sought. Patients who are hospitalized for treatment usually receive empiric antibiotic on admission, and antimicrobial therapy is adjusted accordingly once the etiology has been determined by microbiologic or serologic means. At this time, the use of corticosteroid as an adjunctive treatment for CAP is still controversial, so not all patients with CAP should routinely receive corticosteroids. Because vaccination (PPSV23, PCV13, and influenza vaccine) remains the most effective tool in preventing the development of CAP, clinicians should strive for 100% vaccination rates in persons without contraindications.

        References

        1. Fine MJ, Auble TE, Yealy DM, et al A prediction rule to identify low-risk patients with community-acquired pneumonia. N Engl J Med.1997;336:243-250.

        2. Lim WS, van der Eerden MM, Laing R, et al. Defining community acquired pneumonia severity on presentation to hospital: an international derivation and validation study. Thorax. 2003;58:377-382.

        3. Aujesky D, Auble TE, Yealy DM, et al. Prospective comparison of three validated prediction rules for prognosis in community-acquired pneumonia. Am J Med. 2005;118:384-392.

        4. Arnold FW, Ramirez JA, McDonald LC, Xia EL. Hospitalization for community-acquired pneumonia: the pneumonia severity index vs clinical judgment. Chest. 2003;124:121-124.

        5. Aujesky D, McCausland JB, Whittle J, et al. Reasons why emergency department providers do not rely on the pneumonia severity index to determine the initial site of treatment for patients with pneumonia. Clin Infect Dis. 2009;49:e100-108.

        6. Mandell LA, Wunderink RG, Anzueto A, et al. 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:S27-72.

        7. Charles PG, Wolfe R, Whitby M, et al. SMART-COP: a tool for predicting the need for intensive respiratory or vasopressor support in community-acquired pneumonia. Clin Infect Dis. 2008;47:375-384.

        8. Marti C, Garin N, Grosgurin O, et al. Prediction of severe community-acquired pneumonia: a systematic review and meta-analysis. Crit Care. 2012;16:R141.

        9. Jain S, Self WH, Wunderink RG, et al. Community-acquired pneumonia requiring hospitalization among U.S. adults. N Engl J Med. 2015;373:415-427.

        10. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63:e61-e111.

        11. Wunderink RG, Niederman MS, Kollef MH, et al. Linezolid in methicillin-resistant Staphylococcus aureus nosocomial pneumonia: a randomized, controlled study. Clin Infect Dis. 2012;54:621-629.

        12. Silverman JA, Mortin LI, Vanpraagh AD, et al. Inhibition of daptomycin by pulmonary surfactant: in vitro modeling and clinical impact. J Infect Dis. 2005;191:2149-2152.

        13. El Hajj MS, Turgeon RD, Wilby KJ. Ceftaroline fosamil for community-acquired pneumonia and skin and skin structure infections: a systematic review. Int J Clin Pharm. 2017;39:26-32.

        14. Taboada M, Melnick D, Iaconis JP, et al. Ceftaroline fosamil versus ceftriaxone for the treatment of community-acquired pneumonia: individual patient data meta-analysis of randomized controlled trials. J Antimicrob Chemother. 2016;71:862-870.

        15. Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children: executive summary. Clin Infect Dis. 2011;52:285-292.

        16. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Sauders; 2015:2310-2327.

        17. Teflaro (ceftaroline fosamil) [package insert]. St. Louis, MO: Forest Pharmaceuticals; 2010.

        18. Nuzyra (omadacycline) [package insert]. Boston, MA: Paratek Pharmaceuticals; 2018.

        19. Xenleta (lefamulin) [package insert]. Dublin, Ireland: Nabriva Therapeutics; 2019.

        20. Baddour LM, Yu VL, Klugman KP, et al. Combination antibiotic therapy lowers mortality among severely ill patients with pneumococcal bacteremia. Am J Respir Crit Care Med. 2004;170:440-444.

        21. FDA Drug Safety Communication: FDA warns of increased risk of death with IV antibacterial Tygacil (tigecycline) and approves new boxed warning. www.fda.gov/Drugs/DrugSafety/ucm369580.htm. Accessed 16 September 2019.

        22. Edelstein PR, CR. Legionnaires’ disease and Pontiac fever. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2633.

        23. Hammerschlag MR, Kohlhoff SA, Gaydos, CA. Chlamydia pneumoniae. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2174.

        24. Holzman RS, MS. Mycoplasma pneumoniae and atypical pneumonia. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2183.

        25. Yamada M, Buller R, Bledsoe S, Storch GA. Rising rates of macrolide-resistant Mycoplasma pneumoniae in the central United States. Pediatr Infect Dis J. 2012;31:409-410.

        26. Yi SH, Hatfield KM, Baggs J, et al. Duration of antibiotic use among adults with uncomplicated community-acquired pneumonia requiring hospitalization in the United States. Clin Infect Dis. 2018;66:1333-1341.

        27. Hayashi Y, Paterson DL. Strategies for reduction in duration of antibiotic use in hospitalized patients. Clin Infect Dis. 2011;52:1232-1240.

        28. Akram AR, Chalmers JD, Taylor JK, et al. An evaluation of clinical stability criteria to predict hospital course in community-acquired pneumonia. Clin Microbiol Infect. 2013;19:1174-1180.

        29. Halm EA, Fine MJ, Kapoor WN, et al. Instability on hospital discharge and the risk of adverse outcomes in patients with pneumonia. Arch Intern Med. 2002;162:1278-1284.

        30. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Saunders; 2015:2310-2327.

        31. Roson B, Carratala J, Fernandez-Sabe N, et al. Causes and factors associated with early failure in hospitalized patients with community-acquired pneumonia. Arch Intern Med. 2004;164:502-508.

        32. El-Solh AA, Pietrantoni C, Bhat A, et al. Microbiology of severe aspiration pneumonia in institutionalized elderly. Am J Respir Crit Care Med. 2003;167:1650-1654.

        33. Wan YD, Sun TW, Liu ZQ, et al. Efficacy and safety of corticosteroids for community-acquired pneumonia: a systematic review and meta-analysis. Chest. 2016;149:209-219.

        34. Torres A, Sibila O, Ferrer M, et al. Effect of corticosteroids on treatment failure among hospitalized patients with severe community-acquired pneumonia and high inflammatory response: a randomized clinical trial. JAMA. 2015;313:677-686.

        35. Lloyd M, Karahalios, Janus E, et al. Effectiveness of a bundled intervention including adjunctive corticosteroids on outcomes of hospitalized patients with community-acquired pneumonia: a stepped-wedge randomized clinical trial. JAMA Intern Med. 2019;179:1052-1060.

        36. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200:e45-e67.

        37. McCullers JA. Insights into the interaction between influenza virus and pneumococcus. Clin Microbiol Rev. 2006;19:571-582.

        38. Grohskopf LA, Alyanak E, Broder KR, et al. Prevention and control of seasonal influenza with vaccines: recommendations of the advisory committee on immunization practices - United States, 2019-20 influenza season. MMWR Recomm Rep. 2019;68:1-21.

        39. Rubins JB, Alter M, Loch J, Janoff EN. Determination of antibody responses of elderly adults to all 23 capsular polysaccharides after pneumococcal vaccination. Infect Immun. 1999;67:5979-5984.

        40. Vaccines and preventable diseases. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/vpd/pneumo/hcp/about-vaccine.html. Accessed 16 September 2019.

        41. Bonten MJ, Huijts SM, Bolkenbaas M, et al. Polysaccharide conjugate vaccine against pneumococcal pneumonia in adults. N Engl J Med. 2015;372:1114-1125.

        42. Recommended adult immunization schedule -- United States -- 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/schedules/downloads/adult/adult-combined-schedule.pdf. Accessed 16 September 2019.

        43. Recommended child and adolescent immunization schedule for ages 18 years or younger – United States – 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/schedules/hcp/imz/child-adolescent.html. Accessed 22 September 2019.

        44. Pneumococcal vaccine timing for adults – United States – 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/vpd/pneumo/downloads/pneumo-vaccine-timing.pdf. Accessed 22 September 2019.

        Author and Disclosure Information

        University of North Dakota School of Medicine & Health Sciences, Fargo, ND

        Publications
        Topics
        Sections
        Author and Disclosure Information

        University of North Dakota School of Medicine & Health Sciences, Fargo, ND

        Author and Disclosure Information

        University of North Dakota School of Medicine & Health Sciences, Fargo, ND

        Initial management decisions for patients with community-acquired pneumonia (CAP) will depend on severity of infection, with need for hospitalization being one of the first decisions. Because empiric antibiotics are the mainstay of treatment and the causative organisms are seldom identified, underlying medical conditions and epidemiologic risk factors are considered when selecting an empiric regimen. As with other infections, duration of therapy is not standardized, but rather is guided by clinical improvement. Prevention of pneumonia centers around vaccination and smoking cessation. This article, the second in a 2-part review of CAP in adults, focuses on site of care decision, empiric and directed therapies, length of treatment, and prevention strategies. Evaluation and diagnosis of CAP are discussed in a separate article.

        Site of Care Decision

        For patients diagnosed with CAP, the clinician must decide whether treatment will be done in an outpatient or inpatient setting, and for those in the inpatient setting, whether they can safely be treated on the general medical ward or in the intensive care unit (ICU). Two common scoring systems that can be used to aid the clinician in determining severity of the infection and guide site-of-care decisions are the Pneumonia Severity Index (PSI) and CURB-65 scores.

        The PSI score uses 20 different parameters, including comorbidities, laboratory parameters, and radiographic findings, to stratify patients into 5 mortality risk classes.1 On the basis of associated mortality rates, it has been suggested that risk class I and II patients should be treated as outpatients, risk class III patients should be treated in an observation unit or with a short hospitalization, and risk class IV and V patients should be treated as inpatients.1

        The CURB-65 method of risk stratification is based on 5 clinical parameters: confusion, urea level, respiratory rate, systolic blood pressure, and age ≥ 65 years (Table 1).2,3 A modification to the CURB-65 algorithm tool was CRB-65, which excludes urea nitrogen, making it optimal for making determinations in a clinic-based setting. It should be emphasized that these tools do not take into account other factors that should be used in determining location of treatment, such as stable home, mental illness, or concerns about compliance with medications. In many instances, it is these factors that preclude low-risk patients from being treated as outpatients.4,5 Similarly, these scoring systems have not been validated for immunocompromised patients or those who would qualify as having health care–associated pneumonia.

        PDBR01609002_t1.PNG

        Patients with CURB-65 scores of 4 or 5 are considered to have severe pneumonia, and admission to the ICU should be considered for these patients. Aside from the CURB-65 score, anyone requiring vasopressor support or mechanical ventilation merits admission to the ICU.6 American Thoracic Society (ATS) and Infectious Diseases Society of America (IDSA) guidelines also recommend the use of “minor criteria” for making ICU admission decisions; these include respiratory rate ≥ 30 breaths/minute, PaO2 fraction ≤ 250 mm Hg, multilobar infiltrates, confusion, blood urea nitrogen ≥ 20 mg/dL, leukopenia, thrombocytopenia, hypothermia, and hypotension.6 These factors are associated with increased mortality due to CAP, and ICU admission is indicated if 3 of the minor criteria for severe CAP are present.

        Another clinical calculator that can be used for assessing severity of CAP is SMART-COP (systolic blood pressure, multilobar chest radiography involvement, albumin level, respiratory rate, tachycardia, confusion, oxygenation and arterial pH).7 This scoring system uses 8 weighted criteria to predict which patients will require intensive respiratory or vasopressor support. SMART-COP has a sensitivity of 79% and a specificity of 64% in predicting ICU admission, whereas CURB-65 has a pooled sensitivity of 57.2% and specificity of 77.2%.8

         

         

        Antibiotic Therapy

        Antibiotics are the mainstay of treatment for CAP, with the majority of patients with CAP treated empirically taking into account the site of care, likely pathogen, and antimicrobial resistance issues. Patients with pneumonia who are treated as outpatients usually respond well to empiric antibiotic treatment, and a causative pathogen is not usually sought. Patients who are hospitalized for treatment of CAP usually receive empiric antibiotic on admission. Once the etiology has been determined by microbiologic or serologic means, antimicrobial therapy should be adjusted accordingly. A CDC study found that the burden of viral etiologies was higher than previously thought, with rhinovirus and influenza accounting for 15% of cases and Streptococcus pneumoniae for only 5%.9 This study highlighted the fact that despite advances in molecular techniques, no pathogen is identified for most patients with pneumonia.9 Given the lack of discernable pathogens in the majority of cases, patients should continue to be treated with antibiotics unless a nonbacterial etiology is found.

        Outpatients without comorbidities or risk factors for drug-resistant S. pneumoniae (Table 2)10 can be treated with monotherapy. Hospitalized patients are usually treated with combination intravenous therapy, although non-ICU patients who receive a respiratory fluoroquinolone can be treated orally.

        PDBR01609002_t2.PNG

        As previously mentioned, antibiotic therapy is typically empiric, since neither clinical features nor radiographic features are sufficient to include or exclude infectious etiologies. Epidemiologic risk factors should be considered and, in certain cases, antimicrobial coverage should be expanded to include those entities; for example, treatment of anaerobes in the setting of lung abscess and antipseudomonal antibiotics for patients with bronchiectasis.

        Of concern in the treatment of CAP is the increased prevalence of antimicrobial resistance among S. pneumoniae. The IDSA guidelines report that drug-resistant S. pneumoniae is more common in persons aged < 2 or > 65 years, and those with β-lactam therapy within the previous 3 months, alcoholism, medical comorbidities, immunosuppressive illness or therapy, or exposure to a child who attends a day care center.6

        Staphylococcus aureus should be considered during influenza outbreaks, with either vancomycin or linezolid being the recommended agents in the setting of methicillin-resistant S. aureus (MRSA). In a study comparing vancomycin versus linezolid for nosocomial pneumonia, the all-cause 60-day mortality was similar for both agents.11 Daptomycin, another agent used against MRSA, is not indicated in the setting of pneumonia because daptomycin binds to surfactant, yielding it ineffective in the treatment of pneumonia.12 Ceftaroline is a newer cephalosporin with activity against MRSA; its role in treatment of community-acquired MRSA pneumonia has not been fully elucidated, but it appears to be a useful agent for this indication.13,14 Similarly, other agents known to have antibacterial properties against MRSA, such as trimethoprim/sulfamethoxazole and doxycycline, have not been studied for this indication. Clindamycin has been used to treat MRSA in children, and IDSA guidelines on the treatment of MRSA list clindamycin as an alternative15 if MRSA is known to be sensitive.

        A summary of recommended empiric antibiotic therapy is presented in Table 3.16

        PDBR01609002_t3.PNG

        Three antibiotics were approved by the US Food and Drug Administration (FDA) for the treatment of CAP after the release of the IDSA/ATS guidelines in 2007. Ceftaroline fosamil is a fifth-generation cephalosporin that has coverage for MRSA and was approved in November 2010.17 It can only be administered intravenously and needs dose adjustment for renal function. Omadacycline is a new tetracycline that was approved by the FDA in October 2018.18 It is available in both intravenous injectable and oral forms. No dose adjustment is needed for renal function. Lefamulin is a first-in-class novel pleuromutilin antibiotic which was FDA-approved in August 2019. It can be administered intravenously or orally, with no dosage adjustment necessary in patients with renal impairment.19

         

         

        Antibiotic Therapy for Selected Pathogens

        Streptococcus pneumoniae

        Patients with pneumococcal pneumonia who have penicillin-susceptible strains can be treated with intravenous penicillin (2 or 3 million units every 4 hours) or ceftriaxone. Once a patient meets criteria of stability, they can then be transitioned to oral penicillin, amoxicillin, or clarithromycin. Those with strains with reduced susceptibility can still be treated with penicillin, but at a higher dose (4 million units intravenously [IV] every 4 hours), or a third-generation cephalosporin. Those whose pneumococcal pneumonia is complicated by bacteremia will benefit from dual therapy if severely ill, requiring ICU monitoring. Those not severely ill can be treated with monotherapy.20

        Staphylococcus aureus

        Staphylococcus aureus is more commonly associated with hospital-acquired pneumonia, but it may also be seen during the influenza season and in those with severe necrotizing CAP. Both linezolid and vancomycin can be used to treat MRSA CAP. As noted, ceftaroline has activity against MRSA and is approved for treatment of CAP, but is not approved by the FDA for MRSA CAP treatment. Similarly, tigecycline is approved for CAP and has activity against MRSA, but is not approved for MRSA CAP. Moreover, the FDA has warned of increased risk of death with tigecycline and has a black box warning to that effect.21

        Legionella

        Legionellosis can be treated with tetra¬cyclines, macrolides, or fluoroquinolones. For non-immunocompromised patients with mild pneumonia, any of the listed antibiotics is considered appropriate. However, patients with severe infection or those with immunosuppression should be treated with either levofloxacin or azithromycin for 7 to 10 days.22

        Chlamydophila pneumoniae

        As with other atypical organisms, Chlamydophila pneumoniae can be treated with doxycycline, a macrolide, or respiratory fluoroquinolones. However, length of therapy varies by regimen used; treating with doxycycline 100 mg twice daily generally requires 14 to 21 days, whereas moxifloxacin 400 mg daily requires 10 days.23

        Mycoplasma pneumoniae

        As with C. pneumoniae, length of therapy of Mycoplasma pneumoniae varies by which antimicrobial regimen is used. Shortest courses are seen with the use of macrolides for 5 days, whereas 14 days is considered standard for doxycycline or a respiratory fluoroquinolone.24 It should be noted that there has been increasingly documented resistance to macrolides, with known resistance of 8.2% in the United States.25

         

         

        Duration of Treatment

        Most patients with CAP respond to appropriate therapy within 72 hours. IDSA/ATS guidelines recommend that patients with routine cases of CAP be treated for a minimum of 5 days. Despite this, many patients are treated for an excessive amount of time, with over 70% of patients reported to have received antibiotics for more than 10 days for uncomplicated CAP.26 There are instances that require longer courses of antibiotics, including cases caused by Pseudomonas aeruginosa, S. aureus, and Legionella species and patients with lung abscesses or necrotizing infections, among others.27

        Hospitalized patients do not need to be monitored for an additional day once they have reached clinical stability (Table 4), are able to maintain oral intake, and have normal mentation, provided that other comorbidities are stable and social needs have been met.6 C-reactive protein (CRP) level has been postulated as an additional measure of stability, specifically monitoring for a greater than 50% reduction in CRP; however, this was validated only for those with complicated pneumonia.28 Patients discharged from the hospital with instability have higher risk of readmission or death.29

        PDBR01609002_t4.PNG

        Transition to Oral Therapy

        IDSA/ATS guidelines6 recommend that patients should be transitioned from intravenous to oral antibiotics when they are improving clinically, have stable vital signs, and are able to ingest food/fluids and medications.

        Management of Nonresponders

        Although the majority of patients respond to antibiotics within 72 hours, treatment failure occurs in up to 15% of patients.15 Nonresponding pneumonia is generally seen in 2 patterns: worsening of clinical status despite empiric antibiotics or delay in achieving clinical stability, as defined in Table 4, after 72 hours of treatment.30 Risk factors associated with nonresponding pneumonia31 are:

        • Radiographic: multilobar infiltrates, pleural effusion, cavitation
        • Bacteriologic: MRSA, gram-negative or Legionella pneumonia
        • Severity index: PSI > 90
        • Pharmacologic: incorrect antibiotic choice based on susceptibility

        Patients with acute deterioration of clinical status require prompt transfer to a higher level of care and may require mechanical ventilator support. In those with delay in achieving clinical stability, a question centers on whether the same antibiotics can be continued while doing further radiographic/microbiologic work-up and/or changing antibiotics. History should be reviewed, with particular attention to exposures, travel history, and microbiologic and radiographic data. Clinicians should recall that viruses account for up to 20% of pneumonias and that there are also noninfectious causes that can mimic pyogenic infections.32 If adequate initial cultures were not obtained, they should be obtained; however, care must be taken in reviewing new sets of cultures while on antibiotics, as they may reveal colonization selected out by antibiotics and not a true pathogen. If repeat evaluation is unrevealing, then further evaluation with computed tomography (CT) scan and bronchoscopy with bronchoalveolar lavage and biopsy is warranted. CT scans can show pleural effusions, bronchial obstructions, or a pattern suggestive of cryptogenic pneumonia. A bronchoscopy might yield a microbiologic diagnosis and, when combined with biopsy, can also evaluate for noninfectious causes.

        As with other infections, if escalation of antibiotics is undertaken, clinicians should try to determine the reason for nonresponse. To simply broaden antimicrobial therapy without attempts at establishing a microbiologic or radiographic cause for nonresponse may lead to inappropriate treatment and recurrence of infection. Aside from patients who have bacteremic pneumococcal pneumonia in an ICU setting, there are no published reports pointing to superiority of combination antibiotics.20

         

         

        Other Treatment

        Several agents have been evaluated as adjunctive treatment of pneumonia to decrease the inflammatory response associated with pneumonia; namely, steroids, macrolide antibiotics, and statins. To date, only the use of steroids (methylprednisolone 0.5 mg/kg every 12 hours for 5 days) in those with severe CAP and high initial anti-inflammatory response (CRP > 150) has been shown to decrease treatment failure, decrease risk of acute respiratory distress syndrome, and possibly reduce length of stay and duration of intravenous antibiotics, without effect on mortality or adverse side effects.33,34 However, a recent double-blind randomized study conducted in Australia in which patients admitted with CAP were prescribed prednisolone acetate (50 mg/day for 7 days) and de-escalated from parenteral to oral antibiotics according to standardized criteria revealed no difference in mortality, length of stay, or readmission rates between the corticosteroids group and the control group at 90-day follow-up.35 At this point, corticosteroid as an adjunctive treatment for CAP is still controversial and the new 2019 ATS/IDSA guidelines recommend not routinely using corticosteroids in all patients with CAP.36 Other adjunctive methods have not been found to have significant impact.6

        Prevention of Pneumonia

        Prevention of pneumococcal pneumonia involves vaccinations to prevent infection caused by S. pneumoniae and influenza viruses. As influenza is a risk factor for bacterial infection, specifically with S. pneumoniae, influenza vaccination can help prevent bacterial pneumonia.37 In their most recent recommendations, the CDC continues to recommend routine influenza vaccination for all persons older than age 6 months, unless otherwise contraindicated.38

        There are 2 vaccines for prevention of pneumococcal disease: the pneumococcal polysaccharide vaccine (PPSV23) and a conjugate vaccine (PCV13). Following vaccination with PPSV23, 80% of adults develop antibodies against at least 18 of the 23 serotypes.39 PPSV23 is reported to be protective against invasive pneumococcal infection, although there is no consensus regarding whether PPSV23 leads to decreased rates of pneumonia.40 On the other hand, PCV13 vaccination was associated with prevention of both invasive disease and CAP in adults aged 65 years or older.41 The CDC recommends that all children aged 2 years or younger receive PCV13, and those aged 65 or older receive PCV13 followed by a dose of PPSV23.42,43 The dose of PPSV23 should be given at least 1 year after the dose of PCV13 is administered.44 Persons younger than 65 years with immunocompromising and certain other conditions should also receive vaccination (Table 5).44 Full recommendations, many scenarios, and details on timing of vaccinations can be found at the CDC’s website.

        PDBR01609002_t5.PNG

        Cigarette smoking increases the risk of respiratory infections, as evidenced by smokers accounting for almost half of all patients with invasive pneumococcal disease.11 As this is a modifiable risk factor, smoking cessation should be part of a comprehensive approach toward prevention of pneumonia.

         

         

        Summary

        Most patients with CAP are treated empirically with antibiotics, with therapy selection based on the site of care, likely pathogen, and antimicrobial resistance issues. Those treated as outpatients usually respond well to empiric antibiotic treatment, and a causative pathogen is not usually sought. Patients who are hospitalized for treatment usually receive empiric antibiotic on admission, and antimicrobial therapy is adjusted accordingly once the etiology has been determined by microbiologic or serologic means. At this time, the use of corticosteroid as an adjunctive treatment for CAP is still controversial, so not all patients with CAP should routinely receive corticosteroids. Because vaccination (PPSV23, PCV13, and influenza vaccine) remains the most effective tool in preventing the development of CAP, clinicians should strive for 100% vaccination rates in persons without contraindications.

        Initial management decisions for patients with community-acquired pneumonia (CAP) will depend on severity of infection, with need for hospitalization being one of the first decisions. Because empiric antibiotics are the mainstay of treatment and the causative organisms are seldom identified, underlying medical conditions and epidemiologic risk factors are considered when selecting an empiric regimen. As with other infections, duration of therapy is not standardized, but rather is guided by clinical improvement. Prevention of pneumonia centers around vaccination and smoking cessation. This article, the second in a 2-part review of CAP in adults, focuses on site of care decision, empiric and directed therapies, length of treatment, and prevention strategies. Evaluation and diagnosis of CAP are discussed in a separate article.

        Site of Care Decision

        For patients diagnosed with CAP, the clinician must decide whether treatment will be done in an outpatient or inpatient setting, and for those in the inpatient setting, whether they can safely be treated on the general medical ward or in the intensive care unit (ICU). Two common scoring systems that can be used to aid the clinician in determining severity of the infection and guide site-of-care decisions are the Pneumonia Severity Index (PSI) and CURB-65 scores.

        The PSI score uses 20 different parameters, including comorbidities, laboratory parameters, and radiographic findings, to stratify patients into 5 mortality risk classes.1 On the basis of associated mortality rates, it has been suggested that risk class I and II patients should be treated as outpatients, risk class III patients should be treated in an observation unit or with a short hospitalization, and risk class IV and V patients should be treated as inpatients.1

        The CURB-65 method of risk stratification is based on 5 clinical parameters: confusion, urea level, respiratory rate, systolic blood pressure, and age ≥ 65 years (Table 1).2,3 A modification to the CURB-65 algorithm tool was CRB-65, which excludes urea nitrogen, making it optimal for making determinations in a clinic-based setting. It should be emphasized that these tools do not take into account other factors that should be used in determining location of treatment, such as stable home, mental illness, or concerns about compliance with medications. In many instances, it is these factors that preclude low-risk patients from being treated as outpatients.4,5 Similarly, these scoring systems have not been validated for immunocompromised patients or those who would qualify as having health care–associated pneumonia.

        PDBR01609002_t1.PNG

        Patients with CURB-65 scores of 4 or 5 are considered to have severe pneumonia, and admission to the ICU should be considered for these patients. Aside from the CURB-65 score, anyone requiring vasopressor support or mechanical ventilation merits admission to the ICU.6 American Thoracic Society (ATS) and Infectious Diseases Society of America (IDSA) guidelines also recommend the use of “minor criteria” for making ICU admission decisions; these include respiratory rate ≥ 30 breaths/minute, PaO2 fraction ≤ 250 mm Hg, multilobar infiltrates, confusion, blood urea nitrogen ≥ 20 mg/dL, leukopenia, thrombocytopenia, hypothermia, and hypotension.6 These factors are associated with increased mortality due to CAP, and ICU admission is indicated if 3 of the minor criteria for severe CAP are present.

        Another clinical calculator that can be used for assessing severity of CAP is SMART-COP (systolic blood pressure, multilobar chest radiography involvement, albumin level, respiratory rate, tachycardia, confusion, oxygenation and arterial pH).7 This scoring system uses 8 weighted criteria to predict which patients will require intensive respiratory or vasopressor support. SMART-COP has a sensitivity of 79% and a specificity of 64% in predicting ICU admission, whereas CURB-65 has a pooled sensitivity of 57.2% and specificity of 77.2%.8

         

         

        Antibiotic Therapy

        Antibiotics are the mainstay of treatment for CAP, with the majority of patients with CAP treated empirically taking into account the site of care, likely pathogen, and antimicrobial resistance issues. Patients with pneumonia who are treated as outpatients usually respond well to empiric antibiotic treatment, and a causative pathogen is not usually sought. Patients who are hospitalized for treatment of CAP usually receive empiric antibiotic on admission. Once the etiology has been determined by microbiologic or serologic means, antimicrobial therapy should be adjusted accordingly. A CDC study found that the burden of viral etiologies was higher than previously thought, with rhinovirus and influenza accounting for 15% of cases and Streptococcus pneumoniae for only 5%.9 This study highlighted the fact that despite advances in molecular techniques, no pathogen is identified for most patients with pneumonia.9 Given the lack of discernable pathogens in the majority of cases, patients should continue to be treated with antibiotics unless a nonbacterial etiology is found.

        Outpatients without comorbidities or risk factors for drug-resistant S. pneumoniae (Table 2)10 can be treated with monotherapy. Hospitalized patients are usually treated with combination intravenous therapy, although non-ICU patients who receive a respiratory fluoroquinolone can be treated orally.

        PDBR01609002_t2.PNG

        As previously mentioned, antibiotic therapy is typically empiric, since neither clinical features nor radiographic features are sufficient to include or exclude infectious etiologies. Epidemiologic risk factors should be considered and, in certain cases, antimicrobial coverage should be expanded to include those entities; for example, treatment of anaerobes in the setting of lung abscess and antipseudomonal antibiotics for patients with bronchiectasis.

        Of concern in the treatment of CAP is the increased prevalence of antimicrobial resistance among S. pneumoniae. The IDSA guidelines report that drug-resistant S. pneumoniae is more common in persons aged < 2 or > 65 years, and those with β-lactam therapy within the previous 3 months, alcoholism, medical comorbidities, immunosuppressive illness or therapy, or exposure to a child who attends a day care center.6

        Staphylococcus aureus should be considered during influenza outbreaks, with either vancomycin or linezolid being the recommended agents in the setting of methicillin-resistant S. aureus (MRSA). In a study comparing vancomycin versus linezolid for nosocomial pneumonia, the all-cause 60-day mortality was similar for both agents.11 Daptomycin, another agent used against MRSA, is not indicated in the setting of pneumonia because daptomycin binds to surfactant, yielding it ineffective in the treatment of pneumonia.12 Ceftaroline is a newer cephalosporin with activity against MRSA; its role in treatment of community-acquired MRSA pneumonia has not been fully elucidated, but it appears to be a useful agent for this indication.13,14 Similarly, other agents known to have antibacterial properties against MRSA, such as trimethoprim/sulfamethoxazole and doxycycline, have not been studied for this indication. Clindamycin has been used to treat MRSA in children, and IDSA guidelines on the treatment of MRSA list clindamycin as an alternative15 if MRSA is known to be sensitive.

        A summary of recommended empiric antibiotic therapy is presented in Table 3.16

        PDBR01609002_t3.PNG

        Three antibiotics were approved by the US Food and Drug Administration (FDA) for the treatment of CAP after the release of the IDSA/ATS guidelines in 2007. Ceftaroline fosamil is a fifth-generation cephalosporin that has coverage for MRSA and was approved in November 2010.17 It can only be administered intravenously and needs dose adjustment for renal function. Omadacycline is a new tetracycline that was approved by the FDA in October 2018.18 It is available in both intravenous injectable and oral forms. No dose adjustment is needed for renal function. Lefamulin is a first-in-class novel pleuromutilin antibiotic which was FDA-approved in August 2019. It can be administered intravenously or orally, with no dosage adjustment necessary in patients with renal impairment.19

         

         

        Antibiotic Therapy for Selected Pathogens

        Streptococcus pneumoniae

        Patients with pneumococcal pneumonia who have penicillin-susceptible strains can be treated with intravenous penicillin (2 or 3 million units every 4 hours) or ceftriaxone. Once a patient meets criteria of stability, they can then be transitioned to oral penicillin, amoxicillin, or clarithromycin. Those with strains with reduced susceptibility can still be treated with penicillin, but at a higher dose (4 million units intravenously [IV] every 4 hours), or a third-generation cephalosporin. Those whose pneumococcal pneumonia is complicated by bacteremia will benefit from dual therapy if severely ill, requiring ICU monitoring. Those not severely ill can be treated with monotherapy.20

        Staphylococcus aureus

        Staphylococcus aureus is more commonly associated with hospital-acquired pneumonia, but it may also be seen during the influenza season and in those with severe necrotizing CAP. Both linezolid and vancomycin can be used to treat MRSA CAP. As noted, ceftaroline has activity against MRSA and is approved for treatment of CAP, but is not approved by the FDA for MRSA CAP treatment. Similarly, tigecycline is approved for CAP and has activity against MRSA, but is not approved for MRSA CAP. Moreover, the FDA has warned of increased risk of death with tigecycline and has a black box warning to that effect.21

        Legionella

        Legionellosis can be treated with tetra¬cyclines, macrolides, or fluoroquinolones. For non-immunocompromised patients with mild pneumonia, any of the listed antibiotics is considered appropriate. However, patients with severe infection or those with immunosuppression should be treated with either levofloxacin or azithromycin for 7 to 10 days.22

        Chlamydophila pneumoniae

        As with other atypical organisms, Chlamydophila pneumoniae can be treated with doxycycline, a macrolide, or respiratory fluoroquinolones. However, length of therapy varies by regimen used; treating with doxycycline 100 mg twice daily generally requires 14 to 21 days, whereas moxifloxacin 400 mg daily requires 10 days.23

        Mycoplasma pneumoniae

        As with C. pneumoniae, length of therapy of Mycoplasma pneumoniae varies by which antimicrobial regimen is used. Shortest courses are seen with the use of macrolides for 5 days, whereas 14 days is considered standard for doxycycline or a respiratory fluoroquinolone.24 It should be noted that there has been increasingly documented resistance to macrolides, with known resistance of 8.2% in the United States.25

         

         

        Duration of Treatment

        Most patients with CAP respond to appropriate therapy within 72 hours. IDSA/ATS guidelines recommend that patients with routine cases of CAP be treated for a minimum of 5 days. Despite this, many patients are treated for an excessive amount of time, with over 70% of patients reported to have received antibiotics for more than 10 days for uncomplicated CAP.26 There are instances that require longer courses of antibiotics, including cases caused by Pseudomonas aeruginosa, S. aureus, and Legionella species and patients with lung abscesses or necrotizing infections, among others.27

        Hospitalized patients do not need to be monitored for an additional day once they have reached clinical stability (Table 4), are able to maintain oral intake, and have normal mentation, provided that other comorbidities are stable and social needs have been met.6 C-reactive protein (CRP) level has been postulated as an additional measure of stability, specifically monitoring for a greater than 50% reduction in CRP; however, this was validated only for those with complicated pneumonia.28 Patients discharged from the hospital with instability have higher risk of readmission or death.29

        PDBR01609002_t4.PNG

        Transition to Oral Therapy

        IDSA/ATS guidelines6 recommend that patients should be transitioned from intravenous to oral antibiotics when they are improving clinically, have stable vital signs, and are able to ingest food/fluids and medications.

        Management of Nonresponders

        Although the majority of patients respond to antibiotics within 72 hours, treatment failure occurs in up to 15% of patients.15 Nonresponding pneumonia is generally seen in 2 patterns: worsening of clinical status despite empiric antibiotics or delay in achieving clinical stability, as defined in Table 4, after 72 hours of treatment.30 Risk factors associated with nonresponding pneumonia31 are:

        • Radiographic: multilobar infiltrates, pleural effusion, cavitation
        • Bacteriologic: MRSA, gram-negative or Legionella pneumonia
        • Severity index: PSI > 90
        • Pharmacologic: incorrect antibiotic choice based on susceptibility

        Patients with acute deterioration of clinical status require prompt transfer to a higher level of care and may require mechanical ventilator support. In those with delay in achieving clinical stability, a question centers on whether the same antibiotics can be continued while doing further radiographic/microbiologic work-up and/or changing antibiotics. History should be reviewed, with particular attention to exposures, travel history, and microbiologic and radiographic data. Clinicians should recall that viruses account for up to 20% of pneumonias and that there are also noninfectious causes that can mimic pyogenic infections.32 If adequate initial cultures were not obtained, they should be obtained; however, care must be taken in reviewing new sets of cultures while on antibiotics, as they may reveal colonization selected out by antibiotics and not a true pathogen. If repeat evaluation is unrevealing, then further evaluation with computed tomography (CT) scan and bronchoscopy with bronchoalveolar lavage and biopsy is warranted. CT scans can show pleural effusions, bronchial obstructions, or a pattern suggestive of cryptogenic pneumonia. A bronchoscopy might yield a microbiologic diagnosis and, when combined with biopsy, can also evaluate for noninfectious causes.

        As with other infections, if escalation of antibiotics is undertaken, clinicians should try to determine the reason for nonresponse. To simply broaden antimicrobial therapy without attempts at establishing a microbiologic or radiographic cause for nonresponse may lead to inappropriate treatment and recurrence of infection. Aside from patients who have bacteremic pneumococcal pneumonia in an ICU setting, there are no published reports pointing to superiority of combination antibiotics.20

         

         

        Other Treatment

        Several agents have been evaluated as adjunctive treatment of pneumonia to decrease the inflammatory response associated with pneumonia; namely, steroids, macrolide antibiotics, and statins. To date, only the use of steroids (methylprednisolone 0.5 mg/kg every 12 hours for 5 days) in those with severe CAP and high initial anti-inflammatory response (CRP > 150) has been shown to decrease treatment failure, decrease risk of acute respiratory distress syndrome, and possibly reduce length of stay and duration of intravenous antibiotics, without effect on mortality or adverse side effects.33,34 However, a recent double-blind randomized study conducted in Australia in which patients admitted with CAP were prescribed prednisolone acetate (50 mg/day for 7 days) and de-escalated from parenteral to oral antibiotics according to standardized criteria revealed no difference in mortality, length of stay, or readmission rates between the corticosteroids group and the control group at 90-day follow-up.35 At this point, corticosteroid as an adjunctive treatment for CAP is still controversial and the new 2019 ATS/IDSA guidelines recommend not routinely using corticosteroids in all patients with CAP.36 Other adjunctive methods have not been found to have significant impact.6

        Prevention of Pneumonia

        Prevention of pneumococcal pneumonia involves vaccinations to prevent infection caused by S. pneumoniae and influenza viruses. As influenza is a risk factor for bacterial infection, specifically with S. pneumoniae, influenza vaccination can help prevent bacterial pneumonia.37 In their most recent recommendations, the CDC continues to recommend routine influenza vaccination for all persons older than age 6 months, unless otherwise contraindicated.38

        There are 2 vaccines for prevention of pneumococcal disease: the pneumococcal polysaccharide vaccine (PPSV23) and a conjugate vaccine (PCV13). Following vaccination with PPSV23, 80% of adults develop antibodies against at least 18 of the 23 serotypes.39 PPSV23 is reported to be protective against invasive pneumococcal infection, although there is no consensus regarding whether PPSV23 leads to decreased rates of pneumonia.40 On the other hand, PCV13 vaccination was associated with prevention of both invasive disease and CAP in adults aged 65 years or older.41 The CDC recommends that all children aged 2 years or younger receive PCV13, and those aged 65 or older receive PCV13 followed by a dose of PPSV23.42,43 The dose of PPSV23 should be given at least 1 year after the dose of PCV13 is administered.44 Persons younger than 65 years with immunocompromising and certain other conditions should also receive vaccination (Table 5).44 Full recommendations, many scenarios, and details on timing of vaccinations can be found at the CDC’s website.

        PDBR01609002_t5.PNG

        Cigarette smoking increases the risk of respiratory infections, as evidenced by smokers accounting for almost half of all patients with invasive pneumococcal disease.11 As this is a modifiable risk factor, smoking cessation should be part of a comprehensive approach toward prevention of pneumonia.

         

         

        Summary

        Most patients with CAP are treated empirically with antibiotics, with therapy selection based on the site of care, likely pathogen, and antimicrobial resistance issues. Those treated as outpatients usually respond well to empiric antibiotic treatment, and a causative pathogen is not usually sought. Patients who are hospitalized for treatment usually receive empiric antibiotic on admission, and antimicrobial therapy is adjusted accordingly once the etiology has been determined by microbiologic or serologic means. At this time, the use of corticosteroid as an adjunctive treatment for CAP is still controversial, so not all patients with CAP should routinely receive corticosteroids. Because vaccination (PPSV23, PCV13, and influenza vaccine) remains the most effective tool in preventing the development of CAP, clinicians should strive for 100% vaccination rates in persons without contraindications.

        References

        1. Fine MJ, Auble TE, Yealy DM, et al A prediction rule to identify low-risk patients with community-acquired pneumonia. N Engl J Med.1997;336:243-250.

        2. Lim WS, van der Eerden MM, Laing R, et al. Defining community acquired pneumonia severity on presentation to hospital: an international derivation and validation study. Thorax. 2003;58:377-382.

        3. Aujesky D, Auble TE, Yealy DM, et al. Prospective comparison of three validated prediction rules for prognosis in community-acquired pneumonia. Am J Med. 2005;118:384-392.

        4. Arnold FW, Ramirez JA, McDonald LC, Xia EL. Hospitalization for community-acquired pneumonia: the pneumonia severity index vs clinical judgment. Chest. 2003;124:121-124.

        5. Aujesky D, McCausland JB, Whittle J, et al. Reasons why emergency department providers do not rely on the pneumonia severity index to determine the initial site of treatment for patients with pneumonia. Clin Infect Dis. 2009;49:e100-108.

        6. Mandell LA, Wunderink RG, Anzueto A, et al. 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:S27-72.

        7. Charles PG, Wolfe R, Whitby M, et al. SMART-COP: a tool for predicting the need for intensive respiratory or vasopressor support in community-acquired pneumonia. Clin Infect Dis. 2008;47:375-384.

        8. Marti C, Garin N, Grosgurin O, et al. Prediction of severe community-acquired pneumonia: a systematic review and meta-analysis. Crit Care. 2012;16:R141.

        9. Jain S, Self WH, Wunderink RG, et al. Community-acquired pneumonia requiring hospitalization among U.S. adults. N Engl J Med. 2015;373:415-427.

        10. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63:e61-e111.

        11. Wunderink RG, Niederman MS, Kollef MH, et al. Linezolid in methicillin-resistant Staphylococcus aureus nosocomial pneumonia: a randomized, controlled study. Clin Infect Dis. 2012;54:621-629.

        12. Silverman JA, Mortin LI, Vanpraagh AD, et al. Inhibition of daptomycin by pulmonary surfactant: in vitro modeling and clinical impact. J Infect Dis. 2005;191:2149-2152.

        13. El Hajj MS, Turgeon RD, Wilby KJ. Ceftaroline fosamil for community-acquired pneumonia and skin and skin structure infections: a systematic review. Int J Clin Pharm. 2017;39:26-32.

        14. Taboada M, Melnick D, Iaconis JP, et al. Ceftaroline fosamil versus ceftriaxone for the treatment of community-acquired pneumonia: individual patient data meta-analysis of randomized controlled trials. J Antimicrob Chemother. 2016;71:862-870.

        15. Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children: executive summary. Clin Infect Dis. 2011;52:285-292.

        16. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Sauders; 2015:2310-2327.

        17. Teflaro (ceftaroline fosamil) [package insert]. St. Louis, MO: Forest Pharmaceuticals; 2010.

        18. Nuzyra (omadacycline) [package insert]. Boston, MA: Paratek Pharmaceuticals; 2018.

        19. Xenleta (lefamulin) [package insert]. Dublin, Ireland: Nabriva Therapeutics; 2019.

        20. Baddour LM, Yu VL, Klugman KP, et al. Combination antibiotic therapy lowers mortality among severely ill patients with pneumococcal bacteremia. Am J Respir Crit Care Med. 2004;170:440-444.

        21. FDA Drug Safety Communication: FDA warns of increased risk of death with IV antibacterial Tygacil (tigecycline) and approves new boxed warning. www.fda.gov/Drugs/DrugSafety/ucm369580.htm. Accessed 16 September 2019.

        22. Edelstein PR, CR. Legionnaires’ disease and Pontiac fever. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2633.

        23. Hammerschlag MR, Kohlhoff SA, Gaydos, CA. Chlamydia pneumoniae. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2174.

        24. Holzman RS, MS. Mycoplasma pneumoniae and atypical pneumonia. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2183.

        25. Yamada M, Buller R, Bledsoe S, Storch GA. Rising rates of macrolide-resistant Mycoplasma pneumoniae in the central United States. Pediatr Infect Dis J. 2012;31:409-410.

        26. Yi SH, Hatfield KM, Baggs J, et al. Duration of antibiotic use among adults with uncomplicated community-acquired pneumonia requiring hospitalization in the United States. Clin Infect Dis. 2018;66:1333-1341.

        27. Hayashi Y, Paterson DL. Strategies for reduction in duration of antibiotic use in hospitalized patients. Clin Infect Dis. 2011;52:1232-1240.

        28. Akram AR, Chalmers JD, Taylor JK, et al. An evaluation of clinical stability criteria to predict hospital course in community-acquired pneumonia. Clin Microbiol Infect. 2013;19:1174-1180.

        29. Halm EA, Fine MJ, Kapoor WN, et al. Instability on hospital discharge and the risk of adverse outcomes in patients with pneumonia. Arch Intern Med. 2002;162:1278-1284.

        30. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Saunders; 2015:2310-2327.

        31. Roson B, Carratala J, Fernandez-Sabe N, et al. Causes and factors associated with early failure in hospitalized patients with community-acquired pneumonia. Arch Intern Med. 2004;164:502-508.

        32. El-Solh AA, Pietrantoni C, Bhat A, et al. Microbiology of severe aspiration pneumonia in institutionalized elderly. Am J Respir Crit Care Med. 2003;167:1650-1654.

        33. Wan YD, Sun TW, Liu ZQ, et al. Efficacy and safety of corticosteroids for community-acquired pneumonia: a systematic review and meta-analysis. Chest. 2016;149:209-219.

        34. Torres A, Sibila O, Ferrer M, et al. Effect of corticosteroids on treatment failure among hospitalized patients with severe community-acquired pneumonia and high inflammatory response: a randomized clinical trial. JAMA. 2015;313:677-686.

        35. Lloyd M, Karahalios, Janus E, et al. Effectiveness of a bundled intervention including adjunctive corticosteroids on outcomes of hospitalized patients with community-acquired pneumonia: a stepped-wedge randomized clinical trial. JAMA Intern Med. 2019;179:1052-1060.

        36. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200:e45-e67.

        37. McCullers JA. Insights into the interaction between influenza virus and pneumococcus. Clin Microbiol Rev. 2006;19:571-582.

        38. Grohskopf LA, Alyanak E, Broder KR, et al. Prevention and control of seasonal influenza with vaccines: recommendations of the advisory committee on immunization practices - United States, 2019-20 influenza season. MMWR Recomm Rep. 2019;68:1-21.

        39. Rubins JB, Alter M, Loch J, Janoff EN. Determination of antibody responses of elderly adults to all 23 capsular polysaccharides after pneumococcal vaccination. Infect Immun. 1999;67:5979-5984.

        40. Vaccines and preventable diseases. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/vpd/pneumo/hcp/about-vaccine.html. Accessed 16 September 2019.

        41. Bonten MJ, Huijts SM, Bolkenbaas M, et al. Polysaccharide conjugate vaccine against pneumococcal pneumonia in adults. N Engl J Med. 2015;372:1114-1125.

        42. Recommended adult immunization schedule -- United States -- 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/schedules/downloads/adult/adult-combined-schedule.pdf. Accessed 16 September 2019.

        43. Recommended child and adolescent immunization schedule for ages 18 years or younger – United States – 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/schedules/hcp/imz/child-adolescent.html. Accessed 22 September 2019.

        44. Pneumococcal vaccine timing for adults – United States – 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/vpd/pneumo/downloads/pneumo-vaccine-timing.pdf. Accessed 22 September 2019.

        References

        1. Fine MJ, Auble TE, Yealy DM, et al A prediction rule to identify low-risk patients with community-acquired pneumonia. N Engl J Med.1997;336:243-250.

        2. Lim WS, van der Eerden MM, Laing R, et al. Defining community acquired pneumonia severity on presentation to hospital: an international derivation and validation study. Thorax. 2003;58:377-382.

        3. Aujesky D, Auble TE, Yealy DM, et al. Prospective comparison of three validated prediction rules for prognosis in community-acquired pneumonia. Am J Med. 2005;118:384-392.

        4. Arnold FW, Ramirez JA, McDonald LC, Xia EL. Hospitalization for community-acquired pneumonia: the pneumonia severity index vs clinical judgment. Chest. 2003;124:121-124.

        5. Aujesky D, McCausland JB, Whittle J, et al. Reasons why emergency department providers do not rely on the pneumonia severity index to determine the initial site of treatment for patients with pneumonia. Clin Infect Dis. 2009;49:e100-108.

        6. Mandell LA, Wunderink RG, Anzueto A, et al. 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:S27-72.

        7. Charles PG, Wolfe R, Whitby M, et al. SMART-COP: a tool for predicting the need for intensive respiratory or vasopressor support in community-acquired pneumonia. Clin Infect Dis. 2008;47:375-384.

        8. Marti C, Garin N, Grosgurin O, et al. Prediction of severe community-acquired pneumonia: a systematic review and meta-analysis. Crit Care. 2012;16:R141.

        9. Jain S, Self WH, Wunderink RG, et al. Community-acquired pneumonia requiring hospitalization among U.S. adults. N Engl J Med. 2015;373:415-427.

        10. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63:e61-e111.

        11. Wunderink RG, Niederman MS, Kollef MH, et al. Linezolid in methicillin-resistant Staphylococcus aureus nosocomial pneumonia: a randomized, controlled study. Clin Infect Dis. 2012;54:621-629.

        12. Silverman JA, Mortin LI, Vanpraagh AD, et al. Inhibition of daptomycin by pulmonary surfactant: in vitro modeling and clinical impact. J Infect Dis. 2005;191:2149-2152.

        13. El Hajj MS, Turgeon RD, Wilby KJ. Ceftaroline fosamil for community-acquired pneumonia and skin and skin structure infections: a systematic review. Int J Clin Pharm. 2017;39:26-32.

        14. Taboada M, Melnick D, Iaconis JP, et al. Ceftaroline fosamil versus ceftriaxone for the treatment of community-acquired pneumonia: individual patient data meta-analysis of randomized controlled trials. J Antimicrob Chemother. 2016;71:862-870.

        15. Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children: executive summary. Clin Infect Dis. 2011;52:285-292.

        16. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Sauders; 2015:2310-2327.

        17. Teflaro (ceftaroline fosamil) [package insert]. St. Louis, MO: Forest Pharmaceuticals; 2010.

        18. Nuzyra (omadacycline) [package insert]. Boston, MA: Paratek Pharmaceuticals; 2018.

        19. Xenleta (lefamulin) [package insert]. Dublin, Ireland: Nabriva Therapeutics; 2019.

        20. Baddour LM, Yu VL, Klugman KP, et al. Combination antibiotic therapy lowers mortality among severely ill patients with pneumococcal bacteremia. Am J Respir Crit Care Med. 2004;170:440-444.

        21. FDA Drug Safety Communication: FDA warns of increased risk of death with IV antibacterial Tygacil (tigecycline) and approves new boxed warning. www.fda.gov/Drugs/DrugSafety/ucm369580.htm. Accessed 16 September 2019.

        22. Edelstein PR, CR. Legionnaires’ disease and Pontiac fever. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2633.

        23. Hammerschlag MR, Kohlhoff SA, Gaydos, CA. Chlamydia pneumoniae. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2174.

        24. Holzman RS, MS. Mycoplasma pneumoniae and atypical pneumonia. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:2183.

        25. Yamada M, Buller R, Bledsoe S, Storch GA. Rising rates of macrolide-resistant Mycoplasma pneumoniae in the central United States. Pediatr Infect Dis J. 2012;31:409-410.

        26. Yi SH, Hatfield KM, Baggs J, et al. Duration of antibiotic use among adults with uncomplicated community-acquired pneumonia requiring hospitalization in the United States. Clin Infect Dis. 2018;66:1333-1341.

        27. Hayashi Y, Paterson DL. Strategies for reduction in duration of antibiotic use in hospitalized patients. Clin Infect Dis. 2011;52:1232-1240.

        28. Akram AR, Chalmers JD, Taylor JK, et al. An evaluation of clinical stability criteria to predict hospital course in community-acquired pneumonia. Clin Microbiol Infect. 2013;19:1174-1180.

        29. Halm EA, Fine MJ, Kapoor WN, et al. Instability on hospital discharge and the risk of adverse outcomes in patients with pneumonia. Arch Intern Med. 2002;162:1278-1284.

        30. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Saunders; 2015:2310-2327.

        31. Roson B, Carratala J, Fernandez-Sabe N, et al. Causes and factors associated with early failure in hospitalized patients with community-acquired pneumonia. Arch Intern Med. 2004;164:502-508.

        32. El-Solh AA, Pietrantoni C, Bhat A, et al. Microbiology of severe aspiration pneumonia in institutionalized elderly. Am J Respir Crit Care Med. 2003;167:1650-1654.

        33. Wan YD, Sun TW, Liu ZQ, et al. Efficacy and safety of corticosteroids for community-acquired pneumonia: a systematic review and meta-analysis. Chest. 2016;149:209-219.

        34. Torres A, Sibila O, Ferrer M, et al. Effect of corticosteroids on treatment failure among hospitalized patients with severe community-acquired pneumonia and high inflammatory response: a randomized clinical trial. JAMA. 2015;313:677-686.

        35. Lloyd M, Karahalios, Janus E, et al. Effectiveness of a bundled intervention including adjunctive corticosteroids on outcomes of hospitalized patients with community-acquired pneumonia: a stepped-wedge randomized clinical trial. JAMA Intern Med. 2019;179:1052-1060.

        36. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200:e45-e67.

        37. McCullers JA. Insights into the interaction between influenza virus and pneumococcus. Clin Microbiol Rev. 2006;19:571-582.

        38. Grohskopf LA, Alyanak E, Broder KR, et al. Prevention and control of seasonal influenza with vaccines: recommendations of the advisory committee on immunization practices - United States, 2019-20 influenza season. MMWR Recomm Rep. 2019;68:1-21.

        39. Rubins JB, Alter M, Loch J, Janoff EN. Determination of antibody responses of elderly adults to all 23 capsular polysaccharides after pneumococcal vaccination. Infect Immun. 1999;67:5979-5984.

        40. Vaccines and preventable diseases. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/vpd/pneumo/hcp/about-vaccine.html. Accessed 16 September 2019.

        41. Bonten MJ, Huijts SM, Bolkenbaas M, et al. Polysaccharide conjugate vaccine against pneumococcal pneumonia in adults. N Engl J Med. 2015;372:1114-1125.

        42. Recommended adult immunization schedule -- United States -- 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/schedules/downloads/adult/adult-combined-schedule.pdf. Accessed 16 September 2019.

        43. Recommended child and adolescent immunization schedule for ages 18 years or younger – United States – 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/schedules/hcp/imz/child-adolescent.html. Accessed 22 September 2019.

        44. Pneumococcal vaccine timing for adults – United States – 2019. Centers for Disease Control and Prevention Web site. www.cdc.gov/vaccines/vpd/pneumo/downloads/pneumo-vaccine-timing.pdf. Accessed 22 September 2019.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Community-Acquired Pneumonia: Treatment
        Display Headline
        Community-Acquired Pneumonia: Treatment
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 September;16(3)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Fri, 09/27/2019 - 09:30
        Un-Gate On Date
        Fri, 09/27/2019 - 09:30
        Use ProPublica
        CFC Schedule Remove Status
        Fri, 09/27/2019 - 09:30
        Hide sidebar & use full width
        render the right sidebar.

        Community-Acquired Pneumonia: Evaluation and Diagnosis

        Article Type
        Changed
        Mon, 10/07/2019 - 07:34
        Display Headline
        Community-Acquired Pneumonia: Evaluation and Diagnosis

        Despite advances in medical science, pneumonia remains a major cause of morbidity and mortality. In 2017, 49,157 patients in the United States died from the disease.1 Pneumonia can be classified as community-acquired, hospital-acquired, or ventilator-associated. Another category, healthcare-associated pneumonia, was included in an earlier Infectious Diseases Society of America (IDSA) and American Thoracic Society (ATS) guideline but was removed from the 2016 guideline because there was no clear evidence that patients diagnosed with healthcare-associated pneumonia were at higher risk for harboring multidrug-resistant pathogens.2 This review is the first of 2 articles focusing on the management of community-acquired pneumonia (CAP). Here, we review CAP epidemiology, microbiology, predisposing factors, and diagnosis; current treatment and prevention of CAP are reviewed in a separate article.

        Definition and Epidemiology

        CAP is defined as an acute infection of the lungs that develops in patients who have not been hospitalized recently and have not had regular exposure to the health care system.3 A previously ambulatory patient who is diagnosed with pneumonia within 48 hours after admission also meets the criteria for CAP. Approximately 4 to 5 million cases of CAP are diagnosed in the United States annually.4 About 25% of CAP patients require hospitalization, and about 5% to 10% of these patients are admitted to the intensive care unit (ICU).5 In-hospital mortality is considerable (~10% in population-based studies),6 and 30-day mortality was found to be as high as 23% in a review by File and Marrie.7 CAP also confers a high risk of long-term morbidity and mortality compared with the general population who have never had CAP, irrespective of age.8

        Causative Organisms

        Numerous microorganisms can cause CAP. Common causes and less common causes are delineated in Table 1. Until recently, many studies had demonstrated that pneumococcus was the most common cause of CAP. However, in the CDC Etiology of Pneumonia in the Community (EPIC) study team’s 2015 prospective, multicenter, population-based study, no pathogen was detected in the majority of patients diagnosed with CAP requiring hospitalization. The most common pathogens they detected were rhinovirus (9%), followed by influenza virus (6%) and pneumococcus (5%).9 Factors considered to be contributing to the decrease in the percentage of pneumococcus in patients diagnosed with CAP are the widespread use of pneumococcal vaccine and reduced rates of smoking.10,11

        PDBR01609001_t1.PNG

        Predisposing Factors

        Most people diagnosed with CAP have 1 or more predisposing factors (Table 2).12,13 Patients who develop CAP typically have a combination of these predisposing factors rather than a single factor. Aging, in combination with other risk factors, increases the susceptibility of a person to pneumonia.

        PDBR01609001_t2.PNG

        Clinical Signs and Symptoms

        Symptoms of CAP include fever, chills, rigors, fatigue, anorexia, diaphoresis, dyspnea, cough (with or without sputum production), and pleuritic chest pain. There is no individual symptom or cluster of symptoms that can absolutely differentiate pneumonia from other acute respiratory diseases, including upper and lower respiratory infections. However, patients presenting with the constellation of symptoms of fever ≥ 100°F (37.8°C), productive cough, and tachycardia is more suggestive of pneumonia.14 Abnormal vital signs include fever, hypothermia, tachypnea, tachycardia, and oxygen desaturation. Auscultation of the chest reveals crackles or other adventitious breath sounds. Elderly patients with pneumonia report a significantly lower number of both respiratory and nonrespiratory symptoms compared with younger patients. Clinicians should be aware of this phenomenon to avoid delayed diagnosis and treatment.15

         

         

        Imaging Evaluation

        The presence of a pulmonary consolidation or an infiltrate on chest radiograph is required to diagnose CAP, and a chest radiograph should be obtained when CAP is suspected.16 However, there is no pattern of radiographic abnormalities reliable enough to differentiate infectious pneumonia from noninfectious causes.17

        There are case reports and case series demonstrating false-negative plain chest radiographs in dehydrated patients18 or in patients in a neutropenic state. However, animal studies have shown that dogs challenged with pneumococcus showed abnormal pulmonary shadow, suggestive of pneumonia, regardless of hydration status.19 There is also no reliable scientific evidence to support the notion that severe neutropenia can cause false-negative radiographs because of the inability to develop an acute inflammatory reaction in the lungs.20

        A chest computed tomography (CT) scan is more sensitive than a plain chest radiograph in detecting pneumonia. Therefore, a chest CT should be performed in a patient with negative plain chest radiograph when pneumonia is still highly suspected.21 A chest CT scan is also more sensitive in detecting cavitation, adenopathy, interstitial disease, and empyema. It also has the advantage of better defining anatomical changes than plain films.22

        Because improvement of pulmonary opacities in patients with CAP lags behind clinical improvement, repeating chest imaging studies is not recommended in patients who demonstrate clinical improvement. Clearing of pulmonary infiltrate or consolidation sometimes can take 6 weeks or longer.23

        Laboratory Evaluation

        Generally, the etiologic agent of CAP cannot be determined solely on the basis of clinical signs and symptoms or imaging studies. Although routine microbiological testing for patients suspicious for CAP is not necessary for empirical treatment, determining the etiologic agent of the pneumonia allows the clinician to narrow the antibiotics from a broad-spectrum empirical regimen to specific pathogen-directed therapy. Determination of certain etiologic agents causing the pneumonia can have important public health implications (eg, Mycobacterium tuberculosis and influenza virus).24

        Sputum Gram Stain and Culture

        Sputum Gram stain is an inexpensive test that may identify pathogens that cause CAP (eg, Streptococcus pneumoniae and Haemophilus influenzae). A quality specimen is required. A sputum sample must contain more than 25 neutrophils and less than 10 squamous epithelial cells/low power field on Gram stain to be considered suitable for culture. The sensitivity and specificity of sputum Gram stain and culture are highly variable in different clinical settings (eg, outpatient setting, nursing home, ICU). Reed et al’s meta-analysis of patients diagnosed with CAP in the United States showed the sensitivity and specificity of sputum Gram stain (compared with sputum culture) ranged from 15% to 100% and 11% to 100%, respectively.24 In cases of proven bacteremic pneumococcal pneumonia, positive cultures from sputum samples were positive less than 50% of the time.25

        For patients who cannot provide sputum samples or are intubated, deep-suction aspirate or bronchoalveolar lavage through a bronchoscopic procedure may be necessary to obtain pulmonary secretion for Gram stain and culture. Besides bacterial culture, sputum samples can also be sent for fungal and mycobacterial cultures and acid-fast stain, if deemed clinically necessary.

        The 2019 ATS/IDSA guidelines for diagnosis and treatment of adults with CAP recommend sputum culture in patients with severe disease and in all inpatients empirically treated for MRSA or Pseudomonas aeruginosa.26

         

         

        Blood Culture

        Because the positivity rate of blood culture in patients who are suspected to have pneumonia but not exposed to antimicrobial agents is low (5%–14%), blood cultures are not recommended for all patients with CAP. Another reason for not recommending blood culture is positive culture rarely leads to changes in antibiotic regimen in patients without underlying diseases.27 However, the 2019 ATS/IDSA guidelines recommend blood culture in patients with severe disease and in all inpatients treated empirically for MRSA or P. aeruginosa.26

        A multinational study published in 2008 examined 125 patients with pneumococcal bacteremic CAP versus 1847 patients with non-bacteremic CAP.28 Analysis of the data demonstrated no association between pneumococcal bacteremic CAP and time to clinical stability, length of hospital stay, all-cause mortality, or CAP-related mortality. The authors concluded that pneumococcal bacteremia does not increase the risk of poor outcomes in patients with CAP compared to non-bacteremic patients, and the presence of pneumococcal bacteremia should not deter de-escalation of therapy in clinically stable patients.

        Urinary Antigen Tests

        Urinary antigen tests may assist clinicians in narrowing antibiotic therapy when test results are positive. There are 2 US Food and Drug Administration–approved tests available to clinicians for detecting pneumococcal and Legionella antigen in urine. The test for Legionella pneumophila detects disease due to serogroup 1 only, which accounts for 80% of community-acquired Legionnaires’ disease. The sensitivity and specificity of the Legionella urine antigen test are 90% and 99%, respectively. The pneumococcal urine antigen test is less sensitive and specific than the Legionella urine antigen test (sensitivity 80% and specificity > 90%).29,30

        Advantages of the urinary antigen tests are that they are easily performed, results are available in less than an hour if done in-house, and results are not affected by prior exposure to antibiotics. However, the tests do not meet Clinical Laboratory Improvements Amendments criteria for waiver and must be performed by a technician in the laboratory. A multicenter, prospective surveillance study of hospitalized patients with CAP showed that the 2007 IDSA/ATS guidelines’ recommended indications for S. pneumoniae and L. pneumophila urinary antigen tests do not have sufficient sensitivity and specificity to identify patients with positive tests.31

        Polymerase Chain Reaction

        There are several FDA-approved polymerase chain reaction (PCR) tests commercially available to assist clinicians in diagnosing pneumonia. PCR testing of nasopharyngeal swabs for diagnosis of influenza has become standard in many US medical facilities. The great advantages of using PCR to diagnose influenza are its high sensitivity and specificity and rapid turnaround time. PCR can also be used to detect Legionella species, S. pneumonia, Mycoplasma pneumoniae, Chlamydophila pneumonia, and mycobacterial species.24

        One limitation of using PCR tests on respiratory specimens is that specimens can be contaminated with oral or upper airway flora, so the results must be interpreted with caution, bearing in mind that some of the pathogens isolated may be colonizers of the oral or upper airway flora.32

         

         

        Biologic Markers

        Two biologic markers—procalcitonin and C-reactive protein (CRP)—can be used in conjunction with history, physical examination, laboratory tests, and imaging studies to assist in the diagnosis and treatment of CAP.24 Procalcitonin is a peptide precursor of the hormone calcitonin that is released by parenchymal cells into the bloodstream, resulting in increased serum level in patients with bacterial infections. In contrast, there is no remarkable procalcitonin level increase with viral or noninfectious inflammation. The reference value of procalcitonin in the blood of an adult individual without infection or inflammation is < 0.15 ng/mL. In the blood, procalcitonin has a half-life of 25 to 30 hours. The quantitative immunoluminometric method (LUMI test, Brahms PCT, Berlin, Germany) is the preferred test to use because of its high sensitivity.33 A meta-analysis of 12 studies involving more than 2400 patients with CAP demonstrated that serum procalcitonin does not have sufficient sensitivity or specificity to distinguish between bacterial and nonbacterial pneumonia. The authors concluded that procalcitonin level cannot be used to decide whether an antibiotic should be administered.34

        A 2012 Cochrane meta-analysis that involved 4221 patients with acute respiratory infections (with half of the patients diagnosed with CAP) from 14 prospective trials found the use of procalcitonin test for antibiotic use significantly decreased median antibiotic exposure from 8 to 4 days without an increase in treatment failure, mortality rates in any clinical setting (eg, outpatient clinic, emergency room), or length of hospitalization.35 An update of the 2012 Cochrane review that examined the safety and efficacy of using procalcitonin for starting or stopping antibiotics again demonstrated procalcitonin use was associated with a reduction of antibiotic use (2.4 days).36 A prospective study conducted in France on 100 ICU patients showed that increased procalcitonin from day 1 to day 3 has a poor prognosis factor for severe CAP, whereas decreasing procalcitonin levels is associated with a favorable outcome.37

        Because of conflicting data, the 2019 ATS/IDSA guidelines do not recommend using procalcitonin to determine need for initial antibacterial therapy.26

        CRP is an acute phase protein produced by the liver. CRP level in the blood increases in response to acute infection or inflammation. Use of CRP in assisting diagnosis and guiding treatment of CAP is more limited in part due to its poor specificity. A prospective study conducted on 168 consecutive patients who presented with cough showed that a CRP level > 40 mg/L had a sensitivity and specificity of 70% and 90%, respectively.38

        Summary

        CAP remains a leading cause of hospitalization and death in the 21st century. Traditionally, pneumococcus has been considered the major pathogen causing CAP; however, the 2015 EPIC study found that S. pneumoniae was detected in only 5% of patients diagnosed with CAP. Despite the new findings, it is still recommended that empiric treatment for CAP target common typical bacteria (pneumococcus, H. influenzae, Moraxella catarrhalis) and atypical bacteria (M. pneumonia, C. pneumoniae, L. pneumophila).

        Because diagnosing pneumonia through history and clinical examination is less than 50% sensitive, a chest imaging study (a plain chest radiograph or a chest CT scan) is usually required to make the diagnosis. Laboratory tests, such as sputum Gram stain/culture, blood culture, urinary antigen tests, PCR test, procalcitonin, and CRP are important adjunctive diagnostic modalities to assist in the diagnosis and management of CAP. However, because no single test is sensitive and specific enough to be a stand-alone test, they should be used in conjunction with history, physical examination, and imaging studies.

        References

        1. Centers for Disease Control and Prevention. National Center for Health Statistics. FastStats - Pneumonia. www.cdc.gov/nchs/fastats/pneumonia.htm. Accessed 16 September 2019.

        2. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63:e61-e111.

        3. Musher DM, Thorner AR. Community-acquired pneumonia. N Engl J Med. 2014;371:1619-1628.

        4. Mandell LA. Epidemiology and etiology of community-acquired pneumonia. Infect Dis Clin North Am. 2004;18:761-776.

        5. Hoare Z, Lim WS. Pneumonia: update on diagnosis and management. BMJ. 2006;332:1077-1079.

        6. Johnstone J, Marrie TJ, Eurich DT, Majumdar SR. Effect of pneumococcal vaccination in hospitalized adults with community-acquired pneumonia. Arch Intern Med. 2007;167:1938-1943.

        7. File TM Jr, Marrie TJ. Burden of community-acquired pneumonia in North American adults. Postgrad Med. 2010;122:130-141.

        8. Eurich DT, Marrie TJ, Minhas-Sandhu JK, Majumdar SR. Ten-year mortality after community-acquired pneumonia. a prospective cohort. Am J Respir Crit Care Med. 2015;192:597-604.

        9. Jain S, Self WH, Wunderink RG, et al. Community-acquired pneumonia requiring hospitalization among U.S. adults. N Engl J Med. 2015;373:415-427.

        10. Griffin MR, Zhu Y, Moore MR, et al. U.S. hospitalizations for pneumonia after a decade of pneumococcal vaccination. N Engl J Med. 2013;369:155-163.

        11. Nuorti JP, Butler JC, Farley MM, et al. Cigarette smoking and invasive pneumococcal disease. Active Bacterial Core Surveillance Team. N Engl J Med. 2000;342:681-689.

        12. Almirall J, Serra-Prat M, Bolíbar I, Balasso V. Risk factors for community-acquired pneumonia in adults: a systemic review of observational studies. Respiration. 2017;94:299-311.

        13. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Saunders; 2015:2310-2327.

        14. Diehr P, Wood RW, Bushyhead J, et al. Prediction of pneumonia in outpatients with acute cough--a statistical approach. J Chronic Dis. 1984;37:215-225.

        15. Metlay JP, Schulz R, Li YH, et al. Influence of age on symptoms at presentation in patients with community-acquired pneumonia. Arch Intern Med. 1997;157:1453-1459.

        16. Mandell LA, Wunderink RG, Anzueto A, et al. 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:S27-72.

        17. Jartti A, Rauvala E, Kauma H, et al. Chest imaging findings in hospitalized patients with H1N1 influenza. Acta Radiol. 2011;52:297-304.

        18. Basi SK, Marrie TJ, Huang JQ, Majumdar SR. Patients admitted to hospital with suspected pneumonia and normal chest radiographs: epidemiology, microbiology, and outcomes. Am J Med. 2004;117:305-311.

        19. Caldwell A, Glauser FL, Smith WR, et al. The effects of dehydration on the radiologic and pathologic appearance of experimental canine segmental pneumonia. Am Rev Respir Dis. 1975;112:651-656.

        20. Bartlett JG. Pneumonia. In: Barlett JG, editor. Management of Respiratory Tract Infections. Philadelphia: Lippincott, Williams & Wilkins; 2001:1-122.

        21. Claessens YE, Debray MP, Tubach F, et al. Early chest computed tomography scan to assist diagnosis and guide treatment decision for suspected community-acquired pneumonia. Am J Respir Crit Care Med. 2015;192:974-982.

        22. Wheeler JH, Fishman EK. Computed tomography in the management of chest infections: current status. Clin Infect Dis. 1996;23:232-240.

        23. Chesnutt MP. Pulmonary disorders. In: Papadakis MM, editor. Current Medical Diagnosis and Treatment. New York: McGraw-Hill; 2016:242-320.

        24. Mandell LW. Pneumonia. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:188-201.

        25. Reed WW, Byrd GS, Gates RH Jr, et al. Sputum gram’s stain in community-acquired pneumococcal pneumonia. A meta-analysis. West J Med. 1996;165:197-204.

        26. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200:e45-e67.

        27. Chalasani NP, Valdecanas MA, Gopal AK, et al. Clinical utility of blood cultures in adult patients with community-acquired pneumonia without defined underlying risks. Chest. 1995;108:932-936.

        28. Bordon J, Peyrani P, Brock GN, et al. The presence of pneumococcal bacteremia does not influence clinical outcomes in patients with community-acquired pneumonia: results from the Community-Acquired Pneumonia Organization (CAPO) International Cohort study. Chest. 2008;133:618-624.

        29. Helbig JH, Uldum SA, Bernander S, et al. Clinical utility of urinary antigen detection for diagnosis of community-acquired, travel-associated, and nosocomial legionnaires’ disease. J Clin Microbiol. 2003;41:838-840.

        30. Smith MD, Derrington P, Evans R, et al. Rapid diagnosis of bacteremic pneumococcal infections in adults by using the Binax NOW Streptococcus pneumoniae urinary antigen test: a prospective, controlled clinical evaluation. J Clin Microbiol. 2003;41:2810-2813.

        31. Bellew S, Grijalva CG, Williams DJ, et al. Pneumococcal and Legionella urinary antigen tests in community-acquired pneumonia: Prospective evaluation of indications for testing. Clin Infect Dis. 2019;68:2026-2033.

        32. Johansson N, Kalin M, Tiveljung-Lindell A, et al. Etiology of community-acquired pneumonia: increased microbiological yield with new diagnostic methods. Clin Infect Dis. 2010;50:202-209.

        33. Gilbert DN. Procalcitonin as a biomarker in respiratory tract infection. Clin Infect Dis. 2011;52 Suppl 4:S346-350.

        34. Kamat IS Ramachandran V, Eswaran H, et al. Procalcitonin to distinguish viral from bacterial pneumonia: A systematic review and meta-analysis. Clin Infect Dis. 2019 Jun 25. [Epub ahead of print]

        35. Schuetz P, Muller B, Christ-Crain M, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2012;(9):CD007498.

        36. Schuetz P, Wirz Y, Sager R, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2017;10:CD007498.

        37. Boussekey N, Leroy O, Alfandari S, et al. Procalcitonin kinetics in the prognosis of severe community-acquired pneumonia. Intensive Care Med. 2006;32:469-472.

        38. Flanders SA, Stein J, Shochat G, et al. Performance of a bedside C-reactive protein test in the diagnosis of community-acquired pneumonia in adults with acute cough. Am J Med. 2004;116:529-535.

        Author and Disclosure Information

        University of North Dakota School of Medicine & Health Sciences, Fargo, ND

        Publications
        Topics
        Sections
        Author and Disclosure Information

        University of North Dakota School of Medicine & Health Sciences, Fargo, ND

        Author and Disclosure Information

        University of North Dakota School of Medicine & Health Sciences, Fargo, ND

        Despite advances in medical science, pneumonia remains a major cause of morbidity and mortality. In 2017, 49,157 patients in the United States died from the disease.1 Pneumonia can be classified as community-acquired, hospital-acquired, or ventilator-associated. Another category, healthcare-associated pneumonia, was included in an earlier Infectious Diseases Society of America (IDSA) and American Thoracic Society (ATS) guideline but was removed from the 2016 guideline because there was no clear evidence that patients diagnosed with healthcare-associated pneumonia were at higher risk for harboring multidrug-resistant pathogens.2 This review is the first of 2 articles focusing on the management of community-acquired pneumonia (CAP). Here, we review CAP epidemiology, microbiology, predisposing factors, and diagnosis; current treatment and prevention of CAP are reviewed in a separate article.

        Definition and Epidemiology

        CAP is defined as an acute infection of the lungs that develops in patients who have not been hospitalized recently and have not had regular exposure to the health care system.3 A previously ambulatory patient who is diagnosed with pneumonia within 48 hours after admission also meets the criteria for CAP. Approximately 4 to 5 million cases of CAP are diagnosed in the United States annually.4 About 25% of CAP patients require hospitalization, and about 5% to 10% of these patients are admitted to the intensive care unit (ICU).5 In-hospital mortality is considerable (~10% in population-based studies),6 and 30-day mortality was found to be as high as 23% in a review by File and Marrie.7 CAP also confers a high risk of long-term morbidity and mortality compared with the general population who have never had CAP, irrespective of age.8

        Causative Organisms

        Numerous microorganisms can cause CAP. Common causes and less common causes are delineated in Table 1. Until recently, many studies had demonstrated that pneumococcus was the most common cause of CAP. However, in the CDC Etiology of Pneumonia in the Community (EPIC) study team’s 2015 prospective, multicenter, population-based study, no pathogen was detected in the majority of patients diagnosed with CAP requiring hospitalization. The most common pathogens they detected were rhinovirus (9%), followed by influenza virus (6%) and pneumococcus (5%).9 Factors considered to be contributing to the decrease in the percentage of pneumococcus in patients diagnosed with CAP are the widespread use of pneumococcal vaccine and reduced rates of smoking.10,11

        PDBR01609001_t1.PNG

        Predisposing Factors

        Most people diagnosed with CAP have 1 or more predisposing factors (Table 2).12,13 Patients who develop CAP typically have a combination of these predisposing factors rather than a single factor. Aging, in combination with other risk factors, increases the susceptibility of a person to pneumonia.

        PDBR01609001_t2.PNG

        Clinical Signs and Symptoms

        Symptoms of CAP include fever, chills, rigors, fatigue, anorexia, diaphoresis, dyspnea, cough (with or without sputum production), and pleuritic chest pain. There is no individual symptom or cluster of symptoms that can absolutely differentiate pneumonia from other acute respiratory diseases, including upper and lower respiratory infections. However, patients presenting with the constellation of symptoms of fever ≥ 100°F (37.8°C), productive cough, and tachycardia is more suggestive of pneumonia.14 Abnormal vital signs include fever, hypothermia, tachypnea, tachycardia, and oxygen desaturation. Auscultation of the chest reveals crackles or other adventitious breath sounds. Elderly patients with pneumonia report a significantly lower number of both respiratory and nonrespiratory symptoms compared with younger patients. Clinicians should be aware of this phenomenon to avoid delayed diagnosis and treatment.15

         

         

        Imaging Evaluation

        The presence of a pulmonary consolidation or an infiltrate on chest radiograph is required to diagnose CAP, and a chest radiograph should be obtained when CAP is suspected.16 However, there is no pattern of radiographic abnormalities reliable enough to differentiate infectious pneumonia from noninfectious causes.17

        There are case reports and case series demonstrating false-negative plain chest radiographs in dehydrated patients18 or in patients in a neutropenic state. However, animal studies have shown that dogs challenged with pneumococcus showed abnormal pulmonary shadow, suggestive of pneumonia, regardless of hydration status.19 There is also no reliable scientific evidence to support the notion that severe neutropenia can cause false-negative radiographs because of the inability to develop an acute inflammatory reaction in the lungs.20

        A chest computed tomography (CT) scan is more sensitive than a plain chest radiograph in detecting pneumonia. Therefore, a chest CT should be performed in a patient with negative plain chest radiograph when pneumonia is still highly suspected.21 A chest CT scan is also more sensitive in detecting cavitation, adenopathy, interstitial disease, and empyema. It also has the advantage of better defining anatomical changes than plain films.22

        Because improvement of pulmonary opacities in patients with CAP lags behind clinical improvement, repeating chest imaging studies is not recommended in patients who demonstrate clinical improvement. Clearing of pulmonary infiltrate or consolidation sometimes can take 6 weeks or longer.23

        Laboratory Evaluation

        Generally, the etiologic agent of CAP cannot be determined solely on the basis of clinical signs and symptoms or imaging studies. Although routine microbiological testing for patients suspicious for CAP is not necessary for empirical treatment, determining the etiologic agent of the pneumonia allows the clinician to narrow the antibiotics from a broad-spectrum empirical regimen to specific pathogen-directed therapy. Determination of certain etiologic agents causing the pneumonia can have important public health implications (eg, Mycobacterium tuberculosis and influenza virus).24

        Sputum Gram Stain and Culture

        Sputum Gram stain is an inexpensive test that may identify pathogens that cause CAP (eg, Streptococcus pneumoniae and Haemophilus influenzae). A quality specimen is required. A sputum sample must contain more than 25 neutrophils and less than 10 squamous epithelial cells/low power field on Gram stain to be considered suitable for culture. The sensitivity and specificity of sputum Gram stain and culture are highly variable in different clinical settings (eg, outpatient setting, nursing home, ICU). Reed et al’s meta-analysis of patients diagnosed with CAP in the United States showed the sensitivity and specificity of sputum Gram stain (compared with sputum culture) ranged from 15% to 100% and 11% to 100%, respectively.24 In cases of proven bacteremic pneumococcal pneumonia, positive cultures from sputum samples were positive less than 50% of the time.25

        For patients who cannot provide sputum samples or are intubated, deep-suction aspirate or bronchoalveolar lavage through a bronchoscopic procedure may be necessary to obtain pulmonary secretion for Gram stain and culture. Besides bacterial culture, sputum samples can also be sent for fungal and mycobacterial cultures and acid-fast stain, if deemed clinically necessary.

        The 2019 ATS/IDSA guidelines for diagnosis and treatment of adults with CAP recommend sputum culture in patients with severe disease and in all inpatients empirically treated for MRSA or Pseudomonas aeruginosa.26

         

         

        Blood Culture

        Because the positivity rate of blood culture in patients who are suspected to have pneumonia but not exposed to antimicrobial agents is low (5%–14%), blood cultures are not recommended for all patients with CAP. Another reason for not recommending blood culture is positive culture rarely leads to changes in antibiotic regimen in patients without underlying diseases.27 However, the 2019 ATS/IDSA guidelines recommend blood culture in patients with severe disease and in all inpatients treated empirically for MRSA or P. aeruginosa.26

        A multinational study published in 2008 examined 125 patients with pneumococcal bacteremic CAP versus 1847 patients with non-bacteremic CAP.28 Analysis of the data demonstrated no association between pneumococcal bacteremic CAP and time to clinical stability, length of hospital stay, all-cause mortality, or CAP-related mortality. The authors concluded that pneumococcal bacteremia does not increase the risk of poor outcomes in patients with CAP compared to non-bacteremic patients, and the presence of pneumococcal bacteremia should not deter de-escalation of therapy in clinically stable patients.

        Urinary Antigen Tests

        Urinary antigen tests may assist clinicians in narrowing antibiotic therapy when test results are positive. There are 2 US Food and Drug Administration–approved tests available to clinicians for detecting pneumococcal and Legionella antigen in urine. The test for Legionella pneumophila detects disease due to serogroup 1 only, which accounts for 80% of community-acquired Legionnaires’ disease. The sensitivity and specificity of the Legionella urine antigen test are 90% and 99%, respectively. The pneumococcal urine antigen test is less sensitive and specific than the Legionella urine antigen test (sensitivity 80% and specificity > 90%).29,30

        Advantages of the urinary antigen tests are that they are easily performed, results are available in less than an hour if done in-house, and results are not affected by prior exposure to antibiotics. However, the tests do not meet Clinical Laboratory Improvements Amendments criteria for waiver and must be performed by a technician in the laboratory. A multicenter, prospective surveillance study of hospitalized patients with CAP showed that the 2007 IDSA/ATS guidelines’ recommended indications for S. pneumoniae and L. pneumophila urinary antigen tests do not have sufficient sensitivity and specificity to identify patients with positive tests.31

        Polymerase Chain Reaction

        There are several FDA-approved polymerase chain reaction (PCR) tests commercially available to assist clinicians in diagnosing pneumonia. PCR testing of nasopharyngeal swabs for diagnosis of influenza has become standard in many US medical facilities. The great advantages of using PCR to diagnose influenza are its high sensitivity and specificity and rapid turnaround time. PCR can also be used to detect Legionella species, S. pneumonia, Mycoplasma pneumoniae, Chlamydophila pneumonia, and mycobacterial species.24

        One limitation of using PCR tests on respiratory specimens is that specimens can be contaminated with oral or upper airway flora, so the results must be interpreted with caution, bearing in mind that some of the pathogens isolated may be colonizers of the oral or upper airway flora.32

         

         

        Biologic Markers

        Two biologic markers—procalcitonin and C-reactive protein (CRP)—can be used in conjunction with history, physical examination, laboratory tests, and imaging studies to assist in the diagnosis and treatment of CAP.24 Procalcitonin is a peptide precursor of the hormone calcitonin that is released by parenchymal cells into the bloodstream, resulting in increased serum level in patients with bacterial infections. In contrast, there is no remarkable procalcitonin level increase with viral or noninfectious inflammation. The reference value of procalcitonin in the blood of an adult individual without infection or inflammation is < 0.15 ng/mL. In the blood, procalcitonin has a half-life of 25 to 30 hours. The quantitative immunoluminometric method (LUMI test, Brahms PCT, Berlin, Germany) is the preferred test to use because of its high sensitivity.33 A meta-analysis of 12 studies involving more than 2400 patients with CAP demonstrated that serum procalcitonin does not have sufficient sensitivity or specificity to distinguish between bacterial and nonbacterial pneumonia. The authors concluded that procalcitonin level cannot be used to decide whether an antibiotic should be administered.34

        A 2012 Cochrane meta-analysis that involved 4221 patients with acute respiratory infections (with half of the patients diagnosed with CAP) from 14 prospective trials found the use of procalcitonin test for antibiotic use significantly decreased median antibiotic exposure from 8 to 4 days without an increase in treatment failure, mortality rates in any clinical setting (eg, outpatient clinic, emergency room), or length of hospitalization.35 An update of the 2012 Cochrane review that examined the safety and efficacy of using procalcitonin for starting or stopping antibiotics again demonstrated procalcitonin use was associated with a reduction of antibiotic use (2.4 days).36 A prospective study conducted in France on 100 ICU patients showed that increased procalcitonin from day 1 to day 3 has a poor prognosis factor for severe CAP, whereas decreasing procalcitonin levels is associated with a favorable outcome.37

        Because of conflicting data, the 2019 ATS/IDSA guidelines do not recommend using procalcitonin to determine need for initial antibacterial therapy.26

        CRP is an acute phase protein produced by the liver. CRP level in the blood increases in response to acute infection or inflammation. Use of CRP in assisting diagnosis and guiding treatment of CAP is more limited in part due to its poor specificity. A prospective study conducted on 168 consecutive patients who presented with cough showed that a CRP level > 40 mg/L had a sensitivity and specificity of 70% and 90%, respectively.38

        Summary

        CAP remains a leading cause of hospitalization and death in the 21st century. Traditionally, pneumococcus has been considered the major pathogen causing CAP; however, the 2015 EPIC study found that S. pneumoniae was detected in only 5% of patients diagnosed with CAP. Despite the new findings, it is still recommended that empiric treatment for CAP target common typical bacteria (pneumococcus, H. influenzae, Moraxella catarrhalis) and atypical bacteria (M. pneumonia, C. pneumoniae, L. pneumophila).

        Because diagnosing pneumonia through history and clinical examination is less than 50% sensitive, a chest imaging study (a plain chest radiograph or a chest CT scan) is usually required to make the diagnosis. Laboratory tests, such as sputum Gram stain/culture, blood culture, urinary antigen tests, PCR test, procalcitonin, and CRP are important adjunctive diagnostic modalities to assist in the diagnosis and management of CAP. However, because no single test is sensitive and specific enough to be a stand-alone test, they should be used in conjunction with history, physical examination, and imaging studies.

        Despite advances in medical science, pneumonia remains a major cause of morbidity and mortality. In 2017, 49,157 patients in the United States died from the disease.1 Pneumonia can be classified as community-acquired, hospital-acquired, or ventilator-associated. Another category, healthcare-associated pneumonia, was included in an earlier Infectious Diseases Society of America (IDSA) and American Thoracic Society (ATS) guideline but was removed from the 2016 guideline because there was no clear evidence that patients diagnosed with healthcare-associated pneumonia were at higher risk for harboring multidrug-resistant pathogens.2 This review is the first of 2 articles focusing on the management of community-acquired pneumonia (CAP). Here, we review CAP epidemiology, microbiology, predisposing factors, and diagnosis; current treatment and prevention of CAP are reviewed in a separate article.

        Definition and Epidemiology

        CAP is defined as an acute infection of the lungs that develops in patients who have not been hospitalized recently and have not had regular exposure to the health care system.3 A previously ambulatory patient who is diagnosed with pneumonia within 48 hours after admission also meets the criteria for CAP. Approximately 4 to 5 million cases of CAP are diagnosed in the United States annually.4 About 25% of CAP patients require hospitalization, and about 5% to 10% of these patients are admitted to the intensive care unit (ICU).5 In-hospital mortality is considerable (~10% in population-based studies),6 and 30-day mortality was found to be as high as 23% in a review by File and Marrie.7 CAP also confers a high risk of long-term morbidity and mortality compared with the general population who have never had CAP, irrespective of age.8

        Causative Organisms

        Numerous microorganisms can cause CAP. Common causes and less common causes are delineated in Table 1. Until recently, many studies had demonstrated that pneumococcus was the most common cause of CAP. However, in the CDC Etiology of Pneumonia in the Community (EPIC) study team’s 2015 prospective, multicenter, population-based study, no pathogen was detected in the majority of patients diagnosed with CAP requiring hospitalization. The most common pathogens they detected were rhinovirus (9%), followed by influenza virus (6%) and pneumococcus (5%).9 Factors considered to be contributing to the decrease in the percentage of pneumococcus in patients diagnosed with CAP are the widespread use of pneumococcal vaccine and reduced rates of smoking.10,11

        PDBR01609001_t1.PNG

        Predisposing Factors

        Most people diagnosed with CAP have 1 or more predisposing factors (Table 2).12,13 Patients who develop CAP typically have a combination of these predisposing factors rather than a single factor. Aging, in combination with other risk factors, increases the susceptibility of a person to pneumonia.

        PDBR01609001_t2.PNG

        Clinical Signs and Symptoms

        Symptoms of CAP include fever, chills, rigors, fatigue, anorexia, diaphoresis, dyspnea, cough (with or without sputum production), and pleuritic chest pain. There is no individual symptom or cluster of symptoms that can absolutely differentiate pneumonia from other acute respiratory diseases, including upper and lower respiratory infections. However, patients presenting with the constellation of symptoms of fever ≥ 100°F (37.8°C), productive cough, and tachycardia is more suggestive of pneumonia.14 Abnormal vital signs include fever, hypothermia, tachypnea, tachycardia, and oxygen desaturation. Auscultation of the chest reveals crackles or other adventitious breath sounds. Elderly patients with pneumonia report a significantly lower number of both respiratory and nonrespiratory symptoms compared with younger patients. Clinicians should be aware of this phenomenon to avoid delayed diagnosis and treatment.15

         

         

        Imaging Evaluation

        The presence of a pulmonary consolidation or an infiltrate on chest radiograph is required to diagnose CAP, and a chest radiograph should be obtained when CAP is suspected.16 However, there is no pattern of radiographic abnormalities reliable enough to differentiate infectious pneumonia from noninfectious causes.17

        There are case reports and case series demonstrating false-negative plain chest radiographs in dehydrated patients18 or in patients in a neutropenic state. However, animal studies have shown that dogs challenged with pneumococcus showed abnormal pulmonary shadow, suggestive of pneumonia, regardless of hydration status.19 There is also no reliable scientific evidence to support the notion that severe neutropenia can cause false-negative radiographs because of the inability to develop an acute inflammatory reaction in the lungs.20

        A chest computed tomography (CT) scan is more sensitive than a plain chest radiograph in detecting pneumonia. Therefore, a chest CT should be performed in a patient with negative plain chest radiograph when pneumonia is still highly suspected.21 A chest CT scan is also more sensitive in detecting cavitation, adenopathy, interstitial disease, and empyema. It also has the advantage of better defining anatomical changes than plain films.22

        Because improvement of pulmonary opacities in patients with CAP lags behind clinical improvement, repeating chest imaging studies is not recommended in patients who demonstrate clinical improvement. Clearing of pulmonary infiltrate or consolidation sometimes can take 6 weeks or longer.23

        Laboratory Evaluation

        Generally, the etiologic agent of CAP cannot be determined solely on the basis of clinical signs and symptoms or imaging studies. Although routine microbiological testing for patients suspicious for CAP is not necessary for empirical treatment, determining the etiologic agent of the pneumonia allows the clinician to narrow the antibiotics from a broad-spectrum empirical regimen to specific pathogen-directed therapy. Determination of certain etiologic agents causing the pneumonia can have important public health implications (eg, Mycobacterium tuberculosis and influenza virus).24

        Sputum Gram Stain and Culture

        Sputum Gram stain is an inexpensive test that may identify pathogens that cause CAP (eg, Streptococcus pneumoniae and Haemophilus influenzae). A quality specimen is required. A sputum sample must contain more than 25 neutrophils and less than 10 squamous epithelial cells/low power field on Gram stain to be considered suitable for culture. The sensitivity and specificity of sputum Gram stain and culture are highly variable in different clinical settings (eg, outpatient setting, nursing home, ICU). Reed et al’s meta-analysis of patients diagnosed with CAP in the United States showed the sensitivity and specificity of sputum Gram stain (compared with sputum culture) ranged from 15% to 100% and 11% to 100%, respectively.24 In cases of proven bacteremic pneumococcal pneumonia, positive cultures from sputum samples were positive less than 50% of the time.25

        For patients who cannot provide sputum samples or are intubated, deep-suction aspirate or bronchoalveolar lavage through a bronchoscopic procedure may be necessary to obtain pulmonary secretion for Gram stain and culture. Besides bacterial culture, sputum samples can also be sent for fungal and mycobacterial cultures and acid-fast stain, if deemed clinically necessary.

        The 2019 ATS/IDSA guidelines for diagnosis and treatment of adults with CAP recommend sputum culture in patients with severe disease and in all inpatients empirically treated for MRSA or Pseudomonas aeruginosa.26

         

         

        Blood Culture

        Because the positivity rate of blood culture in patients who are suspected to have pneumonia but not exposed to antimicrobial agents is low (5%–14%), blood cultures are not recommended for all patients with CAP. Another reason for not recommending blood culture is positive culture rarely leads to changes in antibiotic regimen in patients without underlying diseases.27 However, the 2019 ATS/IDSA guidelines recommend blood culture in patients with severe disease and in all inpatients treated empirically for MRSA or P. aeruginosa.26

        A multinational study published in 2008 examined 125 patients with pneumococcal bacteremic CAP versus 1847 patients with non-bacteremic CAP.28 Analysis of the data demonstrated no association between pneumococcal bacteremic CAP and time to clinical stability, length of hospital stay, all-cause mortality, or CAP-related mortality. The authors concluded that pneumococcal bacteremia does not increase the risk of poor outcomes in patients with CAP compared to non-bacteremic patients, and the presence of pneumococcal bacteremia should not deter de-escalation of therapy in clinically stable patients.

        Urinary Antigen Tests

        Urinary antigen tests may assist clinicians in narrowing antibiotic therapy when test results are positive. There are 2 US Food and Drug Administration–approved tests available to clinicians for detecting pneumococcal and Legionella antigen in urine. The test for Legionella pneumophila detects disease due to serogroup 1 only, which accounts for 80% of community-acquired Legionnaires’ disease. The sensitivity and specificity of the Legionella urine antigen test are 90% and 99%, respectively. The pneumococcal urine antigen test is less sensitive and specific than the Legionella urine antigen test (sensitivity 80% and specificity > 90%).29,30

        Advantages of the urinary antigen tests are that they are easily performed, results are available in less than an hour if done in-house, and results are not affected by prior exposure to antibiotics. However, the tests do not meet Clinical Laboratory Improvements Amendments criteria for waiver and must be performed by a technician in the laboratory. A multicenter, prospective surveillance study of hospitalized patients with CAP showed that the 2007 IDSA/ATS guidelines’ recommended indications for S. pneumoniae and L. pneumophila urinary antigen tests do not have sufficient sensitivity and specificity to identify patients with positive tests.31

        Polymerase Chain Reaction

        There are several FDA-approved polymerase chain reaction (PCR) tests commercially available to assist clinicians in diagnosing pneumonia. PCR testing of nasopharyngeal swabs for diagnosis of influenza has become standard in many US medical facilities. The great advantages of using PCR to diagnose influenza are its high sensitivity and specificity and rapid turnaround time. PCR can also be used to detect Legionella species, S. pneumonia, Mycoplasma pneumoniae, Chlamydophila pneumonia, and mycobacterial species.24

        One limitation of using PCR tests on respiratory specimens is that specimens can be contaminated with oral or upper airway flora, so the results must be interpreted with caution, bearing in mind that some of the pathogens isolated may be colonizers of the oral or upper airway flora.32

         

         

        Biologic Markers

        Two biologic markers—procalcitonin and C-reactive protein (CRP)—can be used in conjunction with history, physical examination, laboratory tests, and imaging studies to assist in the diagnosis and treatment of CAP.24 Procalcitonin is a peptide precursor of the hormone calcitonin that is released by parenchymal cells into the bloodstream, resulting in increased serum level in patients with bacterial infections. In contrast, there is no remarkable procalcitonin level increase with viral or noninfectious inflammation. The reference value of procalcitonin in the blood of an adult individual without infection or inflammation is < 0.15 ng/mL. In the blood, procalcitonin has a half-life of 25 to 30 hours. The quantitative immunoluminometric method (LUMI test, Brahms PCT, Berlin, Germany) is the preferred test to use because of its high sensitivity.33 A meta-analysis of 12 studies involving more than 2400 patients with CAP demonstrated that serum procalcitonin does not have sufficient sensitivity or specificity to distinguish between bacterial and nonbacterial pneumonia. The authors concluded that procalcitonin level cannot be used to decide whether an antibiotic should be administered.34

        A 2012 Cochrane meta-analysis that involved 4221 patients with acute respiratory infections (with half of the patients diagnosed with CAP) from 14 prospective trials found the use of procalcitonin test for antibiotic use significantly decreased median antibiotic exposure from 8 to 4 days without an increase in treatment failure, mortality rates in any clinical setting (eg, outpatient clinic, emergency room), or length of hospitalization.35 An update of the 2012 Cochrane review that examined the safety and efficacy of using procalcitonin for starting or stopping antibiotics again demonstrated procalcitonin use was associated with a reduction of antibiotic use (2.4 days).36 A prospective study conducted in France on 100 ICU patients showed that increased procalcitonin from day 1 to day 3 has a poor prognosis factor for severe CAP, whereas decreasing procalcitonin levels is associated with a favorable outcome.37

        Because of conflicting data, the 2019 ATS/IDSA guidelines do not recommend using procalcitonin to determine need for initial antibacterial therapy.26

        CRP is an acute phase protein produced by the liver. CRP level in the blood increases in response to acute infection or inflammation. Use of CRP in assisting diagnosis and guiding treatment of CAP is more limited in part due to its poor specificity. A prospective study conducted on 168 consecutive patients who presented with cough showed that a CRP level > 40 mg/L had a sensitivity and specificity of 70% and 90%, respectively.38

        Summary

        CAP remains a leading cause of hospitalization and death in the 21st century. Traditionally, pneumococcus has been considered the major pathogen causing CAP; however, the 2015 EPIC study found that S. pneumoniae was detected in only 5% of patients diagnosed with CAP. Despite the new findings, it is still recommended that empiric treatment for CAP target common typical bacteria (pneumococcus, H. influenzae, Moraxella catarrhalis) and atypical bacteria (M. pneumonia, C. pneumoniae, L. pneumophila).

        Because diagnosing pneumonia through history and clinical examination is less than 50% sensitive, a chest imaging study (a plain chest radiograph or a chest CT scan) is usually required to make the diagnosis. Laboratory tests, such as sputum Gram stain/culture, blood culture, urinary antigen tests, PCR test, procalcitonin, and CRP are important adjunctive diagnostic modalities to assist in the diagnosis and management of CAP. However, because no single test is sensitive and specific enough to be a stand-alone test, they should be used in conjunction with history, physical examination, and imaging studies.

        References

        1. Centers for Disease Control and Prevention. National Center for Health Statistics. FastStats - Pneumonia. www.cdc.gov/nchs/fastats/pneumonia.htm. Accessed 16 September 2019.

        2. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63:e61-e111.

        3. Musher DM, Thorner AR. Community-acquired pneumonia. N Engl J Med. 2014;371:1619-1628.

        4. Mandell LA. Epidemiology and etiology of community-acquired pneumonia. Infect Dis Clin North Am. 2004;18:761-776.

        5. Hoare Z, Lim WS. Pneumonia: update on diagnosis and management. BMJ. 2006;332:1077-1079.

        6. Johnstone J, Marrie TJ, Eurich DT, Majumdar SR. Effect of pneumococcal vaccination in hospitalized adults with community-acquired pneumonia. Arch Intern Med. 2007;167:1938-1943.

        7. File TM Jr, Marrie TJ. Burden of community-acquired pneumonia in North American adults. Postgrad Med. 2010;122:130-141.

        8. Eurich DT, Marrie TJ, Minhas-Sandhu JK, Majumdar SR. Ten-year mortality after community-acquired pneumonia. a prospective cohort. Am J Respir Crit Care Med. 2015;192:597-604.

        9. Jain S, Self WH, Wunderink RG, et al. Community-acquired pneumonia requiring hospitalization among U.S. adults. N Engl J Med. 2015;373:415-427.

        10. Griffin MR, Zhu Y, Moore MR, et al. U.S. hospitalizations for pneumonia after a decade of pneumococcal vaccination. N Engl J Med. 2013;369:155-163.

        11. Nuorti JP, Butler JC, Farley MM, et al. Cigarette smoking and invasive pneumococcal disease. Active Bacterial Core Surveillance Team. N Engl J Med. 2000;342:681-689.

        12. Almirall J, Serra-Prat M, Bolíbar I, Balasso V. Risk factors for community-acquired pneumonia in adults: a systemic review of observational studies. Respiration. 2017;94:299-311.

        13. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Saunders; 2015:2310-2327.

        14. Diehr P, Wood RW, Bushyhead J, et al. Prediction of pneumonia in outpatients with acute cough--a statistical approach. J Chronic Dis. 1984;37:215-225.

        15. Metlay JP, Schulz R, Li YH, et al. Influence of age on symptoms at presentation in patients with community-acquired pneumonia. Arch Intern Med. 1997;157:1453-1459.

        16. Mandell LA, Wunderink RG, Anzueto A, et al. 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:S27-72.

        17. Jartti A, Rauvala E, Kauma H, et al. Chest imaging findings in hospitalized patients with H1N1 influenza. Acta Radiol. 2011;52:297-304.

        18. Basi SK, Marrie TJ, Huang JQ, Majumdar SR. Patients admitted to hospital with suspected pneumonia and normal chest radiographs: epidemiology, microbiology, and outcomes. Am J Med. 2004;117:305-311.

        19. Caldwell A, Glauser FL, Smith WR, et al. The effects of dehydration on the radiologic and pathologic appearance of experimental canine segmental pneumonia. Am Rev Respir Dis. 1975;112:651-656.

        20. Bartlett JG. Pneumonia. In: Barlett JG, editor. Management of Respiratory Tract Infections. Philadelphia: Lippincott, Williams & Wilkins; 2001:1-122.

        21. Claessens YE, Debray MP, Tubach F, et al. Early chest computed tomography scan to assist diagnosis and guide treatment decision for suspected community-acquired pneumonia. Am J Respir Crit Care Med. 2015;192:974-982.

        22. Wheeler JH, Fishman EK. Computed tomography in the management of chest infections: current status. Clin Infect Dis. 1996;23:232-240.

        23. Chesnutt MP. Pulmonary disorders. In: Papadakis MM, editor. Current Medical Diagnosis and Treatment. New York: McGraw-Hill; 2016:242-320.

        24. Mandell LW. Pneumonia. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:188-201.

        25. Reed WW, Byrd GS, Gates RH Jr, et al. Sputum gram’s stain in community-acquired pneumococcal pneumonia. A meta-analysis. West J Med. 1996;165:197-204.

        26. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200:e45-e67.

        27. Chalasani NP, Valdecanas MA, Gopal AK, et al. Clinical utility of blood cultures in adult patients with community-acquired pneumonia without defined underlying risks. Chest. 1995;108:932-936.

        28. Bordon J, Peyrani P, Brock GN, et al. The presence of pneumococcal bacteremia does not influence clinical outcomes in patients with community-acquired pneumonia: results from the Community-Acquired Pneumonia Organization (CAPO) International Cohort study. Chest. 2008;133:618-624.

        29. Helbig JH, Uldum SA, Bernander S, et al. Clinical utility of urinary antigen detection for diagnosis of community-acquired, travel-associated, and nosocomial legionnaires’ disease. J Clin Microbiol. 2003;41:838-840.

        30. Smith MD, Derrington P, Evans R, et al. Rapid diagnosis of bacteremic pneumococcal infections in adults by using the Binax NOW Streptococcus pneumoniae urinary antigen test: a prospective, controlled clinical evaluation. J Clin Microbiol. 2003;41:2810-2813.

        31. Bellew S, Grijalva CG, Williams DJ, et al. Pneumococcal and Legionella urinary antigen tests in community-acquired pneumonia: Prospective evaluation of indications for testing. Clin Infect Dis. 2019;68:2026-2033.

        32. Johansson N, Kalin M, Tiveljung-Lindell A, et al. Etiology of community-acquired pneumonia: increased microbiological yield with new diagnostic methods. Clin Infect Dis. 2010;50:202-209.

        33. Gilbert DN. Procalcitonin as a biomarker in respiratory tract infection. Clin Infect Dis. 2011;52 Suppl 4:S346-350.

        34. Kamat IS Ramachandran V, Eswaran H, et al. Procalcitonin to distinguish viral from bacterial pneumonia: A systematic review and meta-analysis. Clin Infect Dis. 2019 Jun 25. [Epub ahead of print]

        35. Schuetz P, Muller B, Christ-Crain M, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2012;(9):CD007498.

        36. Schuetz P, Wirz Y, Sager R, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2017;10:CD007498.

        37. Boussekey N, Leroy O, Alfandari S, et al. Procalcitonin kinetics in the prognosis of severe community-acquired pneumonia. Intensive Care Med. 2006;32:469-472.

        38. Flanders SA, Stein J, Shochat G, et al. Performance of a bedside C-reactive protein test in the diagnosis of community-acquired pneumonia in adults with acute cough. Am J Med. 2004;116:529-535.

        References

        1. Centers for Disease Control and Prevention. National Center for Health Statistics. FastStats - Pneumonia. www.cdc.gov/nchs/fastats/pneumonia.htm. Accessed 16 September 2019.

        2. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 Clinical Practice Guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63:e61-e111.

        3. Musher DM, Thorner AR. Community-acquired pneumonia. N Engl J Med. 2014;371:1619-1628.

        4. Mandell LA. Epidemiology and etiology of community-acquired pneumonia. Infect Dis Clin North Am. 2004;18:761-776.

        5. Hoare Z, Lim WS. Pneumonia: update on diagnosis and management. BMJ. 2006;332:1077-1079.

        6. Johnstone J, Marrie TJ, Eurich DT, Majumdar SR. Effect of pneumococcal vaccination in hospitalized adults with community-acquired pneumonia. Arch Intern Med. 2007;167:1938-1943.

        7. File TM Jr, Marrie TJ. Burden of community-acquired pneumonia in North American adults. Postgrad Med. 2010;122:130-141.

        8. Eurich DT, Marrie TJ, Minhas-Sandhu JK, Majumdar SR. Ten-year mortality after community-acquired pneumonia. a prospective cohort. Am J Respir Crit Care Med. 2015;192:597-604.

        9. Jain S, Self WH, Wunderink RG, et al. Community-acquired pneumonia requiring hospitalization among U.S. adults. N Engl J Med. 2015;373:415-427.

        10. Griffin MR, Zhu Y, Moore MR, et al. U.S. hospitalizations for pneumonia after a decade of pneumococcal vaccination. N Engl J Med. 2013;369:155-163.

        11. Nuorti JP, Butler JC, Farley MM, et al. Cigarette smoking and invasive pneumococcal disease. Active Bacterial Core Surveillance Team. N Engl J Med. 2000;342:681-689.

        12. Almirall J, Serra-Prat M, Bolíbar I, Balasso V. Risk factors for community-acquired pneumonia in adults: a systemic review of observational studies. Respiration. 2017;94:299-311.

        13. Janoff EM. Streptococcus pneumonia. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas and Bennett’s Principles and Practice of Infectious Diseases. 8th ed. Philadelphia: Saunders; 2015:2310-2327.

        14. Diehr P, Wood RW, Bushyhead J, et al. Prediction of pneumonia in outpatients with acute cough--a statistical approach. J Chronic Dis. 1984;37:215-225.

        15. Metlay JP, Schulz R, Li YH, et al. Influence of age on symptoms at presentation in patients with community-acquired pneumonia. Arch Intern Med. 1997;157:1453-1459.

        16. Mandell LA, Wunderink RG, Anzueto A, et al. 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:S27-72.

        17. Jartti A, Rauvala E, Kauma H, et al. Chest imaging findings in hospitalized patients with H1N1 influenza. Acta Radiol. 2011;52:297-304.

        18. Basi SK, Marrie TJ, Huang JQ, Majumdar SR. Patients admitted to hospital with suspected pneumonia and normal chest radiographs: epidemiology, microbiology, and outcomes. Am J Med. 2004;117:305-311.

        19. Caldwell A, Glauser FL, Smith WR, et al. The effects of dehydration on the radiologic and pathologic appearance of experimental canine segmental pneumonia. Am Rev Respir Dis. 1975;112:651-656.

        20. Bartlett JG. Pneumonia. In: Barlett JG, editor. Management of Respiratory Tract Infections. Philadelphia: Lippincott, Williams & Wilkins; 2001:1-122.

        21. Claessens YE, Debray MP, Tubach F, et al. Early chest computed tomography scan to assist diagnosis and guide treatment decision for suspected community-acquired pneumonia. Am J Respir Crit Care Med. 2015;192:974-982.

        22. Wheeler JH, Fishman EK. Computed tomography in the management of chest infections: current status. Clin Infect Dis. 1996;23:232-240.

        23. Chesnutt MP. Pulmonary disorders. In: Papadakis MM, editor. Current Medical Diagnosis and Treatment. New York: McGraw-Hill; 2016:242-320.

        24. Mandell LW. Pneumonia. In: Kasper DF, editor. Harrison’s Infectious Diseases. 1st ed. New York: McGraw-Hill; 2010:188-201.

        25. Reed WW, Byrd GS, Gates RH Jr, et al. Sputum gram’s stain in community-acquired pneumococcal pneumonia. A meta-analysis. West J Med. 1996;165:197-204.

        26. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200:e45-e67.

        27. Chalasani NP, Valdecanas MA, Gopal AK, et al. Clinical utility of blood cultures in adult patients with community-acquired pneumonia without defined underlying risks. Chest. 1995;108:932-936.

        28. Bordon J, Peyrani P, Brock GN, et al. The presence of pneumococcal bacteremia does not influence clinical outcomes in patients with community-acquired pneumonia: results from the Community-Acquired Pneumonia Organization (CAPO) International Cohort study. Chest. 2008;133:618-624.

        29. Helbig JH, Uldum SA, Bernander S, et al. Clinical utility of urinary antigen detection for diagnosis of community-acquired, travel-associated, and nosocomial legionnaires’ disease. J Clin Microbiol. 2003;41:838-840.

        30. Smith MD, Derrington P, Evans R, et al. Rapid diagnosis of bacteremic pneumococcal infections in adults by using the Binax NOW Streptococcus pneumoniae urinary antigen test: a prospective, controlled clinical evaluation. J Clin Microbiol. 2003;41:2810-2813.

        31. Bellew S, Grijalva CG, Williams DJ, et al. Pneumococcal and Legionella urinary antigen tests in community-acquired pneumonia: Prospective evaluation of indications for testing. Clin Infect Dis. 2019;68:2026-2033.

        32. Johansson N, Kalin M, Tiveljung-Lindell A, et al. Etiology of community-acquired pneumonia: increased microbiological yield with new diagnostic methods. Clin Infect Dis. 2010;50:202-209.

        33. Gilbert DN. Procalcitonin as a biomarker in respiratory tract infection. Clin Infect Dis. 2011;52 Suppl 4:S346-350.

        34. Kamat IS Ramachandran V, Eswaran H, et al. Procalcitonin to distinguish viral from bacterial pneumonia: A systematic review and meta-analysis. Clin Infect Dis. 2019 Jun 25. [Epub ahead of print]

        35. Schuetz P, Muller B, Christ-Crain M, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2012;(9):CD007498.

        36. Schuetz P, Wirz Y, Sager R, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2017;10:CD007498.

        37. Boussekey N, Leroy O, Alfandari S, et al. Procalcitonin kinetics in the prognosis of severe community-acquired pneumonia. Intensive Care Med. 2006;32:469-472.

        38. Flanders SA, Stein J, Shochat G, et al. Performance of a bedside C-reactive protein test in the diagnosis of community-acquired pneumonia in adults with acute cough. Am J Med. 2004;116:529-535.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Community-Acquired Pneumonia: Evaluation and Diagnosis
        Display Headline
        Community-Acquired Pneumonia: Evaluation and Diagnosis
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 September;16(3)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Fri, 09/27/2019 - 09:00
        Un-Gate On Date
        Fri, 09/27/2019 - 09:00
        Use ProPublica
        CFC Schedule Remove Status
        Fri, 09/27/2019 - 09:00
        Hide sidebar & use full width
        render the right sidebar.

        Malignant Pleural Effusion: Therapeutic Options and Strategies

        Article Type
        Changed
        Wed, 08/28/2019 - 11:54
        Display Headline
        Malignant Pleural Effusion: Therapeutic Options and Strategies

        Malignant pleural effusion (MPE) is a common clinical problem in patients with advanced stage cancer. Each year in the United States, more than 150,000 individuals are diagnosed with MPE, and there are approximately 126,000 admissions for MPE.1-3 Providing effective therapeutic management remains a challenge, and currently available therapeutic interventions are palliative rather than curative. This article, the second in a 2-part review of MPE, focuses on the available management options.

        Therapeutic Thoracentesis

        Evaluation of pleural fluid cytology is a crucial step in the diagnosis and staging of disease. As a result, large-volume fluid removal is often the first therapeutic intervention for patients who present with symptomatic effusions. A patient’s clinical response to therapeutic thoracentesis dictates which additional therapeutic options are appropriate for palliation. Lack of symptom relief suggests that other comorbid conditions or trapped lung physiology may be the primary cause of the patient’s symptoms and discourages more invasive interventions. Radiographic evidence of lung re-expansion after fluid removal is also an important predictor of success for potential pleurodesis.4,5

        There are no absolute contraindications to thoracentesis. However, caution should be used for patients with risk factors that may predispose to complications of pneumothorax and bleeding, such as coagulopathy, treatment with anticoagulation medications, thrombocytopenia, platelet dysfunction (eg, antiplatelet medications, uremia), positive pressure ventilation, and small effusion size. These factors are only relative contraindications, however, as thoracentesis can still be safely performed by experienced operators using guidance technology such as ultrasonography.

        A retrospective review of 1009 ultrasound-guided thoracenteses with risk factors of an international normalized ratio (INR) greater than 1.6, platelet values less than 50,000/μL, or both, reported an overall rate of hemorrhagic complication of 0.4%, with no difference between procedures performed with (n = 303) or without (n = 706) transfusion correction of coagulopathy or thrombocytopenia.6 A similar retrospective evaluation of 1076 ultrasound-guided thoracenteses, including 267 patients with an INR greater than 1.5 and 58 patients with a platelet count less than 50,000/μL, reported a 0% complication rate.7 Small case series have also demonstrated low hemorrhagic complication rates for thoracentesis in patients treated with clopidogrel8,9 and with increased bleeding risk from elevated INR (liver disease or warfarin therapy) and renal disease.10

        Complications from pneumothorax can similarly be affected by patient- and operator-dependent risk factors. Meta-analysis of 24 studies including 6605 thoracenteses demonstrated an overall pneumothorax rate of 6.0%, with 34.1% requiring chest tube insertion.11 Lower pneumothorax rates were associated with the use of ultrasound guidance (odds ratio, 0.3; 95% confidence interval, 0.2-0.7). Experienced operators also had fewer pneumothorax complications, though this factor was not significant in the studies directly comparing this variable. Therapeutic thoracentesis and use of a larger-bore needle were also significantly correlated with pneumothorax, while mechanical ventilation had a nonsignificant trend towards increased risk.

        Although there is no consensus on the volume of pleural fluid that may be safely removed, it is recommended not to remove more than 1.5 L during a procedure in order to avoid precipitating re-expansion pulmonary edema.2,12 However, re-expansion pulmonary edema rates remain low even when larger volumes are removed if the patient remains symptom-free during the procedure and pleural manometry pressure does not exceed 20 cm H2O.13 Patient symptoms alone, however, are neither a sensitive nor specific indicator that pleural pressures exceed 20 cm H2O.14 Use of excessive negative pressure during drainage, such as from a vacuum bottle, should also be avoided. Comparison of suction generated manually with a syringe versus a vacuum bottle suggests decreased complications with manual drainage, though the sample size in the supporting study was small relative to the infrequency of the complications being evaluated.15

        Given the low morbidity and noninvasive nature of the procedure, serial large-volume thoracentesis remains a viable therapeutic intervention for patients who are unable or unwilling to undergo more invasive interventions, especially for patients with a slow fluid re-accumulation rate or who are anticipated to have limited survival. Unfortunately, many symptomatic effusions will recur within a short interval time span, which necessitates repeat procedures.16,17 Therefore, factors such as poor symptom control, patient inconvenience, recurrent procedural risk, and utilization of medical resources need to be considered as well.

         

         

        Tunneled Pleural Catheter

        Tunneled pleural catheters (TPCs) are a potentially permanent and minimally invasive therapy which allow intermittent drainage of pleural fluid (Figure 1). The catheter is tunneled under the skin to prevent infection. A polyester cuff attached to the catheter is positioned within the tunnel and induces fibrosis around the catheter, thereby securing the catheter in place. Placement can be performed under local anesthesia at the patient’s bedside or in an outpatient procedure space. Fluid can then be drained via specialized drainage bottles or bags by the patient, a family member, or visiting home nurse. The catheter can also be removed in the event of a complication or the development of spontaneous pleurodesis.

        PDBR01608002_f1.PNG

        TPCs are an effective palliative management strategy for patients with recurrent effusions and are an efficacious alternative to pleurodesis.18-20 TPCs may be used in patients with poor prognosis or trapped lung or in those in whom prior pleurodesis has failed.21-23 Meta-analysis of 19 studies showed symptomatic improvement in 95.6% of patients, with development of spontaneous pleurodesis in 45.6% of patients (range, 11.8% to 76.4%) after an average of 52 days.24 However, most of the studies included in this analysis were retrospective case series. Development of spontaneous pleurodesis from TPC drainage in prospective, controlled trials has been considerably more modest, supporting a range of approximately 20% to 30% with routine drainage strategies.20,25-27 Spontaneous pleurodesis develops greater rapidity and frequency in patients undergoing daily drainage compared to every-other-day or symptom-directed drainage strategies.25,26 However, there is no appreciable improvement in quality of life scores with a specific drainage strategy. Small case series also demonstrate that TPC drainage may induce spontaneous pleurodesis in some patients initially presenting with trapped lung physiology.22

        Catheter placement can be performed successfully in the vast majority of patients.28 Increased bleeding risk, significant malignancy-related involvement of the skin and chest wall, and pleural loculations can complicate TPC placement. TPC-related complications are relatively uncommon, but include pneumothorax, catheter malfunction and obstruction, and infections including soft tissue and pleural space infections.24 In a multicenter retrospective series of 1021 patients, only 4.9% developed a TPC-related pleural infection.29 Over 94% were successfully managed with antibiotic therapy, and the TPC was able to be preserved in 54%. Staphylococcus aureus was the most common causative organism and was identified in 48% of cases. Of note, spontaneous pleurodesis occurred in 62% of cases following a pleural space infection, which likely occurred as sequelae of the inflammatory nature of the infection. Retrospective analysis suggests that the risk of TPC-related infections is not substantially higher for patients with higher risks of immunosuppression from chemotherapy or hematologic malignancies.30,31 Tumor metastasis along the catheter tract is a rare occurrence (< 1%), but is most notable with mesothelioma, which has an incidence as high as 10%.24,32 In addition, development of pleural loculations can impede fluid drainage and relief of dyspnea. Intrapleural instillation of fibrinolytics can be used to improve drainage and improve symptom palliation.33

        Pleurodesis

        Pleurodesis obliterates the potential pleural space by inducing inflammation and fibrosis, resulting in adherence of the visceral and parietal pleura together. This process can be induced through mechanical abrasion of the pleural surface, introduction of chemical sclerosants, or from prolonged use of a chest tube. Chemical sclerosants are the most commonly used method for MPEs and are introduced through a chest tube or under visual guidance such as medical thoracoscopy or video-assisted thoracoscopic surgery (VATS). The pleurodesis process is thought to occur by induction of a systemic inflammatory response with localized deposition of fibrin.34 Activation of fibroblasts and successful pleurodesis have been correlated with higher basic fibroblast growth factor (bFGF) levels in pleural fluid.35 Increased tumor burden is associated with lower bFGF levels, suggesting a possible mechanism for reduced pleurodesis success in these cases. Corticosteroids may reduce the likelihood of pleurodesis due to a reduction of inflammation, as demonstrated in a rabbit model using talc and doxycycline.36,37 Animal data also suggests that use of nonsteroidal anti-inflammatory drugs may hinder the likelihood of successful pleurodesis, but this has not been observed in humans.38,39

        Patients selected for pleurodesis should have significant symptom relief from large-volume removal of pleural fluid, good functional status, and evidence of full lung re-expansion after thoracentesis. Lack of visceral and parietal pleural apposition will prevent pleural adhesion from developing. As a result, trapped lung is associated with chemical pleurodesis failure and is an absolute contraindication to the procedure.4,5,12 The pleurodesis process typically requires 5 to 7 days, during which time the patient is hospitalized for chest tube drainage and pain control. When pleural fluid output diminishes, the chest tube is removed and the patient can be discharged. Modified protocols are now emerging which may shorten the required hospitalization associated with pleurodesis procedures.

         

         

        Pleurodesis Agents

        A variety of chemical sclerosants have been used for pleurodesis, including talc, bleomycin, tetracycline, doxycycline, iodopovidone, and mepacrine. Published data regarding these agents are heterogenous, with significant variability in reported outcomes. However, systematic review and meta-analysis suggests that talc is likely to have higher success rates compared to other agents or chest tube drainage alone for treatment of MPE.40,41

        Additional factors have been shown to be associated with pleurodesis outcomes. Pleurodesis success is negatively associated with low pleural pH, with receiver operating curve thresholds of 7.28 to 7.34.42,43 Trapped lung, large bulky tumor lining the pleural surfaces, and elevated adenosine deaminase levels are also associated with poor pleurodesis outcomes.4,5,12,35,43 In contrast, pleural fluid output less than 200 mL per day and the presence of EGFR (epidermal growth factor receptor) mutation treated with targeted tyrosine kinase inhibitors are associated with successful pleurodesis.44,45

        The most common complications associated with chemical pleurodesis are fever and pain. Other potential complications include soft tissue infections at the chest tube site and of the pleural space, arrhythmias, cardiac arrest, myocardial infarction, and hypotension. Doxycycline is commonly associated with greater pleuritic pain than talc. Acute respiratory distress syndrome (ARDS), acute pneumonitis, and respiratory failure have been described with talc pleurodesis. ARDS secondary to talc pleurodesis occurs in 1% to 9% of cases, though this may be related to the use of ungraded talc. A prospective description of 558 patients treated with large particle talc (> 5 μm) reported no occurrences of ARDS, suggesting the safety of graded large particle talc.46

        Pleurodesis Methods

        Chest tube thoracostomy is an inpatient procedure performed under local anesthesia or conscious sedation. It can be used for measured, intermittent drainage of large effusions for immediate symptom relief, as well as to demonstrate complete lung re-expansion prior to instillation of a chemical sclerosant. Pleurodesis using a chest tube is performed by instillation of a slurry created by mixing the sclerosing agent of choice with 50 to 100 mL of sterile saline. This slurry is instilled into the pleural cavity through the chest tube. The chest tube is clamped for 1 to 2 hours before being reconnected to suction. Intermittent rotation of the patient has not been shown to improve distribution of the sclerosant or result in better procedural outcomes.47,48 Typically, a 24F to 32F chest tube is used because of the concern about obstruction of smaller bore tubes by fibrin plugs. A noninferiority study comparing 12F to 24F chest tubes for talc pleurodesis demonstrated a higher procedure failure rate with the 12F tube (30% versus 24%) and failed to meet noninferiority criteria.39 However, larger caliber tubes are also associated with greater patient discomfort compared to smaller bore tubes.

        Medical thoracoscopy and VATS are minimally invasive means to visualize the pleural space, obtain visually guided biopsy of the parietal pleura, perform lysis of adhesions, and introduce chemical sclerosants for pleurodesis (Figure 2). Medical thoracoscopy can be performed under local anesthesia with procedural sedation in an endoscopy suite or procedure room.

        PDBR01608002_f2.PNG
        In contrast, VATS is performed in an operating room setting and requires general anesthesia, intubation with a double-lumen endotracheal tube, and multiple trocar incisions. For medical thoracoscopy, the patient is placed in the lateral decubitus position. The medical thoracoscope is introduced into the pleural space through one or more trocars. Trocar sizes range from 5 to 13 mm depending on the type of thoracoscope used. The body of the thoracoscopes may be rigid or semi-rigid (Figure 3). Rigid thoracoscopes have direct (0°) and angled cameras, while semi-rigid thoracoscopes have a flexible tip that can be manipulated similar to a flexible bronchoscope to direct visualization and biopsies. Following the procedure, a chest tube is typically introduced through the trocar insertion site for drainage.

        PDBR01608002_f3.PNG

         

         

        VATS has several distinct and clinically important differences. The equipment is slightly larger but otherwise similar in concept to rigid medical thoracoscopes. A greater number of diagnostic and therapeutic options, such as diagnostic biopsy of lung parenchyma and select hilar lymph nodes, are also possible. However, VATS requires surgical training and is performed in an operating room setting, which necessitates additional ancillary and logistical support. VATS also uses at least 2 trocar insertion sites, requires general anesthesia, and utilizes single-lung ventilation through a double-lumen endotracheal tube. Procedure-related complications for medical thoracoscopy and VATS include pneumothorax, subcutaneous emphysema, fever, and pain.49

        Data comparing talc slurry versus talc poudrage are heterogenous, without clear advantage for either method. Reported rates of successful pleurodesis are generally in the range of 70% to 80% for both methods.19,20,40,50 There is a possible suggestion of benefit with talc poudrage compared to slurry, but data is lacking to support either as a definitive choice in current guidelines.12,51 An advantage of medical thoracoscopy or VATS is that pleural biopsy can be performed simultaneously, if necessary, thereby allowing both diagnostic and therapeutic interventions.52 Visualizing the thoracic cavity may also permit creation of optimal conditions for pleurodesis in select individuals by allowing access to loculated spaces and providing visual confirmation of complete drainage of pleural fluid and uniform distribution of the chemical sclerosant.

        Other Surgical Interventions

        Thoracotomy with decortication is rarely used as treatment of malignant effusions complicated by loculations or trapped lung due to the significantly increased procedural morbidity and mortality. Therefore, it is reserved for the limited population of patients in whom other therapeutic interventions have failed but who otherwise have significant symptoms with a long life expectancy. Mesothelioma is a specific situation in which variations of pleurectomy, such as radical pleurectomy with decortication, lung-sparing total pleurectomy, and extrapleural pneumonectomy (EPP), have been used as front-line therapy. The Mesothelioma and Radical Surgery (MARS) trial, the only randomized, controlled evaluation of EPP, demonstrated decreased median survival in patients treated by EPP compared to controls (14.4 months versus 19.5 months).53 EPP is also associated with high procedure-related morbidity and mortality rates of approximately 50% and 4% to 15%, respectively.54 While successful at achieving pleurodesis, use of EPP as a treatment for mesothelioma is now discouraged.53,55 Less invasive surgical approaches, such as pleurectomy with decortication, are able to palliate symptoms with significantly less operative risk.56

        Management Considerations

        Selection of Therapeutic Interventions

        The ideal management strategy provides both immediate and long-term symptom palliation, has minimal associated morbidity and side effects, minimizes hospitalization time and clinic visits, avoids the risks and inconvenience of recurring procedures, is inexpensive, and minimizes utilization of medical resources. Unfortunately, no single palliation methodology fits these needs for all patients. When evaluating therapeutic options for patients with MPE, it is important to consider factors such as the severity of symptoms, fluid quantity, fluid re-accumulation rate, pleural physiology, functional status, overall prognosis, and anticipated response of the malignancy to therapy. One example management algorithm (Figure 4) demonstrates the impact of these variables on the appropriate treatment options. However, this is a simplified algorithm and the selected palliation strategy should be decided upon after shared decision-making between the patient and physician and should fit within the context of the patient’s desired goals of care. It is also crucial for patients to understand that these therapeutic interventions are palliative rather than curative.

        PDBR01608002_f4.PNG

        When compared directly with pleurodesis, TPC provides similar control of symptoms but with a reduction in hospital length of stay by a median of 3.5 to 5.5 days.19,57 In a nonrandomized trial where patients chose palliation by TPC or talc pleurodesis, more TPC patients had a significant immediate improvement in quality of life and dyspnea after the first 7 days of therapy.58 This is reasonably attributed to the differences between the immediate relief from fluid drainage after TPC placement compared to the time required for pleural symphysis to occur with pleurodesis. However, control of dyspnea symptoms is similar between the 2 strategies after 6 weeks.19 Therefore, both TPC and pleurodesis strategies are viewed as first-line options for patients with expandable lung and no prior palliative interventions for MPE.59

         

         

        Pleural adhesions and trapped lung also pose specific dilemmas. Pleural adhesions can create loculated fluid pockets, thereby complicating drainage by thoracentesis or TPC and hindering dispersal of pleurodesis agents. Adhesiolysis by medical thoracoscopy or VATS may be useful in these patients to free up the pleural space and improve efficacy of long-term drainage options or facilitate pleurodesis. Intrapleural administration of fibrinolytics, such as streptokinase and urokinase, has also been used for treatment of loculated effusions and may improve drainage of pleural fluid and lung re-expansion.60-63 However routine use of intrapleural fibrinolytics with pleurodesis has not been shown to be beneficial. In a randomized comparison using intrapleural urokinase prior to pleurodesis for patients with septated malignant pleural effusions, no difference in pleurodesis outcomes were identified.63 As a result, TPC is the preferred palliation approach for patients with trapped lung physiology.51,59

        Combination Strategies

        Combinations of different therapeutic interventions are being evaluated as a means for patients to achieve long-term benefits from pleurodesis while minimizing hospitalization time. One strategy using simultaneous treatment with thoracoscopic talc poudrage and insertion of a large-bore chest tube and TPC has been shown to permit early removal of the chest tube and discharge home using the TPC for continued daily pleural drainage. This “rapid pleurodesis” strategy has an 80% to 90% successful pleurodesis rate, permitting removal of the TPC at a median of 7 to 10 days.64,65 With this approach, median hospitalization length of stay was approximately 2 days. While there was no control arm in these early reports with limited sample sizes, the pleurodesis success rate and length of hospitalization compare favorably to other published studies. A prospective, randomized trial of TPC versus an outpatient regimen of talc slurry via TPC has also shown promise, with successful pleurodesis after 35 days in 43% of those treated with the combination of talc slurry and TPC compared to only 23% in those treated by TPC alone.27

        Another novel approach to obtain the benefits of both TPC and pleurodesis strategies is the use of drug-eluting TPC to induce inflammation and promote adhesion of the visceral and parietal pleura. An early report of slow-release silver nitrate (AgNO3) –coated TPC demonstrated an encouraging 89% spontaneous pleurodesis rate after a median of 4 days in the small subgroup of patients with fully expandable lung.66 Device-related adverse events were relatively high at 24.6%, though only one was deemed a serious adverse event. Additional studies of these novel and combination strategies are ongoing at this time.

        Costs

        While cost of care is not a consideration in the decision-making for individual patients, it is important from a systems-based perspective. Upfront costs for pleurodesis are generally higher due to the facility and hospitalization costs, whereas TPC have ongoing costs for drainage bottles and supplies. In a prospective, randomized trial of TPC versus talc pleurodesis, there was no appreciable difference in overall costs between the 2 approaches.67 The cost of TPC was significantly less, however, for patients with a shorter survival of less than 14 weeks.

        Readmissions

        Subsequent hospitalization requirements beyond just the initial treatment for a MPE remains another significant consideration for this patient population. A prospective, randomized trial comparing TPC to talc pleurodesis demonstrated a reduction in total all-cause hospital stay for TPC, with a median all-cause hospitalization time of 10 days for patients treated with TPC compared to 12 days for the talc pleurodesis group.20 The primary difference in the number of hospitalization days was due to a difference in effusion-related hospital days (median 1 versus 4 days, respectively), which was primarily comprised of the initial hospitalization. In addition, fewer patients treated with TPC required subsequent ipsilateral invasive procedures (4.1% versus 22.5%, respectively). However, it is important to note that the majority of all-cause hospital days were not effusion-related, demonstrating that this population has a high utilization of acute inpatient services for other reasons related to their advanced malignancy. In a study of regional hospitals in the United States, 38.3% of patients admitted for a primary diagnosis of MPE were readmitted within 30 days.68 There was remarkably little variability in readmission rates among hospitals, despite differences in factors such as institution size, location, patient distribution, and potential practice differences. This suggests that utilization of palliation strategies for MPE are only one component related to hospitalization in this population. Even at the best performing hospitals, there are significant common drivers for readmission that are not addressed. Therefore, additional effort should be focused on addressing aspects of care beyond just the palliation of MPE that predispose this population to requiring frequent treatment in an acute care setting.

         

         

        Conclusion

        MPEs represent advanced stage disease and frequently adversely affect a patient’s quality of life. The treating clinician has access to a variety of therapeutic options, though no single intervention strategy is universally superior in all circumstances. Initial thoracentesis is important in evaluating whether removal of a large volume of fluid provides significant symptom relief and restores functional status. Both talc pleurodesis and TPC provide similar control of symptoms and are first-line approaches for symptomatic patients with MPE and fully expandable lungs. Pleurodesis is associated with greater procedure-related risk and length of hospitalization and is contraindicated in patients with trapped lung, but does not require long-term catheter care or disposable resources. Determination of the appropriate therapeutic management strategy requires careful evaluation of the patient’s clinical situation and informed discussion with the patient to make sure that the treatment plan fits within the context of their goals of medical care.

        References

        1. Antony VB, Loddenkemper R, Astoul P, et al. Management of malignant pleural effusions. Eur Respir J. 2001;18:402-419.

        2. Society AT. Management of malignant pleural effusions. Am J Respir Crit Care Med. 2000;162:1987-2001.

        3. Taghizadeh N, Fortin M, Tremblay A. US hospitalizations for malignant pleural effusions: data from the 2012 National Inpatient Sample. Chest. 2017;151:845-854.

        4. Adler RH, Sayek I. Treatment of malignant pleural effusion: a method using tube thoracostomy and talc. Ann Thorac Surg. 1976;22:8-15.

        5. Villanueva AG, Gray AW, Shahian DM, et al. Efficacy of short term versus long term tube thoracostomy drainage before tetracycline pleurodesis in the treatment of malignant pleural effusions. Thorax. 1994;49:23-25.

        6. Hibbert RM, Atwell TD, Lekah A, et al. Safety of ultrasound-guided thoracentesis in patients with abnormal preprocedural coagulation parameters. Chest. 2013;144:456-463.

        7. Patel MD, Joshi SD. Abnormal preprocedural international normalized ratio and platelet counts are not associated with increased bleeding complications after ultrasound-guided thoracentesis. AJR Am J Roentgenol. 2011;197:W164-168.

        8. Zalt MB, Bechara RI, Parks C, Berkowitz DM. Effect of routine clopidogrel use on bleeding complications after ultrasound-guided thoracentesis. J Bronchology Interv Pulmonol. 2012;19:284-287.

        9. Mahmood K, Shofer SL, Moser BK, et al. Hemorrhagic complications of thoracentesis and small-bore chest tube placement in patients taking clopidogrel. Ann Am Thorac Soc. 2014;11:73-79.

        10. Puchalski JT, Argento AC, Murphy TE, et al. The safety of thoracentesis in patients with uncorrected bleeding risk. Ann Am Thorac Soc. 2013;10:336-341.

        11. Gordon CE, Feller-Kopman D, Balk EM, Smetana GW. Pneumothorax following thoracentesis: a systematic review and meta-analysis. Arch Intern Med. 2010;170:332-339.

        12. Roberts ME, Neville E, Berrisford RG, et al; Group BPDG. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax. 2010;65 Suppl 2:ii32-40.

        13. Feller-Kopman D, Berkowitz D, Boiselle P, Ernst A. Large-volume thoracentesis and the risk of reexpansion pulmonary edema. Ann Thorac Surg. 2007;84:1656-1661.

        14. Feller-Kopman D, Walkey A, Berkowitz D, Ernst A. The relationship of pleural pressure to symptom development during therapeutic thoracentesis. Chest. 2006;129:1556-1560.

        15. Senitko M, Ray AS, Murphy TE, et al. Safety and tolerability of vacuum versus manual drainage during thoracentesis: a randomized trial. J Bronchology Interv Pulmonol. 2019;26:166-171.

        16. Ost DE, Niu J, Zhao H, et al. Quality gaps and comparative effectiveness of management strategies for recurrent malignant pleural effusions. Chest. 2018;153:438-452.

        17. Grosu HB, Molina S, Casal R, et al. Risk factors for pleural effusion recurrence in patients with malignancy. Respirology. 2019;24:76-82.

        18. Musani AI, Haas AR, Seijo L, et al. Outpatient management of malignant pleural effusions with small-bore, tunneled pleural catheters. Respiration. 2004;71:559-566.

        19. Davies HE, Mishra EK, Kahan BC, et al. Effect of an indwelling pleural catheter vs chest tube and talc pleurodesis for relieving dyspnea in patients with malignant pleural effusion: the TIME2 randomized controlled trial. JAMA. 2012;307:2383-2389.

        20. Thomas R, Fysh ETH, Smith NA, et al. Effect of an indwelling pleural catheter vs talc pleurodesis on hospitalization days in patients with malignant pleural effusion: the AMPLE randomized clinical trial. JAMA. 2017;318:1903-1912.

        21. Qureshi RA, Collinson SL, Powell RJ, et al. Management of malignant pleural effusion associated with trapped lung syndrome. Asian Cardiovasc Thorac Ann. 2008;16:120-123.

        22. Efthymiou CA, Masudi T, Thorpe JA, Papagiannopoulos K. Malignant pleural effusion in the presence of trapped lung. Five-year experience of PleurX tunnelled catheters. Interact Cardiovasc Thorac Surg. 2009;9:961-964.

        23. Sioris T, Sihvo E, Salo J, et al. Long-term indwelling pleural catheter (PleurX) for malignant pleural effusion unsuitable for talc pleurodesis. Eur J Surg Oncol. 2009;35:546-551.

        24. Van Meter ME, McKee KY, Kohlwes RJ. Efficacy and safety of tunneled pleural catheters in adults with malignant pleural effusions: a systematic review. J Gen Intern Med. 2011;26:70-76.

        25. Wahidi MM, Reddy C, Yarmus L, et al. Randomized trial of pleural fluid drainage frequency in patients with malignant pleural effusions. the ASAP trial. Am J Respir Crit Care Med. 2017;195:1050-1057.

        26. Muruganandan S, Azzopardi M, Fitzgerald DB, et al. Aggressive versus symptom-guided drainage of malignant pleural effusion via indwelling pleural catheters (AMPLE-2): an open-label randomised trial. Lancet Respir Med. 2018;6:671-680.

        27. Bhatnagar R, Keenan EK, Morley AJ, et al. Outpatient talc administration by indwelling pleural catheter for malignant effusion. N Engl J Med. 2018;378:1313-1322.

        28. Tremblay A, Michaud G. Single-center experience with 250 tunnelled pleural catheter insertions for malignant pleural effusion. Chest. 2006;129:362-368.

        29. Fysh ETH, Tremblay A, Feller-Kopman D, et al. Clinical outcomes of indwelling pleural catheter-related pleural infections: an international multicenter study. Chest. 2013;144:1597-1602.

        30. Morel A, Mishra E, Medley L, et al. Chemotherapy should not be withheld from patients with an indwelling pleural catheter for malignant pleural effusion. Thorax. 2011;66:448-449.

        31. Gilbert CR, Lee HJ, Skalski JH, et al. The use of indwelling tunneled pleural catheters for recurrent pleural effusions in patients with hematologic malignancies: a multicenter study. Chest. 2015;148:752-758.

        32. Thomas R, Budgeon CA, Kuok YJ, et al. Catheter tract metastasis associated with indwelling pleural catheters. Chest. 2014;146:557-562.

        33. Thomas R, Piccolo F, Miller D, et al. Intrapleural fibrinolysis for the treatment of indwelling pleural catheter-related symptomatic loculations: a multicenter observational study. Chest. 2015;148:746-751.

        34. Antony VB. Pathogenesis of malignant pleural effusions and talc pleurodesis. Pneumologie. 1999;53:493-498.

        35. Antony VB, Nasreen N, Mohammed KA, et al. Talc pleurodesis: basic fibroblast growth factor mediates pleural fibrosis. Chest. 2004;126:1522-1528.

        36. Xie C, Teixeira LR, McGovern JP, Light RW. Systemic corticosteroids decrease the effectiveness of talc pleurodesis. Am J Respir Crit Care Med. 1998;157(5 Pt 1):1441-1444.

        37. Teixeira LR, Wu W, Chang DS, Light RW. The effect of corticosteroids on pleurodesis induced by doxycycline in rabbits. Chest. 2002;121:216-219.

        38. Hunt I, Teh E, Southon R, Treasure T. Using non-steroidal anti-inflammatory drugs (NSAIDs) following pleurodesis. Interact Cardiovasc Thorac Surg. 2007;6:102-104.

        39. Rahman NM, Pepperell J, Rehal S, et al. Effect of opioids vs NSAIDs and larger vs smaller chest tube size on pain control and pleurodesis efficacy among patients with malignant pleural effusion: the TIME1 randomized clinical trial. JAMA. 2015;314:2641-2653.

        40. Clive AO, Jones HE, Bhatnagar R, Preston NJ, Maskell N. Interventions for the management of malignant pleural effusions: a network meta-analysis. Cochrane Database Syst Rev. 2016(5):CD010529.

        41. Tan C, Sedrakyan A, Browne J, et al. The evidence on the effectiveness of management for malignant pleural effusion: a systematic review. Eur J Cardiothorac Surg. 2006;29:829-838.

        42. Heffner JE, Nietert PJ, Barbieri C. Pleural fluid pH as a predictor of pleurodesis failure: analysis of primary data. Chest. 2000;117:87-95.

        43. Yildirim H, Metintas M, Ak G, et al. Predictors of talc pleurodesis outcome in patients with malignant pleural effusions. Lung Cancer. 2008;62:139-144.

        44. Aydogmus U, Ozdemir S, Cansever L, et al. Bedside talc pleurodesis for malignant pleural effusion: factors affecting success. Ann Surg Oncol. 2009;16:745-750.

        45. Guo H, Wan Y, Tian G, et al. EGFR mutations predict a favorable outcome for malignant pleural effusion of lung adenocarcinoma with Tarceva therapy. Oncol Rep. 2012;27:880-890.

        46. Janssen JP, Collier G, Astoul P, et al. Safety of pleurodesis with talc poudrage in malignant pleural effusion: a prospective cohort study. Lancet. 2007;369(9572):1535-1539.

        47. Dryzer SR, Allen ML, Strange C, Sahn SA. A comparison of rotation and nonrotation in tetracycline pleurodesis. Chest. 1993;104:1763-1766.

        48. Mager HJ, Maesen B, Verzijlbergen F, Schramel F. Distribution of talc suspension during treatment of malignant pleural effusion with talc pleurodesis. Lung Cancer. 2002;36:77-81.

        49. Hsia D, Musani AI. Interventional pulmonology. Med Clin North Am. 2011;95:1095-1114.

        50. Dresler CM, Olak J, Herndon JE, et al. Phase III intergroup study of talc poudrage vs talc slurry sclerosis for malignant pleural effusion. Chest. 2005;127:909-915.

        51. Bibby AC, Dorn P, Psallidas I, et al. ERS/EACTS statement on the management of malignant pleural effusions. Eur Respir J. 2018;52(1).

        52. Sakuraba M, Masuda K, Hebisawa A, et al. Diagnostic value of thoracoscopic pleural biopsy for pleurisy under local anaesthesia. ANZ J Surg. 2006;76:722-724.

        53. Treasure T, Lang-Lazdunski L, Waller D, et al. Extra-pleural pneumonectomy versus no extra-pleural pneumonectomy for patients with malignant pleural mesothelioma: clinical outcomes of the Mesothelioma and Radical Surgery (MARS) randomised feasibility study. Lancet Oncol. 2011;12:763-772.

        54. Zellos L, Jaklitsch MT, Al-Mourgi MA, Sugarbaker DJ. Complications of extrapleural pneumonectomy. Semin Thorac Cardiovasc Surg. 2007;19:355-359.

        55. Zahid I, Sharif S, Routledge T, Scarci M. Is pleurectomy and decortication superior to palliative care in the treatment of malignant pleural mesothelioma? Interact Cardiovasc Thorac Surg. 2011;12:812-817.

        56. Soysal O, Karaoğlanoğlu N, Demiracan S, et al. Pleurectomy/decortication for palliation in malignant pleural mesothelioma: results of surgery. Eur J Cardiothorac Surg. 1997;11:210-213.

        57. Putnam JB, Light RW, Rodriguez RM, et al. A randomized comparison of indwelling pleural catheter and doxycycline pleurodesis in the management of malignant pleural effusions. Cancer. 1999;86:1992-1999.

        58. Fysh ETH, Waterer GW, Kendall PA, et al. Indwelling pleural catheters reduce inpatient days over pleurodesis for malignant pleural effusion. Chest. 2012;142:394-400.

        59. Feller-Kopman DJ, Reddy CB, DeCamp MM, et al. Management of malignant pleural effusions. An official ATS/STS/STR clinical practice guideline. Am J Respir Crit Care Med. 2018;198:839-849.

        60. Davies CW, Traill ZC, Gleeson FV, Davies RJ. Intrapleural streptokinase in the management of malignant multiloculated pleural effusions. Chest. 1999;115:729-733.

        61. Hsu LH, Soong TC, Feng AC, Liu MC. Intrapleural urokinase for the treatment of loculated malignant pleural effusions and trapped lungs in medically inoperable cancer patients. J Thorac Oncol. 2006;1:460-467.

        62. Okur E, Baysungur V, Tezel C, et al. Streptokinase for malignant pleural effusions: a randomized controlled study. Asian Cardiovasc Thorac Ann. 2011;19:238-243.

        63. Mishra EK, Clive AO, Wills GH, et al. Randomized controlled trial of urokinase versus placebo for nondraining malignant pleural effusion. Am J Respir Crit Care Med. 2018;197:502-508.

        64. Reddy C, Ernst A, Lamb C, Feller-Kopman D. Rapid pleurodesis for malignant pleural effusions: a pilot study. Chest. 2011;139:1419-1423.

        65. Krochmal R, Reddy C, Yarmus L, et al. Patient evaluation for rapid pleurodesis of malignant pleural effusions. J Thorac Dis. 2016;8:2538-2543.

        66. Bhatnagar R, Zahan-Evans N, Kearney C, et al. A novel drug-eluting indwelling pleural catheter for the management of malignant effusions. Am J Respir Crit Care Med. 2018;197:136-138.

        67. Penz ED, Mishra EK, Davies HE, Manns BJ, Miller RF, Rahman NM. Comparing cost of indwelling pleural catheter vs talc pleurodesis for malignant pleural effusion. Chest. 2014;146:991-1000.

        68. Yang TS, Hsia DW, Chang DW. Patient- and hospital-level factors associated with readmission for malignant pleural effusion. J Oncol Pract. 2018;14:e547-e556.

        Author and Disclosure Information

        David Hsia, MD
        Health Sciences Associate Clinical Professor, Division of Respiratory and Critical Care Physiology and Medicine, Harbor-UCLA Medical Center, Torrance, CA

        Ali I. Musani, MD
        Professor of Medicine and Surgery, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Denver, CO

        Publications
        Topics
        Sections
        Author and Disclosure Information

        David Hsia, MD
        Health Sciences Associate Clinical Professor, Division of Respiratory and Critical Care Physiology and Medicine, Harbor-UCLA Medical Center, Torrance, CA

        Ali I. Musani, MD
        Professor of Medicine and Surgery, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Denver, CO

        Author and Disclosure Information

        David Hsia, MD
        Health Sciences Associate Clinical Professor, Division of Respiratory and Critical Care Physiology and Medicine, Harbor-UCLA Medical Center, Torrance, CA

        Ali I. Musani, MD
        Professor of Medicine and Surgery, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Denver, CO

        Malignant pleural effusion (MPE) is a common clinical problem in patients with advanced stage cancer. Each year in the United States, more than 150,000 individuals are diagnosed with MPE, and there are approximately 126,000 admissions for MPE.1-3 Providing effective therapeutic management remains a challenge, and currently available therapeutic interventions are palliative rather than curative. This article, the second in a 2-part review of MPE, focuses on the available management options.

        Therapeutic Thoracentesis

        Evaluation of pleural fluid cytology is a crucial step in the diagnosis and staging of disease. As a result, large-volume fluid removal is often the first therapeutic intervention for patients who present with symptomatic effusions. A patient’s clinical response to therapeutic thoracentesis dictates which additional therapeutic options are appropriate for palliation. Lack of symptom relief suggests that other comorbid conditions or trapped lung physiology may be the primary cause of the patient’s symptoms and discourages more invasive interventions. Radiographic evidence of lung re-expansion after fluid removal is also an important predictor of success for potential pleurodesis.4,5

        There are no absolute contraindications to thoracentesis. However, caution should be used for patients with risk factors that may predispose to complications of pneumothorax and bleeding, such as coagulopathy, treatment with anticoagulation medications, thrombocytopenia, platelet dysfunction (eg, antiplatelet medications, uremia), positive pressure ventilation, and small effusion size. These factors are only relative contraindications, however, as thoracentesis can still be safely performed by experienced operators using guidance technology such as ultrasonography.

        A retrospective review of 1009 ultrasound-guided thoracenteses with risk factors of an international normalized ratio (INR) greater than 1.6, platelet values less than 50,000/μL, or both, reported an overall rate of hemorrhagic complication of 0.4%, with no difference between procedures performed with (n = 303) or without (n = 706) transfusion correction of coagulopathy or thrombocytopenia.6 A similar retrospective evaluation of 1076 ultrasound-guided thoracenteses, including 267 patients with an INR greater than 1.5 and 58 patients with a platelet count less than 50,000/μL, reported a 0% complication rate.7 Small case series have also demonstrated low hemorrhagic complication rates for thoracentesis in patients treated with clopidogrel8,9 and with increased bleeding risk from elevated INR (liver disease or warfarin therapy) and renal disease.10

        Complications from pneumothorax can similarly be affected by patient- and operator-dependent risk factors. Meta-analysis of 24 studies including 6605 thoracenteses demonstrated an overall pneumothorax rate of 6.0%, with 34.1% requiring chest tube insertion.11 Lower pneumothorax rates were associated with the use of ultrasound guidance (odds ratio, 0.3; 95% confidence interval, 0.2-0.7). Experienced operators also had fewer pneumothorax complications, though this factor was not significant in the studies directly comparing this variable. Therapeutic thoracentesis and use of a larger-bore needle were also significantly correlated with pneumothorax, while mechanical ventilation had a nonsignificant trend towards increased risk.

        Although there is no consensus on the volume of pleural fluid that may be safely removed, it is recommended not to remove more than 1.5 L during a procedure in order to avoid precipitating re-expansion pulmonary edema.2,12 However, re-expansion pulmonary edema rates remain low even when larger volumes are removed if the patient remains symptom-free during the procedure and pleural manometry pressure does not exceed 20 cm H2O.13 Patient symptoms alone, however, are neither a sensitive nor specific indicator that pleural pressures exceed 20 cm H2O.14 Use of excessive negative pressure during drainage, such as from a vacuum bottle, should also be avoided. Comparison of suction generated manually with a syringe versus a vacuum bottle suggests decreased complications with manual drainage, though the sample size in the supporting study was small relative to the infrequency of the complications being evaluated.15

        Given the low morbidity and noninvasive nature of the procedure, serial large-volume thoracentesis remains a viable therapeutic intervention for patients who are unable or unwilling to undergo more invasive interventions, especially for patients with a slow fluid re-accumulation rate or who are anticipated to have limited survival. Unfortunately, many symptomatic effusions will recur within a short interval time span, which necessitates repeat procedures.16,17 Therefore, factors such as poor symptom control, patient inconvenience, recurrent procedural risk, and utilization of medical resources need to be considered as well.

         

         

        Tunneled Pleural Catheter

        Tunneled pleural catheters (TPCs) are a potentially permanent and minimally invasive therapy which allow intermittent drainage of pleural fluid (Figure 1). The catheter is tunneled under the skin to prevent infection. A polyester cuff attached to the catheter is positioned within the tunnel and induces fibrosis around the catheter, thereby securing the catheter in place. Placement can be performed under local anesthesia at the patient’s bedside or in an outpatient procedure space. Fluid can then be drained via specialized drainage bottles or bags by the patient, a family member, or visiting home nurse. The catheter can also be removed in the event of a complication or the development of spontaneous pleurodesis.

        PDBR01608002_f1.PNG

        TPCs are an effective palliative management strategy for patients with recurrent effusions and are an efficacious alternative to pleurodesis.18-20 TPCs may be used in patients with poor prognosis or trapped lung or in those in whom prior pleurodesis has failed.21-23 Meta-analysis of 19 studies showed symptomatic improvement in 95.6% of patients, with development of spontaneous pleurodesis in 45.6% of patients (range, 11.8% to 76.4%) after an average of 52 days.24 However, most of the studies included in this analysis were retrospective case series. Development of spontaneous pleurodesis from TPC drainage in prospective, controlled trials has been considerably more modest, supporting a range of approximately 20% to 30% with routine drainage strategies.20,25-27 Spontaneous pleurodesis develops greater rapidity and frequency in patients undergoing daily drainage compared to every-other-day or symptom-directed drainage strategies.25,26 However, there is no appreciable improvement in quality of life scores with a specific drainage strategy. Small case series also demonstrate that TPC drainage may induce spontaneous pleurodesis in some patients initially presenting with trapped lung physiology.22

        Catheter placement can be performed successfully in the vast majority of patients.28 Increased bleeding risk, significant malignancy-related involvement of the skin and chest wall, and pleural loculations can complicate TPC placement. TPC-related complications are relatively uncommon, but include pneumothorax, catheter malfunction and obstruction, and infections including soft tissue and pleural space infections.24 In a multicenter retrospective series of 1021 patients, only 4.9% developed a TPC-related pleural infection.29 Over 94% were successfully managed with antibiotic therapy, and the TPC was able to be preserved in 54%. Staphylococcus aureus was the most common causative organism and was identified in 48% of cases. Of note, spontaneous pleurodesis occurred in 62% of cases following a pleural space infection, which likely occurred as sequelae of the inflammatory nature of the infection. Retrospective analysis suggests that the risk of TPC-related infections is not substantially higher for patients with higher risks of immunosuppression from chemotherapy or hematologic malignancies.30,31 Tumor metastasis along the catheter tract is a rare occurrence (< 1%), but is most notable with mesothelioma, which has an incidence as high as 10%.24,32 In addition, development of pleural loculations can impede fluid drainage and relief of dyspnea. Intrapleural instillation of fibrinolytics can be used to improve drainage and improve symptom palliation.33

        Pleurodesis

        Pleurodesis obliterates the potential pleural space by inducing inflammation and fibrosis, resulting in adherence of the visceral and parietal pleura together. This process can be induced through mechanical abrasion of the pleural surface, introduction of chemical sclerosants, or from prolonged use of a chest tube. Chemical sclerosants are the most commonly used method for MPEs and are introduced through a chest tube or under visual guidance such as medical thoracoscopy or video-assisted thoracoscopic surgery (VATS). The pleurodesis process is thought to occur by induction of a systemic inflammatory response with localized deposition of fibrin.34 Activation of fibroblasts and successful pleurodesis have been correlated with higher basic fibroblast growth factor (bFGF) levels in pleural fluid.35 Increased tumor burden is associated with lower bFGF levels, suggesting a possible mechanism for reduced pleurodesis success in these cases. Corticosteroids may reduce the likelihood of pleurodesis due to a reduction of inflammation, as demonstrated in a rabbit model using talc and doxycycline.36,37 Animal data also suggests that use of nonsteroidal anti-inflammatory drugs may hinder the likelihood of successful pleurodesis, but this has not been observed in humans.38,39

        Patients selected for pleurodesis should have significant symptom relief from large-volume removal of pleural fluid, good functional status, and evidence of full lung re-expansion after thoracentesis. Lack of visceral and parietal pleural apposition will prevent pleural adhesion from developing. As a result, trapped lung is associated with chemical pleurodesis failure and is an absolute contraindication to the procedure.4,5,12 The pleurodesis process typically requires 5 to 7 days, during which time the patient is hospitalized for chest tube drainage and pain control. When pleural fluid output diminishes, the chest tube is removed and the patient can be discharged. Modified protocols are now emerging which may shorten the required hospitalization associated with pleurodesis procedures.

         

         

        Pleurodesis Agents

        A variety of chemical sclerosants have been used for pleurodesis, including talc, bleomycin, tetracycline, doxycycline, iodopovidone, and mepacrine. Published data regarding these agents are heterogenous, with significant variability in reported outcomes. However, systematic review and meta-analysis suggests that talc is likely to have higher success rates compared to other agents or chest tube drainage alone for treatment of MPE.40,41

        Additional factors have been shown to be associated with pleurodesis outcomes. Pleurodesis success is negatively associated with low pleural pH, with receiver operating curve thresholds of 7.28 to 7.34.42,43 Trapped lung, large bulky tumor lining the pleural surfaces, and elevated adenosine deaminase levels are also associated with poor pleurodesis outcomes.4,5,12,35,43 In contrast, pleural fluid output less than 200 mL per day and the presence of EGFR (epidermal growth factor receptor) mutation treated with targeted tyrosine kinase inhibitors are associated with successful pleurodesis.44,45

        The most common complications associated with chemical pleurodesis are fever and pain. Other potential complications include soft tissue infections at the chest tube site and of the pleural space, arrhythmias, cardiac arrest, myocardial infarction, and hypotension. Doxycycline is commonly associated with greater pleuritic pain than talc. Acute respiratory distress syndrome (ARDS), acute pneumonitis, and respiratory failure have been described with talc pleurodesis. ARDS secondary to talc pleurodesis occurs in 1% to 9% of cases, though this may be related to the use of ungraded talc. A prospective description of 558 patients treated with large particle talc (> 5 μm) reported no occurrences of ARDS, suggesting the safety of graded large particle talc.46

        Pleurodesis Methods

        Chest tube thoracostomy is an inpatient procedure performed under local anesthesia or conscious sedation. It can be used for measured, intermittent drainage of large effusions for immediate symptom relief, as well as to demonstrate complete lung re-expansion prior to instillation of a chemical sclerosant. Pleurodesis using a chest tube is performed by instillation of a slurry created by mixing the sclerosing agent of choice with 50 to 100 mL of sterile saline. This slurry is instilled into the pleural cavity through the chest tube. The chest tube is clamped for 1 to 2 hours before being reconnected to suction. Intermittent rotation of the patient has not been shown to improve distribution of the sclerosant or result in better procedural outcomes.47,48 Typically, a 24F to 32F chest tube is used because of the concern about obstruction of smaller bore tubes by fibrin plugs. A noninferiority study comparing 12F to 24F chest tubes for talc pleurodesis demonstrated a higher procedure failure rate with the 12F tube (30% versus 24%) and failed to meet noninferiority criteria.39 However, larger caliber tubes are also associated with greater patient discomfort compared to smaller bore tubes.

        Medical thoracoscopy and VATS are minimally invasive means to visualize the pleural space, obtain visually guided biopsy of the parietal pleura, perform lysis of adhesions, and introduce chemical sclerosants for pleurodesis (Figure 2). Medical thoracoscopy can be performed under local anesthesia with procedural sedation in an endoscopy suite or procedure room.

        PDBR01608002_f2.PNG
        In contrast, VATS is performed in an operating room setting and requires general anesthesia, intubation with a double-lumen endotracheal tube, and multiple trocar incisions. For medical thoracoscopy, the patient is placed in the lateral decubitus position. The medical thoracoscope is introduced into the pleural space through one or more trocars. Trocar sizes range from 5 to 13 mm depending on the type of thoracoscope used. The body of the thoracoscopes may be rigid or semi-rigid (Figure 3). Rigid thoracoscopes have direct (0°) and angled cameras, while semi-rigid thoracoscopes have a flexible tip that can be manipulated similar to a flexible bronchoscope to direct visualization and biopsies. Following the procedure, a chest tube is typically introduced through the trocar insertion site for drainage.

        PDBR01608002_f3.PNG

         

         

        VATS has several distinct and clinically important differences. The equipment is slightly larger but otherwise similar in concept to rigid medical thoracoscopes. A greater number of diagnostic and therapeutic options, such as diagnostic biopsy of lung parenchyma and select hilar lymph nodes, are also possible. However, VATS requires surgical training and is performed in an operating room setting, which necessitates additional ancillary and logistical support. VATS also uses at least 2 trocar insertion sites, requires general anesthesia, and utilizes single-lung ventilation through a double-lumen endotracheal tube. Procedure-related complications for medical thoracoscopy and VATS include pneumothorax, subcutaneous emphysema, fever, and pain.49

        Data comparing talc slurry versus talc poudrage are heterogenous, without clear advantage for either method. Reported rates of successful pleurodesis are generally in the range of 70% to 80% for both methods.19,20,40,50 There is a possible suggestion of benefit with talc poudrage compared to slurry, but data is lacking to support either as a definitive choice in current guidelines.12,51 An advantage of medical thoracoscopy or VATS is that pleural biopsy can be performed simultaneously, if necessary, thereby allowing both diagnostic and therapeutic interventions.52 Visualizing the thoracic cavity may also permit creation of optimal conditions for pleurodesis in select individuals by allowing access to loculated spaces and providing visual confirmation of complete drainage of pleural fluid and uniform distribution of the chemical sclerosant.

        Other Surgical Interventions

        Thoracotomy with decortication is rarely used as treatment of malignant effusions complicated by loculations or trapped lung due to the significantly increased procedural morbidity and mortality. Therefore, it is reserved for the limited population of patients in whom other therapeutic interventions have failed but who otherwise have significant symptoms with a long life expectancy. Mesothelioma is a specific situation in which variations of pleurectomy, such as radical pleurectomy with decortication, lung-sparing total pleurectomy, and extrapleural pneumonectomy (EPP), have been used as front-line therapy. The Mesothelioma and Radical Surgery (MARS) trial, the only randomized, controlled evaluation of EPP, demonstrated decreased median survival in patients treated by EPP compared to controls (14.4 months versus 19.5 months).53 EPP is also associated with high procedure-related morbidity and mortality rates of approximately 50% and 4% to 15%, respectively.54 While successful at achieving pleurodesis, use of EPP as a treatment for mesothelioma is now discouraged.53,55 Less invasive surgical approaches, such as pleurectomy with decortication, are able to palliate symptoms with significantly less operative risk.56

        Management Considerations

        Selection of Therapeutic Interventions

        The ideal management strategy provides both immediate and long-term symptom palliation, has minimal associated morbidity and side effects, minimizes hospitalization time and clinic visits, avoids the risks and inconvenience of recurring procedures, is inexpensive, and minimizes utilization of medical resources. Unfortunately, no single palliation methodology fits these needs for all patients. When evaluating therapeutic options for patients with MPE, it is important to consider factors such as the severity of symptoms, fluid quantity, fluid re-accumulation rate, pleural physiology, functional status, overall prognosis, and anticipated response of the malignancy to therapy. One example management algorithm (Figure 4) demonstrates the impact of these variables on the appropriate treatment options. However, this is a simplified algorithm and the selected palliation strategy should be decided upon after shared decision-making between the patient and physician and should fit within the context of the patient’s desired goals of care. It is also crucial for patients to understand that these therapeutic interventions are palliative rather than curative.

        PDBR01608002_f4.PNG

        When compared directly with pleurodesis, TPC provides similar control of symptoms but with a reduction in hospital length of stay by a median of 3.5 to 5.5 days.19,57 In a nonrandomized trial where patients chose palliation by TPC or talc pleurodesis, more TPC patients had a significant immediate improvement in quality of life and dyspnea after the first 7 days of therapy.58 This is reasonably attributed to the differences between the immediate relief from fluid drainage after TPC placement compared to the time required for pleural symphysis to occur with pleurodesis. However, control of dyspnea symptoms is similar between the 2 strategies after 6 weeks.19 Therefore, both TPC and pleurodesis strategies are viewed as first-line options for patients with expandable lung and no prior palliative interventions for MPE.59

         

         

        Pleural adhesions and trapped lung also pose specific dilemmas. Pleural adhesions can create loculated fluid pockets, thereby complicating drainage by thoracentesis or TPC and hindering dispersal of pleurodesis agents. Adhesiolysis by medical thoracoscopy or VATS may be useful in these patients to free up the pleural space and improve efficacy of long-term drainage options or facilitate pleurodesis. Intrapleural administration of fibrinolytics, such as streptokinase and urokinase, has also been used for treatment of loculated effusions and may improve drainage of pleural fluid and lung re-expansion.60-63 However routine use of intrapleural fibrinolytics with pleurodesis has not been shown to be beneficial. In a randomized comparison using intrapleural urokinase prior to pleurodesis for patients with septated malignant pleural effusions, no difference in pleurodesis outcomes were identified.63 As a result, TPC is the preferred palliation approach for patients with trapped lung physiology.51,59

        Combination Strategies

        Combinations of different therapeutic interventions are being evaluated as a means for patients to achieve long-term benefits from pleurodesis while minimizing hospitalization time. One strategy using simultaneous treatment with thoracoscopic talc poudrage and insertion of a large-bore chest tube and TPC has been shown to permit early removal of the chest tube and discharge home using the TPC for continued daily pleural drainage. This “rapid pleurodesis” strategy has an 80% to 90% successful pleurodesis rate, permitting removal of the TPC at a median of 7 to 10 days.64,65 With this approach, median hospitalization length of stay was approximately 2 days. While there was no control arm in these early reports with limited sample sizes, the pleurodesis success rate and length of hospitalization compare favorably to other published studies. A prospective, randomized trial of TPC versus an outpatient regimen of talc slurry via TPC has also shown promise, with successful pleurodesis after 35 days in 43% of those treated with the combination of talc slurry and TPC compared to only 23% in those treated by TPC alone.27

        Another novel approach to obtain the benefits of both TPC and pleurodesis strategies is the use of drug-eluting TPC to induce inflammation and promote adhesion of the visceral and parietal pleura. An early report of slow-release silver nitrate (AgNO3) –coated TPC demonstrated an encouraging 89% spontaneous pleurodesis rate after a median of 4 days in the small subgroup of patients with fully expandable lung.66 Device-related adverse events were relatively high at 24.6%, though only one was deemed a serious adverse event. Additional studies of these novel and combination strategies are ongoing at this time.

        Costs

        While cost of care is not a consideration in the decision-making for individual patients, it is important from a systems-based perspective. Upfront costs for pleurodesis are generally higher due to the facility and hospitalization costs, whereas TPC have ongoing costs for drainage bottles and supplies. In a prospective, randomized trial of TPC versus talc pleurodesis, there was no appreciable difference in overall costs between the 2 approaches.67 The cost of TPC was significantly less, however, for patients with a shorter survival of less than 14 weeks.

        Readmissions

        Subsequent hospitalization requirements beyond just the initial treatment for a MPE remains another significant consideration for this patient population. A prospective, randomized trial comparing TPC to talc pleurodesis demonstrated a reduction in total all-cause hospital stay for TPC, with a median all-cause hospitalization time of 10 days for patients treated with TPC compared to 12 days for the talc pleurodesis group.20 The primary difference in the number of hospitalization days was due to a difference in effusion-related hospital days (median 1 versus 4 days, respectively), which was primarily comprised of the initial hospitalization. In addition, fewer patients treated with TPC required subsequent ipsilateral invasive procedures (4.1% versus 22.5%, respectively). However, it is important to note that the majority of all-cause hospital days were not effusion-related, demonstrating that this population has a high utilization of acute inpatient services for other reasons related to their advanced malignancy. In a study of regional hospitals in the United States, 38.3% of patients admitted for a primary diagnosis of MPE were readmitted within 30 days.68 There was remarkably little variability in readmission rates among hospitals, despite differences in factors such as institution size, location, patient distribution, and potential practice differences. This suggests that utilization of palliation strategies for MPE are only one component related to hospitalization in this population. Even at the best performing hospitals, there are significant common drivers for readmission that are not addressed. Therefore, additional effort should be focused on addressing aspects of care beyond just the palliation of MPE that predispose this population to requiring frequent treatment in an acute care setting.

         

         

        Conclusion

        MPEs represent advanced stage disease and frequently adversely affect a patient’s quality of life. The treating clinician has access to a variety of therapeutic options, though no single intervention strategy is universally superior in all circumstances. Initial thoracentesis is important in evaluating whether removal of a large volume of fluid provides significant symptom relief and restores functional status. Both talc pleurodesis and TPC provide similar control of symptoms and are first-line approaches for symptomatic patients with MPE and fully expandable lungs. Pleurodesis is associated with greater procedure-related risk and length of hospitalization and is contraindicated in patients with trapped lung, but does not require long-term catheter care or disposable resources. Determination of the appropriate therapeutic management strategy requires careful evaluation of the patient’s clinical situation and informed discussion with the patient to make sure that the treatment plan fits within the context of their goals of medical care.

        Malignant pleural effusion (MPE) is a common clinical problem in patients with advanced stage cancer. Each year in the United States, more than 150,000 individuals are diagnosed with MPE, and there are approximately 126,000 admissions for MPE.1-3 Providing effective therapeutic management remains a challenge, and currently available therapeutic interventions are palliative rather than curative. This article, the second in a 2-part review of MPE, focuses on the available management options.

        Therapeutic Thoracentesis

        Evaluation of pleural fluid cytology is a crucial step in the diagnosis and staging of disease. As a result, large-volume fluid removal is often the first therapeutic intervention for patients who present with symptomatic effusions. A patient’s clinical response to therapeutic thoracentesis dictates which additional therapeutic options are appropriate for palliation. Lack of symptom relief suggests that other comorbid conditions or trapped lung physiology may be the primary cause of the patient’s symptoms and discourages more invasive interventions. Radiographic evidence of lung re-expansion after fluid removal is also an important predictor of success for potential pleurodesis.4,5

        There are no absolute contraindications to thoracentesis. However, caution should be used for patients with risk factors that may predispose to complications of pneumothorax and bleeding, such as coagulopathy, treatment with anticoagulation medications, thrombocytopenia, platelet dysfunction (eg, antiplatelet medications, uremia), positive pressure ventilation, and small effusion size. These factors are only relative contraindications, however, as thoracentesis can still be safely performed by experienced operators using guidance technology such as ultrasonography.

        A retrospective review of 1009 ultrasound-guided thoracenteses with risk factors of an international normalized ratio (INR) greater than 1.6, platelet values less than 50,000/μL, or both, reported an overall rate of hemorrhagic complication of 0.4%, with no difference between procedures performed with (n = 303) or without (n = 706) transfusion correction of coagulopathy or thrombocytopenia.6 A similar retrospective evaluation of 1076 ultrasound-guided thoracenteses, including 267 patients with an INR greater than 1.5 and 58 patients with a platelet count less than 50,000/μL, reported a 0% complication rate.7 Small case series have also demonstrated low hemorrhagic complication rates for thoracentesis in patients treated with clopidogrel8,9 and with increased bleeding risk from elevated INR (liver disease or warfarin therapy) and renal disease.10

        Complications from pneumothorax can similarly be affected by patient- and operator-dependent risk factors. Meta-analysis of 24 studies including 6605 thoracenteses demonstrated an overall pneumothorax rate of 6.0%, with 34.1% requiring chest tube insertion.11 Lower pneumothorax rates were associated with the use of ultrasound guidance (odds ratio, 0.3; 95% confidence interval, 0.2-0.7). Experienced operators also had fewer pneumothorax complications, though this factor was not significant in the studies directly comparing this variable. Therapeutic thoracentesis and use of a larger-bore needle were also significantly correlated with pneumothorax, while mechanical ventilation had a nonsignificant trend towards increased risk.

        Although there is no consensus on the volume of pleural fluid that may be safely removed, it is recommended not to remove more than 1.5 L during a procedure in order to avoid precipitating re-expansion pulmonary edema.2,12 However, re-expansion pulmonary edema rates remain low even when larger volumes are removed if the patient remains symptom-free during the procedure and pleural manometry pressure does not exceed 20 cm H2O.13 Patient symptoms alone, however, are neither a sensitive nor specific indicator that pleural pressures exceed 20 cm H2O.14 Use of excessive negative pressure during drainage, such as from a vacuum bottle, should also be avoided. Comparison of suction generated manually with a syringe versus a vacuum bottle suggests decreased complications with manual drainage, though the sample size in the supporting study was small relative to the infrequency of the complications being evaluated.15

        Given the low morbidity and noninvasive nature of the procedure, serial large-volume thoracentesis remains a viable therapeutic intervention for patients who are unable or unwilling to undergo more invasive interventions, especially for patients with a slow fluid re-accumulation rate or who are anticipated to have limited survival. Unfortunately, many symptomatic effusions will recur within a short interval time span, which necessitates repeat procedures.16,17 Therefore, factors such as poor symptom control, patient inconvenience, recurrent procedural risk, and utilization of medical resources need to be considered as well.

         

         

        Tunneled Pleural Catheter

        Tunneled pleural catheters (TPCs) are a potentially permanent and minimally invasive therapy which allow intermittent drainage of pleural fluid (Figure 1). The catheter is tunneled under the skin to prevent infection. A polyester cuff attached to the catheter is positioned within the tunnel and induces fibrosis around the catheter, thereby securing the catheter in place. Placement can be performed under local anesthesia at the patient’s bedside or in an outpatient procedure space. Fluid can then be drained via specialized drainage bottles or bags by the patient, a family member, or visiting home nurse. The catheter can also be removed in the event of a complication or the development of spontaneous pleurodesis.

        PDBR01608002_f1.PNG

        TPCs are an effective palliative management strategy for patients with recurrent effusions and are an efficacious alternative to pleurodesis.18-20 TPCs may be used in patients with poor prognosis or trapped lung or in those in whom prior pleurodesis has failed.21-23 Meta-analysis of 19 studies showed symptomatic improvement in 95.6% of patients, with development of spontaneous pleurodesis in 45.6% of patients (range, 11.8% to 76.4%) after an average of 52 days.24 However, most of the studies included in this analysis were retrospective case series. Development of spontaneous pleurodesis from TPC drainage in prospective, controlled trials has been considerably more modest, supporting a range of approximately 20% to 30% with routine drainage strategies.20,25-27 Spontaneous pleurodesis develops greater rapidity and frequency in patients undergoing daily drainage compared to every-other-day or symptom-directed drainage strategies.25,26 However, there is no appreciable improvement in quality of life scores with a specific drainage strategy. Small case series also demonstrate that TPC drainage may induce spontaneous pleurodesis in some patients initially presenting with trapped lung physiology.22

        Catheter placement can be performed successfully in the vast majority of patients.28 Increased bleeding risk, significant malignancy-related involvement of the skin and chest wall, and pleural loculations can complicate TPC placement. TPC-related complications are relatively uncommon, but include pneumothorax, catheter malfunction and obstruction, and infections including soft tissue and pleural space infections.24 In a multicenter retrospective series of 1021 patients, only 4.9% developed a TPC-related pleural infection.29 Over 94% were successfully managed with antibiotic therapy, and the TPC was able to be preserved in 54%. Staphylococcus aureus was the most common causative organism and was identified in 48% of cases. Of note, spontaneous pleurodesis occurred in 62% of cases following a pleural space infection, which likely occurred as sequelae of the inflammatory nature of the infection. Retrospective analysis suggests that the risk of TPC-related infections is not substantially higher for patients with higher risks of immunosuppression from chemotherapy or hematologic malignancies.30,31 Tumor metastasis along the catheter tract is a rare occurrence (< 1%), but is most notable with mesothelioma, which has an incidence as high as 10%.24,32 In addition, development of pleural loculations can impede fluid drainage and relief of dyspnea. Intrapleural instillation of fibrinolytics can be used to improve drainage and improve symptom palliation.33

        Pleurodesis

        Pleurodesis obliterates the potential pleural space by inducing inflammation and fibrosis, resulting in adherence of the visceral and parietal pleura together. This process can be induced through mechanical abrasion of the pleural surface, introduction of chemical sclerosants, or from prolonged use of a chest tube. Chemical sclerosants are the most commonly used method for MPEs and are introduced through a chest tube or under visual guidance such as medical thoracoscopy or video-assisted thoracoscopic surgery (VATS). The pleurodesis process is thought to occur by induction of a systemic inflammatory response with localized deposition of fibrin.34 Activation of fibroblasts and successful pleurodesis have been correlated with higher basic fibroblast growth factor (bFGF) levels in pleural fluid.35 Increased tumor burden is associated with lower bFGF levels, suggesting a possible mechanism for reduced pleurodesis success in these cases. Corticosteroids may reduce the likelihood of pleurodesis due to a reduction of inflammation, as demonstrated in a rabbit model using talc and doxycycline.36,37 Animal data also suggests that use of nonsteroidal anti-inflammatory drugs may hinder the likelihood of successful pleurodesis, but this has not been observed in humans.38,39

        Patients selected for pleurodesis should have significant symptom relief from large-volume removal of pleural fluid, good functional status, and evidence of full lung re-expansion after thoracentesis. Lack of visceral and parietal pleural apposition will prevent pleural adhesion from developing. As a result, trapped lung is associated with chemical pleurodesis failure and is an absolute contraindication to the procedure.4,5,12 The pleurodesis process typically requires 5 to 7 days, during which time the patient is hospitalized for chest tube drainage and pain control. When pleural fluid output diminishes, the chest tube is removed and the patient can be discharged. Modified protocols are now emerging which may shorten the required hospitalization associated with pleurodesis procedures.

         

         

        Pleurodesis Agents

        A variety of chemical sclerosants have been used for pleurodesis, including talc, bleomycin, tetracycline, doxycycline, iodopovidone, and mepacrine. Published data regarding these agents are heterogenous, with significant variability in reported outcomes. However, systematic review and meta-analysis suggests that talc is likely to have higher success rates compared to other agents or chest tube drainage alone for treatment of MPE.40,41

        Additional factors have been shown to be associated with pleurodesis outcomes. Pleurodesis success is negatively associated with low pleural pH, with receiver operating curve thresholds of 7.28 to 7.34.42,43 Trapped lung, large bulky tumor lining the pleural surfaces, and elevated adenosine deaminase levels are also associated with poor pleurodesis outcomes.4,5,12,35,43 In contrast, pleural fluid output less than 200 mL per day and the presence of EGFR (epidermal growth factor receptor) mutation treated with targeted tyrosine kinase inhibitors are associated with successful pleurodesis.44,45

        The most common complications associated with chemical pleurodesis are fever and pain. Other potential complications include soft tissue infections at the chest tube site and of the pleural space, arrhythmias, cardiac arrest, myocardial infarction, and hypotension. Doxycycline is commonly associated with greater pleuritic pain than talc. Acute respiratory distress syndrome (ARDS), acute pneumonitis, and respiratory failure have been described with talc pleurodesis. ARDS secondary to talc pleurodesis occurs in 1% to 9% of cases, though this may be related to the use of ungraded talc. A prospective description of 558 patients treated with large particle talc (> 5 μm) reported no occurrences of ARDS, suggesting the safety of graded large particle talc.46

        Pleurodesis Methods

        Chest tube thoracostomy is an inpatient procedure performed under local anesthesia or conscious sedation. It can be used for measured, intermittent drainage of large effusions for immediate symptom relief, as well as to demonstrate complete lung re-expansion prior to instillation of a chemical sclerosant. Pleurodesis using a chest tube is performed by instillation of a slurry created by mixing the sclerosing agent of choice with 50 to 100 mL of sterile saline. This slurry is instilled into the pleural cavity through the chest tube. The chest tube is clamped for 1 to 2 hours before being reconnected to suction. Intermittent rotation of the patient has not been shown to improve distribution of the sclerosant or result in better procedural outcomes.47,48 Typically, a 24F to 32F chest tube is used because of the concern about obstruction of smaller bore tubes by fibrin plugs. A noninferiority study comparing 12F to 24F chest tubes for talc pleurodesis demonstrated a higher procedure failure rate with the 12F tube (30% versus 24%) and failed to meet noninferiority criteria.39 However, larger caliber tubes are also associated with greater patient discomfort compared to smaller bore tubes.

        Medical thoracoscopy and VATS are minimally invasive means to visualize the pleural space, obtain visually guided biopsy of the parietal pleura, perform lysis of adhesions, and introduce chemical sclerosants for pleurodesis (Figure 2). Medical thoracoscopy can be performed under local anesthesia with procedural sedation in an endoscopy suite or procedure room.

        PDBR01608002_f2.PNG
        In contrast, VATS is performed in an operating room setting and requires general anesthesia, intubation with a double-lumen endotracheal tube, and multiple trocar incisions. For medical thoracoscopy, the patient is placed in the lateral decubitus position. The medical thoracoscope is introduced into the pleural space through one or more trocars. Trocar sizes range from 5 to 13 mm depending on the type of thoracoscope used. The body of the thoracoscopes may be rigid or semi-rigid (Figure 3). Rigid thoracoscopes have direct (0°) and angled cameras, while semi-rigid thoracoscopes have a flexible tip that can be manipulated similar to a flexible bronchoscope to direct visualization and biopsies. Following the procedure, a chest tube is typically introduced through the trocar insertion site for drainage.

        PDBR01608002_f3.PNG

         

         

        VATS has several distinct and clinically important differences. The equipment is slightly larger but otherwise similar in concept to rigid medical thoracoscopes. A greater number of diagnostic and therapeutic options, such as diagnostic biopsy of lung parenchyma and select hilar lymph nodes, are also possible. However, VATS requires surgical training and is performed in an operating room setting, which necessitates additional ancillary and logistical support. VATS also uses at least 2 trocar insertion sites, requires general anesthesia, and utilizes single-lung ventilation through a double-lumen endotracheal tube. Procedure-related complications for medical thoracoscopy and VATS include pneumothorax, subcutaneous emphysema, fever, and pain.49

        Data comparing talc slurry versus talc poudrage are heterogenous, without clear advantage for either method. Reported rates of successful pleurodesis are generally in the range of 70% to 80% for both methods.19,20,40,50 There is a possible suggestion of benefit with talc poudrage compared to slurry, but data is lacking to support either as a definitive choice in current guidelines.12,51 An advantage of medical thoracoscopy or VATS is that pleural biopsy can be performed simultaneously, if necessary, thereby allowing both diagnostic and therapeutic interventions.52 Visualizing the thoracic cavity may also permit creation of optimal conditions for pleurodesis in select individuals by allowing access to loculated spaces and providing visual confirmation of complete drainage of pleural fluid and uniform distribution of the chemical sclerosant.

        Other Surgical Interventions

        Thoracotomy with decortication is rarely used as treatment of malignant effusions complicated by loculations or trapped lung due to the significantly increased procedural morbidity and mortality. Therefore, it is reserved for the limited population of patients in whom other therapeutic interventions have failed but who otherwise have significant symptoms with a long life expectancy. Mesothelioma is a specific situation in which variations of pleurectomy, such as radical pleurectomy with decortication, lung-sparing total pleurectomy, and extrapleural pneumonectomy (EPP), have been used as front-line therapy. The Mesothelioma and Radical Surgery (MARS) trial, the only randomized, controlled evaluation of EPP, demonstrated decreased median survival in patients treated by EPP compared to controls (14.4 months versus 19.5 months).53 EPP is also associated with high procedure-related morbidity and mortality rates of approximately 50% and 4% to 15%, respectively.54 While successful at achieving pleurodesis, use of EPP as a treatment for mesothelioma is now discouraged.53,55 Less invasive surgical approaches, such as pleurectomy with decortication, are able to palliate symptoms with significantly less operative risk.56

        Management Considerations

        Selection of Therapeutic Interventions

        The ideal management strategy provides both immediate and long-term symptom palliation, has minimal associated morbidity and side effects, minimizes hospitalization time and clinic visits, avoids the risks and inconvenience of recurring procedures, is inexpensive, and minimizes utilization of medical resources. Unfortunately, no single palliation methodology fits these needs for all patients. When evaluating therapeutic options for patients with MPE, it is important to consider factors such as the severity of symptoms, fluid quantity, fluid re-accumulation rate, pleural physiology, functional status, overall prognosis, and anticipated response of the malignancy to therapy. One example management algorithm (Figure 4) demonstrates the impact of these variables on the appropriate treatment options. However, this is a simplified algorithm and the selected palliation strategy should be decided upon after shared decision-making between the patient and physician and should fit within the context of the patient’s desired goals of care. It is also crucial for patients to understand that these therapeutic interventions are palliative rather than curative.

        PDBR01608002_f4.PNG

        When compared directly with pleurodesis, TPC provides similar control of symptoms but with a reduction in hospital length of stay by a median of 3.5 to 5.5 days.19,57 In a nonrandomized trial where patients chose palliation by TPC or talc pleurodesis, more TPC patients had a significant immediate improvement in quality of life and dyspnea after the first 7 days of therapy.58 This is reasonably attributed to the differences between the immediate relief from fluid drainage after TPC placement compared to the time required for pleural symphysis to occur with pleurodesis. However, control of dyspnea symptoms is similar between the 2 strategies after 6 weeks.19 Therefore, both TPC and pleurodesis strategies are viewed as first-line options for patients with expandable lung and no prior palliative interventions for MPE.59

         

         

        Pleural adhesions and trapped lung also pose specific dilemmas. Pleural adhesions can create loculated fluid pockets, thereby complicating drainage by thoracentesis or TPC and hindering dispersal of pleurodesis agents. Adhesiolysis by medical thoracoscopy or VATS may be useful in these patients to free up the pleural space and improve efficacy of long-term drainage options or facilitate pleurodesis. Intrapleural administration of fibrinolytics, such as streptokinase and urokinase, has also been used for treatment of loculated effusions and may improve drainage of pleural fluid and lung re-expansion.60-63 However routine use of intrapleural fibrinolytics with pleurodesis has not been shown to be beneficial. In a randomized comparison using intrapleural urokinase prior to pleurodesis for patients with septated malignant pleural effusions, no difference in pleurodesis outcomes were identified.63 As a result, TPC is the preferred palliation approach for patients with trapped lung physiology.51,59

        Combination Strategies

        Combinations of different therapeutic interventions are being evaluated as a means for patients to achieve long-term benefits from pleurodesis while minimizing hospitalization time. One strategy using simultaneous treatment with thoracoscopic talc poudrage and insertion of a large-bore chest tube and TPC has been shown to permit early removal of the chest tube and discharge home using the TPC for continued daily pleural drainage. This “rapid pleurodesis” strategy has an 80% to 90% successful pleurodesis rate, permitting removal of the TPC at a median of 7 to 10 days.64,65 With this approach, median hospitalization length of stay was approximately 2 days. While there was no control arm in these early reports with limited sample sizes, the pleurodesis success rate and length of hospitalization compare favorably to other published studies. A prospective, randomized trial of TPC versus an outpatient regimen of talc slurry via TPC has also shown promise, with successful pleurodesis after 35 days in 43% of those treated with the combination of talc slurry and TPC compared to only 23% in those treated by TPC alone.27

        Another novel approach to obtain the benefits of both TPC and pleurodesis strategies is the use of drug-eluting TPC to induce inflammation and promote adhesion of the visceral and parietal pleura. An early report of slow-release silver nitrate (AgNO3) –coated TPC demonstrated an encouraging 89% spontaneous pleurodesis rate after a median of 4 days in the small subgroup of patients with fully expandable lung.66 Device-related adverse events were relatively high at 24.6%, though only one was deemed a serious adverse event. Additional studies of these novel and combination strategies are ongoing at this time.

        Costs

        While cost of care is not a consideration in the decision-making for individual patients, it is important from a systems-based perspective. Upfront costs for pleurodesis are generally higher due to the facility and hospitalization costs, whereas TPC have ongoing costs for drainage bottles and supplies. In a prospective, randomized trial of TPC versus talc pleurodesis, there was no appreciable difference in overall costs between the 2 approaches.67 The cost of TPC was significantly less, however, for patients with a shorter survival of less than 14 weeks.

        Readmissions

        Subsequent hospitalization requirements beyond just the initial treatment for a MPE remains another significant consideration for this patient population. A prospective, randomized trial comparing TPC to talc pleurodesis demonstrated a reduction in total all-cause hospital stay for TPC, with a median all-cause hospitalization time of 10 days for patients treated with TPC compared to 12 days for the talc pleurodesis group.20 The primary difference in the number of hospitalization days was due to a difference in effusion-related hospital days (median 1 versus 4 days, respectively), which was primarily comprised of the initial hospitalization. In addition, fewer patients treated with TPC required subsequent ipsilateral invasive procedures (4.1% versus 22.5%, respectively). However, it is important to note that the majority of all-cause hospital days were not effusion-related, demonstrating that this population has a high utilization of acute inpatient services for other reasons related to their advanced malignancy. In a study of regional hospitals in the United States, 38.3% of patients admitted for a primary diagnosis of MPE were readmitted within 30 days.68 There was remarkably little variability in readmission rates among hospitals, despite differences in factors such as institution size, location, patient distribution, and potential practice differences. This suggests that utilization of palliation strategies for MPE are only one component related to hospitalization in this population. Even at the best performing hospitals, there are significant common drivers for readmission that are not addressed. Therefore, additional effort should be focused on addressing aspects of care beyond just the palliation of MPE that predispose this population to requiring frequent treatment in an acute care setting.

         

         

        Conclusion

        MPEs represent advanced stage disease and frequently adversely affect a patient’s quality of life. The treating clinician has access to a variety of therapeutic options, though no single intervention strategy is universally superior in all circumstances. Initial thoracentesis is important in evaluating whether removal of a large volume of fluid provides significant symptom relief and restores functional status. Both talc pleurodesis and TPC provide similar control of symptoms and are first-line approaches for symptomatic patients with MPE and fully expandable lungs. Pleurodesis is associated with greater procedure-related risk and length of hospitalization and is contraindicated in patients with trapped lung, but does not require long-term catheter care or disposable resources. Determination of the appropriate therapeutic management strategy requires careful evaluation of the patient’s clinical situation and informed discussion with the patient to make sure that the treatment plan fits within the context of their goals of medical care.

        References

        1. Antony VB, Loddenkemper R, Astoul P, et al. Management of malignant pleural effusions. Eur Respir J. 2001;18:402-419.

        2. Society AT. Management of malignant pleural effusions. Am J Respir Crit Care Med. 2000;162:1987-2001.

        3. Taghizadeh N, Fortin M, Tremblay A. US hospitalizations for malignant pleural effusions: data from the 2012 National Inpatient Sample. Chest. 2017;151:845-854.

        4. Adler RH, Sayek I. Treatment of malignant pleural effusion: a method using tube thoracostomy and talc. Ann Thorac Surg. 1976;22:8-15.

        5. Villanueva AG, Gray AW, Shahian DM, et al. Efficacy of short term versus long term tube thoracostomy drainage before tetracycline pleurodesis in the treatment of malignant pleural effusions. Thorax. 1994;49:23-25.

        6. Hibbert RM, Atwell TD, Lekah A, et al. Safety of ultrasound-guided thoracentesis in patients with abnormal preprocedural coagulation parameters. Chest. 2013;144:456-463.

        7. Patel MD, Joshi SD. Abnormal preprocedural international normalized ratio and platelet counts are not associated with increased bleeding complications after ultrasound-guided thoracentesis. AJR Am J Roentgenol. 2011;197:W164-168.

        8. Zalt MB, Bechara RI, Parks C, Berkowitz DM. Effect of routine clopidogrel use on bleeding complications after ultrasound-guided thoracentesis. J Bronchology Interv Pulmonol. 2012;19:284-287.

        9. Mahmood K, Shofer SL, Moser BK, et al. Hemorrhagic complications of thoracentesis and small-bore chest tube placement in patients taking clopidogrel. Ann Am Thorac Soc. 2014;11:73-79.

        10. Puchalski JT, Argento AC, Murphy TE, et al. The safety of thoracentesis in patients with uncorrected bleeding risk. Ann Am Thorac Soc. 2013;10:336-341.

        11. Gordon CE, Feller-Kopman D, Balk EM, Smetana GW. Pneumothorax following thoracentesis: a systematic review and meta-analysis. Arch Intern Med. 2010;170:332-339.

        12. Roberts ME, Neville E, Berrisford RG, et al; Group BPDG. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax. 2010;65 Suppl 2:ii32-40.

        13. Feller-Kopman D, Berkowitz D, Boiselle P, Ernst A. Large-volume thoracentesis and the risk of reexpansion pulmonary edema. Ann Thorac Surg. 2007;84:1656-1661.

        14. Feller-Kopman D, Walkey A, Berkowitz D, Ernst A. The relationship of pleural pressure to symptom development during therapeutic thoracentesis. Chest. 2006;129:1556-1560.

        15. Senitko M, Ray AS, Murphy TE, et al. Safety and tolerability of vacuum versus manual drainage during thoracentesis: a randomized trial. J Bronchology Interv Pulmonol. 2019;26:166-171.

        16. Ost DE, Niu J, Zhao H, et al. Quality gaps and comparative effectiveness of management strategies for recurrent malignant pleural effusions. Chest. 2018;153:438-452.

        17. Grosu HB, Molina S, Casal R, et al. Risk factors for pleural effusion recurrence in patients with malignancy. Respirology. 2019;24:76-82.

        18. Musani AI, Haas AR, Seijo L, et al. Outpatient management of malignant pleural effusions with small-bore, tunneled pleural catheters. Respiration. 2004;71:559-566.

        19. Davies HE, Mishra EK, Kahan BC, et al. Effect of an indwelling pleural catheter vs chest tube and talc pleurodesis for relieving dyspnea in patients with malignant pleural effusion: the TIME2 randomized controlled trial. JAMA. 2012;307:2383-2389.

        20. Thomas R, Fysh ETH, Smith NA, et al. Effect of an indwelling pleural catheter vs talc pleurodesis on hospitalization days in patients with malignant pleural effusion: the AMPLE randomized clinical trial. JAMA. 2017;318:1903-1912.

        21. Qureshi RA, Collinson SL, Powell RJ, et al. Management of malignant pleural effusion associated with trapped lung syndrome. Asian Cardiovasc Thorac Ann. 2008;16:120-123.

        22. Efthymiou CA, Masudi T, Thorpe JA, Papagiannopoulos K. Malignant pleural effusion in the presence of trapped lung. Five-year experience of PleurX tunnelled catheters. Interact Cardiovasc Thorac Surg. 2009;9:961-964.

        23. Sioris T, Sihvo E, Salo J, et al. Long-term indwelling pleural catheter (PleurX) for malignant pleural effusion unsuitable for talc pleurodesis. Eur J Surg Oncol. 2009;35:546-551.

        24. Van Meter ME, McKee KY, Kohlwes RJ. Efficacy and safety of tunneled pleural catheters in adults with malignant pleural effusions: a systematic review. J Gen Intern Med. 2011;26:70-76.

        25. Wahidi MM, Reddy C, Yarmus L, et al. Randomized trial of pleural fluid drainage frequency in patients with malignant pleural effusions. the ASAP trial. Am J Respir Crit Care Med. 2017;195:1050-1057.

        26. Muruganandan S, Azzopardi M, Fitzgerald DB, et al. Aggressive versus symptom-guided drainage of malignant pleural effusion via indwelling pleural catheters (AMPLE-2): an open-label randomised trial. Lancet Respir Med. 2018;6:671-680.

        27. Bhatnagar R, Keenan EK, Morley AJ, et al. Outpatient talc administration by indwelling pleural catheter for malignant effusion. N Engl J Med. 2018;378:1313-1322.

        28. Tremblay A, Michaud G. Single-center experience with 250 tunnelled pleural catheter insertions for malignant pleural effusion. Chest. 2006;129:362-368.

        29. Fysh ETH, Tremblay A, Feller-Kopman D, et al. Clinical outcomes of indwelling pleural catheter-related pleural infections: an international multicenter study. Chest. 2013;144:1597-1602.

        30. Morel A, Mishra E, Medley L, et al. Chemotherapy should not be withheld from patients with an indwelling pleural catheter for malignant pleural effusion. Thorax. 2011;66:448-449.

        31. Gilbert CR, Lee HJ, Skalski JH, et al. The use of indwelling tunneled pleural catheters for recurrent pleural effusions in patients with hematologic malignancies: a multicenter study. Chest. 2015;148:752-758.

        32. Thomas R, Budgeon CA, Kuok YJ, et al. Catheter tract metastasis associated with indwelling pleural catheters. Chest. 2014;146:557-562.

        33. Thomas R, Piccolo F, Miller D, et al. Intrapleural fibrinolysis for the treatment of indwelling pleural catheter-related symptomatic loculations: a multicenter observational study. Chest. 2015;148:746-751.

        34. Antony VB. Pathogenesis of malignant pleural effusions and talc pleurodesis. Pneumologie. 1999;53:493-498.

        35. Antony VB, Nasreen N, Mohammed KA, et al. Talc pleurodesis: basic fibroblast growth factor mediates pleural fibrosis. Chest. 2004;126:1522-1528.

        36. Xie C, Teixeira LR, McGovern JP, Light RW. Systemic corticosteroids decrease the effectiveness of talc pleurodesis. Am J Respir Crit Care Med. 1998;157(5 Pt 1):1441-1444.

        37. Teixeira LR, Wu W, Chang DS, Light RW. The effect of corticosteroids on pleurodesis induced by doxycycline in rabbits. Chest. 2002;121:216-219.

        38. Hunt I, Teh E, Southon R, Treasure T. Using non-steroidal anti-inflammatory drugs (NSAIDs) following pleurodesis. Interact Cardiovasc Thorac Surg. 2007;6:102-104.

        39. Rahman NM, Pepperell J, Rehal S, et al. Effect of opioids vs NSAIDs and larger vs smaller chest tube size on pain control and pleurodesis efficacy among patients with malignant pleural effusion: the TIME1 randomized clinical trial. JAMA. 2015;314:2641-2653.

        40. Clive AO, Jones HE, Bhatnagar R, Preston NJ, Maskell N. Interventions for the management of malignant pleural effusions: a network meta-analysis. Cochrane Database Syst Rev. 2016(5):CD010529.

        41. Tan C, Sedrakyan A, Browne J, et al. The evidence on the effectiveness of management for malignant pleural effusion: a systematic review. Eur J Cardiothorac Surg. 2006;29:829-838.

        42. Heffner JE, Nietert PJ, Barbieri C. Pleural fluid pH as a predictor of pleurodesis failure: analysis of primary data. Chest. 2000;117:87-95.

        43. Yildirim H, Metintas M, Ak G, et al. Predictors of talc pleurodesis outcome in patients with malignant pleural effusions. Lung Cancer. 2008;62:139-144.

        44. Aydogmus U, Ozdemir S, Cansever L, et al. Bedside talc pleurodesis for malignant pleural effusion: factors affecting success. Ann Surg Oncol. 2009;16:745-750.

        45. Guo H, Wan Y, Tian G, et al. EGFR mutations predict a favorable outcome for malignant pleural effusion of lung adenocarcinoma with Tarceva therapy. Oncol Rep. 2012;27:880-890.

        46. Janssen JP, Collier G, Astoul P, et al. Safety of pleurodesis with talc poudrage in malignant pleural effusion: a prospective cohort study. Lancet. 2007;369(9572):1535-1539.

        47. Dryzer SR, Allen ML, Strange C, Sahn SA. A comparison of rotation and nonrotation in tetracycline pleurodesis. Chest. 1993;104:1763-1766.

        48. Mager HJ, Maesen B, Verzijlbergen F, Schramel F. Distribution of talc suspension during treatment of malignant pleural effusion with talc pleurodesis. Lung Cancer. 2002;36:77-81.

        49. Hsia D, Musani AI. Interventional pulmonology. Med Clin North Am. 2011;95:1095-1114.

        50. Dresler CM, Olak J, Herndon JE, et al. Phase III intergroup study of talc poudrage vs talc slurry sclerosis for malignant pleural effusion. Chest. 2005;127:909-915.

        51. Bibby AC, Dorn P, Psallidas I, et al. ERS/EACTS statement on the management of malignant pleural effusions. Eur Respir J. 2018;52(1).

        52. Sakuraba M, Masuda K, Hebisawa A, et al. Diagnostic value of thoracoscopic pleural biopsy for pleurisy under local anaesthesia. ANZ J Surg. 2006;76:722-724.

        53. Treasure T, Lang-Lazdunski L, Waller D, et al. Extra-pleural pneumonectomy versus no extra-pleural pneumonectomy for patients with malignant pleural mesothelioma: clinical outcomes of the Mesothelioma and Radical Surgery (MARS) randomised feasibility study. Lancet Oncol. 2011;12:763-772.

        54. Zellos L, Jaklitsch MT, Al-Mourgi MA, Sugarbaker DJ. Complications of extrapleural pneumonectomy. Semin Thorac Cardiovasc Surg. 2007;19:355-359.

        55. Zahid I, Sharif S, Routledge T, Scarci M. Is pleurectomy and decortication superior to palliative care in the treatment of malignant pleural mesothelioma? Interact Cardiovasc Thorac Surg. 2011;12:812-817.

        56. Soysal O, Karaoğlanoğlu N, Demiracan S, et al. Pleurectomy/decortication for palliation in malignant pleural mesothelioma: results of surgery. Eur J Cardiothorac Surg. 1997;11:210-213.

        57. Putnam JB, Light RW, Rodriguez RM, et al. A randomized comparison of indwelling pleural catheter and doxycycline pleurodesis in the management of malignant pleural effusions. Cancer. 1999;86:1992-1999.

        58. Fysh ETH, Waterer GW, Kendall PA, et al. Indwelling pleural catheters reduce inpatient days over pleurodesis for malignant pleural effusion. Chest. 2012;142:394-400.

        59. Feller-Kopman DJ, Reddy CB, DeCamp MM, et al. Management of malignant pleural effusions. An official ATS/STS/STR clinical practice guideline. Am J Respir Crit Care Med. 2018;198:839-849.

        60. Davies CW, Traill ZC, Gleeson FV, Davies RJ. Intrapleural streptokinase in the management of malignant multiloculated pleural effusions. Chest. 1999;115:729-733.

        61. Hsu LH, Soong TC, Feng AC, Liu MC. Intrapleural urokinase for the treatment of loculated malignant pleural effusions and trapped lungs in medically inoperable cancer patients. J Thorac Oncol. 2006;1:460-467.

        62. Okur E, Baysungur V, Tezel C, et al. Streptokinase for malignant pleural effusions: a randomized controlled study. Asian Cardiovasc Thorac Ann. 2011;19:238-243.

        63. Mishra EK, Clive AO, Wills GH, et al. Randomized controlled trial of urokinase versus placebo for nondraining malignant pleural effusion. Am J Respir Crit Care Med. 2018;197:502-508.

        64. Reddy C, Ernst A, Lamb C, Feller-Kopman D. Rapid pleurodesis for malignant pleural effusions: a pilot study. Chest. 2011;139:1419-1423.

        65. Krochmal R, Reddy C, Yarmus L, et al. Patient evaluation for rapid pleurodesis of malignant pleural effusions. J Thorac Dis. 2016;8:2538-2543.

        66. Bhatnagar R, Zahan-Evans N, Kearney C, et al. A novel drug-eluting indwelling pleural catheter for the management of malignant effusions. Am J Respir Crit Care Med. 2018;197:136-138.

        67. Penz ED, Mishra EK, Davies HE, Manns BJ, Miller RF, Rahman NM. Comparing cost of indwelling pleural catheter vs talc pleurodesis for malignant pleural effusion. Chest. 2014;146:991-1000.

        68. Yang TS, Hsia DW, Chang DW. Patient- and hospital-level factors associated with readmission for malignant pleural effusion. J Oncol Pract. 2018;14:e547-e556.

        References

        1. Antony VB, Loddenkemper R, Astoul P, et al. Management of malignant pleural effusions. Eur Respir J. 2001;18:402-419.

        2. Society AT. Management of malignant pleural effusions. Am J Respir Crit Care Med. 2000;162:1987-2001.

        3. Taghizadeh N, Fortin M, Tremblay A. US hospitalizations for malignant pleural effusions: data from the 2012 National Inpatient Sample. Chest. 2017;151:845-854.

        4. Adler RH, Sayek I. Treatment of malignant pleural effusion: a method using tube thoracostomy and talc. Ann Thorac Surg. 1976;22:8-15.

        5. Villanueva AG, Gray AW, Shahian DM, et al. Efficacy of short term versus long term tube thoracostomy drainage before tetracycline pleurodesis in the treatment of malignant pleural effusions. Thorax. 1994;49:23-25.

        6. Hibbert RM, Atwell TD, Lekah A, et al. Safety of ultrasound-guided thoracentesis in patients with abnormal preprocedural coagulation parameters. Chest. 2013;144:456-463.

        7. Patel MD, Joshi SD. Abnormal preprocedural international normalized ratio and platelet counts are not associated with increased bleeding complications after ultrasound-guided thoracentesis. AJR Am J Roentgenol. 2011;197:W164-168.

        8. Zalt MB, Bechara RI, Parks C, Berkowitz DM. Effect of routine clopidogrel use on bleeding complications after ultrasound-guided thoracentesis. J Bronchology Interv Pulmonol. 2012;19:284-287.

        9. Mahmood K, Shofer SL, Moser BK, et al. Hemorrhagic complications of thoracentesis and small-bore chest tube placement in patients taking clopidogrel. Ann Am Thorac Soc. 2014;11:73-79.

        10. Puchalski JT, Argento AC, Murphy TE, et al. The safety of thoracentesis in patients with uncorrected bleeding risk. Ann Am Thorac Soc. 2013;10:336-341.

        11. Gordon CE, Feller-Kopman D, Balk EM, Smetana GW. Pneumothorax following thoracentesis: a systematic review and meta-analysis. Arch Intern Med. 2010;170:332-339.

        12. Roberts ME, Neville E, Berrisford RG, et al; Group BPDG. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax. 2010;65 Suppl 2:ii32-40.

        13. Feller-Kopman D, Berkowitz D, Boiselle P, Ernst A. Large-volume thoracentesis and the risk of reexpansion pulmonary edema. Ann Thorac Surg. 2007;84:1656-1661.

        14. Feller-Kopman D, Walkey A, Berkowitz D, Ernst A. The relationship of pleural pressure to symptom development during therapeutic thoracentesis. Chest. 2006;129:1556-1560.

        15. Senitko M, Ray AS, Murphy TE, et al. Safety and tolerability of vacuum versus manual drainage during thoracentesis: a randomized trial. J Bronchology Interv Pulmonol. 2019;26:166-171.

        16. Ost DE, Niu J, Zhao H, et al. Quality gaps and comparative effectiveness of management strategies for recurrent malignant pleural effusions. Chest. 2018;153:438-452.

        17. Grosu HB, Molina S, Casal R, et al. Risk factors for pleural effusion recurrence in patients with malignancy. Respirology. 2019;24:76-82.

        18. Musani AI, Haas AR, Seijo L, et al. Outpatient management of malignant pleural effusions with small-bore, tunneled pleural catheters. Respiration. 2004;71:559-566.

        19. Davies HE, Mishra EK, Kahan BC, et al. Effect of an indwelling pleural catheter vs chest tube and talc pleurodesis for relieving dyspnea in patients with malignant pleural effusion: the TIME2 randomized controlled trial. JAMA. 2012;307:2383-2389.

        20. Thomas R, Fysh ETH, Smith NA, et al. Effect of an indwelling pleural catheter vs talc pleurodesis on hospitalization days in patients with malignant pleural effusion: the AMPLE randomized clinical trial. JAMA. 2017;318:1903-1912.

        21. Qureshi RA, Collinson SL, Powell RJ, et al. Management of malignant pleural effusion associated with trapped lung syndrome. Asian Cardiovasc Thorac Ann. 2008;16:120-123.

        22. Efthymiou CA, Masudi T, Thorpe JA, Papagiannopoulos K. Malignant pleural effusion in the presence of trapped lung. Five-year experience of PleurX tunnelled catheters. Interact Cardiovasc Thorac Surg. 2009;9:961-964.

        23. Sioris T, Sihvo E, Salo J, et al. Long-term indwelling pleural catheter (PleurX) for malignant pleural effusion unsuitable for talc pleurodesis. Eur J Surg Oncol. 2009;35:546-551.

        24. Van Meter ME, McKee KY, Kohlwes RJ. Efficacy and safety of tunneled pleural catheters in adults with malignant pleural effusions: a systematic review. J Gen Intern Med. 2011;26:70-76.

        25. Wahidi MM, Reddy C, Yarmus L, et al. Randomized trial of pleural fluid drainage frequency in patients with malignant pleural effusions. the ASAP trial. Am J Respir Crit Care Med. 2017;195:1050-1057.

        26. Muruganandan S, Azzopardi M, Fitzgerald DB, et al. Aggressive versus symptom-guided drainage of malignant pleural effusion via indwelling pleural catheters (AMPLE-2): an open-label randomised trial. Lancet Respir Med. 2018;6:671-680.

        27. Bhatnagar R, Keenan EK, Morley AJ, et al. Outpatient talc administration by indwelling pleural catheter for malignant effusion. N Engl J Med. 2018;378:1313-1322.

        28. Tremblay A, Michaud G. Single-center experience with 250 tunnelled pleural catheter insertions for malignant pleural effusion. Chest. 2006;129:362-368.

        29. Fysh ETH, Tremblay A, Feller-Kopman D, et al. Clinical outcomes of indwelling pleural catheter-related pleural infections: an international multicenter study. Chest. 2013;144:1597-1602.

        30. Morel A, Mishra E, Medley L, et al. Chemotherapy should not be withheld from patients with an indwelling pleural catheter for malignant pleural effusion. Thorax. 2011;66:448-449.

        31. Gilbert CR, Lee HJ, Skalski JH, et al. The use of indwelling tunneled pleural catheters for recurrent pleural effusions in patients with hematologic malignancies: a multicenter study. Chest. 2015;148:752-758.

        32. Thomas R, Budgeon CA, Kuok YJ, et al. Catheter tract metastasis associated with indwelling pleural catheters. Chest. 2014;146:557-562.

        33. Thomas R, Piccolo F, Miller D, et al. Intrapleural fibrinolysis for the treatment of indwelling pleural catheter-related symptomatic loculations: a multicenter observational study. Chest. 2015;148:746-751.

        34. Antony VB. Pathogenesis of malignant pleural effusions and talc pleurodesis. Pneumologie. 1999;53:493-498.

        35. Antony VB, Nasreen N, Mohammed KA, et al. Talc pleurodesis: basic fibroblast growth factor mediates pleural fibrosis. Chest. 2004;126:1522-1528.

        36. Xie C, Teixeira LR, McGovern JP, Light RW. Systemic corticosteroids decrease the effectiveness of talc pleurodesis. Am J Respir Crit Care Med. 1998;157(5 Pt 1):1441-1444.

        37. Teixeira LR, Wu W, Chang DS, Light RW. The effect of corticosteroids on pleurodesis induced by doxycycline in rabbits. Chest. 2002;121:216-219.

        38. Hunt I, Teh E, Southon R, Treasure T. Using non-steroidal anti-inflammatory drugs (NSAIDs) following pleurodesis. Interact Cardiovasc Thorac Surg. 2007;6:102-104.

        39. Rahman NM, Pepperell J, Rehal S, et al. Effect of opioids vs NSAIDs and larger vs smaller chest tube size on pain control and pleurodesis efficacy among patients with malignant pleural effusion: the TIME1 randomized clinical trial. JAMA. 2015;314:2641-2653.

        40. Clive AO, Jones HE, Bhatnagar R, Preston NJ, Maskell N. Interventions for the management of malignant pleural effusions: a network meta-analysis. Cochrane Database Syst Rev. 2016(5):CD010529.

        41. Tan C, Sedrakyan A, Browne J, et al. The evidence on the effectiveness of management for malignant pleural effusion: a systematic review. Eur J Cardiothorac Surg. 2006;29:829-838.

        42. Heffner JE, Nietert PJ, Barbieri C. Pleural fluid pH as a predictor of pleurodesis failure: analysis of primary data. Chest. 2000;117:87-95.

        43. Yildirim H, Metintas M, Ak G, et al. Predictors of talc pleurodesis outcome in patients with malignant pleural effusions. Lung Cancer. 2008;62:139-144.

        44. Aydogmus U, Ozdemir S, Cansever L, et al. Bedside talc pleurodesis for malignant pleural effusion: factors affecting success. Ann Surg Oncol. 2009;16:745-750.

        45. Guo H, Wan Y, Tian G, et al. EGFR mutations predict a favorable outcome for malignant pleural effusion of lung adenocarcinoma with Tarceva therapy. Oncol Rep. 2012;27:880-890.

        46. Janssen JP, Collier G, Astoul P, et al. Safety of pleurodesis with talc poudrage in malignant pleural effusion: a prospective cohort study. Lancet. 2007;369(9572):1535-1539.

        47. Dryzer SR, Allen ML, Strange C, Sahn SA. A comparison of rotation and nonrotation in tetracycline pleurodesis. Chest. 1993;104:1763-1766.

        48. Mager HJ, Maesen B, Verzijlbergen F, Schramel F. Distribution of talc suspension during treatment of malignant pleural effusion with talc pleurodesis. Lung Cancer. 2002;36:77-81.

        49. Hsia D, Musani AI. Interventional pulmonology. Med Clin North Am. 2011;95:1095-1114.

        50. Dresler CM, Olak J, Herndon JE, et al. Phase III intergroup study of talc poudrage vs talc slurry sclerosis for malignant pleural effusion. Chest. 2005;127:909-915.

        51. Bibby AC, Dorn P, Psallidas I, et al. ERS/EACTS statement on the management of malignant pleural effusions. Eur Respir J. 2018;52(1).

        52. Sakuraba M, Masuda K, Hebisawa A, et al. Diagnostic value of thoracoscopic pleural biopsy for pleurisy under local anaesthesia. ANZ J Surg. 2006;76:722-724.

        53. Treasure T, Lang-Lazdunski L, Waller D, et al. Extra-pleural pneumonectomy versus no extra-pleural pneumonectomy for patients with malignant pleural mesothelioma: clinical outcomes of the Mesothelioma and Radical Surgery (MARS) randomised feasibility study. Lancet Oncol. 2011;12:763-772.

        54. Zellos L, Jaklitsch MT, Al-Mourgi MA, Sugarbaker DJ. Complications of extrapleural pneumonectomy. Semin Thorac Cardiovasc Surg. 2007;19:355-359.

        55. Zahid I, Sharif S, Routledge T, Scarci M. Is pleurectomy and decortication superior to palliative care in the treatment of malignant pleural mesothelioma? Interact Cardiovasc Thorac Surg. 2011;12:812-817.

        56. Soysal O, Karaoğlanoğlu N, Demiracan S, et al. Pleurectomy/decortication for palliation in malignant pleural mesothelioma: results of surgery. Eur J Cardiothorac Surg. 1997;11:210-213.

        57. Putnam JB, Light RW, Rodriguez RM, et al. A randomized comparison of indwelling pleural catheter and doxycycline pleurodesis in the management of malignant pleural effusions. Cancer. 1999;86:1992-1999.

        58. Fysh ETH, Waterer GW, Kendall PA, et al. Indwelling pleural catheters reduce inpatient days over pleurodesis for malignant pleural effusion. Chest. 2012;142:394-400.

        59. Feller-Kopman DJ, Reddy CB, DeCamp MM, et al. Management of malignant pleural effusions. An official ATS/STS/STR clinical practice guideline. Am J Respir Crit Care Med. 2018;198:839-849.

        60. Davies CW, Traill ZC, Gleeson FV, Davies RJ. Intrapleural streptokinase in the management of malignant multiloculated pleural effusions. Chest. 1999;115:729-733.

        61. Hsu LH, Soong TC, Feng AC, Liu MC. Intrapleural urokinase for the treatment of loculated malignant pleural effusions and trapped lungs in medically inoperable cancer patients. J Thorac Oncol. 2006;1:460-467.

        62. Okur E, Baysungur V, Tezel C, et al. Streptokinase for malignant pleural effusions: a randomized controlled study. Asian Cardiovasc Thorac Ann. 2011;19:238-243.

        63. Mishra EK, Clive AO, Wills GH, et al. Randomized controlled trial of urokinase versus placebo for nondraining malignant pleural effusion. Am J Respir Crit Care Med. 2018;197:502-508.

        64. Reddy C, Ernst A, Lamb C, Feller-Kopman D. Rapid pleurodesis for malignant pleural effusions: a pilot study. Chest. 2011;139:1419-1423.

        65. Krochmal R, Reddy C, Yarmus L, et al. Patient evaluation for rapid pleurodesis of malignant pleural effusions. J Thorac Dis. 2016;8:2538-2543.

        66. Bhatnagar R, Zahan-Evans N, Kearney C, et al. A novel drug-eluting indwelling pleural catheter for the management of malignant effusions. Am J Respir Crit Care Med. 2018;197:136-138.

        67. Penz ED, Mishra EK, Davies HE, Manns BJ, Miller RF, Rahman NM. Comparing cost of indwelling pleural catheter vs talc pleurodesis for malignant pleural effusion. Chest. 2014;146:991-1000.

        68. Yang TS, Hsia DW, Chang DW. Patient- and hospital-level factors associated with readmission for malignant pleural effusion. J Oncol Pract. 2018;14:e547-e556.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Malignant Pleural Effusion: Therapeutic Options and Strategies
        Display Headline
        Malignant Pleural Effusion: Therapeutic Options and Strategies
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 August;16(2)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Tue, 08/27/2019 - 10:45
        Un-Gate On Date
        Tue, 08/27/2019 - 10:45
        Use ProPublica
        CFC Schedule Remove Status
        Tue, 08/27/2019 - 10:45
        Hide sidebar & use full width
        render the right sidebar.

        Malignant Pleural Effusion: Evaluation and Diagnosis

        Article Type
        Changed
        Wed, 08/28/2019 - 11:54
        Display Headline
        Malignant Pleural Effusion: Evaluation and Diagnosis

        Accumulation of pleural fluid is a common clinical problem associated with malignancy. Malignant pleural effusions (MPEs) are the second most common cause of a pleural exudate, with more than 150,000 patients diagnosed annually in the United States alone.1,2 MPEs represent advanced disease and are generally a poor prognostic indicator. Median survival for patients with MPE ranges from 3 to 12 months and depends on the tumor origin.3 In addition, MPEs are a frequent cause of dyspnea and discomfort, which adversely affect a patient’s quality of life. This group of patients requires substantial medical support to manage the burden of their disease, and providing effective therapeutic management remains a challenge. In the United States, there are approximately 126,000 admissions for MPE annually, with a median length of stay of 5.5 days.4 Thirty-day readmission rates are almost 40%, which is approximately 1.5 times higher than for acute myocardial infarction and 2 times higher than for congestive heart failure.5 In addition, palliative measures for patients with MPE are probably underutilized.6

        This review is the first of 2 articles focusing on the management of MPE. Here, we discuss the pathophysiology of this disease process and provide an overview of the evaluation and diagnosis of MPE; available therapeutic options for the management of MPE are reviewed in a separate article.

        Pathogenesis and Etiology

        Normally, the thoracic cavity contains less than 15 mL of pleural fluid. Therefore, the visceral and parietal pleura are usually in close proximity to each other and the space between them is a potential space. Negative intrapleural pressures generated during regular breathing create a gradient for fluid movement into the pleural space from the parietal pleura dictated by Starling forces. Pleural fluid normally has low protein content and is primarily drained back into lymphatics through stomata lining the parietal pleura.7 This system’s ability to remove pleural fluid exceeds normal fluid production by 20- to 30-fold, suggesting that accumulation of excess pleural fluid requires a combination of increased fluid production and/or impaired fluid removal.8

        Several mechanisms have been associated with the development of MPE. Pleural involvement by malignancy may occur from direct invasion of the pleural cavity by tumor (eg, lung cancer, breast cancer, chest wall neoplasms) or hematogenous spread of tumor to the pleura (eg, metastasis, non-Hodgkin lymphoma).9,10 Pleural malignancies can produce cytokine and inflammatory mediators, which may directly increase fluid production or indirectly alter vascular permeability.11,12 Tumor cells can also disrupt lymphatic drainage by occluding either pleural stomata or downstream lymphatic drainage. However, tumor involvement of the pleura does not always result in the development of an effusion and is only associated with fluid accumulation in approximately 60% of cases.13,14 MPE have also been strongly associated with mediastinal metastases, likely resulting from obstruction of mediastinal lymphatics.13,15,16

        Pleural effusions with negative fluid cytology and pleural biopsies may result from secondary effects of tumor burden without direct pleural involvement and are referred to as paramalignant effusions. Common causes include thoracic duct obstruction (eg, Hodgkin lymphoma), bronchial obstruction, pneumonia, atelectasis, pulmonary embolism, trapped lung, and effects related to radiation or chemotherapy.15

        Lung cancer is the most frequent cause of MPE and accounts for approximately one-third of cases. Other common primary tumor sites include breast, lymphoma, ovary, and gastrointestinal. Combined, these etiologies comprise about 75% of cases (Table 1).4,5 Females comprise a greater percentage of patients with MPE mainly due to the prevalence of ovarian and breast cancer. Mesothelioma-related effusions may be more prevalent in certain parts of the world due to associated exposure to asbestos.17 The primary tumor origin remains unknown in approximately 10% of cases.4

        pdbr01608001_t1.png

        Clinical Presentation and Response to Therapeutic Drainage

        More than 75% of patients with MPE are symptomatic. Dyspnea is the most common symptom and is present in more than half of patients.15 The mechanism of dyspnea caused by large effusions may not be solely due to impaired lung volumes or gas exchange. Other associated factors include decreased chest wall compliance, mediastinal shift causing decreased volume of the contralateral lung, paradoxical motion of the diaphragm, inefficient muscle length-tension relationships resulting from the stretch of respiratory muscles, and reflex stimulation from the lungs and chest wall.18-20 Other common presenting symptoms include cough, orthopnea, and chest pain. Hemoptysis suggests endobronchial involvement of the large airways. And, given the advanced nature of most MPEs, patients may also present with weight loss and cachexia.

         

         

        A patient’s degree of symptom palliation and physiologic improvement in response to large-volume fluid removal is important to assess as these are important clinical factors that will influence management decision-making. Upwards of 50% of patients will not have significant palliation because they may be symptom-limited by other comorbid conditions, generalized deconditioning, or incomplete lung re-expansion. Presence of impaired lung compliance during fluid removal is also important to recognize. A trapped lung refers to a lung that cannot expand completely after removal of pleural fluid. Trapped lung may result from pleural-based malignancies or metastases, loculations and adhesions, or bronchial obstruction. Trapped lung is associated with high elastance (Pel) affecting pleural pressure-volume relationships (Figure 1). While clinically often considered together, some authors differentiate the category of incomplete lung expansion into 2 subgroups. In this context, the term trapped lung is used specifically to describe a mature, fibrous membrane that prevents lung re-expansion and is caused by a prior inflammatory pleural condition.21Entrapped lung describes incomplete lung expansion resulting from an active disease process, such as malignancy, ongoing infection, or rheumatologic pleurisy. Differences in pleural manometry can be seen in the 2 subgroups. Pleural manometry can be helpful to monitor for the generation of high negative intrapleural pressures during fluid removal, with negative pressures in excess of –19 cm H2O being suggestive of trapped lung physiology.22 However routine use of pleural manometry has not been shown to avoid the development of procedure-related chest discomfort that develops when the lung is unable to expand in response to the removal of fluid.23

        PDBR01608001_f1.PNG

        Pleural Fluid Analysis and Pleural Biopsy

        While most MPEs are protein-rich exudates, approximately 2% to 5% may be transudates.24,25 MPEs often appear hemorrhagic, so a ratio of pleural fluid to blood serum hematocrit greater than 0.5 is used to distinguish a true hemothorax from bloody-appearing pleural fluid.26 The cell count may be lymphocyte-predominant, but other cell types, such as eosinophils, do not exclude malignancy.27 Fluid may have a low glucose concentration and pH as well.

        Thoracentesis with pleural fluid cytology evaluation is the most common method of diagnosis. The diagnostic sensitivity of fluid cytology ranges from 62% to 90%, with variability resulting from the extent of disease and etiology of the primary malignancy.1 If the initial pleural fluid analysis is not diagnostic, repeat thoracentesis can improve the diagnostic yield, but subsequent sampling has diminishing utility. In one series, diagnosis of malignancy was made by fluid cytology analysis in 65% of patients from the initial thoracentesis, 27% from a second procedure, but only 5% from a third procedure.28 At least 50 to 60 mL of pleural fluid should be obtained for pleural fluid cytology, but analysis of significantly larger volumes may not appreciably improve diagnostic yield.29,30

        In addition to diagnostic yield, adequate sample cellularity to test for genetic driver mutations has become increasingly important given the rapid development of targeted therapies that are now available. The relative paucity of malignant cells in pleural fluid compared to other types of biopsies can make MPEs difficult to analyze for molecular markers. Newer generation assays have increased sensitivity, with one series reporting that pleural fluid was sufficient in 71.4% of cases to analyze for a panel comprised of EGFR, KRAS, BRAF, ALK, and ROS1 mutations.31 Similarly, fluid analysis from patients with MPEs demonstrated that 71.3% had at least 100 tumor cells, which permitted evaluation for PD-L1, with a concordance of 0.78 when compared to matched parenchymal lung biopsies from the same patient.32

        In contrast, pleural biopsy methods may be useful to increase the diagnostic yield when pleural fluid analysis is insufficient. Closed needle biopsy may marginally improve diagnostic yields for malignancy over pleural fluid analysis alone. Diagnostic sensitivity may improve with the use of point-of-care ultrasonography to guide needle placement.33,34 The true value of closed needle biopsy is seen in situations in which there is a high pretest probability to diagnose an alternative disseminated pleural process, such as in tuberculosis, where the diagnostic yield increases substantially with closed needle biopsy of the pleura.33 Otherwise, the diagnosis of lung cancer and mesothelioma is superior with visually guided pleural biopsies, such as medical thoracoscopy or video-assisted thoracoscopic surgery (VATS), with diagnostic yields over 90%.33,35 Testing for genetic driver mutations in pleural biopsies is also substantially improved, with sample adequacy of 90% to 95% for most molecular markers.36,37 Despite the advantages, pleural biopsies are generally reserved for cases when pleural fluid analysis is insufficient or when performed in conjunction with palliative therapeutic interventions due to the increased invasive nature of the procedure.

         

         

        Predictors of Recurrence and Prognosis

        Not all MPEs will progress in size or become symptomatic, and predicting which patients will develop symptoms from their effusions is difficult. Pleural effusions will develop in only a minority of patients with lung cancer, and only a small subset will progress and require therapeutic intervention.38,39 Therefore, management guidelines for malignant pleural effusions discourage empiric intervention for patients with small, asymptomatic effusions.40 However, patients with larger, symptomatic effusions are more likely to have significant and rapid fluid recurrence. In a series of 988 symptomatic patients undergoing drainage, 30% had fluid recurrence within 15 days, 40% within 30 days, 45% within 60 days, and 48% within 90 days.41 Factors associated with fluid recurrence included radiographic size of the effusion, requirement for a larger amount of fluid to be initially drained, and higher pleural fluid lactate dehydrogenase (LDH) level. Negative cytology was associated with lower likelihood for recurrence.

        Prognostication of life expectancy is another important clinical assessment which impacts medical decision-making when weighing the risk and benefits of different palliation options. Patient performance status, pleural fluid LDH, serum neutrophil-to-lymphocyte ratio, and tumor origin are independently associated with prognosis in a validated scoring system (Table 2).3 In this study, the overall median survival of patients with MPE was approximately 4.5 months, while the median survival for patients with mesothelioma was 11.3 months, 6.6 months for breast cancer, and 2.5 months for lung cancer and other malignancies. When stratified based on the combination of these 4 variables, patients in the high-risk group had a median survival of just 44 days compared to 130 days for the moderate-risk group and 319 days for the low-risk group. Additional, more complex prediction systems for survival and response to MPE therapies are now emerging and may provide clinicians and patients with additional information useful in medical decision-making.42

        PDBR01608001_t2.PNG

        Conclusion

        MPEs represent advanced stage disease and frequently adversely affect a pa­tient’s quality of life. Ideal therapeutic options, discussed in the second part of this review, should effectively palliate symptoms, provide long-term relief, be minimally invasive with few side effects, minimize hospitalization and reliance on medical assistance, and be cost-effective.

        References

        1. Antony VB, Loddenkemper R, Astoul P, et al. Management of malignant pleural effusions. Eur Respir J. 2001;18:402-419.

        2. Society AT. Management of malignant pleural effusions. Am J Respir Crit Care Med. 2000;162:1987-2001.

        3. Clive AO, Kahan BC, Hooper CE, et al. Predicting survival in malignant pleural effusion: development and validation of the LENT prognostic score. Thorax. 2014;69:1098-1104.

        4. Taghizadeh N, Fortin M, Tremblay A. US hospitalizations for malignant pleural effusions: data from the 2012 National Inpatient Sample. Chest. 2017;151:845-854.

        5. Yang TS, Hsia DW, Chang DW. Patient- and hospital-level factors associated with readmission for malignant pleural effusion. J Oncol Pract. 2018;14:e547-e556.

        6. Ost DE, Niu J, Zhao H, et al. Quality gaps and comparative effectiveness of management strategies for recurrent malignant pleural effusions. Chest. 2018;153:438-452.

        7. Miserocchi G. Physiology and pathophysiology of pleural fluid turnover. Eur Respir J. 1997;10:219-225.

        8. Sahn SA. State of the art. The pleura. Am Rev Respir Dis. 1988;138:184-234.

        9. Khaleeq G, Musani AI. Emerging paradigms in the management of malignant pleural effusions. Respir Med. 2008;102:939-948.

        10. Das DK. Serous effusions in malignant lymphomas: a review. Diagn Cytopathol. 2006;34:335-347.

        11. Qian Q, Zhan P, Sun WK, et al. Vascular endothelial growth factor and soluble intercellular adhesion molecule-1 in lung adenocarcinoma with malignant pleural effusion: correlations with patient survival and pleural effusion control. Neoplasma. 2012;59:433-439.

        12. Kraft A, Weindel K, Ochs A, et al. Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer. 1999;85:178-187.

        13. Meyer PC. Metastatic carcinoma of the pleura. Thorax. 1966;21:437-443.

        14. Light RW, Hamm H. Malignant pleural effusion: would the real cause please stand up? Eur Respir J. 1997;10:1701-1702.

        15. Chernow B, Sahn SA. Carcinomatous involvement of the pleura: an analysis of 96 patients. Am J Med. 1977;63:695-702.

        16. Musani AI, Haas AR, Seijo L, et al. Outpatient management of malignant pleural effusions with small-bore, tunneled pleural catheters. Respiration. 2004;71:559-566.

        17. Roberts ME, Neville E, Berrisford RG, et al; Group BPDG. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax. 2010;65 Suppl 2:ii32-40.

        18. Estenne M, Yernault JC, De Troyer A. Mechanism of relief of dyspnea after thoracocentesis in patients with large pleural effusions. Am J Med. 1983;74:813-819.

        19. Brown NE, Zamel N, Aberman A. Changes in pulmonary mechanics and gas exchange following thoracocentesis. Chest. 1978;74:540-542.

        20. Wang LM, Cherng JM, Wang JS. Improved lung function after thoracocentesis in patients with paradoxical movement of a hemidiaphragm secondary to a large pleural effusion. Respirology. 2007;12:719-723.

        21. Huggins JT, Doelken P, Sahn SA. The unexpandable lung. F1000 Med Rep. 2010;2:77.

        22. Lan RS, Lo SK, Chuang ML, Yang CT, Tsao TC, Lee CH. Elastance of the pleural space: a predictor for the outcome of pleurodesis in patients with malignant pleural effusion. Ann Intern Med. 1997;126:768-774.

        23. Lentz RJ, Lerner AD, Pannu JK, et al. Routine monitoring with pleural manometry during therapeutic large-volume thoracentesis to prevent pleural-pressure-related complications: a multicentre, single-blind randomised controlled trial. Lancet Respir Med. 2019;7:447-455.

        24. Porcel JM, Alvarez M, Salud A, Vives M. Should a cytologic study be ordered in transudative pleural effusions? Chest. 1999;116:1836-1837.

        25. Ryu JS, Ryu ST, Kim YS, et al. What is the clinical significance of transudative malignant pleural effusion? Korean J Intern Med. 2003;18:230-233.

        26. Boersma WG, Stigt JA, Smit HJ. Treatment of haemothorax. Respir Med. 2010;104:1583-1587.

        27. Light RW, Erozan YS, Ball WC. Cells in pleural fluid. Their value in differential diagnosis. Arch Intern Med. 1973;132:854-860.

        28. Garcia LW, Ducatman BS, Wang HH. The value of multiple fluid specimens in the cytological diagnosis of malignancy. Mod Pathol. 1994;7:665-668.

        29. Swiderek J, Morcos S, Donthireddy V, et al. Prospective study to determine the volume of pleural fluid required to diagnose malignancy. Chest. 2010;137:68-73.

        30. Abouzgheib W, Bartter T, Dagher H, Pratter M, Klump W. A prospective study of the volume of pleural fluid required for accurate diagnosis of malignant pleural effusion. Chest. 2009;135:999-1001.

        31. DeMaio A, Clarke JM, Dash R, et al. Yield of malignant pleural effusion for detection of oncogenic driver mutations in lung adenocarcinoma. J Bronchology Interv Pulmonol. 2019;26:96-101.

        32. Grosu HB, Arriola A, Stewart J, et al. PD-L1 detection in histology specimens and matched pleural fluid cell blocks of patients with NSCLC. Respirology. 2019 Jun 17. doi: 10.1111/resp.13614.

        33. Koegelenberg CF, Diacon AH. Pleural controversy: close needle pleural biopsy or thoracoscopy-which first? Respirology. 2011;16:738-746.

        34. McLaughlin KM, Kerr KM, Currie GP. Closed pleural biopsy to diagnose mesothelioma: dead or alive? Lung Cancer. 2009;65:388-389.

        35. Miyoshi S, Sasada S, Izumo T, et al. Diagnostic utility of pleural fluid cell block versus pleural biopsy collected by flex-rigid pleuroscopy for malignant pleural disease: a single center retrospective analysis. PLoS One. 2016;11:e0167186.

        36. Vanderlaan PA, Yamaguchi N, Folch E, et al. Success and failure rates of tumor genotyping techniques in routine pathological samples with non-small-cell lung cancer. Lung Cancer. 2014;84:39-44.

        37. Albanna AS, Kasymjanova G, Robitaille C, et al. Comparison of the yield of different diagnostic procedures for cellular differentiation and genetic profiling of non-small-cell lung cancer. J Thorac Oncol. 2014;9:1120-1125.

        38. Tremblay A RS, Berthiaume L, and Michaud G. Natural history of asymptomatic pleural effusions in lung cancer patients. J Bronchol. 2007;14:98-100.

        39. Porcel JM, Gasol A, Bielsa S, et al. Clinical features and survival of lung cancer patients with pleural effusions. Respirology. 2015;20:654-659.

        40. Feller-Kopman DJ, Reddy CB, DeCamp MM, et al. Management of malignant pleural effusions. An official ATS/STS/STR clinical practice guideline. Am J Respir Crit Care Med. 2018;198:839-849.

        41. Grosu HB, Molina S, Casal R, et al. Risk factors for pleural effusion recurrence in patients with malignancy. Respirology. 2019;24:76-82.

        42. Psallidas I, Kanellakis NI, Gerry S, et al. Development and validation of response markers to predict survival and pleurodesis success in patients with malignant pleural effusion (PROMISE): a multicohort analysis. Lancet Oncol. 2018;19:930-939.

        Author and Disclosure Information

        David Hsia, MD
        Health Sciences Associate Clinical Professor, Division of Respiratory and Critical Care Physiology and Medicine, Harbor-UCLA Medical Center, Torrance, CA

        Ali I. Musani, MD
        Professor of Medicine and Surgery, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Denver, CO

        Publications
        Topics
        Sections
        Author and Disclosure Information

        David Hsia, MD
        Health Sciences Associate Clinical Professor, Division of Respiratory and Critical Care Physiology and Medicine, Harbor-UCLA Medical Center, Torrance, CA

        Ali I. Musani, MD
        Professor of Medicine and Surgery, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Denver, CO

        Author and Disclosure Information

        David Hsia, MD
        Health Sciences Associate Clinical Professor, Division of Respiratory and Critical Care Physiology and Medicine, Harbor-UCLA Medical Center, Torrance, CA

        Ali I. Musani, MD
        Professor of Medicine and Surgery, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Denver, CO

        Accumulation of pleural fluid is a common clinical problem associated with malignancy. Malignant pleural effusions (MPEs) are the second most common cause of a pleural exudate, with more than 150,000 patients diagnosed annually in the United States alone.1,2 MPEs represent advanced disease and are generally a poor prognostic indicator. Median survival for patients with MPE ranges from 3 to 12 months and depends on the tumor origin.3 In addition, MPEs are a frequent cause of dyspnea and discomfort, which adversely affect a patient’s quality of life. This group of patients requires substantial medical support to manage the burden of their disease, and providing effective therapeutic management remains a challenge. In the United States, there are approximately 126,000 admissions for MPE annually, with a median length of stay of 5.5 days.4 Thirty-day readmission rates are almost 40%, which is approximately 1.5 times higher than for acute myocardial infarction and 2 times higher than for congestive heart failure.5 In addition, palliative measures for patients with MPE are probably underutilized.6

        This review is the first of 2 articles focusing on the management of MPE. Here, we discuss the pathophysiology of this disease process and provide an overview of the evaluation and diagnosis of MPE; available therapeutic options for the management of MPE are reviewed in a separate article.

        Pathogenesis and Etiology

        Normally, the thoracic cavity contains less than 15 mL of pleural fluid. Therefore, the visceral and parietal pleura are usually in close proximity to each other and the space between them is a potential space. Negative intrapleural pressures generated during regular breathing create a gradient for fluid movement into the pleural space from the parietal pleura dictated by Starling forces. Pleural fluid normally has low protein content and is primarily drained back into lymphatics through stomata lining the parietal pleura.7 This system’s ability to remove pleural fluid exceeds normal fluid production by 20- to 30-fold, suggesting that accumulation of excess pleural fluid requires a combination of increased fluid production and/or impaired fluid removal.8

        Several mechanisms have been associated with the development of MPE. Pleural involvement by malignancy may occur from direct invasion of the pleural cavity by tumor (eg, lung cancer, breast cancer, chest wall neoplasms) or hematogenous spread of tumor to the pleura (eg, metastasis, non-Hodgkin lymphoma).9,10 Pleural malignancies can produce cytokine and inflammatory mediators, which may directly increase fluid production or indirectly alter vascular permeability.11,12 Tumor cells can also disrupt lymphatic drainage by occluding either pleural stomata or downstream lymphatic drainage. However, tumor involvement of the pleura does not always result in the development of an effusion and is only associated with fluid accumulation in approximately 60% of cases.13,14 MPE have also been strongly associated with mediastinal metastases, likely resulting from obstruction of mediastinal lymphatics.13,15,16

        Pleural effusions with negative fluid cytology and pleural biopsies may result from secondary effects of tumor burden without direct pleural involvement and are referred to as paramalignant effusions. Common causes include thoracic duct obstruction (eg, Hodgkin lymphoma), bronchial obstruction, pneumonia, atelectasis, pulmonary embolism, trapped lung, and effects related to radiation or chemotherapy.15

        Lung cancer is the most frequent cause of MPE and accounts for approximately one-third of cases. Other common primary tumor sites include breast, lymphoma, ovary, and gastrointestinal. Combined, these etiologies comprise about 75% of cases (Table 1).4,5 Females comprise a greater percentage of patients with MPE mainly due to the prevalence of ovarian and breast cancer. Mesothelioma-related effusions may be more prevalent in certain parts of the world due to associated exposure to asbestos.17 The primary tumor origin remains unknown in approximately 10% of cases.4

        pdbr01608001_t1.png

        Clinical Presentation and Response to Therapeutic Drainage

        More than 75% of patients with MPE are symptomatic. Dyspnea is the most common symptom and is present in more than half of patients.15 The mechanism of dyspnea caused by large effusions may not be solely due to impaired lung volumes or gas exchange. Other associated factors include decreased chest wall compliance, mediastinal shift causing decreased volume of the contralateral lung, paradoxical motion of the diaphragm, inefficient muscle length-tension relationships resulting from the stretch of respiratory muscles, and reflex stimulation from the lungs and chest wall.18-20 Other common presenting symptoms include cough, orthopnea, and chest pain. Hemoptysis suggests endobronchial involvement of the large airways. And, given the advanced nature of most MPEs, patients may also present with weight loss and cachexia.

         

         

        A patient’s degree of symptom palliation and physiologic improvement in response to large-volume fluid removal is important to assess as these are important clinical factors that will influence management decision-making. Upwards of 50% of patients will not have significant palliation because they may be symptom-limited by other comorbid conditions, generalized deconditioning, or incomplete lung re-expansion. Presence of impaired lung compliance during fluid removal is also important to recognize. A trapped lung refers to a lung that cannot expand completely after removal of pleural fluid. Trapped lung may result from pleural-based malignancies or metastases, loculations and adhesions, or bronchial obstruction. Trapped lung is associated with high elastance (Pel) affecting pleural pressure-volume relationships (Figure 1). While clinically often considered together, some authors differentiate the category of incomplete lung expansion into 2 subgroups. In this context, the term trapped lung is used specifically to describe a mature, fibrous membrane that prevents lung re-expansion and is caused by a prior inflammatory pleural condition.21Entrapped lung describes incomplete lung expansion resulting from an active disease process, such as malignancy, ongoing infection, or rheumatologic pleurisy. Differences in pleural manometry can be seen in the 2 subgroups. Pleural manometry can be helpful to monitor for the generation of high negative intrapleural pressures during fluid removal, with negative pressures in excess of –19 cm H2O being suggestive of trapped lung physiology.22 However routine use of pleural manometry has not been shown to avoid the development of procedure-related chest discomfort that develops when the lung is unable to expand in response to the removal of fluid.23

        PDBR01608001_f1.PNG

        Pleural Fluid Analysis and Pleural Biopsy

        While most MPEs are protein-rich exudates, approximately 2% to 5% may be transudates.24,25 MPEs often appear hemorrhagic, so a ratio of pleural fluid to blood serum hematocrit greater than 0.5 is used to distinguish a true hemothorax from bloody-appearing pleural fluid.26 The cell count may be lymphocyte-predominant, but other cell types, such as eosinophils, do not exclude malignancy.27 Fluid may have a low glucose concentration and pH as well.

        Thoracentesis with pleural fluid cytology evaluation is the most common method of diagnosis. The diagnostic sensitivity of fluid cytology ranges from 62% to 90%, with variability resulting from the extent of disease and etiology of the primary malignancy.1 If the initial pleural fluid analysis is not diagnostic, repeat thoracentesis can improve the diagnostic yield, but subsequent sampling has diminishing utility. In one series, diagnosis of malignancy was made by fluid cytology analysis in 65% of patients from the initial thoracentesis, 27% from a second procedure, but only 5% from a third procedure.28 At least 50 to 60 mL of pleural fluid should be obtained for pleural fluid cytology, but analysis of significantly larger volumes may not appreciably improve diagnostic yield.29,30

        In addition to diagnostic yield, adequate sample cellularity to test for genetic driver mutations has become increasingly important given the rapid development of targeted therapies that are now available. The relative paucity of malignant cells in pleural fluid compared to other types of biopsies can make MPEs difficult to analyze for molecular markers. Newer generation assays have increased sensitivity, with one series reporting that pleural fluid was sufficient in 71.4% of cases to analyze for a panel comprised of EGFR, KRAS, BRAF, ALK, and ROS1 mutations.31 Similarly, fluid analysis from patients with MPEs demonstrated that 71.3% had at least 100 tumor cells, which permitted evaluation for PD-L1, with a concordance of 0.78 when compared to matched parenchymal lung biopsies from the same patient.32

        In contrast, pleural biopsy methods may be useful to increase the diagnostic yield when pleural fluid analysis is insufficient. Closed needle biopsy may marginally improve diagnostic yields for malignancy over pleural fluid analysis alone. Diagnostic sensitivity may improve with the use of point-of-care ultrasonography to guide needle placement.33,34 The true value of closed needle biopsy is seen in situations in which there is a high pretest probability to diagnose an alternative disseminated pleural process, such as in tuberculosis, where the diagnostic yield increases substantially with closed needle biopsy of the pleura.33 Otherwise, the diagnosis of lung cancer and mesothelioma is superior with visually guided pleural biopsies, such as medical thoracoscopy or video-assisted thoracoscopic surgery (VATS), with diagnostic yields over 90%.33,35 Testing for genetic driver mutations in pleural biopsies is also substantially improved, with sample adequacy of 90% to 95% for most molecular markers.36,37 Despite the advantages, pleural biopsies are generally reserved for cases when pleural fluid analysis is insufficient or when performed in conjunction with palliative therapeutic interventions due to the increased invasive nature of the procedure.

         

         

        Predictors of Recurrence and Prognosis

        Not all MPEs will progress in size or become symptomatic, and predicting which patients will develop symptoms from their effusions is difficult. Pleural effusions will develop in only a minority of patients with lung cancer, and only a small subset will progress and require therapeutic intervention.38,39 Therefore, management guidelines for malignant pleural effusions discourage empiric intervention for patients with small, asymptomatic effusions.40 However, patients with larger, symptomatic effusions are more likely to have significant and rapid fluid recurrence. In a series of 988 symptomatic patients undergoing drainage, 30% had fluid recurrence within 15 days, 40% within 30 days, 45% within 60 days, and 48% within 90 days.41 Factors associated with fluid recurrence included radiographic size of the effusion, requirement for a larger amount of fluid to be initially drained, and higher pleural fluid lactate dehydrogenase (LDH) level. Negative cytology was associated with lower likelihood for recurrence.

        Prognostication of life expectancy is another important clinical assessment which impacts medical decision-making when weighing the risk and benefits of different palliation options. Patient performance status, pleural fluid LDH, serum neutrophil-to-lymphocyte ratio, and tumor origin are independently associated with prognosis in a validated scoring system (Table 2).3 In this study, the overall median survival of patients with MPE was approximately 4.5 months, while the median survival for patients with mesothelioma was 11.3 months, 6.6 months for breast cancer, and 2.5 months for lung cancer and other malignancies. When stratified based on the combination of these 4 variables, patients in the high-risk group had a median survival of just 44 days compared to 130 days for the moderate-risk group and 319 days for the low-risk group. Additional, more complex prediction systems for survival and response to MPE therapies are now emerging and may provide clinicians and patients with additional information useful in medical decision-making.42

        PDBR01608001_t2.PNG

        Conclusion

        MPEs represent advanced stage disease and frequently adversely affect a pa­tient’s quality of life. Ideal therapeutic options, discussed in the second part of this review, should effectively palliate symptoms, provide long-term relief, be minimally invasive with few side effects, minimize hospitalization and reliance on medical assistance, and be cost-effective.

        Accumulation of pleural fluid is a common clinical problem associated with malignancy. Malignant pleural effusions (MPEs) are the second most common cause of a pleural exudate, with more than 150,000 patients diagnosed annually in the United States alone.1,2 MPEs represent advanced disease and are generally a poor prognostic indicator. Median survival for patients with MPE ranges from 3 to 12 months and depends on the tumor origin.3 In addition, MPEs are a frequent cause of dyspnea and discomfort, which adversely affect a patient’s quality of life. This group of patients requires substantial medical support to manage the burden of their disease, and providing effective therapeutic management remains a challenge. In the United States, there are approximately 126,000 admissions for MPE annually, with a median length of stay of 5.5 days.4 Thirty-day readmission rates are almost 40%, which is approximately 1.5 times higher than for acute myocardial infarction and 2 times higher than for congestive heart failure.5 In addition, palliative measures for patients with MPE are probably underutilized.6

        This review is the first of 2 articles focusing on the management of MPE. Here, we discuss the pathophysiology of this disease process and provide an overview of the evaluation and diagnosis of MPE; available therapeutic options for the management of MPE are reviewed in a separate article.

        Pathogenesis and Etiology

        Normally, the thoracic cavity contains less than 15 mL of pleural fluid. Therefore, the visceral and parietal pleura are usually in close proximity to each other and the space between them is a potential space. Negative intrapleural pressures generated during regular breathing create a gradient for fluid movement into the pleural space from the parietal pleura dictated by Starling forces. Pleural fluid normally has low protein content and is primarily drained back into lymphatics through stomata lining the parietal pleura.7 This system’s ability to remove pleural fluid exceeds normal fluid production by 20- to 30-fold, suggesting that accumulation of excess pleural fluid requires a combination of increased fluid production and/or impaired fluid removal.8

        Several mechanisms have been associated with the development of MPE. Pleural involvement by malignancy may occur from direct invasion of the pleural cavity by tumor (eg, lung cancer, breast cancer, chest wall neoplasms) or hematogenous spread of tumor to the pleura (eg, metastasis, non-Hodgkin lymphoma).9,10 Pleural malignancies can produce cytokine and inflammatory mediators, which may directly increase fluid production or indirectly alter vascular permeability.11,12 Tumor cells can also disrupt lymphatic drainage by occluding either pleural stomata or downstream lymphatic drainage. However, tumor involvement of the pleura does not always result in the development of an effusion and is only associated with fluid accumulation in approximately 60% of cases.13,14 MPE have also been strongly associated with mediastinal metastases, likely resulting from obstruction of mediastinal lymphatics.13,15,16

        Pleural effusions with negative fluid cytology and pleural biopsies may result from secondary effects of tumor burden without direct pleural involvement and are referred to as paramalignant effusions. Common causes include thoracic duct obstruction (eg, Hodgkin lymphoma), bronchial obstruction, pneumonia, atelectasis, pulmonary embolism, trapped lung, and effects related to radiation or chemotherapy.15

        Lung cancer is the most frequent cause of MPE and accounts for approximately one-third of cases. Other common primary tumor sites include breast, lymphoma, ovary, and gastrointestinal. Combined, these etiologies comprise about 75% of cases (Table 1).4,5 Females comprise a greater percentage of patients with MPE mainly due to the prevalence of ovarian and breast cancer. Mesothelioma-related effusions may be more prevalent in certain parts of the world due to associated exposure to asbestos.17 The primary tumor origin remains unknown in approximately 10% of cases.4

        pdbr01608001_t1.png

        Clinical Presentation and Response to Therapeutic Drainage

        More than 75% of patients with MPE are symptomatic. Dyspnea is the most common symptom and is present in more than half of patients.15 The mechanism of dyspnea caused by large effusions may not be solely due to impaired lung volumes or gas exchange. Other associated factors include decreased chest wall compliance, mediastinal shift causing decreased volume of the contralateral lung, paradoxical motion of the diaphragm, inefficient muscle length-tension relationships resulting from the stretch of respiratory muscles, and reflex stimulation from the lungs and chest wall.18-20 Other common presenting symptoms include cough, orthopnea, and chest pain. Hemoptysis suggests endobronchial involvement of the large airways. And, given the advanced nature of most MPEs, patients may also present with weight loss and cachexia.

         

         

        A patient’s degree of symptom palliation and physiologic improvement in response to large-volume fluid removal is important to assess as these are important clinical factors that will influence management decision-making. Upwards of 50% of patients will not have significant palliation because they may be symptom-limited by other comorbid conditions, generalized deconditioning, or incomplete lung re-expansion. Presence of impaired lung compliance during fluid removal is also important to recognize. A trapped lung refers to a lung that cannot expand completely after removal of pleural fluid. Trapped lung may result from pleural-based malignancies or metastases, loculations and adhesions, or bronchial obstruction. Trapped lung is associated with high elastance (Pel) affecting pleural pressure-volume relationships (Figure 1). While clinically often considered together, some authors differentiate the category of incomplete lung expansion into 2 subgroups. In this context, the term trapped lung is used specifically to describe a mature, fibrous membrane that prevents lung re-expansion and is caused by a prior inflammatory pleural condition.21Entrapped lung describes incomplete lung expansion resulting from an active disease process, such as malignancy, ongoing infection, or rheumatologic pleurisy. Differences in pleural manometry can be seen in the 2 subgroups. Pleural manometry can be helpful to monitor for the generation of high negative intrapleural pressures during fluid removal, with negative pressures in excess of –19 cm H2O being suggestive of trapped lung physiology.22 However routine use of pleural manometry has not been shown to avoid the development of procedure-related chest discomfort that develops when the lung is unable to expand in response to the removal of fluid.23

        PDBR01608001_f1.PNG

        Pleural Fluid Analysis and Pleural Biopsy

        While most MPEs are protein-rich exudates, approximately 2% to 5% may be transudates.24,25 MPEs often appear hemorrhagic, so a ratio of pleural fluid to blood serum hematocrit greater than 0.5 is used to distinguish a true hemothorax from bloody-appearing pleural fluid.26 The cell count may be lymphocyte-predominant, but other cell types, such as eosinophils, do not exclude malignancy.27 Fluid may have a low glucose concentration and pH as well.

        Thoracentesis with pleural fluid cytology evaluation is the most common method of diagnosis. The diagnostic sensitivity of fluid cytology ranges from 62% to 90%, with variability resulting from the extent of disease and etiology of the primary malignancy.1 If the initial pleural fluid analysis is not diagnostic, repeat thoracentesis can improve the diagnostic yield, but subsequent sampling has diminishing utility. In one series, diagnosis of malignancy was made by fluid cytology analysis in 65% of patients from the initial thoracentesis, 27% from a second procedure, but only 5% from a third procedure.28 At least 50 to 60 mL of pleural fluid should be obtained for pleural fluid cytology, but analysis of significantly larger volumes may not appreciably improve diagnostic yield.29,30

        In addition to diagnostic yield, adequate sample cellularity to test for genetic driver mutations has become increasingly important given the rapid development of targeted therapies that are now available. The relative paucity of malignant cells in pleural fluid compared to other types of biopsies can make MPEs difficult to analyze for molecular markers. Newer generation assays have increased sensitivity, with one series reporting that pleural fluid was sufficient in 71.4% of cases to analyze for a panel comprised of EGFR, KRAS, BRAF, ALK, and ROS1 mutations.31 Similarly, fluid analysis from patients with MPEs demonstrated that 71.3% had at least 100 tumor cells, which permitted evaluation for PD-L1, with a concordance of 0.78 when compared to matched parenchymal lung biopsies from the same patient.32

        In contrast, pleural biopsy methods may be useful to increase the diagnostic yield when pleural fluid analysis is insufficient. Closed needle biopsy may marginally improve diagnostic yields for malignancy over pleural fluid analysis alone. Diagnostic sensitivity may improve with the use of point-of-care ultrasonography to guide needle placement.33,34 The true value of closed needle biopsy is seen in situations in which there is a high pretest probability to diagnose an alternative disseminated pleural process, such as in tuberculosis, where the diagnostic yield increases substantially with closed needle biopsy of the pleura.33 Otherwise, the diagnosis of lung cancer and mesothelioma is superior with visually guided pleural biopsies, such as medical thoracoscopy or video-assisted thoracoscopic surgery (VATS), with diagnostic yields over 90%.33,35 Testing for genetic driver mutations in pleural biopsies is also substantially improved, with sample adequacy of 90% to 95% for most molecular markers.36,37 Despite the advantages, pleural biopsies are generally reserved for cases when pleural fluid analysis is insufficient or when performed in conjunction with palliative therapeutic interventions due to the increased invasive nature of the procedure.

         

         

        Predictors of Recurrence and Prognosis

        Not all MPEs will progress in size or become symptomatic, and predicting which patients will develop symptoms from their effusions is difficult. Pleural effusions will develop in only a minority of patients with lung cancer, and only a small subset will progress and require therapeutic intervention.38,39 Therefore, management guidelines for malignant pleural effusions discourage empiric intervention for patients with small, asymptomatic effusions.40 However, patients with larger, symptomatic effusions are more likely to have significant and rapid fluid recurrence. In a series of 988 symptomatic patients undergoing drainage, 30% had fluid recurrence within 15 days, 40% within 30 days, 45% within 60 days, and 48% within 90 days.41 Factors associated with fluid recurrence included radiographic size of the effusion, requirement for a larger amount of fluid to be initially drained, and higher pleural fluid lactate dehydrogenase (LDH) level. Negative cytology was associated with lower likelihood for recurrence.

        Prognostication of life expectancy is another important clinical assessment which impacts medical decision-making when weighing the risk and benefits of different palliation options. Patient performance status, pleural fluid LDH, serum neutrophil-to-lymphocyte ratio, and tumor origin are independently associated with prognosis in a validated scoring system (Table 2).3 In this study, the overall median survival of patients with MPE was approximately 4.5 months, while the median survival for patients with mesothelioma was 11.3 months, 6.6 months for breast cancer, and 2.5 months for lung cancer and other malignancies. When stratified based on the combination of these 4 variables, patients in the high-risk group had a median survival of just 44 days compared to 130 days for the moderate-risk group and 319 days for the low-risk group. Additional, more complex prediction systems for survival and response to MPE therapies are now emerging and may provide clinicians and patients with additional information useful in medical decision-making.42

        PDBR01608001_t2.PNG

        Conclusion

        MPEs represent advanced stage disease and frequently adversely affect a pa­tient’s quality of life. Ideal therapeutic options, discussed in the second part of this review, should effectively palliate symptoms, provide long-term relief, be minimally invasive with few side effects, minimize hospitalization and reliance on medical assistance, and be cost-effective.

        References

        1. Antony VB, Loddenkemper R, Astoul P, et al. Management of malignant pleural effusions. Eur Respir J. 2001;18:402-419.

        2. Society AT. Management of malignant pleural effusions. Am J Respir Crit Care Med. 2000;162:1987-2001.

        3. Clive AO, Kahan BC, Hooper CE, et al. Predicting survival in malignant pleural effusion: development and validation of the LENT prognostic score. Thorax. 2014;69:1098-1104.

        4. Taghizadeh N, Fortin M, Tremblay A. US hospitalizations for malignant pleural effusions: data from the 2012 National Inpatient Sample. Chest. 2017;151:845-854.

        5. Yang TS, Hsia DW, Chang DW. Patient- and hospital-level factors associated with readmission for malignant pleural effusion. J Oncol Pract. 2018;14:e547-e556.

        6. Ost DE, Niu J, Zhao H, et al. Quality gaps and comparative effectiveness of management strategies for recurrent malignant pleural effusions. Chest. 2018;153:438-452.

        7. Miserocchi G. Physiology and pathophysiology of pleural fluid turnover. Eur Respir J. 1997;10:219-225.

        8. Sahn SA. State of the art. The pleura. Am Rev Respir Dis. 1988;138:184-234.

        9. Khaleeq G, Musani AI. Emerging paradigms in the management of malignant pleural effusions. Respir Med. 2008;102:939-948.

        10. Das DK. Serous effusions in malignant lymphomas: a review. Diagn Cytopathol. 2006;34:335-347.

        11. Qian Q, Zhan P, Sun WK, et al. Vascular endothelial growth factor and soluble intercellular adhesion molecule-1 in lung adenocarcinoma with malignant pleural effusion: correlations with patient survival and pleural effusion control. Neoplasma. 2012;59:433-439.

        12. Kraft A, Weindel K, Ochs A, et al. Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer. 1999;85:178-187.

        13. Meyer PC. Metastatic carcinoma of the pleura. Thorax. 1966;21:437-443.

        14. Light RW, Hamm H. Malignant pleural effusion: would the real cause please stand up? Eur Respir J. 1997;10:1701-1702.

        15. Chernow B, Sahn SA. Carcinomatous involvement of the pleura: an analysis of 96 patients. Am J Med. 1977;63:695-702.

        16. Musani AI, Haas AR, Seijo L, et al. Outpatient management of malignant pleural effusions with small-bore, tunneled pleural catheters. Respiration. 2004;71:559-566.

        17. Roberts ME, Neville E, Berrisford RG, et al; Group BPDG. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax. 2010;65 Suppl 2:ii32-40.

        18. Estenne M, Yernault JC, De Troyer A. Mechanism of relief of dyspnea after thoracocentesis in patients with large pleural effusions. Am J Med. 1983;74:813-819.

        19. Brown NE, Zamel N, Aberman A. Changes in pulmonary mechanics and gas exchange following thoracocentesis. Chest. 1978;74:540-542.

        20. Wang LM, Cherng JM, Wang JS. Improved lung function after thoracocentesis in patients with paradoxical movement of a hemidiaphragm secondary to a large pleural effusion. Respirology. 2007;12:719-723.

        21. Huggins JT, Doelken P, Sahn SA. The unexpandable lung. F1000 Med Rep. 2010;2:77.

        22. Lan RS, Lo SK, Chuang ML, Yang CT, Tsao TC, Lee CH. Elastance of the pleural space: a predictor for the outcome of pleurodesis in patients with malignant pleural effusion. Ann Intern Med. 1997;126:768-774.

        23. Lentz RJ, Lerner AD, Pannu JK, et al. Routine monitoring with pleural manometry during therapeutic large-volume thoracentesis to prevent pleural-pressure-related complications: a multicentre, single-blind randomised controlled trial. Lancet Respir Med. 2019;7:447-455.

        24. Porcel JM, Alvarez M, Salud A, Vives M. Should a cytologic study be ordered in transudative pleural effusions? Chest. 1999;116:1836-1837.

        25. Ryu JS, Ryu ST, Kim YS, et al. What is the clinical significance of transudative malignant pleural effusion? Korean J Intern Med. 2003;18:230-233.

        26. Boersma WG, Stigt JA, Smit HJ. Treatment of haemothorax. Respir Med. 2010;104:1583-1587.

        27. Light RW, Erozan YS, Ball WC. Cells in pleural fluid. Their value in differential diagnosis. Arch Intern Med. 1973;132:854-860.

        28. Garcia LW, Ducatman BS, Wang HH. The value of multiple fluid specimens in the cytological diagnosis of malignancy. Mod Pathol. 1994;7:665-668.

        29. Swiderek J, Morcos S, Donthireddy V, et al. Prospective study to determine the volume of pleural fluid required to diagnose malignancy. Chest. 2010;137:68-73.

        30. Abouzgheib W, Bartter T, Dagher H, Pratter M, Klump W. A prospective study of the volume of pleural fluid required for accurate diagnosis of malignant pleural effusion. Chest. 2009;135:999-1001.

        31. DeMaio A, Clarke JM, Dash R, et al. Yield of malignant pleural effusion for detection of oncogenic driver mutations in lung adenocarcinoma. J Bronchology Interv Pulmonol. 2019;26:96-101.

        32. Grosu HB, Arriola A, Stewart J, et al. PD-L1 detection in histology specimens and matched pleural fluid cell blocks of patients with NSCLC. Respirology. 2019 Jun 17. doi: 10.1111/resp.13614.

        33. Koegelenberg CF, Diacon AH. Pleural controversy: close needle pleural biopsy or thoracoscopy-which first? Respirology. 2011;16:738-746.

        34. McLaughlin KM, Kerr KM, Currie GP. Closed pleural biopsy to diagnose mesothelioma: dead or alive? Lung Cancer. 2009;65:388-389.

        35. Miyoshi S, Sasada S, Izumo T, et al. Diagnostic utility of pleural fluid cell block versus pleural biopsy collected by flex-rigid pleuroscopy for malignant pleural disease: a single center retrospective analysis. PLoS One. 2016;11:e0167186.

        36. Vanderlaan PA, Yamaguchi N, Folch E, et al. Success and failure rates of tumor genotyping techniques in routine pathological samples with non-small-cell lung cancer. Lung Cancer. 2014;84:39-44.

        37. Albanna AS, Kasymjanova G, Robitaille C, et al. Comparison of the yield of different diagnostic procedures for cellular differentiation and genetic profiling of non-small-cell lung cancer. J Thorac Oncol. 2014;9:1120-1125.

        38. Tremblay A RS, Berthiaume L, and Michaud G. Natural history of asymptomatic pleural effusions in lung cancer patients. J Bronchol. 2007;14:98-100.

        39. Porcel JM, Gasol A, Bielsa S, et al. Clinical features and survival of lung cancer patients with pleural effusions. Respirology. 2015;20:654-659.

        40. Feller-Kopman DJ, Reddy CB, DeCamp MM, et al. Management of malignant pleural effusions. An official ATS/STS/STR clinical practice guideline. Am J Respir Crit Care Med. 2018;198:839-849.

        41. Grosu HB, Molina S, Casal R, et al. Risk factors for pleural effusion recurrence in patients with malignancy. Respirology. 2019;24:76-82.

        42. Psallidas I, Kanellakis NI, Gerry S, et al. Development and validation of response markers to predict survival and pleurodesis success in patients with malignant pleural effusion (PROMISE): a multicohort analysis. Lancet Oncol. 2018;19:930-939.

        References

        1. Antony VB, Loddenkemper R, Astoul P, et al. Management of malignant pleural effusions. Eur Respir J. 2001;18:402-419.

        2. Society AT. Management of malignant pleural effusions. Am J Respir Crit Care Med. 2000;162:1987-2001.

        3. Clive AO, Kahan BC, Hooper CE, et al. Predicting survival in malignant pleural effusion: development and validation of the LENT prognostic score. Thorax. 2014;69:1098-1104.

        4. Taghizadeh N, Fortin M, Tremblay A. US hospitalizations for malignant pleural effusions: data from the 2012 National Inpatient Sample. Chest. 2017;151:845-854.

        5. Yang TS, Hsia DW, Chang DW. Patient- and hospital-level factors associated with readmission for malignant pleural effusion. J Oncol Pract. 2018;14:e547-e556.

        6. Ost DE, Niu J, Zhao H, et al. Quality gaps and comparative effectiveness of management strategies for recurrent malignant pleural effusions. Chest. 2018;153:438-452.

        7. Miserocchi G. Physiology and pathophysiology of pleural fluid turnover. Eur Respir J. 1997;10:219-225.

        8. Sahn SA. State of the art. The pleura. Am Rev Respir Dis. 1988;138:184-234.

        9. Khaleeq G, Musani AI. Emerging paradigms in the management of malignant pleural effusions. Respir Med. 2008;102:939-948.

        10. Das DK. Serous effusions in malignant lymphomas: a review. Diagn Cytopathol. 2006;34:335-347.

        11. Qian Q, Zhan P, Sun WK, et al. Vascular endothelial growth factor and soluble intercellular adhesion molecule-1 in lung adenocarcinoma with malignant pleural effusion: correlations with patient survival and pleural effusion control. Neoplasma. 2012;59:433-439.

        12. Kraft A, Weindel K, Ochs A, et al. Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer. 1999;85:178-187.

        13. Meyer PC. Metastatic carcinoma of the pleura. Thorax. 1966;21:437-443.

        14. Light RW, Hamm H. Malignant pleural effusion: would the real cause please stand up? Eur Respir J. 1997;10:1701-1702.

        15. Chernow B, Sahn SA. Carcinomatous involvement of the pleura: an analysis of 96 patients. Am J Med. 1977;63:695-702.

        16. Musani AI, Haas AR, Seijo L, et al. Outpatient management of malignant pleural effusions with small-bore, tunneled pleural catheters. Respiration. 2004;71:559-566.

        17. Roberts ME, Neville E, Berrisford RG, et al; Group BPDG. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax. 2010;65 Suppl 2:ii32-40.

        18. Estenne M, Yernault JC, De Troyer A. Mechanism of relief of dyspnea after thoracocentesis in patients with large pleural effusions. Am J Med. 1983;74:813-819.

        19. Brown NE, Zamel N, Aberman A. Changes in pulmonary mechanics and gas exchange following thoracocentesis. Chest. 1978;74:540-542.

        20. Wang LM, Cherng JM, Wang JS. Improved lung function after thoracocentesis in patients with paradoxical movement of a hemidiaphragm secondary to a large pleural effusion. Respirology. 2007;12:719-723.

        21. Huggins JT, Doelken P, Sahn SA. The unexpandable lung. F1000 Med Rep. 2010;2:77.

        22. Lan RS, Lo SK, Chuang ML, Yang CT, Tsao TC, Lee CH. Elastance of the pleural space: a predictor for the outcome of pleurodesis in patients with malignant pleural effusion. Ann Intern Med. 1997;126:768-774.

        23. Lentz RJ, Lerner AD, Pannu JK, et al. Routine monitoring with pleural manometry during therapeutic large-volume thoracentesis to prevent pleural-pressure-related complications: a multicentre, single-blind randomised controlled trial. Lancet Respir Med. 2019;7:447-455.

        24. Porcel JM, Alvarez M, Salud A, Vives M. Should a cytologic study be ordered in transudative pleural effusions? Chest. 1999;116:1836-1837.

        25. Ryu JS, Ryu ST, Kim YS, et al. What is the clinical significance of transudative malignant pleural effusion? Korean J Intern Med. 2003;18:230-233.

        26. Boersma WG, Stigt JA, Smit HJ. Treatment of haemothorax. Respir Med. 2010;104:1583-1587.

        27. Light RW, Erozan YS, Ball WC. Cells in pleural fluid. Their value in differential diagnosis. Arch Intern Med. 1973;132:854-860.

        28. Garcia LW, Ducatman BS, Wang HH. The value of multiple fluid specimens in the cytological diagnosis of malignancy. Mod Pathol. 1994;7:665-668.

        29. Swiderek J, Morcos S, Donthireddy V, et al. Prospective study to determine the volume of pleural fluid required to diagnose malignancy. Chest. 2010;137:68-73.

        30. Abouzgheib W, Bartter T, Dagher H, Pratter M, Klump W. A prospective study of the volume of pleural fluid required for accurate diagnosis of malignant pleural effusion. Chest. 2009;135:999-1001.

        31. DeMaio A, Clarke JM, Dash R, et al. Yield of malignant pleural effusion for detection of oncogenic driver mutations in lung adenocarcinoma. J Bronchology Interv Pulmonol. 2019;26:96-101.

        32. Grosu HB, Arriola A, Stewart J, et al. PD-L1 detection in histology specimens and matched pleural fluid cell blocks of patients with NSCLC. Respirology. 2019 Jun 17. doi: 10.1111/resp.13614.

        33. Koegelenberg CF, Diacon AH. Pleural controversy: close needle pleural biopsy or thoracoscopy-which first? Respirology. 2011;16:738-746.

        34. McLaughlin KM, Kerr KM, Currie GP. Closed pleural biopsy to diagnose mesothelioma: dead or alive? Lung Cancer. 2009;65:388-389.

        35. Miyoshi S, Sasada S, Izumo T, et al. Diagnostic utility of pleural fluid cell block versus pleural biopsy collected by flex-rigid pleuroscopy for malignant pleural disease: a single center retrospective analysis. PLoS One. 2016;11:e0167186.

        36. Vanderlaan PA, Yamaguchi N, Folch E, et al. Success and failure rates of tumor genotyping techniques in routine pathological samples with non-small-cell lung cancer. Lung Cancer. 2014;84:39-44.

        37. Albanna AS, Kasymjanova G, Robitaille C, et al. Comparison of the yield of different diagnostic procedures for cellular differentiation and genetic profiling of non-small-cell lung cancer. J Thorac Oncol. 2014;9:1120-1125.

        38. Tremblay A RS, Berthiaume L, and Michaud G. Natural history of asymptomatic pleural effusions in lung cancer patients. J Bronchol. 2007;14:98-100.

        39. Porcel JM, Gasol A, Bielsa S, et al. Clinical features and survival of lung cancer patients with pleural effusions. Respirology. 2015;20:654-659.

        40. Feller-Kopman DJ, Reddy CB, DeCamp MM, et al. Management of malignant pleural effusions. An official ATS/STS/STR clinical practice guideline. Am J Respir Crit Care Med. 2018;198:839-849.

        41. Grosu HB, Molina S, Casal R, et al. Risk factors for pleural effusion recurrence in patients with malignancy. Respirology. 2019;24:76-82.

        42. Psallidas I, Kanellakis NI, Gerry S, et al. Development and validation of response markers to predict survival and pleurodesis success in patients with malignant pleural effusion (PROMISE): a multicohort analysis. Lancet Oncol. 2018;19:930-939.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Malignant Pleural Effusion: Evaluation and Diagnosis
        Display Headline
        Malignant Pleural Effusion: Evaluation and Diagnosis
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 August;16(2)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Tue, 08/27/2019 - 10:00
        Un-Gate On Date
        Tue, 08/27/2019 - 10:00
        Use ProPublica
        CFC Schedule Remove Status
        Tue, 08/27/2019 - 10:00
        Hide sidebar & use full width
        render the right sidebar.

        Stable COPD: Initiating and Optimizing Therapy

        Article Type
        Changed
        Wed, 07/17/2019 - 14:54
        Display Headline
        Stable COPD: Initiating and Optimizing Therapy

        Chronic obstructive pulmonary disease (COPD) is a systemic inflammatory disease characterized by irreversible obstructive ventilatory defects.1-4 It is a major cause of morbidity and mortality, affecting 5% of the population in the United States and ranking as the third leading cause of death in 2008.5,6 The goals in COPD management are to provide symptom relief, improve the quality of life, preserve lung function, and reduce the frequency of exacerbations and mortality. In this 3-part review, we discuss the management of stable COPD in the context of 3 common clinical scenarios: initiating and optimizing therapy, managing acute exacerbations, and managing advanced disease.

        Case Presentation

        A 65-year-old man with COPD underwent pulmonary function testing (PFT), which demonstrated an obstructive ventilatory defect: forced expiratory volume in 1 second/forced vital capacity ratio (FEV1/FVC), 0.45; FEV1, 2 L (65% of predicted); and diffusing capacity of the lung for carbon monoxide, 15 mL/min/mm Hg (65% of predicted). He has dyspnea with strenuous exercise but is comfortable at rest and with minimal exercise. He has had 1 exacerbation in the past year, and this was treated on an outpatient basis with steroids and antibiotics. His medication regimen includes inhaled tiotropium once daily and inhaled albuterol as needed that he uses roughly twice a week.

        What determines the appropriate therapy for a given COPD patient?

        COPD management is guided by disease severity that is measured using a multimodal staging system developed by the Global Initiative for Chronic Obstructive Lung Disease (GOLD). The initial classification adopted by the GOLD 2011 report encompassed 4 categories based on symptoms, number of exacerbations, and degree of airflow limitation on PFT. However, in 2017 the GOLD ABCD classification was modified to consider only symptoms and risk of exacerbation in classifying patients, regardless of performance on spirometry and FEV1 (Figure 1).7,8 This approach was intended to make therapy more individualized based on the patient clinical profile. The Table provides a summary of the recommended treatments according to classification based on the GOLD 2017 report.

        pdbr01607001_f1.png

        The patient in our clinical scenario can be classified as GOLD category B.

        pdbr01607001_t1.png

        What is the approach to building a pharmacologic regimen for the patient with COPD?

        The backbone of the pharmacologic regimen for COPD includes short- and long-acting bronchodilators. They are usually given in an inhaled form to maximize local effects on the lungs and minimize systemic side effects. There are 2 main classes of bronchodilators, beta-agonists and muscarinic antagonists, and each targets specific receptors on the surface of airway smooth muscle cells. Beta- agonists work by stimulating beta-2 receptors, resulting in bronchodilation, while muscarinic antagonists work by blocking the bronchoconstrictor action of M3 muscarinic receptors. Inhaled corticosteroids can be added to long-acting bronchodilator therapy but cannot be used as stand-alone therapy. Theophylline is an oral bronchodilator that is used infrequently due to its narrow therapeutic index, toxicity, and multiple drug interactions.

        Figure 2 presents an approach to building a treatment plan for the patient with stable COPD.

        pdbr01607001_f2.png

        Who should be on short-acting bronchodilators? What is the best agent? Should it be scheduled or used as needed?

        All patients with COPD should be an on inhaled short-acting bronchodilator as needed for relief of symptoms.7 Both short-acting beta-agonists (albuterol and levalbuterol) and short-acting muscarinic antagonists (ipratropium) have been shown in clinical trials and meta-analyses to improve symptoms and lung function in patients with stable COPD9,10 and seem to have comparative efficacy when compared head-to-head in trials.11 However, the airway bronchodilator effect achieved by both classes seems to be additive when used in combination and is also associated with fewer exacerbations compared to albuterol alone.12 On the other hand, adding albuterol to ipratropium increased the bronchodilator response but did not reduce the exacerbation rate.11-13 Inhaled short-acting beta-agonists when used as needed rather than scheduled are associated with less medication use without any significant difference in symptoms or lung function.14

        The side effects related to using recommended doses of a short-acting bronchodilator are minimal. In retrospective studies, short-acting beta-agonists increased the risk of severe cardiac arrhythmias.15 Levalbuterol, the active enantiomer of albuterol (R-albuterol) developed for the theoretical benefits of reduced tachycardia, increased tolerability, and better or equal efficacy compared to racemic albuterol, failed to show a clinically significant difference in inducing tachycardia.16 Beta-agonist overuse is associated with tremor and in severe cases hypokalemia, which happens mainly when patients try to achieve maximal bronchodilation; the clinically used doses of beta agonists are associated with fewer side effects but achieve less than maximal bronchodilation.17 Ipratropium can produce systemic anticholinergic side effects, urinary retention being the most clinically significant, especially when combined with long-acting anticholinergic agents.18

         

         

        In light of the above discussion, a combination of a short-acting beta-agonist and a muscarinic antagonist is recommended in all patients with COPD, unless the patient is on a long-acting muscarinic antagonist (LAMA).7,18 In the latter case, a short-acting beta agonist used as a rescue inhaler is the best option. In our patient, albuterol was the choice for his short-acting bronchodilator, as he was using the LAMA tiotropium.

        Are short-acting bronchodilators enough? What do we use for maintenance therapy?

        All patients with COPD who are category B or higher according to the modified GOLD staging system should be on a long-acting bronchodilator:7,19 either a long-acting beta-agonist (LABA) or a LAMA. Long-acting bronchodilators work on the same receptors as their short-acting counterparts but have structural differences. Salmeterol is the prototype long-acting selective beta-2 agonist. It is structurally similar to albuterol but has an elongated side chain that allows it to bind firmly to the area of beta receptors and stimulate them repetitively, resulting in an extended-duration of action.20 Tiotropium on the other hand is a quaternary ammonium of ipratropium that is a nonselective muscarinic antagonist.21 Compared to ipratropium, tiotropium dissociates more quickly from M2 receptors, which is responsible for the undesired anticholinergic effects, while at the same time it binds M1 and M3 receptors for a prolonged time, resulting in an extended duration of action.21 Revefenacin is a new lung-selective LAMA that is under development and has shown promise among those with moderate to very severe COPD. Results are only limited to phase 3 trials, and clinical studies are still underway.22

        The currently available LABAs include salmeterol, formoterol, arformoterol, olodaterol, and indacaterol. The last 2 have the advantage of once-daily dosing rather than twice daily.23,24 LABAs have been shown to improve lung function, exacerbation rate, and quality of life in multiple clinical trials.23,25 Vilanterol is another LABA that has a long duration of action and can be used once daily,26 but is only available in a combination with umeclidinium, a LAMA. Several LAMAs are approved for use in COPD, including the prototype tiotropium, in addition to aclidinium, umeclidinium, and glycopyrronium. These have been shown in clinical trials to improve lung function, symptoms, and exacerbation rate.27-30

        Patients can be started on either a LAMA or LABA depending on the individual patient's needs and the agent's adverse effects.7 Both have comparable adverse effects and efficacy, as detailed below. Concerning adverse effects, there is conflicting data concerning an association of cardiovascular events with both classes of long-acting bronchodilators. While clinical trials failed to show an increased risk,25,31,32 several retrospective studies showed an increased risk of emergency room visits and hospitalizations due to tachyarrhythmias, heart failure, myocardial infarction, and stroke upon initiation of long-acting bronchodilators.33,34 There was no difference in risk for adverse cardiovascular events between LABA and LAMA in 1 Canadian study, and slightly more with LABA in a study using an American database.33,34 Wang et al reported that the risk of cardiovascular adverse effects, defined as hospitalizations and emergency room visits from heart failure, arrythmia, stroke, or ischemia, was 1.5 times the baseline risk in the first 30 days of starting a LABA or LAMA.35 The risk was subsequently the same as baseline or even lower after that period. Urinary retention is another possible complication of LAMA supported by evidence from meta-analyses and retrospective studies, but not clinical trials; the possibility of urinary retention should be discussed with patients upon initiation.36,37 Concerns about increased mortality with the soft mist formulation of tiotropium were put to rest by the Tiotropium Safety and Performance in Respimat (TIOSPIR) trial, which showed no increased mortality compared to Handihaler.38

        As far as efficacy and benefits, tiotropium and salmeterol were compared head-to-head in a clinical trial, and tiotropium increased the time before developing first exacerbation and decreased the overall rate of exacerbations.39 No difference in hospitalization rate or mortality was noted in 1 meta-analysis, although tiotropium was more effective in reducing exacerbations.40 The choice of agent should be made based on patient comorbidities and side effects. For example, an elderly patient with severe benign prostatic hyperplasia and urinary retention should try a LABA, while a LAMA would be a better first agent for a patient with severe tachycardia induced by albuterol.

         

         

        What is the role of inhaled corticosteroids in COPD?

        Inhaled corticosteroids (ICS) are believed to work in COPD by reducing airway inflammation.41 ICS should not be used alone for COPD management and are always combined with a LABA.7 Several ICS formulations are approved for use in COPD, including budesonide and fluticasone. ICS has been shown to decrease symptoms and exacerbations, with modest effect on lung function and no change in mortality.42 Side effects include oral candidiasis, dysphonia, and skin bruising.43 There is also an increased risk of pneumonia.44 ICS are best reserved for patients with a component of asthma or asthma–COPD overlap syndrome (ACOS).45 ACOS is characterized by persistent airflow limitation with several features usually associated with asthma and several features usually associated with COPD.46

        What if the patient is still symptomatic on a LABA or LAMA?

        For patients whose symptoms are not controlled on one class of LABA, recommendations are to add a bronchodilator from the other class.7 There are also multiple combined LAMA-LABA inhalers that are approved in the United States and can possibly improve adherence to therapy. These include tiotropium-olodaterol, umeclidinium-vilanterol, glycopyrronium-indacaterol, and glycopyrrolate-formoterol. In a large systematic review and meta-analysis comparing LABA-LAMA combination to either agent alone, there was a modest improvement in post-bronchodilator FEV1 and quality of life, with no change in hospital admissions, mortality, or adverse effects.47 Interestingly, adding tiotropium to LABA reduced exacerbations, although adding LABA to tiotropium did not.47

        Current guidelines recommend that patients in GOLD categories C and D who are not well controlled should receive a combination of LABA-ICS.7 However, a new randomized trial showed better reduction of exacerbations and decreased occurrence of pneumonia in patients receiving LAMA-LABA compared to LABA-ICS.48 In light of this new evidence, it is prudent to use a LAMA-LABA combination before adding ICS.

        Triple therapy with LAMA, LABA, and ICS is a common approach for patients with severe uncontrolled disease and has been shown to decrease exacerbations and improve quality of life.7,49 Adding tiotropium to LABA-ICS decreased exacerbations and improved quality of life and airflow in the landmark UPLIFT trial.27 In another clinical trial, triple therapy with LAMA, LABA, and ICS compared to tiotropium alone decreased severe exacerbations, pre-bronchodilator FEV1, and morning symptoms.50 A combination of triple therapy with fluticasone furoate, umeclidinium, and vilanterol was recently noted to result in a lower rate of moderate or severe COPD exacerbations, preserve lung function, and maintain health-related quality of life, as compared with fluticasone furoate/vilanterol or umeclidinium/vilanterol combination therapy among those with symptomatic COPD with a history of exacerbations.51

        Is there a role for theophylline? Other agents?

        Theophylline

        Theophylline is an oral adenosine diphosphate antagonist with indirect adrenergic activity, which is responsible for the bronchodilator therapeutic effect in patients with obstructive lung disease. It is also thought to work by an additional mechanism that decreases inflammation in the airways.52 Theophylline has a serious adverse-effect profile that includes ventricular arrhythmias, seizures, vomiting, and tremor.53 It is metabolized in the liver and has multiple drug interactions and a narrow therapeutic index. It has been shown to improve lung function, gas exchange and symptoms in meta-analysis and clinical trials.54,55

         

         

        In light of the nature of the adverse effects and the wide array of safer and more effective pharmacologic agents available, theophylline should be avoided early on in the treatment of COPD. Its use can be justified as an add-on therapy in patients with refractory disease on triple therapy for symptomatic relief.53 If used, the therapeutic range of theophylline for COPD is 8 to 12 mcg/mL peak level measured 3 to 7 hours after morning dose, and this level is usually achieved using a daily dose of 10 mg per kilogram of body weight for nonobese patients.56

        Systemic Steroids

        Oral steroids are used in COPD exacerbations but should never be used chronically in COPD patients, regardless of disease severity, as they increase morbidity and mortality without improving symptoms or lung function.57,58 The dose of systemic steroids should be tapered and finally discontinued.

        Mucolytics

        Classes of mucolytics include thiol derivatives, inhaled dornase alfa, hypertonic saline, and iodine preparations. Thiol derivatives such as N-acetylcysteine are the most widely studied.59 There is no consistent evidence of beneficial role of mucolytics in COPD patients.7,59 The PANTHEON trial showed decreased exacerbations with N-acetylcysteine (1.16 exacerbations per patient-year compared to 1.49 exacerbations per patient-year in the placebo group; risk ratio, 0.78; 95% CI, 0.67-0.90; P = 0.001) but had methodologic issues including high drop-out rate, exclusion of patients on oxygen, and a large of proportion of nonsmokers.60

        Long-Term Antibiotics

        There is no role for long-term antibiotics in the management of COPD.7 Macrolides are an exception but are used for their anti-inflammatory effects rather than their antibiotic effects. They should be reserved for patients with frequent exacerbations on optimal therapy and will be discussed later in the review.61

        What nonpharmacologic treatments are recommended for COPD patients?

        Smoking cessation, oxygen therapy for severe hypoxemia (resting O2 saturation ≤ 88% or PaO2 ≤ 55 mm Hg), vaccination for influenza and pneumococcus, and appropriate nutrition should be provided in all COPD patients. Pulmonary rehabilitation is indicated for patients in GOLD categories B, C, and D.7 It improves symptoms, quality of life, exercise tolerance, and health care utilization. Beneficial effects last for about 2 years.62,63

         

         

        What other diagnoses should be considered in patients who continue to be symptomatic on optimal therapy?

        Other diseases that share the same risk factors as COPD and can contribute to dyspnea, including coronary heart disease, heart failure, thromboembolic disease, and pulmonary hypertension, should be considered. In addition, all patients with refractory disease should have a careful assessment of their inhaler technique, continued smoking, need for oxygen therapy, and associated deconditioning.

        [embed:render:related:node:204720]

        [embed:render:related:node:204724]

        References

        1. Segreti A, Stirpe E, Rogliani P, Cazzola M. Defining phenotypes in COPD: an aid to personalized healthcare. Mol Diagn Ther. 2014;18:381-388.

        2. Han MK, Agusti A, Calverley PM, et al. Chronic obstructive pulmonary disease phenotypes: the future of COPD. Am J Respir Crit Care Med. 2010;182:598-604.

        3. Aubier M, Marthan R, Berger P, et al. [COPD and inflammation: statement from a French expert group: inflammation and remodelling mechanisms]. Rev Mal Respir. 2010;27:1254-1266.

        4. Wang ZL. Evolving role of systemic inflammation in comorbidities of chronic obstructive pulmonary disease. Chin Med J (Engl). 2010;123:3467-3478.

        5. Buist AS, McBurnie MA, Vollmer WM, et al. International variation in the prevalence of COPD (the BOLD Study): a population-based prevalence study. Lancet. 2007;370:741-750.

        6. Miniño AM, Murphy SL, Xu J, Kochanek KD. Deaths: final data for 2008. Natl Vital Stat Rep. 2011;59:1-126.

        7. Global Initiative for Chronic Obstructive Lung Disease (GOLD): Global strategy for the diagnosis, management, and prevention of COPD 2017. www.goldcopd.org. Accessed July 9, 2019.

        8. Jones PW, Harding G, Berry P, et al. Development and first validation of the COPD Assessment Test. Eur Respir J. 2009;34:648-654.

        9. Wadbo M, Löfdahl CG, Larsson K, et al. Effects of formoterol and ipratropium bromide in COPD: a 3-month placebo-controlled study. Eur Respir J. 2002;20:1138-1146.

        10. Ram FS, Sestini P. Regular inhaled short acting beta2 agonists for the management of stable chronic obstructive pulmonary disease: Cochrane systematic review and meta-analysis. Thorax. 2003;58:580-584.

        11. Colice GL. Nebulized bronchodilators for outpatient management of stable chronic obstructive pulmonary disease. Am J Med. 1996;100(1A):11S-8S.

        12. In chronic obstructive pulmonary disease, a combination of ipratropium and albuterol is more effective than either agent alone. An 85-day multicenter trial. COMBIVENT Inhalation Aerosol Study Group. Chest. 1994;105:1411-1419.

        13. Friedman M, Serby CW, Menjoge SS, et al. Pharmacoeconomic evaluation of a combination of ipratropium plus albuterol compared with ipratropium alone and albuterol alone in COPD. Chest. 1999;115:635-641.

        14. Cook D, Guyatt G, Wong E, et al. Regular versus as-needed short-acting inhaled beta-agonist therapy for chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;163:85-90.

        15. Wilchesky M, Ernst P, Brophy JM, et al. Bronchodilator use and the risk of arrhythmia in COPD: part 2: reassessment in the larger Quebec cohort. Chest. 2012;142:305-311.

        16. Scott VL, Frazee LA. Retrospective comparison of nebulized levalbuterol and albuterol for adverse events in patients with acute airflow obstruction. Am J Ther. 2003;10:341-347.

        17. Wong CS, Pavord ID, Williams J, et al. Bronchodilator, cardiovascular, and hypokalaemic effects of fenoterol, salbutamol, and terbutaline in asthma. Lancet. 1990;336:1396-1399.

        18. Cole JM, Sheehan AH, Jordan JK. Concomitant use of ipratropium and tiotropium in chronic obstructive pulmonary disease. Ann Pharmacother. 2012;46:1717-1721.

        19. Qaseem A, Wilt TJ, Weinberger SE, et al. Diagnosis and management of stable chronic obstructive pulmonary disease: a clinical practice guideline update from the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society. Ann Intern Med. 2011;155:179-191.

        20. Pearlman DS, Chervinsky P, LaForce C, et al. A comparison of salmeterol with albuterol in the treatment of mild-to-moderate asthma. N Engl J Med. 1992;327:1420-1425.

        21. Takahashi T, Belvisi MG, Patel H, et al. Effect of Ba 679 BR, a novel long-acting anticholinergic agent, on cholinergic neurotransmission in guinea pig and human airways. Am J Respir Crit Care Med. 1994;150(6 Pt 1):1640-1645.

        22. Ferguson GT, Feldman G, Pudi KK, et al. improvements in lung function with nebulized revefenacin in the treatment of patients with moderate to very severe COPD: results from two replicate phase III clinical trials. Chronic Obstr Pulm Dis. 2019;6:154-165.

        23. Donohue JF, Fogarty C, Lötvall J, et al. Once-daily bronchodilators for chronic obstructive pulmonary disease: indacaterol versus tiotropium. Am J Respir Crit Care Med. 2010;182:155-162.

        24. Koch A, Pizzichini E, Hamilton A, et al. Lung function efficacy and symptomatic benefit of olodaterol once daily delivered via Respimat versus placebo and formoterol twice daily in patients with GOLD 2-4 COPD: results from two replicate 48-week studies. Int J Chron Obstruct Pulmon Dis. 2014;9:697-714.

        25. Calverley PM, Anderson JA, Celli B, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356:775-789.

        26. Hanania NA, Feldman G, Zachgo W, et al. The efficacy and safety of the novel long-acting β2 agonist vilanterol in patients with COPD: a randomized placebo-controlled trial. Chest. 2012;142:119-127.

        27. Tashkin DP, Celli B, Senn S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543-1554.

        28. Decramer ML, Chapman KR, Dahl R, et al. Once-daily indacaterol versus tiotropium for patients with severe chronic obstructive pulmonary disease (INVIGORATE): a randomised, blinded, parallel-group study. Lancet Respir Med. 2013;1:524-533.

        29. Jones PW, Singh D, Bateman ED, et al. Efficacy and safety of twice-daily aclidinium bromide in COPD patients: the ATTAIN study. Eur Respir J. 2012;40:830-836.

        30. D’Urzo A, Ferguson GT, van Noord JA, et al. Efficacy and safety of once-daily NVA237 in patients with moderate-to-severe COPD: the GLOW1 trial. Respir Res. 2011;12:156.

        31. Antoniu SA. UPLIFT Study: the effects of long-term therapy with inhaled tiotropium in chronic obstructive pulmonary disease. Evaluation of: Tashkin DP, Celli B, Senn S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543-1554. Expert Opin Pharmacother. 2009;10:719–22.

        32. Nelson HS, Gross NJ, Levine B, et al. Cardiac safety profile of nebulized formoterol in adults with COPD: a 12-week, multicenter, randomized, double- blind, double-dummy, placebo- and active-controlled trial. Clin Ther. 2007;29:2167-2178.

        33. Gershon A, Croxford R, Calzavara A, et al. Cardiovascular safety of inhaled long-acting bronchodilators in individuals with chronic obstructive pulmonary disease. JAMA Intern Med. 2013;173:1175-1185.

        34. Aljaafareh A, Valle JR, Lin YL, et al. Risk of cardiovascular events after initiation of long-acting bronchodilators in patients with chronic obstructive lung disease: A population-based study. SAGE Open Med. 2016;4:2050312116671337.

        35. Wang MT, Liou JT, Lin CW, et al. Association of cardiovascular risk with inhaled long-acting bronchodilators in patients with chronic obstructive pulmonary disease: a nested case-Control Study. JAMA Intern Med. 2018;178:229-238.

        36. O’Connor AB. Tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2009;360:185-186.

        37. Kesten S, Jara M, Wentworth C, Lanes S. Pooled clinical trial analysis of tiotropium safety. Chest. 2006;130:1695-1703.

        38. Wise RA, Anzueto A, Cotton D, et al. Tiotropium Respimat inhaler and the risk of death in COPD. N Engl J Med. 2013;369:1491-1501.

        39. Vogelmeier C, Hederer B, Glaab T, et al. Tiotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med. 2011;364:1093-1103.

        40. Chong J, Karner C, Poole P. Tiotropium versus long-acting beta-agonists for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012(9):CD009157.

        41. Gan WQ, Man SF, Sin DD. Effects of inhaled corticosteroids on sputum cell counts in stable chronic obstructive pulmonary disease: a systematic review and a meta-analysis. BMC Pulm Med. 2005;5:3.

        42. Yang IA, Clarke MS, Sim EH, Fong KM. Inhaled corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012(7):CD002991.

        43. Roland NJ, Bhalla RK, Earis J. The local side effects of inhaled corticosteroids: current understanding and review of the literature. Chest. 2004;126:213-219.

        44. Drummond MB, Dasenbrook EC, Pitz MW, et al. Inhaled corticosteroids in patients with stable chronic obstructive pulmonary disease: a systematic review and meta-analysis. JAMA. 2008;300:2407-2416.

        45. Lee SY, Park HY, Kim EK, et al. Combination therapy of inhaled steroids and long-acting beta2-agonists in asthma-COPD overlap syndrome. Int J Chron Obstruct Pulmon Dis. 2016;11:2797-2803.

        46. Postma DS, Rabe KF. The asthma-COPD overlap syndrome. N Engl J Med. 2015;373:1241-1249.

        47. Farne HA, Cates CJ. Long-acting beta2-agonist in addition to tiotropium versus either tiotropium or long-acting beta2-agonist alone for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015:CD008989.

        48. Wedzicha JA, Banerji D, Chapman KR, et al. Indacaterol-glycopyrronium versus salmeterol-fluticasone for COPD. N Engl J Med. 2016;374:2222-2234.

        49. Aaron SD, Vandemheen KL, Fergusson D, et al. Tiotropium in combination with placebo, salmeterol, or fluticasone-salmeterol for treatment of chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med. 2007;146:545-555.

        50. Welte T, Miravitlles M, Hernandez P, et al. Efficacy and tolerability of budesonide/formoterol added to tiotropium in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:741-750.

        51. Lipson DA, Barnhart, Brealey N, et al; IMPACT Investigators. Once-daily single-inhaler triple versus dual therapy in patients with COPD. N Engl J Med. 2018;378:1671-1680.

        52. Gallelli L, Falcone D, Cannataro R, et al. Theophylline action on primary human bronchial epithelial cells under proinflammatory stimuli and steroidal drugs: a therapeutic rationale approach. Drug Des Devel Ther. 2017;11:265-272.

        53. Paloucek FP, Rodvold KA. Evaluation of theophylline overdoses and toxicities. Ann Emerg Med. 1988;17:135-144.

        54. Ram FS, Jones PW, Castro AA, et al. Oral theophylline for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2002(4):CD003902.

        55. Murciano D, Auclair MH, Pariente R, Aubier M. A randomized, controlled trial of theophylline in patients with severe chronic obstructive pulmonary disease. N Engl J Med. 1989;320:1521-1525.

        56. Devereux G, Cotton S, Barnes P, et al. Use of low-dose oral theophylline as an adjunct to inhaled corticosteroids in preventing exacerbations of chronic obstructive pulmonary disease: study protocol for a randomised controlled trial. Trials. 2015;16:267.

        57. Walters JA, Walters EH, Wood-Baker R. Oral corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005(3):CD005374.

        58. Horita N, Miyazawa N, Morita S, et al. Evidence suggesting that oral corticosteroids increase mortality in stable chronic obstructive pulmonary disease. Respir Res. 2014;15:37.

        59. Poole P, Chong J, Cates CJ. Mucolytic agents versus placebo for chronic bronchitis or chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015(7):CD001287.

        60. Zheng JP, Wen FQ, Bai CX, et al. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): a randomised, double-blind placebo-controlled trial. Lancet Respir Med. 2014;2:187-194.

        61. Seemungal TA, Wilkinson TM, Hurst JR, et al. Long-term erythromycin therapy is associated with decreased chronic obstructive pulmonary disease exacerbations. Am J Respir Crit Care Med. 2008;178:1139-1147.

        62. Ries AL, Kaplan RM, Limberg TM, Prewitt LM. Effects of pulmonary rehabilitation on physiologic and psychosocial outcomes in patients with chronic obstructive pulmonary disease. Ann Intern Med. 1995;122:823-832.

        63. Güell R, Casan P, Belda J, et al. Long-term effects of outpatient rehabilitation of COPD: a randomized trial. Chest. 2000;117:976-983.

        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Publications
        Topics
        Sections
        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Chronic obstructive pulmonary disease (COPD) is a systemic inflammatory disease characterized by irreversible obstructive ventilatory defects.1-4 It is a major cause of morbidity and mortality, affecting 5% of the population in the United States and ranking as the third leading cause of death in 2008.5,6 The goals in COPD management are to provide symptom relief, improve the quality of life, preserve lung function, and reduce the frequency of exacerbations and mortality. In this 3-part review, we discuss the management of stable COPD in the context of 3 common clinical scenarios: initiating and optimizing therapy, managing acute exacerbations, and managing advanced disease.

        Case Presentation

        A 65-year-old man with COPD underwent pulmonary function testing (PFT), which demonstrated an obstructive ventilatory defect: forced expiratory volume in 1 second/forced vital capacity ratio (FEV1/FVC), 0.45; FEV1, 2 L (65% of predicted); and diffusing capacity of the lung for carbon monoxide, 15 mL/min/mm Hg (65% of predicted). He has dyspnea with strenuous exercise but is comfortable at rest and with minimal exercise. He has had 1 exacerbation in the past year, and this was treated on an outpatient basis with steroids and antibiotics. His medication regimen includes inhaled tiotropium once daily and inhaled albuterol as needed that he uses roughly twice a week.

        What determines the appropriate therapy for a given COPD patient?

        COPD management is guided by disease severity that is measured using a multimodal staging system developed by the Global Initiative for Chronic Obstructive Lung Disease (GOLD). The initial classification adopted by the GOLD 2011 report encompassed 4 categories based on symptoms, number of exacerbations, and degree of airflow limitation on PFT. However, in 2017 the GOLD ABCD classification was modified to consider only symptoms and risk of exacerbation in classifying patients, regardless of performance on spirometry and FEV1 (Figure 1).7,8 This approach was intended to make therapy more individualized based on the patient clinical profile. The Table provides a summary of the recommended treatments according to classification based on the GOLD 2017 report.

        pdbr01607001_f1.png

        The patient in our clinical scenario can be classified as GOLD category B.

        pdbr01607001_t1.png

        What is the approach to building a pharmacologic regimen for the patient with COPD?

        The backbone of the pharmacologic regimen for COPD includes short- and long-acting bronchodilators. They are usually given in an inhaled form to maximize local effects on the lungs and minimize systemic side effects. There are 2 main classes of bronchodilators, beta-agonists and muscarinic antagonists, and each targets specific receptors on the surface of airway smooth muscle cells. Beta- agonists work by stimulating beta-2 receptors, resulting in bronchodilation, while muscarinic antagonists work by blocking the bronchoconstrictor action of M3 muscarinic receptors. Inhaled corticosteroids can be added to long-acting bronchodilator therapy but cannot be used as stand-alone therapy. Theophylline is an oral bronchodilator that is used infrequently due to its narrow therapeutic index, toxicity, and multiple drug interactions.

        Figure 2 presents an approach to building a treatment plan for the patient with stable COPD.

        pdbr01607001_f2.png

        Who should be on short-acting bronchodilators? What is the best agent? Should it be scheduled or used as needed?

        All patients with COPD should be an on inhaled short-acting bronchodilator as needed for relief of symptoms.7 Both short-acting beta-agonists (albuterol and levalbuterol) and short-acting muscarinic antagonists (ipratropium) have been shown in clinical trials and meta-analyses to improve symptoms and lung function in patients with stable COPD9,10 and seem to have comparative efficacy when compared head-to-head in trials.11 However, the airway bronchodilator effect achieved by both classes seems to be additive when used in combination and is also associated with fewer exacerbations compared to albuterol alone.12 On the other hand, adding albuterol to ipratropium increased the bronchodilator response but did not reduce the exacerbation rate.11-13 Inhaled short-acting beta-agonists when used as needed rather than scheduled are associated with less medication use without any significant difference in symptoms or lung function.14

        The side effects related to using recommended doses of a short-acting bronchodilator are minimal. In retrospective studies, short-acting beta-agonists increased the risk of severe cardiac arrhythmias.15 Levalbuterol, the active enantiomer of albuterol (R-albuterol) developed for the theoretical benefits of reduced tachycardia, increased tolerability, and better or equal efficacy compared to racemic albuterol, failed to show a clinically significant difference in inducing tachycardia.16 Beta-agonist overuse is associated with tremor and in severe cases hypokalemia, which happens mainly when patients try to achieve maximal bronchodilation; the clinically used doses of beta agonists are associated with fewer side effects but achieve less than maximal bronchodilation.17 Ipratropium can produce systemic anticholinergic side effects, urinary retention being the most clinically significant, especially when combined with long-acting anticholinergic agents.18

         

         

        In light of the above discussion, a combination of a short-acting beta-agonist and a muscarinic antagonist is recommended in all patients with COPD, unless the patient is on a long-acting muscarinic antagonist (LAMA).7,18 In the latter case, a short-acting beta agonist used as a rescue inhaler is the best option. In our patient, albuterol was the choice for his short-acting bronchodilator, as he was using the LAMA tiotropium.

        Are short-acting bronchodilators enough? What do we use for maintenance therapy?

        All patients with COPD who are category B or higher according to the modified GOLD staging system should be on a long-acting bronchodilator:7,19 either a long-acting beta-agonist (LABA) or a LAMA. Long-acting bronchodilators work on the same receptors as their short-acting counterparts but have structural differences. Salmeterol is the prototype long-acting selective beta-2 agonist. It is structurally similar to albuterol but has an elongated side chain that allows it to bind firmly to the area of beta receptors and stimulate them repetitively, resulting in an extended-duration of action.20 Tiotropium on the other hand is a quaternary ammonium of ipratropium that is a nonselective muscarinic antagonist.21 Compared to ipratropium, tiotropium dissociates more quickly from M2 receptors, which is responsible for the undesired anticholinergic effects, while at the same time it binds M1 and M3 receptors for a prolonged time, resulting in an extended duration of action.21 Revefenacin is a new lung-selective LAMA that is under development and has shown promise among those with moderate to very severe COPD. Results are only limited to phase 3 trials, and clinical studies are still underway.22

        The currently available LABAs include salmeterol, formoterol, arformoterol, olodaterol, and indacaterol. The last 2 have the advantage of once-daily dosing rather than twice daily.23,24 LABAs have been shown to improve lung function, exacerbation rate, and quality of life in multiple clinical trials.23,25 Vilanterol is another LABA that has a long duration of action and can be used once daily,26 but is only available in a combination with umeclidinium, a LAMA. Several LAMAs are approved for use in COPD, including the prototype tiotropium, in addition to aclidinium, umeclidinium, and glycopyrronium. These have been shown in clinical trials to improve lung function, symptoms, and exacerbation rate.27-30

        Patients can be started on either a LAMA or LABA depending on the individual patient's needs and the agent's adverse effects.7 Both have comparable adverse effects and efficacy, as detailed below. Concerning adverse effects, there is conflicting data concerning an association of cardiovascular events with both classes of long-acting bronchodilators. While clinical trials failed to show an increased risk,25,31,32 several retrospective studies showed an increased risk of emergency room visits and hospitalizations due to tachyarrhythmias, heart failure, myocardial infarction, and stroke upon initiation of long-acting bronchodilators.33,34 There was no difference in risk for adverse cardiovascular events between LABA and LAMA in 1 Canadian study, and slightly more with LABA in a study using an American database.33,34 Wang et al reported that the risk of cardiovascular adverse effects, defined as hospitalizations and emergency room visits from heart failure, arrythmia, stroke, or ischemia, was 1.5 times the baseline risk in the first 30 days of starting a LABA or LAMA.35 The risk was subsequently the same as baseline or even lower after that period. Urinary retention is another possible complication of LAMA supported by evidence from meta-analyses and retrospective studies, but not clinical trials; the possibility of urinary retention should be discussed with patients upon initiation.36,37 Concerns about increased mortality with the soft mist formulation of tiotropium were put to rest by the Tiotropium Safety and Performance in Respimat (TIOSPIR) trial, which showed no increased mortality compared to Handihaler.38

        As far as efficacy and benefits, tiotropium and salmeterol were compared head-to-head in a clinical trial, and tiotropium increased the time before developing first exacerbation and decreased the overall rate of exacerbations.39 No difference in hospitalization rate or mortality was noted in 1 meta-analysis, although tiotropium was more effective in reducing exacerbations.40 The choice of agent should be made based on patient comorbidities and side effects. For example, an elderly patient with severe benign prostatic hyperplasia and urinary retention should try a LABA, while a LAMA would be a better first agent for a patient with severe tachycardia induced by albuterol.

         

         

        What is the role of inhaled corticosteroids in COPD?

        Inhaled corticosteroids (ICS) are believed to work in COPD by reducing airway inflammation.41 ICS should not be used alone for COPD management and are always combined with a LABA.7 Several ICS formulations are approved for use in COPD, including budesonide and fluticasone. ICS has been shown to decrease symptoms and exacerbations, with modest effect on lung function and no change in mortality.42 Side effects include oral candidiasis, dysphonia, and skin bruising.43 There is also an increased risk of pneumonia.44 ICS are best reserved for patients with a component of asthma or asthma–COPD overlap syndrome (ACOS).45 ACOS is characterized by persistent airflow limitation with several features usually associated with asthma and several features usually associated with COPD.46

        What if the patient is still symptomatic on a LABA or LAMA?

        For patients whose symptoms are not controlled on one class of LABA, recommendations are to add a bronchodilator from the other class.7 There are also multiple combined LAMA-LABA inhalers that are approved in the United States and can possibly improve adherence to therapy. These include tiotropium-olodaterol, umeclidinium-vilanterol, glycopyrronium-indacaterol, and glycopyrrolate-formoterol. In a large systematic review and meta-analysis comparing LABA-LAMA combination to either agent alone, there was a modest improvement in post-bronchodilator FEV1 and quality of life, with no change in hospital admissions, mortality, or adverse effects.47 Interestingly, adding tiotropium to LABA reduced exacerbations, although adding LABA to tiotropium did not.47

        Current guidelines recommend that patients in GOLD categories C and D who are not well controlled should receive a combination of LABA-ICS.7 However, a new randomized trial showed better reduction of exacerbations and decreased occurrence of pneumonia in patients receiving LAMA-LABA compared to LABA-ICS.48 In light of this new evidence, it is prudent to use a LAMA-LABA combination before adding ICS.

        Triple therapy with LAMA, LABA, and ICS is a common approach for patients with severe uncontrolled disease and has been shown to decrease exacerbations and improve quality of life.7,49 Adding tiotropium to LABA-ICS decreased exacerbations and improved quality of life and airflow in the landmark UPLIFT trial.27 In another clinical trial, triple therapy with LAMA, LABA, and ICS compared to tiotropium alone decreased severe exacerbations, pre-bronchodilator FEV1, and morning symptoms.50 A combination of triple therapy with fluticasone furoate, umeclidinium, and vilanterol was recently noted to result in a lower rate of moderate or severe COPD exacerbations, preserve lung function, and maintain health-related quality of life, as compared with fluticasone furoate/vilanterol or umeclidinium/vilanterol combination therapy among those with symptomatic COPD with a history of exacerbations.51

        Is there a role for theophylline? Other agents?

        Theophylline

        Theophylline is an oral adenosine diphosphate antagonist with indirect adrenergic activity, which is responsible for the bronchodilator therapeutic effect in patients with obstructive lung disease. It is also thought to work by an additional mechanism that decreases inflammation in the airways.52 Theophylline has a serious adverse-effect profile that includes ventricular arrhythmias, seizures, vomiting, and tremor.53 It is metabolized in the liver and has multiple drug interactions and a narrow therapeutic index. It has been shown to improve lung function, gas exchange and symptoms in meta-analysis and clinical trials.54,55

         

         

        In light of the nature of the adverse effects and the wide array of safer and more effective pharmacologic agents available, theophylline should be avoided early on in the treatment of COPD. Its use can be justified as an add-on therapy in patients with refractory disease on triple therapy for symptomatic relief.53 If used, the therapeutic range of theophylline for COPD is 8 to 12 mcg/mL peak level measured 3 to 7 hours after morning dose, and this level is usually achieved using a daily dose of 10 mg per kilogram of body weight for nonobese patients.56

        Systemic Steroids

        Oral steroids are used in COPD exacerbations but should never be used chronically in COPD patients, regardless of disease severity, as they increase morbidity and mortality without improving symptoms or lung function.57,58 The dose of systemic steroids should be tapered and finally discontinued.

        Mucolytics

        Classes of mucolytics include thiol derivatives, inhaled dornase alfa, hypertonic saline, and iodine preparations. Thiol derivatives such as N-acetylcysteine are the most widely studied.59 There is no consistent evidence of beneficial role of mucolytics in COPD patients.7,59 The PANTHEON trial showed decreased exacerbations with N-acetylcysteine (1.16 exacerbations per patient-year compared to 1.49 exacerbations per patient-year in the placebo group; risk ratio, 0.78; 95% CI, 0.67-0.90; P = 0.001) but had methodologic issues including high drop-out rate, exclusion of patients on oxygen, and a large of proportion of nonsmokers.60

        Long-Term Antibiotics

        There is no role for long-term antibiotics in the management of COPD.7 Macrolides are an exception but are used for their anti-inflammatory effects rather than their antibiotic effects. They should be reserved for patients with frequent exacerbations on optimal therapy and will be discussed later in the review.61

        What nonpharmacologic treatments are recommended for COPD patients?

        Smoking cessation, oxygen therapy for severe hypoxemia (resting O2 saturation ≤ 88% or PaO2 ≤ 55 mm Hg), vaccination for influenza and pneumococcus, and appropriate nutrition should be provided in all COPD patients. Pulmonary rehabilitation is indicated for patients in GOLD categories B, C, and D.7 It improves symptoms, quality of life, exercise tolerance, and health care utilization. Beneficial effects last for about 2 years.62,63

         

         

        What other diagnoses should be considered in patients who continue to be symptomatic on optimal therapy?

        Other diseases that share the same risk factors as COPD and can contribute to dyspnea, including coronary heart disease, heart failure, thromboembolic disease, and pulmonary hypertension, should be considered. In addition, all patients with refractory disease should have a careful assessment of their inhaler technique, continued smoking, need for oxygen therapy, and associated deconditioning.

        [embed:render:related:node:204720]

        [embed:render:related:node:204724]

        Chronic obstructive pulmonary disease (COPD) is a systemic inflammatory disease characterized by irreversible obstructive ventilatory defects.1-4 It is a major cause of morbidity and mortality, affecting 5% of the population in the United States and ranking as the third leading cause of death in 2008.5,6 The goals in COPD management are to provide symptom relief, improve the quality of life, preserve lung function, and reduce the frequency of exacerbations and mortality. In this 3-part review, we discuss the management of stable COPD in the context of 3 common clinical scenarios: initiating and optimizing therapy, managing acute exacerbations, and managing advanced disease.

        Case Presentation

        A 65-year-old man with COPD underwent pulmonary function testing (PFT), which demonstrated an obstructive ventilatory defect: forced expiratory volume in 1 second/forced vital capacity ratio (FEV1/FVC), 0.45; FEV1, 2 L (65% of predicted); and diffusing capacity of the lung for carbon monoxide, 15 mL/min/mm Hg (65% of predicted). He has dyspnea with strenuous exercise but is comfortable at rest and with minimal exercise. He has had 1 exacerbation in the past year, and this was treated on an outpatient basis with steroids and antibiotics. His medication regimen includes inhaled tiotropium once daily and inhaled albuterol as needed that he uses roughly twice a week.

        What determines the appropriate therapy for a given COPD patient?

        COPD management is guided by disease severity that is measured using a multimodal staging system developed by the Global Initiative for Chronic Obstructive Lung Disease (GOLD). The initial classification adopted by the GOLD 2011 report encompassed 4 categories based on symptoms, number of exacerbations, and degree of airflow limitation on PFT. However, in 2017 the GOLD ABCD classification was modified to consider only symptoms and risk of exacerbation in classifying patients, regardless of performance on spirometry and FEV1 (Figure 1).7,8 This approach was intended to make therapy more individualized based on the patient clinical profile. The Table provides a summary of the recommended treatments according to classification based on the GOLD 2017 report.

        pdbr01607001_f1.png

        The patient in our clinical scenario can be classified as GOLD category B.

        pdbr01607001_t1.png

        What is the approach to building a pharmacologic regimen for the patient with COPD?

        The backbone of the pharmacologic regimen for COPD includes short- and long-acting bronchodilators. They are usually given in an inhaled form to maximize local effects on the lungs and minimize systemic side effects. There are 2 main classes of bronchodilators, beta-agonists and muscarinic antagonists, and each targets specific receptors on the surface of airway smooth muscle cells. Beta- agonists work by stimulating beta-2 receptors, resulting in bronchodilation, while muscarinic antagonists work by blocking the bronchoconstrictor action of M3 muscarinic receptors. Inhaled corticosteroids can be added to long-acting bronchodilator therapy but cannot be used as stand-alone therapy. Theophylline is an oral bronchodilator that is used infrequently due to its narrow therapeutic index, toxicity, and multiple drug interactions.

        Figure 2 presents an approach to building a treatment plan for the patient with stable COPD.

        pdbr01607001_f2.png

        Who should be on short-acting bronchodilators? What is the best agent? Should it be scheduled or used as needed?

        All patients with COPD should be an on inhaled short-acting bronchodilator as needed for relief of symptoms.7 Both short-acting beta-agonists (albuterol and levalbuterol) and short-acting muscarinic antagonists (ipratropium) have been shown in clinical trials and meta-analyses to improve symptoms and lung function in patients with stable COPD9,10 and seem to have comparative efficacy when compared head-to-head in trials.11 However, the airway bronchodilator effect achieved by both classes seems to be additive when used in combination and is also associated with fewer exacerbations compared to albuterol alone.12 On the other hand, adding albuterol to ipratropium increased the bronchodilator response but did not reduce the exacerbation rate.11-13 Inhaled short-acting beta-agonists when used as needed rather than scheduled are associated with less medication use without any significant difference in symptoms or lung function.14

        The side effects related to using recommended doses of a short-acting bronchodilator are minimal. In retrospective studies, short-acting beta-agonists increased the risk of severe cardiac arrhythmias.15 Levalbuterol, the active enantiomer of albuterol (R-albuterol) developed for the theoretical benefits of reduced tachycardia, increased tolerability, and better or equal efficacy compared to racemic albuterol, failed to show a clinically significant difference in inducing tachycardia.16 Beta-agonist overuse is associated with tremor and in severe cases hypokalemia, which happens mainly when patients try to achieve maximal bronchodilation; the clinically used doses of beta agonists are associated with fewer side effects but achieve less than maximal bronchodilation.17 Ipratropium can produce systemic anticholinergic side effects, urinary retention being the most clinically significant, especially when combined with long-acting anticholinergic agents.18

         

         

        In light of the above discussion, a combination of a short-acting beta-agonist and a muscarinic antagonist is recommended in all patients with COPD, unless the patient is on a long-acting muscarinic antagonist (LAMA).7,18 In the latter case, a short-acting beta agonist used as a rescue inhaler is the best option. In our patient, albuterol was the choice for his short-acting bronchodilator, as he was using the LAMA tiotropium.

        Are short-acting bronchodilators enough? What do we use for maintenance therapy?

        All patients with COPD who are category B or higher according to the modified GOLD staging system should be on a long-acting bronchodilator:7,19 either a long-acting beta-agonist (LABA) or a LAMA. Long-acting bronchodilators work on the same receptors as their short-acting counterparts but have structural differences. Salmeterol is the prototype long-acting selective beta-2 agonist. It is structurally similar to albuterol but has an elongated side chain that allows it to bind firmly to the area of beta receptors and stimulate them repetitively, resulting in an extended-duration of action.20 Tiotropium on the other hand is a quaternary ammonium of ipratropium that is a nonselective muscarinic antagonist.21 Compared to ipratropium, tiotropium dissociates more quickly from M2 receptors, which is responsible for the undesired anticholinergic effects, while at the same time it binds M1 and M3 receptors for a prolonged time, resulting in an extended duration of action.21 Revefenacin is a new lung-selective LAMA that is under development and has shown promise among those with moderate to very severe COPD. Results are only limited to phase 3 trials, and clinical studies are still underway.22

        The currently available LABAs include salmeterol, formoterol, arformoterol, olodaterol, and indacaterol. The last 2 have the advantage of once-daily dosing rather than twice daily.23,24 LABAs have been shown to improve lung function, exacerbation rate, and quality of life in multiple clinical trials.23,25 Vilanterol is another LABA that has a long duration of action and can be used once daily,26 but is only available in a combination with umeclidinium, a LAMA. Several LAMAs are approved for use in COPD, including the prototype tiotropium, in addition to aclidinium, umeclidinium, and glycopyrronium. These have been shown in clinical trials to improve lung function, symptoms, and exacerbation rate.27-30

        Patients can be started on either a LAMA or LABA depending on the individual patient's needs and the agent's adverse effects.7 Both have comparable adverse effects and efficacy, as detailed below. Concerning adverse effects, there is conflicting data concerning an association of cardiovascular events with both classes of long-acting bronchodilators. While clinical trials failed to show an increased risk,25,31,32 several retrospective studies showed an increased risk of emergency room visits and hospitalizations due to tachyarrhythmias, heart failure, myocardial infarction, and stroke upon initiation of long-acting bronchodilators.33,34 There was no difference in risk for adverse cardiovascular events between LABA and LAMA in 1 Canadian study, and slightly more with LABA in a study using an American database.33,34 Wang et al reported that the risk of cardiovascular adverse effects, defined as hospitalizations and emergency room visits from heart failure, arrythmia, stroke, or ischemia, was 1.5 times the baseline risk in the first 30 days of starting a LABA or LAMA.35 The risk was subsequently the same as baseline or even lower after that period. Urinary retention is another possible complication of LAMA supported by evidence from meta-analyses and retrospective studies, but not clinical trials; the possibility of urinary retention should be discussed with patients upon initiation.36,37 Concerns about increased mortality with the soft mist formulation of tiotropium were put to rest by the Tiotropium Safety and Performance in Respimat (TIOSPIR) trial, which showed no increased mortality compared to Handihaler.38

        As far as efficacy and benefits, tiotropium and salmeterol were compared head-to-head in a clinical trial, and tiotropium increased the time before developing first exacerbation and decreased the overall rate of exacerbations.39 No difference in hospitalization rate or mortality was noted in 1 meta-analysis, although tiotropium was more effective in reducing exacerbations.40 The choice of agent should be made based on patient comorbidities and side effects. For example, an elderly patient with severe benign prostatic hyperplasia and urinary retention should try a LABA, while a LAMA would be a better first agent for a patient with severe tachycardia induced by albuterol.

         

         

        What is the role of inhaled corticosteroids in COPD?

        Inhaled corticosteroids (ICS) are believed to work in COPD by reducing airway inflammation.41 ICS should not be used alone for COPD management and are always combined with a LABA.7 Several ICS formulations are approved for use in COPD, including budesonide and fluticasone. ICS has been shown to decrease symptoms and exacerbations, with modest effect on lung function and no change in mortality.42 Side effects include oral candidiasis, dysphonia, and skin bruising.43 There is also an increased risk of pneumonia.44 ICS are best reserved for patients with a component of asthma or asthma–COPD overlap syndrome (ACOS).45 ACOS is characterized by persistent airflow limitation with several features usually associated with asthma and several features usually associated with COPD.46

        What if the patient is still symptomatic on a LABA or LAMA?

        For patients whose symptoms are not controlled on one class of LABA, recommendations are to add a bronchodilator from the other class.7 There are also multiple combined LAMA-LABA inhalers that are approved in the United States and can possibly improve adherence to therapy. These include tiotropium-olodaterol, umeclidinium-vilanterol, glycopyrronium-indacaterol, and glycopyrrolate-formoterol. In a large systematic review and meta-analysis comparing LABA-LAMA combination to either agent alone, there was a modest improvement in post-bronchodilator FEV1 and quality of life, with no change in hospital admissions, mortality, or adverse effects.47 Interestingly, adding tiotropium to LABA reduced exacerbations, although adding LABA to tiotropium did not.47

        Current guidelines recommend that patients in GOLD categories C and D who are not well controlled should receive a combination of LABA-ICS.7 However, a new randomized trial showed better reduction of exacerbations and decreased occurrence of pneumonia in patients receiving LAMA-LABA compared to LABA-ICS.48 In light of this new evidence, it is prudent to use a LAMA-LABA combination before adding ICS.

        Triple therapy with LAMA, LABA, and ICS is a common approach for patients with severe uncontrolled disease and has been shown to decrease exacerbations and improve quality of life.7,49 Adding tiotropium to LABA-ICS decreased exacerbations and improved quality of life and airflow in the landmark UPLIFT trial.27 In another clinical trial, triple therapy with LAMA, LABA, and ICS compared to tiotropium alone decreased severe exacerbations, pre-bronchodilator FEV1, and morning symptoms.50 A combination of triple therapy with fluticasone furoate, umeclidinium, and vilanterol was recently noted to result in a lower rate of moderate or severe COPD exacerbations, preserve lung function, and maintain health-related quality of life, as compared with fluticasone furoate/vilanterol or umeclidinium/vilanterol combination therapy among those with symptomatic COPD with a history of exacerbations.51

        Is there a role for theophylline? Other agents?

        Theophylline

        Theophylline is an oral adenosine diphosphate antagonist with indirect adrenergic activity, which is responsible for the bronchodilator therapeutic effect in patients with obstructive lung disease. It is also thought to work by an additional mechanism that decreases inflammation in the airways.52 Theophylline has a serious adverse-effect profile that includes ventricular arrhythmias, seizures, vomiting, and tremor.53 It is metabolized in the liver and has multiple drug interactions and a narrow therapeutic index. It has been shown to improve lung function, gas exchange and symptoms in meta-analysis and clinical trials.54,55

         

         

        In light of the nature of the adverse effects and the wide array of safer and more effective pharmacologic agents available, theophylline should be avoided early on in the treatment of COPD. Its use can be justified as an add-on therapy in patients with refractory disease on triple therapy for symptomatic relief.53 If used, the therapeutic range of theophylline for COPD is 8 to 12 mcg/mL peak level measured 3 to 7 hours after morning dose, and this level is usually achieved using a daily dose of 10 mg per kilogram of body weight for nonobese patients.56

        Systemic Steroids

        Oral steroids are used in COPD exacerbations but should never be used chronically in COPD patients, regardless of disease severity, as they increase morbidity and mortality without improving symptoms or lung function.57,58 The dose of systemic steroids should be tapered and finally discontinued.

        Mucolytics

        Classes of mucolytics include thiol derivatives, inhaled dornase alfa, hypertonic saline, and iodine preparations. Thiol derivatives such as N-acetylcysteine are the most widely studied.59 There is no consistent evidence of beneficial role of mucolytics in COPD patients.7,59 The PANTHEON trial showed decreased exacerbations with N-acetylcysteine (1.16 exacerbations per patient-year compared to 1.49 exacerbations per patient-year in the placebo group; risk ratio, 0.78; 95% CI, 0.67-0.90; P = 0.001) but had methodologic issues including high drop-out rate, exclusion of patients on oxygen, and a large of proportion of nonsmokers.60

        Long-Term Antibiotics

        There is no role for long-term antibiotics in the management of COPD.7 Macrolides are an exception but are used for their anti-inflammatory effects rather than their antibiotic effects. They should be reserved for patients with frequent exacerbations on optimal therapy and will be discussed later in the review.61

        What nonpharmacologic treatments are recommended for COPD patients?

        Smoking cessation, oxygen therapy for severe hypoxemia (resting O2 saturation ≤ 88% or PaO2 ≤ 55 mm Hg), vaccination for influenza and pneumococcus, and appropriate nutrition should be provided in all COPD patients. Pulmonary rehabilitation is indicated for patients in GOLD categories B, C, and D.7 It improves symptoms, quality of life, exercise tolerance, and health care utilization. Beneficial effects last for about 2 years.62,63

         

         

        What other diagnoses should be considered in patients who continue to be symptomatic on optimal therapy?

        Other diseases that share the same risk factors as COPD and can contribute to dyspnea, including coronary heart disease, heart failure, thromboembolic disease, and pulmonary hypertension, should be considered. In addition, all patients with refractory disease should have a careful assessment of their inhaler technique, continued smoking, need for oxygen therapy, and associated deconditioning.

        [embed:render:related:node:204720]

        [embed:render:related:node:204724]

        References

        1. Segreti A, Stirpe E, Rogliani P, Cazzola M. Defining phenotypes in COPD: an aid to personalized healthcare. Mol Diagn Ther. 2014;18:381-388.

        2. Han MK, Agusti A, Calverley PM, et al. Chronic obstructive pulmonary disease phenotypes: the future of COPD. Am J Respir Crit Care Med. 2010;182:598-604.

        3. Aubier M, Marthan R, Berger P, et al. [COPD and inflammation: statement from a French expert group: inflammation and remodelling mechanisms]. Rev Mal Respir. 2010;27:1254-1266.

        4. Wang ZL. Evolving role of systemic inflammation in comorbidities of chronic obstructive pulmonary disease. Chin Med J (Engl). 2010;123:3467-3478.

        5. Buist AS, McBurnie MA, Vollmer WM, et al. International variation in the prevalence of COPD (the BOLD Study): a population-based prevalence study. Lancet. 2007;370:741-750.

        6. Miniño AM, Murphy SL, Xu J, Kochanek KD. Deaths: final data for 2008. Natl Vital Stat Rep. 2011;59:1-126.

        7. Global Initiative for Chronic Obstructive Lung Disease (GOLD): Global strategy for the diagnosis, management, and prevention of COPD 2017. www.goldcopd.org. Accessed July 9, 2019.

        8. Jones PW, Harding G, Berry P, et al. Development and first validation of the COPD Assessment Test. Eur Respir J. 2009;34:648-654.

        9. Wadbo M, Löfdahl CG, Larsson K, et al. Effects of formoterol and ipratropium bromide in COPD: a 3-month placebo-controlled study. Eur Respir J. 2002;20:1138-1146.

        10. Ram FS, Sestini P. Regular inhaled short acting beta2 agonists for the management of stable chronic obstructive pulmonary disease: Cochrane systematic review and meta-analysis. Thorax. 2003;58:580-584.

        11. Colice GL. Nebulized bronchodilators for outpatient management of stable chronic obstructive pulmonary disease. Am J Med. 1996;100(1A):11S-8S.

        12. In chronic obstructive pulmonary disease, a combination of ipratropium and albuterol is more effective than either agent alone. An 85-day multicenter trial. COMBIVENT Inhalation Aerosol Study Group. Chest. 1994;105:1411-1419.

        13. Friedman M, Serby CW, Menjoge SS, et al. Pharmacoeconomic evaluation of a combination of ipratropium plus albuterol compared with ipratropium alone and albuterol alone in COPD. Chest. 1999;115:635-641.

        14. Cook D, Guyatt G, Wong E, et al. Regular versus as-needed short-acting inhaled beta-agonist therapy for chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;163:85-90.

        15. Wilchesky M, Ernst P, Brophy JM, et al. Bronchodilator use and the risk of arrhythmia in COPD: part 2: reassessment in the larger Quebec cohort. Chest. 2012;142:305-311.

        16. Scott VL, Frazee LA. Retrospective comparison of nebulized levalbuterol and albuterol for adverse events in patients with acute airflow obstruction. Am J Ther. 2003;10:341-347.

        17. Wong CS, Pavord ID, Williams J, et al. Bronchodilator, cardiovascular, and hypokalaemic effects of fenoterol, salbutamol, and terbutaline in asthma. Lancet. 1990;336:1396-1399.

        18. Cole JM, Sheehan AH, Jordan JK. Concomitant use of ipratropium and tiotropium in chronic obstructive pulmonary disease. Ann Pharmacother. 2012;46:1717-1721.

        19. Qaseem A, Wilt TJ, Weinberger SE, et al. Diagnosis and management of stable chronic obstructive pulmonary disease: a clinical practice guideline update from the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society. Ann Intern Med. 2011;155:179-191.

        20. Pearlman DS, Chervinsky P, LaForce C, et al. A comparison of salmeterol with albuterol in the treatment of mild-to-moderate asthma. N Engl J Med. 1992;327:1420-1425.

        21. Takahashi T, Belvisi MG, Patel H, et al. Effect of Ba 679 BR, a novel long-acting anticholinergic agent, on cholinergic neurotransmission in guinea pig and human airways. Am J Respir Crit Care Med. 1994;150(6 Pt 1):1640-1645.

        22. Ferguson GT, Feldman G, Pudi KK, et al. improvements in lung function with nebulized revefenacin in the treatment of patients with moderate to very severe COPD: results from two replicate phase III clinical trials. Chronic Obstr Pulm Dis. 2019;6:154-165.

        23. Donohue JF, Fogarty C, Lötvall J, et al. Once-daily bronchodilators for chronic obstructive pulmonary disease: indacaterol versus tiotropium. Am J Respir Crit Care Med. 2010;182:155-162.

        24. Koch A, Pizzichini E, Hamilton A, et al. Lung function efficacy and symptomatic benefit of olodaterol once daily delivered via Respimat versus placebo and formoterol twice daily in patients with GOLD 2-4 COPD: results from two replicate 48-week studies. Int J Chron Obstruct Pulmon Dis. 2014;9:697-714.

        25. Calverley PM, Anderson JA, Celli B, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356:775-789.

        26. Hanania NA, Feldman G, Zachgo W, et al. The efficacy and safety of the novel long-acting β2 agonist vilanterol in patients with COPD: a randomized placebo-controlled trial. Chest. 2012;142:119-127.

        27. Tashkin DP, Celli B, Senn S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543-1554.

        28. Decramer ML, Chapman KR, Dahl R, et al. Once-daily indacaterol versus tiotropium for patients with severe chronic obstructive pulmonary disease (INVIGORATE): a randomised, blinded, parallel-group study. Lancet Respir Med. 2013;1:524-533.

        29. Jones PW, Singh D, Bateman ED, et al. Efficacy and safety of twice-daily aclidinium bromide in COPD patients: the ATTAIN study. Eur Respir J. 2012;40:830-836.

        30. D’Urzo A, Ferguson GT, van Noord JA, et al. Efficacy and safety of once-daily NVA237 in patients with moderate-to-severe COPD: the GLOW1 trial. Respir Res. 2011;12:156.

        31. Antoniu SA. UPLIFT Study: the effects of long-term therapy with inhaled tiotropium in chronic obstructive pulmonary disease. Evaluation of: Tashkin DP, Celli B, Senn S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543-1554. Expert Opin Pharmacother. 2009;10:719–22.

        32. Nelson HS, Gross NJ, Levine B, et al. Cardiac safety profile of nebulized formoterol in adults with COPD: a 12-week, multicenter, randomized, double- blind, double-dummy, placebo- and active-controlled trial. Clin Ther. 2007;29:2167-2178.

        33. Gershon A, Croxford R, Calzavara A, et al. Cardiovascular safety of inhaled long-acting bronchodilators in individuals with chronic obstructive pulmonary disease. JAMA Intern Med. 2013;173:1175-1185.

        34. Aljaafareh A, Valle JR, Lin YL, et al. Risk of cardiovascular events after initiation of long-acting bronchodilators in patients with chronic obstructive lung disease: A population-based study. SAGE Open Med. 2016;4:2050312116671337.

        35. Wang MT, Liou JT, Lin CW, et al. Association of cardiovascular risk with inhaled long-acting bronchodilators in patients with chronic obstructive pulmonary disease: a nested case-Control Study. JAMA Intern Med. 2018;178:229-238.

        36. O’Connor AB. Tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2009;360:185-186.

        37. Kesten S, Jara M, Wentworth C, Lanes S. Pooled clinical trial analysis of tiotropium safety. Chest. 2006;130:1695-1703.

        38. Wise RA, Anzueto A, Cotton D, et al. Tiotropium Respimat inhaler and the risk of death in COPD. N Engl J Med. 2013;369:1491-1501.

        39. Vogelmeier C, Hederer B, Glaab T, et al. Tiotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med. 2011;364:1093-1103.

        40. Chong J, Karner C, Poole P. Tiotropium versus long-acting beta-agonists for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012(9):CD009157.

        41. Gan WQ, Man SF, Sin DD. Effects of inhaled corticosteroids on sputum cell counts in stable chronic obstructive pulmonary disease: a systematic review and a meta-analysis. BMC Pulm Med. 2005;5:3.

        42. Yang IA, Clarke MS, Sim EH, Fong KM. Inhaled corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012(7):CD002991.

        43. Roland NJ, Bhalla RK, Earis J. The local side effects of inhaled corticosteroids: current understanding and review of the literature. Chest. 2004;126:213-219.

        44. Drummond MB, Dasenbrook EC, Pitz MW, et al. Inhaled corticosteroids in patients with stable chronic obstructive pulmonary disease: a systematic review and meta-analysis. JAMA. 2008;300:2407-2416.

        45. Lee SY, Park HY, Kim EK, et al. Combination therapy of inhaled steroids and long-acting beta2-agonists in asthma-COPD overlap syndrome. Int J Chron Obstruct Pulmon Dis. 2016;11:2797-2803.

        46. Postma DS, Rabe KF. The asthma-COPD overlap syndrome. N Engl J Med. 2015;373:1241-1249.

        47. Farne HA, Cates CJ. Long-acting beta2-agonist in addition to tiotropium versus either tiotropium or long-acting beta2-agonist alone for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015:CD008989.

        48. Wedzicha JA, Banerji D, Chapman KR, et al. Indacaterol-glycopyrronium versus salmeterol-fluticasone for COPD. N Engl J Med. 2016;374:2222-2234.

        49. Aaron SD, Vandemheen KL, Fergusson D, et al. Tiotropium in combination with placebo, salmeterol, or fluticasone-salmeterol for treatment of chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med. 2007;146:545-555.

        50. Welte T, Miravitlles M, Hernandez P, et al. Efficacy and tolerability of budesonide/formoterol added to tiotropium in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:741-750.

        51. Lipson DA, Barnhart, Brealey N, et al; IMPACT Investigators. Once-daily single-inhaler triple versus dual therapy in patients with COPD. N Engl J Med. 2018;378:1671-1680.

        52. Gallelli L, Falcone D, Cannataro R, et al. Theophylline action on primary human bronchial epithelial cells under proinflammatory stimuli and steroidal drugs: a therapeutic rationale approach. Drug Des Devel Ther. 2017;11:265-272.

        53. Paloucek FP, Rodvold KA. Evaluation of theophylline overdoses and toxicities. Ann Emerg Med. 1988;17:135-144.

        54. Ram FS, Jones PW, Castro AA, et al. Oral theophylline for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2002(4):CD003902.

        55. Murciano D, Auclair MH, Pariente R, Aubier M. A randomized, controlled trial of theophylline in patients with severe chronic obstructive pulmonary disease. N Engl J Med. 1989;320:1521-1525.

        56. Devereux G, Cotton S, Barnes P, et al. Use of low-dose oral theophylline as an adjunct to inhaled corticosteroids in preventing exacerbations of chronic obstructive pulmonary disease: study protocol for a randomised controlled trial. Trials. 2015;16:267.

        57. Walters JA, Walters EH, Wood-Baker R. Oral corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005(3):CD005374.

        58. Horita N, Miyazawa N, Morita S, et al. Evidence suggesting that oral corticosteroids increase mortality in stable chronic obstructive pulmonary disease. Respir Res. 2014;15:37.

        59. Poole P, Chong J, Cates CJ. Mucolytic agents versus placebo for chronic bronchitis or chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015(7):CD001287.

        60. Zheng JP, Wen FQ, Bai CX, et al. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): a randomised, double-blind placebo-controlled trial. Lancet Respir Med. 2014;2:187-194.

        61. Seemungal TA, Wilkinson TM, Hurst JR, et al. Long-term erythromycin therapy is associated with decreased chronic obstructive pulmonary disease exacerbations. Am J Respir Crit Care Med. 2008;178:1139-1147.

        62. Ries AL, Kaplan RM, Limberg TM, Prewitt LM. Effects of pulmonary rehabilitation on physiologic and psychosocial outcomes in patients with chronic obstructive pulmonary disease. Ann Intern Med. 1995;122:823-832.

        63. Güell R, Casan P, Belda J, et al. Long-term effects of outpatient rehabilitation of COPD: a randomized trial. Chest. 2000;117:976-983.

        References

        1. Segreti A, Stirpe E, Rogliani P, Cazzola M. Defining phenotypes in COPD: an aid to personalized healthcare. Mol Diagn Ther. 2014;18:381-388.

        2. Han MK, Agusti A, Calverley PM, et al. Chronic obstructive pulmonary disease phenotypes: the future of COPD. Am J Respir Crit Care Med. 2010;182:598-604.

        3. Aubier M, Marthan R, Berger P, et al. [COPD and inflammation: statement from a French expert group: inflammation and remodelling mechanisms]. Rev Mal Respir. 2010;27:1254-1266.

        4. Wang ZL. Evolving role of systemic inflammation in comorbidities of chronic obstructive pulmonary disease. Chin Med J (Engl). 2010;123:3467-3478.

        5. Buist AS, McBurnie MA, Vollmer WM, et al. International variation in the prevalence of COPD (the BOLD Study): a population-based prevalence study. Lancet. 2007;370:741-750.

        6. Miniño AM, Murphy SL, Xu J, Kochanek KD. Deaths: final data for 2008. Natl Vital Stat Rep. 2011;59:1-126.

        7. Global Initiative for Chronic Obstructive Lung Disease (GOLD): Global strategy for the diagnosis, management, and prevention of COPD 2017. www.goldcopd.org. Accessed July 9, 2019.

        8. Jones PW, Harding G, Berry P, et al. Development and first validation of the COPD Assessment Test. Eur Respir J. 2009;34:648-654.

        9. Wadbo M, Löfdahl CG, Larsson K, et al. Effects of formoterol and ipratropium bromide in COPD: a 3-month placebo-controlled study. Eur Respir J. 2002;20:1138-1146.

        10. Ram FS, Sestini P. Regular inhaled short acting beta2 agonists for the management of stable chronic obstructive pulmonary disease: Cochrane systematic review and meta-analysis. Thorax. 2003;58:580-584.

        11. Colice GL. Nebulized bronchodilators for outpatient management of stable chronic obstructive pulmonary disease. Am J Med. 1996;100(1A):11S-8S.

        12. In chronic obstructive pulmonary disease, a combination of ipratropium and albuterol is more effective than either agent alone. An 85-day multicenter trial. COMBIVENT Inhalation Aerosol Study Group. Chest. 1994;105:1411-1419.

        13. Friedman M, Serby CW, Menjoge SS, et al. Pharmacoeconomic evaluation of a combination of ipratropium plus albuterol compared with ipratropium alone and albuterol alone in COPD. Chest. 1999;115:635-641.

        14. Cook D, Guyatt G, Wong E, et al. Regular versus as-needed short-acting inhaled beta-agonist therapy for chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;163:85-90.

        15. Wilchesky M, Ernst P, Brophy JM, et al. Bronchodilator use and the risk of arrhythmia in COPD: part 2: reassessment in the larger Quebec cohort. Chest. 2012;142:305-311.

        16. Scott VL, Frazee LA. Retrospective comparison of nebulized levalbuterol and albuterol for adverse events in patients with acute airflow obstruction. Am J Ther. 2003;10:341-347.

        17. Wong CS, Pavord ID, Williams J, et al. Bronchodilator, cardiovascular, and hypokalaemic effects of fenoterol, salbutamol, and terbutaline in asthma. Lancet. 1990;336:1396-1399.

        18. Cole JM, Sheehan AH, Jordan JK. Concomitant use of ipratropium and tiotropium in chronic obstructive pulmonary disease. Ann Pharmacother. 2012;46:1717-1721.

        19. Qaseem A, Wilt TJ, Weinberger SE, et al. Diagnosis and management of stable chronic obstructive pulmonary disease: a clinical practice guideline update from the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society. Ann Intern Med. 2011;155:179-191.

        20. Pearlman DS, Chervinsky P, LaForce C, et al. A comparison of salmeterol with albuterol in the treatment of mild-to-moderate asthma. N Engl J Med. 1992;327:1420-1425.

        21. Takahashi T, Belvisi MG, Patel H, et al. Effect of Ba 679 BR, a novel long-acting anticholinergic agent, on cholinergic neurotransmission in guinea pig and human airways. Am J Respir Crit Care Med. 1994;150(6 Pt 1):1640-1645.

        22. Ferguson GT, Feldman G, Pudi KK, et al. improvements in lung function with nebulized revefenacin in the treatment of patients with moderate to very severe COPD: results from two replicate phase III clinical trials. Chronic Obstr Pulm Dis. 2019;6:154-165.

        23. Donohue JF, Fogarty C, Lötvall J, et al. Once-daily bronchodilators for chronic obstructive pulmonary disease: indacaterol versus tiotropium. Am J Respir Crit Care Med. 2010;182:155-162.

        24. Koch A, Pizzichini E, Hamilton A, et al. Lung function efficacy and symptomatic benefit of olodaterol once daily delivered via Respimat versus placebo and formoterol twice daily in patients with GOLD 2-4 COPD: results from two replicate 48-week studies. Int J Chron Obstruct Pulmon Dis. 2014;9:697-714.

        25. Calverley PM, Anderson JA, Celli B, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356:775-789.

        26. Hanania NA, Feldman G, Zachgo W, et al. The efficacy and safety of the novel long-acting β2 agonist vilanterol in patients with COPD: a randomized placebo-controlled trial. Chest. 2012;142:119-127.

        27. Tashkin DP, Celli B, Senn S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543-1554.

        28. Decramer ML, Chapman KR, Dahl R, et al. Once-daily indacaterol versus tiotropium for patients with severe chronic obstructive pulmonary disease (INVIGORATE): a randomised, blinded, parallel-group study. Lancet Respir Med. 2013;1:524-533.

        29. Jones PW, Singh D, Bateman ED, et al. Efficacy and safety of twice-daily aclidinium bromide in COPD patients: the ATTAIN study. Eur Respir J. 2012;40:830-836.

        30. D’Urzo A, Ferguson GT, van Noord JA, et al. Efficacy and safety of once-daily NVA237 in patients with moderate-to-severe COPD: the GLOW1 trial. Respir Res. 2011;12:156.

        31. Antoniu SA. UPLIFT Study: the effects of long-term therapy with inhaled tiotropium in chronic obstructive pulmonary disease. Evaluation of: Tashkin DP, Celli B, Senn S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543-1554. Expert Opin Pharmacother. 2009;10:719–22.

        32. Nelson HS, Gross NJ, Levine B, et al. Cardiac safety profile of nebulized formoterol in adults with COPD: a 12-week, multicenter, randomized, double- blind, double-dummy, placebo- and active-controlled trial. Clin Ther. 2007;29:2167-2178.

        33. Gershon A, Croxford R, Calzavara A, et al. Cardiovascular safety of inhaled long-acting bronchodilators in individuals with chronic obstructive pulmonary disease. JAMA Intern Med. 2013;173:1175-1185.

        34. Aljaafareh A, Valle JR, Lin YL, et al. Risk of cardiovascular events after initiation of long-acting bronchodilators in patients with chronic obstructive lung disease: A population-based study. SAGE Open Med. 2016;4:2050312116671337.

        35. Wang MT, Liou JT, Lin CW, et al. Association of cardiovascular risk with inhaled long-acting bronchodilators in patients with chronic obstructive pulmonary disease: a nested case-Control Study. JAMA Intern Med. 2018;178:229-238.

        36. O’Connor AB. Tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2009;360:185-186.

        37. Kesten S, Jara M, Wentworth C, Lanes S. Pooled clinical trial analysis of tiotropium safety. Chest. 2006;130:1695-1703.

        38. Wise RA, Anzueto A, Cotton D, et al. Tiotropium Respimat inhaler and the risk of death in COPD. N Engl J Med. 2013;369:1491-1501.

        39. Vogelmeier C, Hederer B, Glaab T, et al. Tiotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med. 2011;364:1093-1103.

        40. Chong J, Karner C, Poole P. Tiotropium versus long-acting beta-agonists for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012(9):CD009157.

        41. Gan WQ, Man SF, Sin DD. Effects of inhaled corticosteroids on sputum cell counts in stable chronic obstructive pulmonary disease: a systematic review and a meta-analysis. BMC Pulm Med. 2005;5:3.

        42. Yang IA, Clarke MS, Sim EH, Fong KM. Inhaled corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2012(7):CD002991.

        43. Roland NJ, Bhalla RK, Earis J. The local side effects of inhaled corticosteroids: current understanding and review of the literature. Chest. 2004;126:213-219.

        44. Drummond MB, Dasenbrook EC, Pitz MW, et al. Inhaled corticosteroids in patients with stable chronic obstructive pulmonary disease: a systematic review and meta-analysis. JAMA. 2008;300:2407-2416.

        45. Lee SY, Park HY, Kim EK, et al. Combination therapy of inhaled steroids and long-acting beta2-agonists in asthma-COPD overlap syndrome. Int J Chron Obstruct Pulmon Dis. 2016;11:2797-2803.

        46. Postma DS, Rabe KF. The asthma-COPD overlap syndrome. N Engl J Med. 2015;373:1241-1249.

        47. Farne HA, Cates CJ. Long-acting beta2-agonist in addition to tiotropium versus either tiotropium or long-acting beta2-agonist alone for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015:CD008989.

        48. Wedzicha JA, Banerji D, Chapman KR, et al. Indacaterol-glycopyrronium versus salmeterol-fluticasone for COPD. N Engl J Med. 2016;374:2222-2234.

        49. Aaron SD, Vandemheen KL, Fergusson D, et al. Tiotropium in combination with placebo, salmeterol, or fluticasone-salmeterol for treatment of chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med. 2007;146:545-555.

        50. Welte T, Miravitlles M, Hernandez P, et al. Efficacy and tolerability of budesonide/formoterol added to tiotropium in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:741-750.

        51. Lipson DA, Barnhart, Brealey N, et al; IMPACT Investigators. Once-daily single-inhaler triple versus dual therapy in patients with COPD. N Engl J Med. 2018;378:1671-1680.

        52. Gallelli L, Falcone D, Cannataro R, et al. Theophylline action on primary human bronchial epithelial cells under proinflammatory stimuli and steroidal drugs: a therapeutic rationale approach. Drug Des Devel Ther. 2017;11:265-272.

        53. Paloucek FP, Rodvold KA. Evaluation of theophylline overdoses and toxicities. Ann Emerg Med. 1988;17:135-144.

        54. Ram FS, Jones PW, Castro AA, et al. Oral theophylline for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2002(4):CD003902.

        55. Murciano D, Auclair MH, Pariente R, Aubier M. A randomized, controlled trial of theophylline in patients with severe chronic obstructive pulmonary disease. N Engl J Med. 1989;320:1521-1525.

        56. Devereux G, Cotton S, Barnes P, et al. Use of low-dose oral theophylline as an adjunct to inhaled corticosteroids in preventing exacerbations of chronic obstructive pulmonary disease: study protocol for a randomised controlled trial. Trials. 2015;16:267.

        57. Walters JA, Walters EH, Wood-Baker R. Oral corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005(3):CD005374.

        58. Horita N, Miyazawa N, Morita S, et al. Evidence suggesting that oral corticosteroids increase mortality in stable chronic obstructive pulmonary disease. Respir Res. 2014;15:37.

        59. Poole P, Chong J, Cates CJ. Mucolytic agents versus placebo for chronic bronchitis or chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015(7):CD001287.

        60. Zheng JP, Wen FQ, Bai CX, et al. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): a randomised, double-blind placebo-controlled trial. Lancet Respir Med. 2014;2:187-194.

        61. Seemungal TA, Wilkinson TM, Hurst JR, et al. Long-term erythromycin therapy is associated with decreased chronic obstructive pulmonary disease exacerbations. Am J Respir Crit Care Med. 2008;178:1139-1147.

        62. Ries AL, Kaplan RM, Limberg TM, Prewitt LM. Effects of pulmonary rehabilitation on physiologic and psychosocial outcomes in patients with chronic obstructive pulmonary disease. Ann Intern Med. 1995;122:823-832.

        63. Güell R, Casan P, Belda J, et al. Long-term effects of outpatient rehabilitation of COPD: a randomized trial. Chest. 2000;117:976-983.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Stable COPD: Initiating and Optimizing Therapy
        Display Headline
        Stable COPD: Initiating and Optimizing Therapy
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 July;16(1)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Tue, 07/16/2019 - 10:45
        Un-Gate On Date
        Tue, 07/16/2019 - 10:45
        Use ProPublica
        CFC Schedule Remove Status
        Tue, 07/16/2019 - 10:45
        Hide sidebar & use full width
        render the right sidebar.

        Stable COPD: Managing Acute Exacerbations

        Article Type
        Changed
        Wed, 07/17/2019 - 14:54
        Display Headline
        Stable COPD: Managing Acute Exacerbations

        Case Presentation

        A 70-year-old man with severe chronic obstructive pulmonary disease (COPD) on oxygen therapy and obstructive sleep apnea treated with nocturnal continuous positive airway pressure was seen in the pulmonary clinic for evaluation of his dyspnea. He was symptomatic with minimal activity and had chronic cough with some sputum production. He had been hospitalized 3 times over the past 12 months and had been to the emergency department (ED) the same number of times for dyspnea. Pertinent medications included as-needed albuterol inhaler, inhaled steroids, and tiotropium 18 mcg inhaled daily. He demonstrated good inhaler technique. On examination, his vital signs were pulse 99 beats/min, oxygen saturation 94% on 2 L/min of oxygen by nasal cannula, blood pressure 126/72 mm Hg, respiratory rate 15 breaths/min, and body mass index 35 kg/m2. He appeared chronically ill but in no acute distress. No wheezing or rales were heard. He had no lower extremity edema. The remainder of the exam was within normal limits. His last pulmonary function test demonstrated moderate obstruction with significant bronchodilator response to 2 puffs of albuterol. The side effects of chronic steroid therapy were impressed upon the patient and 500 mg of roflumilast was started daily. Over the course of the next 3 months, he had no further exacerbations. Roflumilast was continued. He has not required any further hospitalizations, ED visits, or oral steroid use since the last clinic visit.

        What is the significance of acute exacerbations of COPD?

        Acute exacerbation of COPD (AECOPD) is a frequently observed complication for many patients with COPD.1,2 AECOPD is associated with accelerated disease progression, augmented decline in health status and quality of life, and increased mortality.3 Exacerbations account for most of the costs associated with COPD. Estimates suggest that the aggregate costs associated with the treatment of AECOPD are between $3.2 and $3.8 billion, and that annual health care costs are 10-fold greater for patients with COPD associated with acute exacerbations than for patients with COPD but without exacerbations.4 Hence, any intervention that could potentially minimize or prevent this complication will have far-reaching benefits to patients with COPD as well as provide significant cost saving.

        How is AECOPD defined?

        COPD exacerbation is defined as a baseline change of the patient’s dyspnea, cough, and/or sputum that is acute in onset, and may warrant a change in regular medication in a patient with underlying COPD.5 Exacerbation in clinical trials has been defined on the basis of whether an increase in the level of care beyond regular care is required primarily in the hospital or ED.6 Frequent exacerbations are defined as 3 symptom-defined exacerbations per year or 2 per year if defined by the need for therapy with corticosteroids, antibiotics, or both.7

        What is the underlying pathophysiology?

        AECOPD is associated with enhanced upper and lower airway and systemic inflammation. The bronchial mucosa of stable COPD patients have increased numbers of CD8+ lymphocytes and macrophages. In mild AECOPD, eosinophils are increased in the bronchial mucosa and modest elevation of neutrophils, T lymphocytes (CD3), and TNF-α positive cells has also been reported.2 With more severe AECOPD, airway neutrophils are increased. Oxidative stress is a key factor in the development of airway inflammation in COPD.1 Patients with severe exacerbations have augmented large airway interleukin-8 (IL-8) levels and increased oxidative stress as demonstrated by markers such as hydrogen peroxide and 8-isoprostane.6

        How do acute exacerbations affect the course of the disease?

        In general, as the severity of the underlying COPD increases, exacerbations become both more severe and more frequent. Patients with frequent exacerbations have a worse quality of life than patients with a history of less frequent exacerbations.8 Frequent exacerbations have also been linked to a decline in lung function, with studies suggesting that there might be a decline of 7 mL in forced expiratory volume in 1 second (FEV1) per lower respiratory tract infection per year,9,10 and approximately 8 mL per year in patients with frequent exacerbations as compared to those with sporadic exacerbations.11

        What are the triggers for COPD exacerbation?

        Respiratory infections are estimated to trigger approximately two-thirds of exacerbations.2 Viral and bacterial infections cause most exacerbations. The effect of the infective triggers is to increase inflammation, cause bronchoconstriction, edema, and mucus production, with a resultant increase in dynamic hyperinflation.12 Thus, any intervention that reduces inflammation in COPD reduces the number and severity of exacerbations, whereas bronchodilators have an impact on exacerbation by their effects on reducing dynamic hyperinflation. The triggers for the one-third of exacerbations not triggered by infection are postulated to be related to other medical conditions, including pulmonary embolism, aspiration, heart failure, and myocardial ischemia.6

         

         

        What are the pharmacologic options available for prevention of AECOPD?

        In recognition of the importance of preventing COPD exacerbations, the American College of Chest Physicians and Canadian Thoracic Society5 have published an evidence-informed clinical guideline specifically examining the prevention of AECOPD, with the goal of assisting clinicians in providing optimal management for COPD patients. The following pharmacologic agents have been recognized as being effective at reducing the frequency of acute exacerbations without any impact on the severity of COPD itself.

        Roflumilast

        Phosphodiesterase 4 (PDE4) inhibition appears to have inflammatory-modulating properties in the airways, although the exact mechanism of action is unclear. Some have proposed that it reduces inflammation by inhibiting the breakdown of intracellular cyclic adenosine monophosphate.13 In 2 large clinical trials,14,15 daily use of a PDE4 inhibitor (roflumilast) showed a significant (15%–18%) reduction in yearly AECOPD incidence (approximate number needed to treat: 4). This benefit was seen in patients with GOLD stage 3–4 disease (FEV1 < 50% predicted) with the chronic bronchitic phenotype and who had experienced at least 1 exacerbation in the previous year.

        Importantly, these clinical trials specifically prohibited the use of inhaled corticosteroids (ICS) and long-acting muscarinic antagonists (LAMAs). Thus, it remains unclear if PDE4 inhibition should be used as an add-on to ICS/LAMA therapy in patients who continue to have frequent AECOPD or whether PDE4 inhibition could be used instead of these standard therapies in patients with well-controlled daily symptoms without ICS or LAMA therapy but who experience frequent exacerbations.

        Of note, earlier trials with roflumilast included patients with ICS and LAMA use.14,16 These trials were focused on FEV1 improvement and found no benefit. It was only in post ad hoc analyses that a reduction in AECOPD in patients with frequent exacerbations was found among those taking roflumilast, regardless of ICS or LAMA use.17 While roflumilast has documented benefit in improving lung function and reducing the rate of exacerbations, it has not been reported to decrease hospitalizations.4 This indicates that although the drug reduces the total number of exacerbations, it may not be as useful in preventing episodes of severe exacerbations of COPD.

        Although PDE4 inhibitors are easy to administer (a once-daily pill), they are associated with significant gastrointestinal side effects (diarrhea, nausea, reduced appetite), weight loss, headache, and sleep disturbance.18 Adverse effects tend to occur early during treatment, are reversible, and lessen over time with treatment.6 Studies reported an average unexplained weight loss of 2 kg, and monitoring weight during treatment is advised. In addition, it is important to avoid roflumilast in underweight patients. Roflumilast should also be used with caution in depressed patients.5

        N-acetylcysteine

        N-acetylcysteine (NAC) reduces the viscosity of respiratory secretions as a result of the cleavage of the disulfide bonds and has been studied as a mucolytic agent to aid in the elimination of respiratory secretions.19 Oral NAC is quickly absorbed and is rapidly present in an active form in lung tissue and respiratory secretions after ingestion. NAC is well-tolerated except for occasional patients with GI adverse effects. The role of NAC in preventing AECOPD has been studied for more than 3 decades,20-22 although the largest clinical trial to date was reported in 2014.23 Taken together, the combined data demonstrate a significant reduction in the rate of COPD exacerbations associated with the use of NAC when compared with placebo (odds ratio [OR], 0.61; 95% confidence interval [CI], 0.37-0.99). Clinical guidelines suggest that in patients with moderate to severe COPD (FEV1/forced vital capacity ratio < 0.7, and FEV1 < 80% predicted) receiving maintenance bronchodilator therapy combined with ICS and history of 2 more exacerbations in the previous 2 years, treatment with oral NAC can be administered to prevent AECOPD.

         

         

        Macrolides

        Continuous prophylactic use of antibiotics in older studies had no effect on the frequency of AECOPD.24,25 But it is known that macrolide antibiotics have several antimicrobial, anti-inflammatory and immunomodulating effects and have been used for many years in the management of other chronic airway disease, including diffuse pan-bronchiolitis and cystic fibrosis.5 One recent study showed that the use of once-daily generic azithromycin 5 days per week appeared to have an impact on AECOPD incidence.26 In this study, the rate of AECOPD was reduced from 1.83 to 1.48 exacerbations per patient-year (relative risk, 0.83; 95% CI, 0.72–0.95; P = 0.01). Azithromycin also prevented severe AECOPD. Greater benefit was obtained with milder forms of the disease and in the elderly. Azithromycin did not appear to provide any benefit in those who continued to smoke (hazard ratio, 0.99).27 Other studies have shown that azithromycin was associated with an increased incidence of bacterial resistance and impaired hearing.28 Overall data from the available clinical trials are robust and demonstrate that regular macrolide therapy definitely reduces the risk of AECOPD. Due to potential adverse effects, however, macrolide therapy is an option rather than a strong recommendation.5 The prescribing clinician also needs to consider potential of prolongation of the QT interval.26

        Immunostimulants

        Immunostimulants have also been reported to reduce frequency of AECOPD.29,30 Bacterial lysates, reconstituted mixtures of bacterial antigens present in the lower airways of COPD patients, act as immunostimulants through the induction of cellular maturation, stimulating lymphocyte chemotaxis and increasing opsonization when administered to individuals with COPD.6 Studies have demonstrated a reduction in the severe complications of exacerbations and hospital admissions in COPD patients with OM-85, a detoxified oral immunoactive bacterial extract.29,30 However, most of these trials were conducted prior to the routine use of long-acting bronchodilators and ICS in COPD. A study that evaluated the efficacy of ismigen, a bacterial lysate, in reducing AECOPD31 found no difference in the exacerbation rate between ismigen and placebo or the time to first exacerbation. Additional studies are needed to examine the long-term effects of this therapy in patients receiving currently recommended COPD maintenance therapy.6

        β-Blockers

        Observational studies of β-blocker use in preventing AECOPD have yielded encouraging results, with one study showing a reduction in AECOPD risk (incidence risk ratio, 0.73; 95% CI, 0.60–0.90) in patients receiving β-blockers versus those not on β-blockers.32 Based on these findings, a clinical trial investigating the impact of metoprolol on risk of AECOPD is ongoing.33

        Proton Pump Inhibitors

        Gastroesophageal reflux disease is an independent risk factor for exacerbations.34 Two small, single-center studies,35,36 have shown that use of lansoprazole decreases the risk and frequency of AECOPD. However, data from the Predicting Outcome using Systemic Markers in Severe Exacerbations of COPD (PROMISE-COPD) study,6 which was a multicenter prospective observational study, suggested that patients with stable COPD receiving a proton pump inhibitor were at high risk of frequent and severe exacerbations.37 Thus, at this stage, their definitive role needs to be defined, possibly with a randomized, placebo-controlled study.

        [embed:render:related:node:204714]

        [embed:render:related:node:204724]

        References

        1. Wedzicha JA, Singh R, Mackay AJ. Acute COPD exacerbations. Clin Chest Med. 2014;35:157-163.

        2. Wedzicha JA, Seemungal TAR. COPD exacerbations: defining their cause and prevention. Lancet. 2007;370:786-796.

        3. Spencer S, Calverley PMA, Burge PS, Jones PW. Impact of preventing exacerbations on deterioration of health status in COPD. Eur Respir J. 2004;23:698-702.

        4. Blanchette CM, Gross NJ, Altman P. Rising costs of COPD and the potential for maintenance therapy to slow the trend. Am Health Drug Benef. 2014;7:98.

        5. Criner GJ, Bourbeau J, Diekemper RL, et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest. 2015;147:894-942.

        6. Vogelmeier CF, Criner GJ, Martinez FJ, et al. Global strategy for the diagnosis, management and prevention of chronic obstructive lung disease 2017 report. Respirology. 2017;22:575-601.

        7. Wedzicha JA, Brill SE, Allinson JP, Donaldson GC. Mechanisms and impact of the frequent exacerbator phenotype in chronic obstructive pulmonary disease. BMC Med. 2013;11:181.

        8. Seemungal TAR, Donaldson GC, Paul EA, et al. Effect of exacerbation on quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1418-1422.

        9. Ries AL, Kaplan RM, Limberg TM, Prewitt LM. Effects of pulmonary rehabilitation on physiologic and psychosocial outcomes in patients with chronic obstructive pulmonary disease. Ann Intern Med. 1995;122:823-832.

        10. Kanner RE, Anthonisen NR, Connett JE. Lower respiratory illnesses promote FEV1 decline in current smokers but not ex-smokers with mild chronic obstructive pulmonary disease: results from the lung health study. Am J Respir Crit Care Med. 2001;164:358-364.

        11. Donaldson GC, Seemungal TAR, Bhowmik A, Wedzicha JA. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57:847-852.

        12. Papi A, Bellettato CM, Braccioni F, et al. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006;173:1114-1121.

        13. Rabe KF. Update on roflumilast, a phosphodiesterase 4 inhibitor for the treatment of chronic obstructive pulmonary disease. Br J Pharmacol. 2011;163:53-67.

        14. Calverley PMA, Rabe KF, Goehring U-M, et al. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet. 2009;374:685-694.

        15. Fabbri LM, Calverley PMA, Izquierdo-Alonso JL, et al. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with long-acting bronchodilators: two randomised clinical trials. Lancet. 2009;374:695-703.

        16. Lee S, Hui DSC, Mahayiddin AA, et al. Roflumilast in Asian patients with COPD: a randomized placebo-controlled trial. Respirology. 2011;16:1249-1257.

        17. Calverley PM, Martinez FJ, Fabbri LM, et al. Does roflumilast decrease exacerbations in severe COPD patients not controlled by inhaled combination therapy? The REACT study protocol. Int J Chron Obstruct Pulmon Dis. 2012;7:375-382.

        18. Chong J, Leung B, Poole P. Phosphodiesterase 4 inhibitors for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2013(11):CD002309.

        19. Sheffner AL, Medler EM, Jacobs LW, Sarett HP. The in vitro reduction in viscosity of human tracheobronchial secretions by acetylcysteine. Am Rev Respir Dis. 1964;90:721-729.

        20. Boman G, Bäcker U, Larsson S, et al. Oral acetylcysteine reduces exacerbation rate in chronic bronchitis: report of a trial organized by the Swedish Society for Pulmonary Diseases. Eur J Respir Dis. 1983;64:405-415.

        21. Grassi C, Morandini GC. A controlled trial of intermittent oral acetylcysteine in the long-term treatment of chronic bronchitis. Eur J Clin Pharmacol. 1976;9:393-396.

        22. Hansen NCG, Skriver A, Brorsen-Riis L, et al. Orally administered N-acetylcysteine may improve general well-being in patients with mild chronic bronchitis. Respir Med. 1994;88:531-535.

        23. Zheng JP, Wen FQ, Bai CX, et al. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): a randomised, double-blind placebo-controlled trial. Lancet Respir Med. 2014;2:187-194.

        24. Francis RS, Spicer CC. Chemotherapy in chronic bronchitis: Influence of daily penicillin and tetracycline on exacerbations and their cost: A report to the research committee of the British Tuberculosis Association by Their Chronic Bronchitis Subcommittee. BMJ. 1960;1:297-303.

        25. Francis RS, May JR, Spicer CC. Chemotherapy of bronchitis. BMJ. 1961;2:979.

        26. Albert RK, Connett J, Bailey WC, et al. Azithromycin for prevention of exacerbations of COPD. N Engl J Med. 2011;365:689-698.

        27. Han MK, Tayob N, Murray S, et al. Predictors of chronic obstructive pulmonary disease exacerbation reduction in response to daily azithromycin therapy. Am J Respir Crit Care Med. 2014;189:1503-1508.

        28. Uzun S, Djamin RS, Kluytmans JAJW, et al. Azithromycin maintenance treatment in patients with frequent exacerbations of chronic obstructive pulmonary disease (COLUMBUS): a randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2014;2:361-368.

        29. Collet JP, Shapiro S, Ernst P, et al. Effects of an immunostimulating agent on acute exacerbations and hospitalizations in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1997;156:1719-1724.

        30. Jing LI. Protective effect of a bacterial extract against acute exacerbation in patients with chronic bronchitis accompanied by chronic obstructive pulmonary. Age. 2004;67:828-834.

        31. Braido F, Tarantini F, Ghiglione V, et al. Bacterial lysate in the prevention of acute exacerbation of COPD and in respiratory recurrent infections. Int J Chron Obstruct Pulmon Dis. 2007;2:335.

        32. Bhatt SP, Wells JM, Kinney GL, et al. β-Blockers are associated with a reduction in COPD exacerbations. Thorax. 2016;71:8-14.

        33. Bhatt SP, Connett JE, Voelker H, et al. β-Blockers for the prevention of acute exacerbations of chronic obstructive pulmonary disease (βLOCK COPD): a randomised controlled study protocol. BMJ Open. 2016;6:e012292.

        34. Hurst JR, Vestbo J, Anzueto A, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363:1128-1138.

        35. Sasaki T, Nakayama K, Yasuda H, et al. A randomized, single-blind study of lansoprazole for the prevention of exacerbations of chronic obstructive pulmonary disease in older patients. J Am Geriatr Soc. 2009;57:1453-1457.

        36. Xiong W, Zhang Qs, Zhao W, et al. A 12-month follow-up study on the preventive effect of oral lansoprazole on acute exacerbation of chronic obstructive pulmonary disease. Int J Exper Pathol. 2016;97:107-113.

        37. Baumeler L, Papakonstantinou E, Milenkovic B, et al. Therapy with proton-pump inhibitors for gastroesophageal reflux disease does not reduce the risk for severe exacerbations in COPD. Respirology. 2016;21:883-890.

        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Publications
        Topics
        Sections
        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Case Presentation

        A 70-year-old man with severe chronic obstructive pulmonary disease (COPD) on oxygen therapy and obstructive sleep apnea treated with nocturnal continuous positive airway pressure was seen in the pulmonary clinic for evaluation of his dyspnea. He was symptomatic with minimal activity and had chronic cough with some sputum production. He had been hospitalized 3 times over the past 12 months and had been to the emergency department (ED) the same number of times for dyspnea. Pertinent medications included as-needed albuterol inhaler, inhaled steroids, and tiotropium 18 mcg inhaled daily. He demonstrated good inhaler technique. On examination, his vital signs were pulse 99 beats/min, oxygen saturation 94% on 2 L/min of oxygen by nasal cannula, blood pressure 126/72 mm Hg, respiratory rate 15 breaths/min, and body mass index 35 kg/m2. He appeared chronically ill but in no acute distress. No wheezing or rales were heard. He had no lower extremity edema. The remainder of the exam was within normal limits. His last pulmonary function test demonstrated moderate obstruction with significant bronchodilator response to 2 puffs of albuterol. The side effects of chronic steroid therapy were impressed upon the patient and 500 mg of roflumilast was started daily. Over the course of the next 3 months, he had no further exacerbations. Roflumilast was continued. He has not required any further hospitalizations, ED visits, or oral steroid use since the last clinic visit.

        What is the significance of acute exacerbations of COPD?

        Acute exacerbation of COPD (AECOPD) is a frequently observed complication for many patients with COPD.1,2 AECOPD is associated with accelerated disease progression, augmented decline in health status and quality of life, and increased mortality.3 Exacerbations account for most of the costs associated with COPD. Estimates suggest that the aggregate costs associated with the treatment of AECOPD are between $3.2 and $3.8 billion, and that annual health care costs are 10-fold greater for patients with COPD associated with acute exacerbations than for patients with COPD but without exacerbations.4 Hence, any intervention that could potentially minimize or prevent this complication will have far-reaching benefits to patients with COPD as well as provide significant cost saving.

        How is AECOPD defined?

        COPD exacerbation is defined as a baseline change of the patient’s dyspnea, cough, and/or sputum that is acute in onset, and may warrant a change in regular medication in a patient with underlying COPD.5 Exacerbation in clinical trials has been defined on the basis of whether an increase in the level of care beyond regular care is required primarily in the hospital or ED.6 Frequent exacerbations are defined as 3 symptom-defined exacerbations per year or 2 per year if defined by the need for therapy with corticosteroids, antibiotics, or both.7

        What is the underlying pathophysiology?

        AECOPD is associated with enhanced upper and lower airway and systemic inflammation. The bronchial mucosa of stable COPD patients have increased numbers of CD8+ lymphocytes and macrophages. In mild AECOPD, eosinophils are increased in the bronchial mucosa and modest elevation of neutrophils, T lymphocytes (CD3), and TNF-α positive cells has also been reported.2 With more severe AECOPD, airway neutrophils are increased. Oxidative stress is a key factor in the development of airway inflammation in COPD.1 Patients with severe exacerbations have augmented large airway interleukin-8 (IL-8) levels and increased oxidative stress as demonstrated by markers such as hydrogen peroxide and 8-isoprostane.6

        How do acute exacerbations affect the course of the disease?

        In general, as the severity of the underlying COPD increases, exacerbations become both more severe and more frequent. Patients with frequent exacerbations have a worse quality of life than patients with a history of less frequent exacerbations.8 Frequent exacerbations have also been linked to a decline in lung function, with studies suggesting that there might be a decline of 7 mL in forced expiratory volume in 1 second (FEV1) per lower respiratory tract infection per year,9,10 and approximately 8 mL per year in patients with frequent exacerbations as compared to those with sporadic exacerbations.11

        What are the triggers for COPD exacerbation?

        Respiratory infections are estimated to trigger approximately two-thirds of exacerbations.2 Viral and bacterial infections cause most exacerbations. The effect of the infective triggers is to increase inflammation, cause bronchoconstriction, edema, and mucus production, with a resultant increase in dynamic hyperinflation.12 Thus, any intervention that reduces inflammation in COPD reduces the number and severity of exacerbations, whereas bronchodilators have an impact on exacerbation by their effects on reducing dynamic hyperinflation. The triggers for the one-third of exacerbations not triggered by infection are postulated to be related to other medical conditions, including pulmonary embolism, aspiration, heart failure, and myocardial ischemia.6

         

         

        What are the pharmacologic options available for prevention of AECOPD?

        In recognition of the importance of preventing COPD exacerbations, the American College of Chest Physicians and Canadian Thoracic Society5 have published an evidence-informed clinical guideline specifically examining the prevention of AECOPD, with the goal of assisting clinicians in providing optimal management for COPD patients. The following pharmacologic agents have been recognized as being effective at reducing the frequency of acute exacerbations without any impact on the severity of COPD itself.

        Roflumilast

        Phosphodiesterase 4 (PDE4) inhibition appears to have inflammatory-modulating properties in the airways, although the exact mechanism of action is unclear. Some have proposed that it reduces inflammation by inhibiting the breakdown of intracellular cyclic adenosine monophosphate.13 In 2 large clinical trials,14,15 daily use of a PDE4 inhibitor (roflumilast) showed a significant (15%–18%) reduction in yearly AECOPD incidence (approximate number needed to treat: 4). This benefit was seen in patients with GOLD stage 3–4 disease (FEV1 < 50% predicted) with the chronic bronchitic phenotype and who had experienced at least 1 exacerbation in the previous year.

        Importantly, these clinical trials specifically prohibited the use of inhaled corticosteroids (ICS) and long-acting muscarinic antagonists (LAMAs). Thus, it remains unclear if PDE4 inhibition should be used as an add-on to ICS/LAMA therapy in patients who continue to have frequent AECOPD or whether PDE4 inhibition could be used instead of these standard therapies in patients with well-controlled daily symptoms without ICS or LAMA therapy but who experience frequent exacerbations.

        Of note, earlier trials with roflumilast included patients with ICS and LAMA use.14,16 These trials were focused on FEV1 improvement and found no benefit. It was only in post ad hoc analyses that a reduction in AECOPD in patients with frequent exacerbations was found among those taking roflumilast, regardless of ICS or LAMA use.17 While roflumilast has documented benefit in improving lung function and reducing the rate of exacerbations, it has not been reported to decrease hospitalizations.4 This indicates that although the drug reduces the total number of exacerbations, it may not be as useful in preventing episodes of severe exacerbations of COPD.

        Although PDE4 inhibitors are easy to administer (a once-daily pill), they are associated with significant gastrointestinal side effects (diarrhea, nausea, reduced appetite), weight loss, headache, and sleep disturbance.18 Adverse effects tend to occur early during treatment, are reversible, and lessen over time with treatment.6 Studies reported an average unexplained weight loss of 2 kg, and monitoring weight during treatment is advised. In addition, it is important to avoid roflumilast in underweight patients. Roflumilast should also be used with caution in depressed patients.5

        N-acetylcysteine

        N-acetylcysteine (NAC) reduces the viscosity of respiratory secretions as a result of the cleavage of the disulfide bonds and has been studied as a mucolytic agent to aid in the elimination of respiratory secretions.19 Oral NAC is quickly absorbed and is rapidly present in an active form in lung tissue and respiratory secretions after ingestion. NAC is well-tolerated except for occasional patients with GI adverse effects. The role of NAC in preventing AECOPD has been studied for more than 3 decades,20-22 although the largest clinical trial to date was reported in 2014.23 Taken together, the combined data demonstrate a significant reduction in the rate of COPD exacerbations associated with the use of NAC when compared with placebo (odds ratio [OR], 0.61; 95% confidence interval [CI], 0.37-0.99). Clinical guidelines suggest that in patients with moderate to severe COPD (FEV1/forced vital capacity ratio < 0.7, and FEV1 < 80% predicted) receiving maintenance bronchodilator therapy combined with ICS and history of 2 more exacerbations in the previous 2 years, treatment with oral NAC can be administered to prevent AECOPD.

         

         

        Macrolides

        Continuous prophylactic use of antibiotics in older studies had no effect on the frequency of AECOPD.24,25 But it is known that macrolide antibiotics have several antimicrobial, anti-inflammatory and immunomodulating effects and have been used for many years in the management of other chronic airway disease, including diffuse pan-bronchiolitis and cystic fibrosis.5 One recent study showed that the use of once-daily generic azithromycin 5 days per week appeared to have an impact on AECOPD incidence.26 In this study, the rate of AECOPD was reduced from 1.83 to 1.48 exacerbations per patient-year (relative risk, 0.83; 95% CI, 0.72–0.95; P = 0.01). Azithromycin also prevented severe AECOPD. Greater benefit was obtained with milder forms of the disease and in the elderly. Azithromycin did not appear to provide any benefit in those who continued to smoke (hazard ratio, 0.99).27 Other studies have shown that azithromycin was associated with an increased incidence of bacterial resistance and impaired hearing.28 Overall data from the available clinical trials are robust and demonstrate that regular macrolide therapy definitely reduces the risk of AECOPD. Due to potential adverse effects, however, macrolide therapy is an option rather than a strong recommendation.5 The prescribing clinician also needs to consider potential of prolongation of the QT interval.26

        Immunostimulants

        Immunostimulants have also been reported to reduce frequency of AECOPD.29,30 Bacterial lysates, reconstituted mixtures of bacterial antigens present in the lower airways of COPD patients, act as immunostimulants through the induction of cellular maturation, stimulating lymphocyte chemotaxis and increasing opsonization when administered to individuals with COPD.6 Studies have demonstrated a reduction in the severe complications of exacerbations and hospital admissions in COPD patients with OM-85, a detoxified oral immunoactive bacterial extract.29,30 However, most of these trials were conducted prior to the routine use of long-acting bronchodilators and ICS in COPD. A study that evaluated the efficacy of ismigen, a bacterial lysate, in reducing AECOPD31 found no difference in the exacerbation rate between ismigen and placebo or the time to first exacerbation. Additional studies are needed to examine the long-term effects of this therapy in patients receiving currently recommended COPD maintenance therapy.6

        β-Blockers

        Observational studies of β-blocker use in preventing AECOPD have yielded encouraging results, with one study showing a reduction in AECOPD risk (incidence risk ratio, 0.73; 95% CI, 0.60–0.90) in patients receiving β-blockers versus those not on β-blockers.32 Based on these findings, a clinical trial investigating the impact of metoprolol on risk of AECOPD is ongoing.33

        Proton Pump Inhibitors

        Gastroesophageal reflux disease is an independent risk factor for exacerbations.34 Two small, single-center studies,35,36 have shown that use of lansoprazole decreases the risk and frequency of AECOPD. However, data from the Predicting Outcome using Systemic Markers in Severe Exacerbations of COPD (PROMISE-COPD) study,6 which was a multicenter prospective observational study, suggested that patients with stable COPD receiving a proton pump inhibitor were at high risk of frequent and severe exacerbations.37 Thus, at this stage, their definitive role needs to be defined, possibly with a randomized, placebo-controlled study.

        [embed:render:related:node:204714]

        [embed:render:related:node:204724]

        Case Presentation

        A 70-year-old man with severe chronic obstructive pulmonary disease (COPD) on oxygen therapy and obstructive sleep apnea treated with nocturnal continuous positive airway pressure was seen in the pulmonary clinic for evaluation of his dyspnea. He was symptomatic with minimal activity and had chronic cough with some sputum production. He had been hospitalized 3 times over the past 12 months and had been to the emergency department (ED) the same number of times for dyspnea. Pertinent medications included as-needed albuterol inhaler, inhaled steroids, and tiotropium 18 mcg inhaled daily. He demonstrated good inhaler technique. On examination, his vital signs were pulse 99 beats/min, oxygen saturation 94% on 2 L/min of oxygen by nasal cannula, blood pressure 126/72 mm Hg, respiratory rate 15 breaths/min, and body mass index 35 kg/m2. He appeared chronically ill but in no acute distress. No wheezing or rales were heard. He had no lower extremity edema. The remainder of the exam was within normal limits. His last pulmonary function test demonstrated moderate obstruction with significant bronchodilator response to 2 puffs of albuterol. The side effects of chronic steroid therapy were impressed upon the patient and 500 mg of roflumilast was started daily. Over the course of the next 3 months, he had no further exacerbations. Roflumilast was continued. He has not required any further hospitalizations, ED visits, or oral steroid use since the last clinic visit.

        What is the significance of acute exacerbations of COPD?

        Acute exacerbation of COPD (AECOPD) is a frequently observed complication for many patients with COPD.1,2 AECOPD is associated with accelerated disease progression, augmented decline in health status and quality of life, and increased mortality.3 Exacerbations account for most of the costs associated with COPD. Estimates suggest that the aggregate costs associated with the treatment of AECOPD are between $3.2 and $3.8 billion, and that annual health care costs are 10-fold greater for patients with COPD associated with acute exacerbations than for patients with COPD but without exacerbations.4 Hence, any intervention that could potentially minimize or prevent this complication will have far-reaching benefits to patients with COPD as well as provide significant cost saving.

        How is AECOPD defined?

        COPD exacerbation is defined as a baseline change of the patient’s dyspnea, cough, and/or sputum that is acute in onset, and may warrant a change in regular medication in a patient with underlying COPD.5 Exacerbation in clinical trials has been defined on the basis of whether an increase in the level of care beyond regular care is required primarily in the hospital or ED.6 Frequent exacerbations are defined as 3 symptom-defined exacerbations per year or 2 per year if defined by the need for therapy with corticosteroids, antibiotics, or both.7

        What is the underlying pathophysiology?

        AECOPD is associated with enhanced upper and lower airway and systemic inflammation. The bronchial mucosa of stable COPD patients have increased numbers of CD8+ lymphocytes and macrophages. In mild AECOPD, eosinophils are increased in the bronchial mucosa and modest elevation of neutrophils, T lymphocytes (CD3), and TNF-α positive cells has also been reported.2 With more severe AECOPD, airway neutrophils are increased. Oxidative stress is a key factor in the development of airway inflammation in COPD.1 Patients with severe exacerbations have augmented large airway interleukin-8 (IL-8) levels and increased oxidative stress as demonstrated by markers such as hydrogen peroxide and 8-isoprostane.6

        How do acute exacerbations affect the course of the disease?

        In general, as the severity of the underlying COPD increases, exacerbations become both more severe and more frequent. Patients with frequent exacerbations have a worse quality of life than patients with a history of less frequent exacerbations.8 Frequent exacerbations have also been linked to a decline in lung function, with studies suggesting that there might be a decline of 7 mL in forced expiratory volume in 1 second (FEV1) per lower respiratory tract infection per year,9,10 and approximately 8 mL per year in patients with frequent exacerbations as compared to those with sporadic exacerbations.11

        What are the triggers for COPD exacerbation?

        Respiratory infections are estimated to trigger approximately two-thirds of exacerbations.2 Viral and bacterial infections cause most exacerbations. The effect of the infective triggers is to increase inflammation, cause bronchoconstriction, edema, and mucus production, with a resultant increase in dynamic hyperinflation.12 Thus, any intervention that reduces inflammation in COPD reduces the number and severity of exacerbations, whereas bronchodilators have an impact on exacerbation by their effects on reducing dynamic hyperinflation. The triggers for the one-third of exacerbations not triggered by infection are postulated to be related to other medical conditions, including pulmonary embolism, aspiration, heart failure, and myocardial ischemia.6

         

         

        What are the pharmacologic options available for prevention of AECOPD?

        In recognition of the importance of preventing COPD exacerbations, the American College of Chest Physicians and Canadian Thoracic Society5 have published an evidence-informed clinical guideline specifically examining the prevention of AECOPD, with the goal of assisting clinicians in providing optimal management for COPD patients. The following pharmacologic agents have been recognized as being effective at reducing the frequency of acute exacerbations without any impact on the severity of COPD itself.

        Roflumilast

        Phosphodiesterase 4 (PDE4) inhibition appears to have inflammatory-modulating properties in the airways, although the exact mechanism of action is unclear. Some have proposed that it reduces inflammation by inhibiting the breakdown of intracellular cyclic adenosine monophosphate.13 In 2 large clinical trials,14,15 daily use of a PDE4 inhibitor (roflumilast) showed a significant (15%–18%) reduction in yearly AECOPD incidence (approximate number needed to treat: 4). This benefit was seen in patients with GOLD stage 3–4 disease (FEV1 < 50% predicted) with the chronic bronchitic phenotype and who had experienced at least 1 exacerbation in the previous year.

        Importantly, these clinical trials specifically prohibited the use of inhaled corticosteroids (ICS) and long-acting muscarinic antagonists (LAMAs). Thus, it remains unclear if PDE4 inhibition should be used as an add-on to ICS/LAMA therapy in patients who continue to have frequent AECOPD or whether PDE4 inhibition could be used instead of these standard therapies in patients with well-controlled daily symptoms without ICS or LAMA therapy but who experience frequent exacerbations.

        Of note, earlier trials with roflumilast included patients with ICS and LAMA use.14,16 These trials were focused on FEV1 improvement and found no benefit. It was only in post ad hoc analyses that a reduction in AECOPD in patients with frequent exacerbations was found among those taking roflumilast, regardless of ICS or LAMA use.17 While roflumilast has documented benefit in improving lung function and reducing the rate of exacerbations, it has not been reported to decrease hospitalizations.4 This indicates that although the drug reduces the total number of exacerbations, it may not be as useful in preventing episodes of severe exacerbations of COPD.

        Although PDE4 inhibitors are easy to administer (a once-daily pill), they are associated with significant gastrointestinal side effects (diarrhea, nausea, reduced appetite), weight loss, headache, and sleep disturbance.18 Adverse effects tend to occur early during treatment, are reversible, and lessen over time with treatment.6 Studies reported an average unexplained weight loss of 2 kg, and monitoring weight during treatment is advised. In addition, it is important to avoid roflumilast in underweight patients. Roflumilast should also be used with caution in depressed patients.5

        N-acetylcysteine

        N-acetylcysteine (NAC) reduces the viscosity of respiratory secretions as a result of the cleavage of the disulfide bonds and has been studied as a mucolytic agent to aid in the elimination of respiratory secretions.19 Oral NAC is quickly absorbed and is rapidly present in an active form in lung tissue and respiratory secretions after ingestion. NAC is well-tolerated except for occasional patients with GI adverse effects. The role of NAC in preventing AECOPD has been studied for more than 3 decades,20-22 although the largest clinical trial to date was reported in 2014.23 Taken together, the combined data demonstrate a significant reduction in the rate of COPD exacerbations associated with the use of NAC when compared with placebo (odds ratio [OR], 0.61; 95% confidence interval [CI], 0.37-0.99). Clinical guidelines suggest that in patients with moderate to severe COPD (FEV1/forced vital capacity ratio < 0.7, and FEV1 < 80% predicted) receiving maintenance bronchodilator therapy combined with ICS and history of 2 more exacerbations in the previous 2 years, treatment with oral NAC can be administered to prevent AECOPD.

         

         

        Macrolides

        Continuous prophylactic use of antibiotics in older studies had no effect on the frequency of AECOPD.24,25 But it is known that macrolide antibiotics have several antimicrobial, anti-inflammatory and immunomodulating effects and have been used for many years in the management of other chronic airway disease, including diffuse pan-bronchiolitis and cystic fibrosis.5 One recent study showed that the use of once-daily generic azithromycin 5 days per week appeared to have an impact on AECOPD incidence.26 In this study, the rate of AECOPD was reduced from 1.83 to 1.48 exacerbations per patient-year (relative risk, 0.83; 95% CI, 0.72–0.95; P = 0.01). Azithromycin also prevented severe AECOPD. Greater benefit was obtained with milder forms of the disease and in the elderly. Azithromycin did not appear to provide any benefit in those who continued to smoke (hazard ratio, 0.99).27 Other studies have shown that azithromycin was associated with an increased incidence of bacterial resistance and impaired hearing.28 Overall data from the available clinical trials are robust and demonstrate that regular macrolide therapy definitely reduces the risk of AECOPD. Due to potential adverse effects, however, macrolide therapy is an option rather than a strong recommendation.5 The prescribing clinician also needs to consider potential of prolongation of the QT interval.26

        Immunostimulants

        Immunostimulants have also been reported to reduce frequency of AECOPD.29,30 Bacterial lysates, reconstituted mixtures of bacterial antigens present in the lower airways of COPD patients, act as immunostimulants through the induction of cellular maturation, stimulating lymphocyte chemotaxis and increasing opsonization when administered to individuals with COPD.6 Studies have demonstrated a reduction in the severe complications of exacerbations and hospital admissions in COPD patients with OM-85, a detoxified oral immunoactive bacterial extract.29,30 However, most of these trials were conducted prior to the routine use of long-acting bronchodilators and ICS in COPD. A study that evaluated the efficacy of ismigen, a bacterial lysate, in reducing AECOPD31 found no difference in the exacerbation rate between ismigen and placebo or the time to first exacerbation. Additional studies are needed to examine the long-term effects of this therapy in patients receiving currently recommended COPD maintenance therapy.6

        β-Blockers

        Observational studies of β-blocker use in preventing AECOPD have yielded encouraging results, with one study showing a reduction in AECOPD risk (incidence risk ratio, 0.73; 95% CI, 0.60–0.90) in patients receiving β-blockers versus those not on β-blockers.32 Based on these findings, a clinical trial investigating the impact of metoprolol on risk of AECOPD is ongoing.33

        Proton Pump Inhibitors

        Gastroesophageal reflux disease is an independent risk factor for exacerbations.34 Two small, single-center studies,35,36 have shown that use of lansoprazole decreases the risk and frequency of AECOPD. However, data from the Predicting Outcome using Systemic Markers in Severe Exacerbations of COPD (PROMISE-COPD) study,6 which was a multicenter prospective observational study, suggested that patients with stable COPD receiving a proton pump inhibitor were at high risk of frequent and severe exacerbations.37 Thus, at this stage, their definitive role needs to be defined, possibly with a randomized, placebo-controlled study.

        [embed:render:related:node:204714]

        [embed:render:related:node:204724]

        References

        1. Wedzicha JA, Singh R, Mackay AJ. Acute COPD exacerbations. Clin Chest Med. 2014;35:157-163.

        2. Wedzicha JA, Seemungal TAR. COPD exacerbations: defining their cause and prevention. Lancet. 2007;370:786-796.

        3. Spencer S, Calverley PMA, Burge PS, Jones PW. Impact of preventing exacerbations on deterioration of health status in COPD. Eur Respir J. 2004;23:698-702.

        4. Blanchette CM, Gross NJ, Altman P. Rising costs of COPD and the potential for maintenance therapy to slow the trend. Am Health Drug Benef. 2014;7:98.

        5. Criner GJ, Bourbeau J, Diekemper RL, et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest. 2015;147:894-942.

        6. Vogelmeier CF, Criner GJ, Martinez FJ, et al. Global strategy for the diagnosis, management and prevention of chronic obstructive lung disease 2017 report. Respirology. 2017;22:575-601.

        7. Wedzicha JA, Brill SE, Allinson JP, Donaldson GC. Mechanisms and impact of the frequent exacerbator phenotype in chronic obstructive pulmonary disease. BMC Med. 2013;11:181.

        8. Seemungal TAR, Donaldson GC, Paul EA, et al. Effect of exacerbation on quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1418-1422.

        9. Ries AL, Kaplan RM, Limberg TM, Prewitt LM. Effects of pulmonary rehabilitation on physiologic and psychosocial outcomes in patients with chronic obstructive pulmonary disease. Ann Intern Med. 1995;122:823-832.

        10. Kanner RE, Anthonisen NR, Connett JE. Lower respiratory illnesses promote FEV1 decline in current smokers but not ex-smokers with mild chronic obstructive pulmonary disease: results from the lung health study. Am J Respir Crit Care Med. 2001;164:358-364.

        11. Donaldson GC, Seemungal TAR, Bhowmik A, Wedzicha JA. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57:847-852.

        12. Papi A, Bellettato CM, Braccioni F, et al. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006;173:1114-1121.

        13. Rabe KF. Update on roflumilast, a phosphodiesterase 4 inhibitor for the treatment of chronic obstructive pulmonary disease. Br J Pharmacol. 2011;163:53-67.

        14. Calverley PMA, Rabe KF, Goehring U-M, et al. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet. 2009;374:685-694.

        15. Fabbri LM, Calverley PMA, Izquierdo-Alonso JL, et al. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with long-acting bronchodilators: two randomised clinical trials. Lancet. 2009;374:695-703.

        16. Lee S, Hui DSC, Mahayiddin AA, et al. Roflumilast in Asian patients with COPD: a randomized placebo-controlled trial. Respirology. 2011;16:1249-1257.

        17. Calverley PM, Martinez FJ, Fabbri LM, et al. Does roflumilast decrease exacerbations in severe COPD patients not controlled by inhaled combination therapy? The REACT study protocol. Int J Chron Obstruct Pulmon Dis. 2012;7:375-382.

        18. Chong J, Leung B, Poole P. Phosphodiesterase 4 inhibitors for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2013(11):CD002309.

        19. Sheffner AL, Medler EM, Jacobs LW, Sarett HP. The in vitro reduction in viscosity of human tracheobronchial secretions by acetylcysteine. Am Rev Respir Dis. 1964;90:721-729.

        20. Boman G, Bäcker U, Larsson S, et al. Oral acetylcysteine reduces exacerbation rate in chronic bronchitis: report of a trial organized by the Swedish Society for Pulmonary Diseases. Eur J Respir Dis. 1983;64:405-415.

        21. Grassi C, Morandini GC. A controlled trial of intermittent oral acetylcysteine in the long-term treatment of chronic bronchitis. Eur J Clin Pharmacol. 1976;9:393-396.

        22. Hansen NCG, Skriver A, Brorsen-Riis L, et al. Orally administered N-acetylcysteine may improve general well-being in patients with mild chronic bronchitis. Respir Med. 1994;88:531-535.

        23. Zheng JP, Wen FQ, Bai CX, et al. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): a randomised, double-blind placebo-controlled trial. Lancet Respir Med. 2014;2:187-194.

        24. Francis RS, Spicer CC. Chemotherapy in chronic bronchitis: Influence of daily penicillin and tetracycline on exacerbations and their cost: A report to the research committee of the British Tuberculosis Association by Their Chronic Bronchitis Subcommittee. BMJ. 1960;1:297-303.

        25. Francis RS, May JR, Spicer CC. Chemotherapy of bronchitis. BMJ. 1961;2:979.

        26. Albert RK, Connett J, Bailey WC, et al. Azithromycin for prevention of exacerbations of COPD. N Engl J Med. 2011;365:689-698.

        27. Han MK, Tayob N, Murray S, et al. Predictors of chronic obstructive pulmonary disease exacerbation reduction in response to daily azithromycin therapy. Am J Respir Crit Care Med. 2014;189:1503-1508.

        28. Uzun S, Djamin RS, Kluytmans JAJW, et al. Azithromycin maintenance treatment in patients with frequent exacerbations of chronic obstructive pulmonary disease (COLUMBUS): a randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2014;2:361-368.

        29. Collet JP, Shapiro S, Ernst P, et al. Effects of an immunostimulating agent on acute exacerbations and hospitalizations in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1997;156:1719-1724.

        30. Jing LI. Protective effect of a bacterial extract against acute exacerbation in patients with chronic bronchitis accompanied by chronic obstructive pulmonary. Age. 2004;67:828-834.

        31. Braido F, Tarantini F, Ghiglione V, et al. Bacterial lysate in the prevention of acute exacerbation of COPD and in respiratory recurrent infections. Int J Chron Obstruct Pulmon Dis. 2007;2:335.

        32. Bhatt SP, Wells JM, Kinney GL, et al. β-Blockers are associated with a reduction in COPD exacerbations. Thorax. 2016;71:8-14.

        33. Bhatt SP, Connett JE, Voelker H, et al. β-Blockers for the prevention of acute exacerbations of chronic obstructive pulmonary disease (βLOCK COPD): a randomised controlled study protocol. BMJ Open. 2016;6:e012292.

        34. Hurst JR, Vestbo J, Anzueto A, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363:1128-1138.

        35. Sasaki T, Nakayama K, Yasuda H, et al. A randomized, single-blind study of lansoprazole for the prevention of exacerbations of chronic obstructive pulmonary disease in older patients. J Am Geriatr Soc. 2009;57:1453-1457.

        36. Xiong W, Zhang Qs, Zhao W, et al. A 12-month follow-up study on the preventive effect of oral lansoprazole on acute exacerbation of chronic obstructive pulmonary disease. Int J Exper Pathol. 2016;97:107-113.

        37. Baumeler L, Papakonstantinou E, Milenkovic B, et al. Therapy with proton-pump inhibitors for gastroesophageal reflux disease does not reduce the risk for severe exacerbations in COPD. Respirology. 2016;21:883-890.

        References

        1. Wedzicha JA, Singh R, Mackay AJ. Acute COPD exacerbations. Clin Chest Med. 2014;35:157-163.

        2. Wedzicha JA, Seemungal TAR. COPD exacerbations: defining their cause and prevention. Lancet. 2007;370:786-796.

        3. Spencer S, Calverley PMA, Burge PS, Jones PW. Impact of preventing exacerbations on deterioration of health status in COPD. Eur Respir J. 2004;23:698-702.

        4. Blanchette CM, Gross NJ, Altman P. Rising costs of COPD and the potential for maintenance therapy to slow the trend. Am Health Drug Benef. 2014;7:98.

        5. Criner GJ, Bourbeau J, Diekemper RL, et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest. 2015;147:894-942.

        6. Vogelmeier CF, Criner GJ, Martinez FJ, et al. Global strategy for the diagnosis, management and prevention of chronic obstructive lung disease 2017 report. Respirology. 2017;22:575-601.

        7. Wedzicha JA, Brill SE, Allinson JP, Donaldson GC. Mechanisms and impact of the frequent exacerbator phenotype in chronic obstructive pulmonary disease. BMC Med. 2013;11:181.

        8. Seemungal TAR, Donaldson GC, Paul EA, et al. Effect of exacerbation on quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1418-1422.

        9. Ries AL, Kaplan RM, Limberg TM, Prewitt LM. Effects of pulmonary rehabilitation on physiologic and psychosocial outcomes in patients with chronic obstructive pulmonary disease. Ann Intern Med. 1995;122:823-832.

        10. Kanner RE, Anthonisen NR, Connett JE. Lower respiratory illnesses promote FEV1 decline in current smokers but not ex-smokers with mild chronic obstructive pulmonary disease: results from the lung health study. Am J Respir Crit Care Med. 2001;164:358-364.

        11. Donaldson GC, Seemungal TAR, Bhowmik A, Wedzicha JA. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57:847-852.

        12. Papi A, Bellettato CM, Braccioni F, et al. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006;173:1114-1121.

        13. Rabe KF. Update on roflumilast, a phosphodiesterase 4 inhibitor for the treatment of chronic obstructive pulmonary disease. Br J Pharmacol. 2011;163:53-67.

        14. Calverley PMA, Rabe KF, Goehring U-M, et al. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet. 2009;374:685-694.

        15. Fabbri LM, Calverley PMA, Izquierdo-Alonso JL, et al. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with long-acting bronchodilators: two randomised clinical trials. Lancet. 2009;374:695-703.

        16. Lee S, Hui DSC, Mahayiddin AA, et al. Roflumilast in Asian patients with COPD: a randomized placebo-controlled trial. Respirology. 2011;16:1249-1257.

        17. Calverley PM, Martinez FJ, Fabbri LM, et al. Does roflumilast decrease exacerbations in severe COPD patients not controlled by inhaled combination therapy? The REACT study protocol. Int J Chron Obstruct Pulmon Dis. 2012;7:375-382.

        18. Chong J, Leung B, Poole P. Phosphodiesterase 4 inhibitors for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2013(11):CD002309.

        19. Sheffner AL, Medler EM, Jacobs LW, Sarett HP. The in vitro reduction in viscosity of human tracheobronchial secretions by acetylcysteine. Am Rev Respir Dis. 1964;90:721-729.

        20. Boman G, Bäcker U, Larsson S, et al. Oral acetylcysteine reduces exacerbation rate in chronic bronchitis: report of a trial organized by the Swedish Society for Pulmonary Diseases. Eur J Respir Dis. 1983;64:405-415.

        21. Grassi C, Morandini GC. A controlled trial of intermittent oral acetylcysteine in the long-term treatment of chronic bronchitis. Eur J Clin Pharmacol. 1976;9:393-396.

        22. Hansen NCG, Skriver A, Brorsen-Riis L, et al. Orally administered N-acetylcysteine may improve general well-being in patients with mild chronic bronchitis. Respir Med. 1994;88:531-535.

        23. Zheng JP, Wen FQ, Bai CX, et al. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): a randomised, double-blind placebo-controlled trial. Lancet Respir Med. 2014;2:187-194.

        24. Francis RS, Spicer CC. Chemotherapy in chronic bronchitis: Influence of daily penicillin and tetracycline on exacerbations and their cost: A report to the research committee of the British Tuberculosis Association by Their Chronic Bronchitis Subcommittee. BMJ. 1960;1:297-303.

        25. Francis RS, May JR, Spicer CC. Chemotherapy of bronchitis. BMJ. 1961;2:979.

        26. Albert RK, Connett J, Bailey WC, et al. Azithromycin for prevention of exacerbations of COPD. N Engl J Med. 2011;365:689-698.

        27. Han MK, Tayob N, Murray S, et al. Predictors of chronic obstructive pulmonary disease exacerbation reduction in response to daily azithromycin therapy. Am J Respir Crit Care Med. 2014;189:1503-1508.

        28. Uzun S, Djamin RS, Kluytmans JAJW, et al. Azithromycin maintenance treatment in patients with frequent exacerbations of chronic obstructive pulmonary disease (COLUMBUS): a randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2014;2:361-368.

        29. Collet JP, Shapiro S, Ernst P, et al. Effects of an immunostimulating agent on acute exacerbations and hospitalizations in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1997;156:1719-1724.

        30. Jing LI. Protective effect of a bacterial extract against acute exacerbation in patients with chronic bronchitis accompanied by chronic obstructive pulmonary. Age. 2004;67:828-834.

        31. Braido F, Tarantini F, Ghiglione V, et al. Bacterial lysate in the prevention of acute exacerbation of COPD and in respiratory recurrent infections. Int J Chron Obstruct Pulmon Dis. 2007;2:335.

        32. Bhatt SP, Wells JM, Kinney GL, et al. β-Blockers are associated with a reduction in COPD exacerbations. Thorax. 2016;71:8-14.

        33. Bhatt SP, Connett JE, Voelker H, et al. β-Blockers for the prevention of acute exacerbations of chronic obstructive pulmonary disease (βLOCK COPD): a randomised controlled study protocol. BMJ Open. 2016;6:e012292.

        34. Hurst JR, Vestbo J, Anzueto A, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363:1128-1138.

        35. Sasaki T, Nakayama K, Yasuda H, et al. A randomized, single-blind study of lansoprazole for the prevention of exacerbations of chronic obstructive pulmonary disease in older patients. J Am Geriatr Soc. 2009;57:1453-1457.

        36. Xiong W, Zhang Qs, Zhao W, et al. A 12-month follow-up study on the preventive effect of oral lansoprazole on acute exacerbation of chronic obstructive pulmonary disease. Int J Exper Pathol. 2016;97:107-113.

        37. Baumeler L, Papakonstantinou E, Milenkovic B, et al. Therapy with proton-pump inhibitors for gastroesophageal reflux disease does not reduce the risk for severe exacerbations in COPD. Respirology. 2016;21:883-890.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Stable COPD: Managing Acute Exacerbations
        Display Headline
        Stable COPD: Managing Acute Exacerbations
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 July;16(1)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Tue, 07/16/2019 - 11:00
        Un-Gate On Date
        Tue, 07/16/2019 - 11:00
        Use ProPublica
        CFC Schedule Remove Status
        Tue, 07/16/2019 - 11:00
        Hide sidebar & use full width
        render the right sidebar.

        Stable COPD: Managing Advanced Disease

        Article Type
        Changed
        Wed, 07/17/2019 - 14:46
        Display Headline
        Stable COPD: Managing Advanced Disease

        Case Presentation

        A 65-year-old man with severe chronic obstructive disease (COPD; forced expiratory volume in 1 second/forced vital capacity ratio [FEV1/FVC], 27; FEV1 25% of predicted; residual volume 170% of predicted for his age and height) was seen in the pulmonary clinic. His medications include a long-acting beta agonist (LABA)/long-acting muscarinic antagonist (LAMA) combination that he uses twice daily as advised. He uses his rescue albuterol inhaler roughly once a week. The patient complains of severe disabling shortness of breath with exertion and severe limitation of his quality of life because of his inability to lead a normal active life. He is on 2 L/min of oxygen at all times. He has received pulmonary rehabilitation in hopes of improving his quality of life but can only climb a flight of stairs before he must stop to rest. He asks about options but does not want to consider lung transplantation today. His most recent chest computed tomography (CT) scan demonstrates upper lobe predominant emphysematous changes with no masses or nodules.

        What are the patient's options at this time?

        Lung volume reduction surgery (LVRS) attempts to reduce space-occupying severely diseased, hyperexpanded lung, thus allowing the relatively normal adjoining lung parenchyma to expand into the vacated space and function effectively.1 Hence, such therapies are suitable for patients with emphysematous lungs and not those with bronchitic-predominant COPD. LVRS offers a greater chance of improvement in exercise capacity, lung function, quality of life, and dyspnea in the correctly chosen patient population, as compared with pharmacologic management alone.2 However, the procedure is associated with risks, including higher short-term morbidity and mortality.2 Patients with predominantly upper-lobe emphysema and a low maximal workload after rehabilitation were noted to have lower mortality, a greater probability of improvement in exercise capacity, and a greater probability of improvement in symptoms if they underwent surgery compared to medical therapy alone.2 On the contrary, patients with predominantly non–upper-lobe emphysema and a high maximal workload after rehabilitation had higher mortality if they underwent surgery compared to receiving medical therapy alone.2 Thus, a subgroup of patients with homogeneous emphysema symmetrically affecting the upper and lower lobes are considered to be unlikely to benefit from this surgery.2,3

        Valves and other methods of lung volume reduction such as coils, sealants, intrapulmonary vents, and thermal vapor in the bronchi or subsegmental airways have emerged as new techniques for nonsurgical lung volume reduction.4-9 Endobronchial-valve therapy is associated with improvement in lung function and with clinical benefits that are greatest in the presence of heterogeneous lung involvement. This works by the same principle as LVRS, by reduction of the most severely diseased lung units, expansion of the more viable, less emphysematous lung results in substantial improvements in lung mechanics.10,11 The most important complications of this procedure include pneumonia, pneumothorax, hemoptysis, and increased frequency of COPD exacerbation in the following 30 days. The fact that a high-heterogeneity subgroup had greater improvements in both the FEV1 and distance on the 6-minute walk test than did patients with lower heterogeneity supports the use of quantitative high-resolution computed tomography (HRCT) in selecting patients for endobronchial-valve therapy.12 The HRCT scans also help in identifying those with complete fissures, a marker of lack of collateral ventilation (CV+) between different lobes. Presence of CV+ state predicts failure of endobronchial valve and all forms of endoscopic LVRS.13 Bronchoscopic thermal vapor ablation (BTVA) therapy can potentially work on a subsegmental level and be successful for treatment of emphysema with lack of intact fissures on CT scans. Other methods that have the potential to be effective in those with collateral ventilation would be endoscopic coil therapy and polymeric lung volume reduction.11,14 Unfortunately, there are no randomized controlled trial data demonstrating clinically meaningful improvement following coil therapy or polymeric lung volume reduction in this CV+ patient population. Vapor therapy is perhaps the only technique that has been found to be effective in upper lobe predominant emphysema even with CV+ status.13

        Our patient has evidence of air trapping and emphysema based on a high residual volume. A CT scan of the chest can determine the nature of the emphysema (heterogeneous versus homogenous) and based on these findings, further determination of the best strategy for lung volume reduction can be made.

        Is there a role for long-term oxygen therapy?

        Long-term oxygen therapy (LTOT) used for more than 15 hours a day is thought to reduce mortality among patients with COPD and severe resting hypoxemia.15-18 More recent studies have failed to show similar beneficial effects of LTOT. A recent study examined the effects of LTOT in randomized fashion and determined that supplemental oxygen for patients with stable COPD and resting or exercise-induced moderate desaturation did not affect the time to death or first hospitalization, time to first COPD exacerbation, time to first hospitalization for a COPD exacerbation, the rate of all hospitalizations, the rate of all COPD exacerbations, or changes in measures of quality of life, depression, anxiety, or functional status.19

        Our patient is currently on long-term oxygen therapy and in spite of some uncertainty as to its benefit, it is prudent to order oxygen therapy until further clarification is available.

        What is the role of pulmonary rehabilitation?

        Pulmonary rehabilitation is an established treatment for patients with chronic lung disease.20 Benefits include improvement in exercise tolerance, symptoms, and quality of life, with a reduction in the use of health care resources.21 A Spanish population-based cohort study that looked at the influence of regular physical activity on COPD showed that patients who reported low, moderate, or high physical activity had a lower risk of COPD admissions and all-cause mortality than patients with very low physical activity after adjusting for all confounders.22

         

         

        As previously mentioned, patients in GOLD categories B, C, and D should be offered pulmonary rehabilitation as part of their treatment.23 The ideal patient is one who is not too sick to undergo rehabilitation and is motivated to improve his or her quality of life.

        What is the current scope of lung transplantation in the management of severe COPD?

        There is an indisputable role for lung transplantation in end-stage COPD. However, lung transplantation does not benefit all COPD patients. There is a subset of patients for whom the treatment provides a survival benefit. It has been reported that 79% of patients with an FEV1 < 16% predicted will survive at least 1 additional year after transplant, but only 11% of patients with an FEV1 > 25% will do so.24 The pre-transplant BODE (body mass index, airflow obstruction/FEV1, dyspnea, and exercise capacity) index score is used to identify patients who will benefit from lung transplantation.25,26 International guidelines for the selection of lung transplant candidates identify the following patient characteristics:27

        • The disease is progressive, despite maximal treatment including medication, pulmonary rehabilitation, and oxygen therapy;
        • The patient is not a candidate for endoscopic or surgical LVRS;
        • BODE index is 5 to 6;
        • The PCO2 is greater than 50 mm Hg (6.6 kPa) and/or PO2 is less than 60 mm Hg (8 kPa);
        • FEV1 is 25% predicted.

        The perioperative mortality of lung transplantation surgery has been reduced to less than 10%. Risk of complications from surgery in the perioperative period, such as bronchial dehiscence, infectious complications, and acute rejection, have also been reduced but do occur. Chronic allograft dysfunction and the risk of lung cancer in cases of single lung transplant should be discussed with the patient before surgery.28

        How can we incorporate palliative care into the management plan for patients with COPD?

        Among patients with end-stage COPD on home oxygen therapy who have required mechanical ventilation for an exacerbation, only 55% are alive at 1 year.29 COPD patients at high risk of death within the next year of life as well as patients with refractory symptoms and unmet needs are candidates for early palliative care. Palliative care and palliative care specialists can aid in reducing symptom burden and improving quality of life among these patients and their family members, and palliative care is recommended by multiple international societies for patients with advanced COPD.30,31 In spite of these recommendations, the utilization of palliative care resources has been dismally low.32,33 Improving physician-patient communication regarding palliative services and patients’ unmet care needs will help ensure that COPD patients receive adequate palliative care services at the appropriate time.

        Conclusion

        COPD is a leading cause of morbidity and mortality in the United States and represents a significant economic burden for both individuals and society. The goals in COPD management are to provide symptom relief, improve the quality of life, preserve lung function, and reduce the frequency of exacerbations and mortality. COPD management is guided by disease severity that is measured using the GOLD multimodal staging system and requires a multidisciplinary approach. Several classes of medication are available for treatment, and a step-wise approach should be applied in building an effective pharmacologic regimen. In addition to pharmacologic therapies, nonpharmacologic therapies, including smoking cessation, vaccinations, proper nutrition, and maintaining physical activity, are an important part of long-term management. Those who continue to be symptomatic despite appropriate maximal therapy may be candidates for lung volume reduction. Palliative care services for COPD patients, which can aid in reducing symptom burden and improving quality of life, should not be overlooked.

        [embed:render:related:node:204714]

        [embed:render:related:node:204720]

        References

        1. Sabanathan A, Sabanathan S, Shah R, Richardson J. Lung volume reduction surgery for emphysema: a review. J Cardiovasc Surg. 1998;39:237.

        2. Group NETTR. Patients at high risk of death after lung-volume–reduction surgery. N Engl J Med. 2001;345:1075-1083.

        3. Group NETTR. A randomized trial comparing lung-volume–reduction surgery with medical therapy for severe emphysema. N Engl J Med. 2003;348:2059-2073.

        4. Decker MR, Leverson GE, Jaoude WA, Maloney JD. Lung volume reduction surgery since the National Emphysema Treatment Trial: study of Society of Thoracic Surgeons database. J Thorac Cardiovasc Surg. 2014;148:2651-2658.

        5. Deslée G, Mal H, Dutau H, et al. Lung volume reduction coil treatment vs usual care in patients with severe emphysema: the REVOLENS randomized clinical trial. JAMA. 2016;315:175-184.

        6. Hartman JE, Klooster K, Gortzak K, et al. Long-term follow-up after bronchoscopic lung volume reduction treatment with coils in patients with severe emphysema. Respirology. 2015;20:319-326.

        7. Snell GI, Hopkins P, Westall G, et al. A feasibility and safety study of bronchoscopic thermal vapor ablation: a novel emphysema therapy. Ann Thorac Surg. 2009;88:1993-1998.

        8. Ingenito EP, Berger RL, Henderson AC, et al. Bronchoscopic lung volume reduction using tissue engineering principles. Am J Respir Crit Care Med. 2003;167:771-778.

        9. Ingenito EP, Loring SH, Moy ML, et al. Comparison of physiological and radiological screening for lung volume reduction surgery. Am J Respir Crit Care Med. 2001;163:1068-1073.

        10. Shah P, Slebos D, Cardoso P, et al. Bronchoscopic lung-volume reduction with Exhale airway stents for emphysema (EASE trial): randomised, sham-controlled, multicentre trial. Lancet. 2011;378:997-1005.

        11. Sciurba FC, Ernst A, Herth FJ, et al. A randomized study of endobronchial valves for advanced emphysema. N Engl J Med. 2010;363:1233-1244.

        12. Wan IY, Toma TP, Geddes DM, et al. Bronchoscopic lung volume reduction for end-stage emphysema: report on the first 98 patients. Chest. 2006;129:518-526.

        13. Gompelmann D, Eberhardt R, Schuhmann M, et al. Lung volume reduction with vapor ablation in the presence of incomplete fissures: 12-month results from the STEP-UP randomized controlled study. Respiration. 2016;92:397-403.

        14. Come CE, Kramer MR, Dransfield MT, et al. A randomised trial of lung sealant versus medical therapy for advanced emphysema. Eur Respir J. 2015;46:651-662.

        15. Group NOTT. Continuous or nocturnal oxygen therapy in hypoxemic chronic obstructive lung disease: a clinical trial. Ann Intern Med. 1980;93:391-398.

        16. Council M. Long term domiciliary oxygen therapy in chronic hypoxic cor pulmonale complicating chronic bronchitis and emphysema: Report of the Medical Research Council Working Party. Lancet. 1981;1:681-686.

        17. Qaseem A, Wilt TJ, Weinberger SE, et al. Diagnosis and management of stable chronic obstructive pulmonary disease: a clinical practice guideline update from the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society. Ann Intern Med. 2011;155:179-191.

        18. Vestbo J, Hurd SS, Agustí AG, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187:347-365.

        19. Group L-TOTTR. A randomized trial of long-term oxygen for COPD with moderate desaturation. N Engl J Med. 2016;375:1617-1627.

        20. McCarthy B, Casey D, Devane D, et al. Pulmonary rehabilitation for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015(2):CD003793.

        21. Griffiths TL, Burr ML, Campbell IA, et al. Results at 1 year of outpatient multidisciplinary pulmonary rehabilitation: a randomised controlled trial. Lancet. 2000;355:362-368.

        22. Garcia-Aymerich J, Lange P, Benet M, et al. Regular physical activity reduces hospital admission and mortality in chronic obstructive pulmonary disease: a population based cohort study. Thorax. 2006;61:772-778.

        23. Global Initiative for Chronic Obstructive Lung Disease (GOLD): Global strategy for the diagnosis, management, and prevention of COPD 2017. www.goldcopd.org. Accessed July 9, 2019.

        24. Thabut G, Ravaud P, Christie JD, et al. Determinants of the survival benefit of lung transplantation in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;177:1156-1163.

        25. Lahzami S, Bridevaux PO, Soccal PM, et al. Survival impact of lung transplantation for COPD. Eur Respir J. 2010;36:74-80.

        26. Cerón Navarro J, de Aguiar Quevedo K, Ansótegui Barrera E, et al. Functional outcomes after lung transplant in chronic obstructive pulmonary disease. Arch Bronconeumol. 2015;51:109-114.

        27. Weill D, Benden C, Corris PA, et al. A consensus document for the selection of lung transplant candidates: 2014--an update from the Pulmonary Transplantation Council of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant. 2015;34:1-15.

        28. Minai OA, Shah S, Mazzone P, et al. Bronchogenic carcinoma after lung transplantation: characteristics and outcomes. J Thorac Oncol. 2008;3:1404-1409.

        29. Hajizadeh N, Goldfeld K, Crothers K. What happens to patients with COPD with long-term oxygen treatment who receive mechanical ventilation for COPD exacerbation? A 1-year retrospective follow- up study. Thorax. 2015;70:294-296.

        30. Siouta N, van Beek K, Preston N, et al. Towards integration of palliative care in patients with chronic heart failure and chronic obstructive pulmonary disease: a systematic literature review of European guidelines and pathways. BMC Palliat Care. 2016;15:18.

        31. Celli BR, MacNee W; ATS/ERS Task Force. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper. Eur Respir J. 2004;23:932-946.

        32. Szekendi MK, Vaughn J, Lal A, et al. The prevalence of inpatients at thirty-three U.S. hospitals appropriate for and receiving referral to palliative care. J Palliat Med. 2016;19:360-372.

        33. Rush B, Hertz P, Bond A, et al. Use of palliative care in patients with end-stage COPD and receiving home oxygen: national trends and barriers to care in the United States. Chest. 2017;151:41-46.

        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Publications
        Topics
        Sections
        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Author and Disclosure Information

        From the University of Florida, Gainesville, FL (Dr. Aljaafareh and Dr. Fakih), and Parkview Regional Medical Center, Fort Wayne, IN (Dr. Biswas).

        Case Presentation

        A 65-year-old man with severe chronic obstructive disease (COPD; forced expiratory volume in 1 second/forced vital capacity ratio [FEV1/FVC], 27; FEV1 25% of predicted; residual volume 170% of predicted for his age and height) was seen in the pulmonary clinic. His medications include a long-acting beta agonist (LABA)/long-acting muscarinic antagonist (LAMA) combination that he uses twice daily as advised. He uses his rescue albuterol inhaler roughly once a week. The patient complains of severe disabling shortness of breath with exertion and severe limitation of his quality of life because of his inability to lead a normal active life. He is on 2 L/min of oxygen at all times. He has received pulmonary rehabilitation in hopes of improving his quality of life but can only climb a flight of stairs before he must stop to rest. He asks about options but does not want to consider lung transplantation today. His most recent chest computed tomography (CT) scan demonstrates upper lobe predominant emphysematous changes with no masses or nodules.

        What are the patient's options at this time?

        Lung volume reduction surgery (LVRS) attempts to reduce space-occupying severely diseased, hyperexpanded lung, thus allowing the relatively normal adjoining lung parenchyma to expand into the vacated space and function effectively.1 Hence, such therapies are suitable for patients with emphysematous lungs and not those with bronchitic-predominant COPD. LVRS offers a greater chance of improvement in exercise capacity, lung function, quality of life, and dyspnea in the correctly chosen patient population, as compared with pharmacologic management alone.2 However, the procedure is associated with risks, including higher short-term morbidity and mortality.2 Patients with predominantly upper-lobe emphysema and a low maximal workload after rehabilitation were noted to have lower mortality, a greater probability of improvement in exercise capacity, and a greater probability of improvement in symptoms if they underwent surgery compared to medical therapy alone.2 On the contrary, patients with predominantly non–upper-lobe emphysema and a high maximal workload after rehabilitation had higher mortality if they underwent surgery compared to receiving medical therapy alone.2 Thus, a subgroup of patients with homogeneous emphysema symmetrically affecting the upper and lower lobes are considered to be unlikely to benefit from this surgery.2,3

        Valves and other methods of lung volume reduction such as coils, sealants, intrapulmonary vents, and thermal vapor in the bronchi or subsegmental airways have emerged as new techniques for nonsurgical lung volume reduction.4-9 Endobronchial-valve therapy is associated with improvement in lung function and with clinical benefits that are greatest in the presence of heterogeneous lung involvement. This works by the same principle as LVRS, by reduction of the most severely diseased lung units, expansion of the more viable, less emphysematous lung results in substantial improvements in lung mechanics.10,11 The most important complications of this procedure include pneumonia, pneumothorax, hemoptysis, and increased frequency of COPD exacerbation in the following 30 days. The fact that a high-heterogeneity subgroup had greater improvements in both the FEV1 and distance on the 6-minute walk test than did patients with lower heterogeneity supports the use of quantitative high-resolution computed tomography (HRCT) in selecting patients for endobronchial-valve therapy.12 The HRCT scans also help in identifying those with complete fissures, a marker of lack of collateral ventilation (CV+) between different lobes. Presence of CV+ state predicts failure of endobronchial valve and all forms of endoscopic LVRS.13 Bronchoscopic thermal vapor ablation (BTVA) therapy can potentially work on a subsegmental level and be successful for treatment of emphysema with lack of intact fissures on CT scans. Other methods that have the potential to be effective in those with collateral ventilation would be endoscopic coil therapy and polymeric lung volume reduction.11,14 Unfortunately, there are no randomized controlled trial data demonstrating clinically meaningful improvement following coil therapy or polymeric lung volume reduction in this CV+ patient population. Vapor therapy is perhaps the only technique that has been found to be effective in upper lobe predominant emphysema even with CV+ status.13

        Our patient has evidence of air trapping and emphysema based on a high residual volume. A CT scan of the chest can determine the nature of the emphysema (heterogeneous versus homogenous) and based on these findings, further determination of the best strategy for lung volume reduction can be made.

        Is there a role for long-term oxygen therapy?

        Long-term oxygen therapy (LTOT) used for more than 15 hours a day is thought to reduce mortality among patients with COPD and severe resting hypoxemia.15-18 More recent studies have failed to show similar beneficial effects of LTOT. A recent study examined the effects of LTOT in randomized fashion and determined that supplemental oxygen for patients with stable COPD and resting or exercise-induced moderate desaturation did not affect the time to death or first hospitalization, time to first COPD exacerbation, time to first hospitalization for a COPD exacerbation, the rate of all hospitalizations, the rate of all COPD exacerbations, or changes in measures of quality of life, depression, anxiety, or functional status.19

        Our patient is currently on long-term oxygen therapy and in spite of some uncertainty as to its benefit, it is prudent to order oxygen therapy until further clarification is available.

        What is the role of pulmonary rehabilitation?

        Pulmonary rehabilitation is an established treatment for patients with chronic lung disease.20 Benefits include improvement in exercise tolerance, symptoms, and quality of life, with a reduction in the use of health care resources.21 A Spanish population-based cohort study that looked at the influence of regular physical activity on COPD showed that patients who reported low, moderate, or high physical activity had a lower risk of COPD admissions and all-cause mortality than patients with very low physical activity after adjusting for all confounders.22

         

         

        As previously mentioned, patients in GOLD categories B, C, and D should be offered pulmonary rehabilitation as part of their treatment.23 The ideal patient is one who is not too sick to undergo rehabilitation and is motivated to improve his or her quality of life.

        What is the current scope of lung transplantation in the management of severe COPD?

        There is an indisputable role for lung transplantation in end-stage COPD. However, lung transplantation does not benefit all COPD patients. There is a subset of patients for whom the treatment provides a survival benefit. It has been reported that 79% of patients with an FEV1 < 16% predicted will survive at least 1 additional year after transplant, but only 11% of patients with an FEV1 > 25% will do so.24 The pre-transplant BODE (body mass index, airflow obstruction/FEV1, dyspnea, and exercise capacity) index score is used to identify patients who will benefit from lung transplantation.25,26 International guidelines for the selection of lung transplant candidates identify the following patient characteristics:27

        • The disease is progressive, despite maximal treatment including medication, pulmonary rehabilitation, and oxygen therapy;
        • The patient is not a candidate for endoscopic or surgical LVRS;
        • BODE index is 5 to 6;
        • The PCO2 is greater than 50 mm Hg (6.6 kPa) and/or PO2 is less than 60 mm Hg (8 kPa);
        • FEV1 is 25% predicted.

        The perioperative mortality of lung transplantation surgery has been reduced to less than 10%. Risk of complications from surgery in the perioperative period, such as bronchial dehiscence, infectious complications, and acute rejection, have also been reduced but do occur. Chronic allograft dysfunction and the risk of lung cancer in cases of single lung transplant should be discussed with the patient before surgery.28

        How can we incorporate palliative care into the management plan for patients with COPD?

        Among patients with end-stage COPD on home oxygen therapy who have required mechanical ventilation for an exacerbation, only 55% are alive at 1 year.29 COPD patients at high risk of death within the next year of life as well as patients with refractory symptoms and unmet needs are candidates for early palliative care. Palliative care and palliative care specialists can aid in reducing symptom burden and improving quality of life among these patients and their family members, and palliative care is recommended by multiple international societies for patients with advanced COPD.30,31 In spite of these recommendations, the utilization of palliative care resources has been dismally low.32,33 Improving physician-patient communication regarding palliative services and patients’ unmet care needs will help ensure that COPD patients receive adequate palliative care services at the appropriate time.

        Conclusion

        COPD is a leading cause of morbidity and mortality in the United States and represents a significant economic burden for both individuals and society. The goals in COPD management are to provide symptom relief, improve the quality of life, preserve lung function, and reduce the frequency of exacerbations and mortality. COPD management is guided by disease severity that is measured using the GOLD multimodal staging system and requires a multidisciplinary approach. Several classes of medication are available for treatment, and a step-wise approach should be applied in building an effective pharmacologic regimen. In addition to pharmacologic therapies, nonpharmacologic therapies, including smoking cessation, vaccinations, proper nutrition, and maintaining physical activity, are an important part of long-term management. Those who continue to be symptomatic despite appropriate maximal therapy may be candidates for lung volume reduction. Palliative care services for COPD patients, which can aid in reducing symptom burden and improving quality of life, should not be overlooked.

        [embed:render:related:node:204714]

        [embed:render:related:node:204720]

        Case Presentation

        A 65-year-old man with severe chronic obstructive disease (COPD; forced expiratory volume in 1 second/forced vital capacity ratio [FEV1/FVC], 27; FEV1 25% of predicted; residual volume 170% of predicted for his age and height) was seen in the pulmonary clinic. His medications include a long-acting beta agonist (LABA)/long-acting muscarinic antagonist (LAMA) combination that he uses twice daily as advised. He uses his rescue albuterol inhaler roughly once a week. The patient complains of severe disabling shortness of breath with exertion and severe limitation of his quality of life because of his inability to lead a normal active life. He is on 2 L/min of oxygen at all times. He has received pulmonary rehabilitation in hopes of improving his quality of life but can only climb a flight of stairs before he must stop to rest. He asks about options but does not want to consider lung transplantation today. His most recent chest computed tomography (CT) scan demonstrates upper lobe predominant emphysematous changes with no masses or nodules.

        What are the patient's options at this time?

        Lung volume reduction surgery (LVRS) attempts to reduce space-occupying severely diseased, hyperexpanded lung, thus allowing the relatively normal adjoining lung parenchyma to expand into the vacated space and function effectively.1 Hence, such therapies are suitable for patients with emphysematous lungs and not those with bronchitic-predominant COPD. LVRS offers a greater chance of improvement in exercise capacity, lung function, quality of life, and dyspnea in the correctly chosen patient population, as compared with pharmacologic management alone.2 However, the procedure is associated with risks, including higher short-term morbidity and mortality.2 Patients with predominantly upper-lobe emphysema and a low maximal workload after rehabilitation were noted to have lower mortality, a greater probability of improvement in exercise capacity, and a greater probability of improvement in symptoms if they underwent surgery compared to medical therapy alone.2 On the contrary, patients with predominantly non–upper-lobe emphysema and a high maximal workload after rehabilitation had higher mortality if they underwent surgery compared to receiving medical therapy alone.2 Thus, a subgroup of patients with homogeneous emphysema symmetrically affecting the upper and lower lobes are considered to be unlikely to benefit from this surgery.2,3

        Valves and other methods of lung volume reduction such as coils, sealants, intrapulmonary vents, and thermal vapor in the bronchi or subsegmental airways have emerged as new techniques for nonsurgical lung volume reduction.4-9 Endobronchial-valve therapy is associated with improvement in lung function and with clinical benefits that are greatest in the presence of heterogeneous lung involvement. This works by the same principle as LVRS, by reduction of the most severely diseased lung units, expansion of the more viable, less emphysematous lung results in substantial improvements in lung mechanics.10,11 The most important complications of this procedure include pneumonia, pneumothorax, hemoptysis, and increased frequency of COPD exacerbation in the following 30 days. The fact that a high-heterogeneity subgroup had greater improvements in both the FEV1 and distance on the 6-minute walk test than did patients with lower heterogeneity supports the use of quantitative high-resolution computed tomography (HRCT) in selecting patients for endobronchial-valve therapy.12 The HRCT scans also help in identifying those with complete fissures, a marker of lack of collateral ventilation (CV+) between different lobes. Presence of CV+ state predicts failure of endobronchial valve and all forms of endoscopic LVRS.13 Bronchoscopic thermal vapor ablation (BTVA) therapy can potentially work on a subsegmental level and be successful for treatment of emphysema with lack of intact fissures on CT scans. Other methods that have the potential to be effective in those with collateral ventilation would be endoscopic coil therapy and polymeric lung volume reduction.11,14 Unfortunately, there are no randomized controlled trial data demonstrating clinically meaningful improvement following coil therapy or polymeric lung volume reduction in this CV+ patient population. Vapor therapy is perhaps the only technique that has been found to be effective in upper lobe predominant emphysema even with CV+ status.13

        Our patient has evidence of air trapping and emphysema based on a high residual volume. A CT scan of the chest can determine the nature of the emphysema (heterogeneous versus homogenous) and based on these findings, further determination of the best strategy for lung volume reduction can be made.

        Is there a role for long-term oxygen therapy?

        Long-term oxygen therapy (LTOT) used for more than 15 hours a day is thought to reduce mortality among patients with COPD and severe resting hypoxemia.15-18 More recent studies have failed to show similar beneficial effects of LTOT. A recent study examined the effects of LTOT in randomized fashion and determined that supplemental oxygen for patients with stable COPD and resting or exercise-induced moderate desaturation did not affect the time to death or first hospitalization, time to first COPD exacerbation, time to first hospitalization for a COPD exacerbation, the rate of all hospitalizations, the rate of all COPD exacerbations, or changes in measures of quality of life, depression, anxiety, or functional status.19

        Our patient is currently on long-term oxygen therapy and in spite of some uncertainty as to its benefit, it is prudent to order oxygen therapy until further clarification is available.

        What is the role of pulmonary rehabilitation?

        Pulmonary rehabilitation is an established treatment for patients with chronic lung disease.20 Benefits include improvement in exercise tolerance, symptoms, and quality of life, with a reduction in the use of health care resources.21 A Spanish population-based cohort study that looked at the influence of regular physical activity on COPD showed that patients who reported low, moderate, or high physical activity had a lower risk of COPD admissions and all-cause mortality than patients with very low physical activity after adjusting for all confounders.22

         

         

        As previously mentioned, patients in GOLD categories B, C, and D should be offered pulmonary rehabilitation as part of their treatment.23 The ideal patient is one who is not too sick to undergo rehabilitation and is motivated to improve his or her quality of life.

        What is the current scope of lung transplantation in the management of severe COPD?

        There is an indisputable role for lung transplantation in end-stage COPD. However, lung transplantation does not benefit all COPD patients. There is a subset of patients for whom the treatment provides a survival benefit. It has been reported that 79% of patients with an FEV1 < 16% predicted will survive at least 1 additional year after transplant, but only 11% of patients with an FEV1 > 25% will do so.24 The pre-transplant BODE (body mass index, airflow obstruction/FEV1, dyspnea, and exercise capacity) index score is used to identify patients who will benefit from lung transplantation.25,26 International guidelines for the selection of lung transplant candidates identify the following patient characteristics:27

        • The disease is progressive, despite maximal treatment including medication, pulmonary rehabilitation, and oxygen therapy;
        • The patient is not a candidate for endoscopic or surgical LVRS;
        • BODE index is 5 to 6;
        • The PCO2 is greater than 50 mm Hg (6.6 kPa) and/or PO2 is less than 60 mm Hg (8 kPa);
        • FEV1 is 25% predicted.

        The perioperative mortality of lung transplantation surgery has been reduced to less than 10%. Risk of complications from surgery in the perioperative period, such as bronchial dehiscence, infectious complications, and acute rejection, have also been reduced but do occur. Chronic allograft dysfunction and the risk of lung cancer in cases of single lung transplant should be discussed with the patient before surgery.28

        How can we incorporate palliative care into the management plan for patients with COPD?

        Among patients with end-stage COPD on home oxygen therapy who have required mechanical ventilation for an exacerbation, only 55% are alive at 1 year.29 COPD patients at high risk of death within the next year of life as well as patients with refractory symptoms and unmet needs are candidates for early palliative care. Palliative care and palliative care specialists can aid in reducing symptom burden and improving quality of life among these patients and their family members, and palliative care is recommended by multiple international societies for patients with advanced COPD.30,31 In spite of these recommendations, the utilization of palliative care resources has been dismally low.32,33 Improving physician-patient communication regarding palliative services and patients’ unmet care needs will help ensure that COPD patients receive adequate palliative care services at the appropriate time.

        Conclusion

        COPD is a leading cause of morbidity and mortality in the United States and represents a significant economic burden for both individuals and society. The goals in COPD management are to provide symptom relief, improve the quality of life, preserve lung function, and reduce the frequency of exacerbations and mortality. COPD management is guided by disease severity that is measured using the GOLD multimodal staging system and requires a multidisciplinary approach. Several classes of medication are available for treatment, and a step-wise approach should be applied in building an effective pharmacologic regimen. In addition to pharmacologic therapies, nonpharmacologic therapies, including smoking cessation, vaccinations, proper nutrition, and maintaining physical activity, are an important part of long-term management. Those who continue to be symptomatic despite appropriate maximal therapy may be candidates for lung volume reduction. Palliative care services for COPD patients, which can aid in reducing symptom burden and improving quality of life, should not be overlooked.

        [embed:render:related:node:204714]

        [embed:render:related:node:204720]

        References

        1. Sabanathan A, Sabanathan S, Shah R, Richardson J. Lung volume reduction surgery for emphysema: a review. J Cardiovasc Surg. 1998;39:237.

        2. Group NETTR. Patients at high risk of death after lung-volume–reduction surgery. N Engl J Med. 2001;345:1075-1083.

        3. Group NETTR. A randomized trial comparing lung-volume–reduction surgery with medical therapy for severe emphysema. N Engl J Med. 2003;348:2059-2073.

        4. Decker MR, Leverson GE, Jaoude WA, Maloney JD. Lung volume reduction surgery since the National Emphysema Treatment Trial: study of Society of Thoracic Surgeons database. J Thorac Cardiovasc Surg. 2014;148:2651-2658.

        5. Deslée G, Mal H, Dutau H, et al. Lung volume reduction coil treatment vs usual care in patients with severe emphysema: the REVOLENS randomized clinical trial. JAMA. 2016;315:175-184.

        6. Hartman JE, Klooster K, Gortzak K, et al. Long-term follow-up after bronchoscopic lung volume reduction treatment with coils in patients with severe emphysema. Respirology. 2015;20:319-326.

        7. Snell GI, Hopkins P, Westall G, et al. A feasibility and safety study of bronchoscopic thermal vapor ablation: a novel emphysema therapy. Ann Thorac Surg. 2009;88:1993-1998.

        8. Ingenito EP, Berger RL, Henderson AC, et al. Bronchoscopic lung volume reduction using tissue engineering principles. Am J Respir Crit Care Med. 2003;167:771-778.

        9. Ingenito EP, Loring SH, Moy ML, et al. Comparison of physiological and radiological screening for lung volume reduction surgery. Am J Respir Crit Care Med. 2001;163:1068-1073.

        10. Shah P, Slebos D, Cardoso P, et al. Bronchoscopic lung-volume reduction with Exhale airway stents for emphysema (EASE trial): randomised, sham-controlled, multicentre trial. Lancet. 2011;378:997-1005.

        11. Sciurba FC, Ernst A, Herth FJ, et al. A randomized study of endobronchial valves for advanced emphysema. N Engl J Med. 2010;363:1233-1244.

        12. Wan IY, Toma TP, Geddes DM, et al. Bronchoscopic lung volume reduction for end-stage emphysema: report on the first 98 patients. Chest. 2006;129:518-526.

        13. Gompelmann D, Eberhardt R, Schuhmann M, et al. Lung volume reduction with vapor ablation in the presence of incomplete fissures: 12-month results from the STEP-UP randomized controlled study. Respiration. 2016;92:397-403.

        14. Come CE, Kramer MR, Dransfield MT, et al. A randomised trial of lung sealant versus medical therapy for advanced emphysema. Eur Respir J. 2015;46:651-662.

        15. Group NOTT. Continuous or nocturnal oxygen therapy in hypoxemic chronic obstructive lung disease: a clinical trial. Ann Intern Med. 1980;93:391-398.

        16. Council M. Long term domiciliary oxygen therapy in chronic hypoxic cor pulmonale complicating chronic bronchitis and emphysema: Report of the Medical Research Council Working Party. Lancet. 1981;1:681-686.

        17. Qaseem A, Wilt TJ, Weinberger SE, et al. Diagnosis and management of stable chronic obstructive pulmonary disease: a clinical practice guideline update from the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society. Ann Intern Med. 2011;155:179-191.

        18. Vestbo J, Hurd SS, Agustí AG, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187:347-365.

        19. Group L-TOTTR. A randomized trial of long-term oxygen for COPD with moderate desaturation. N Engl J Med. 2016;375:1617-1627.

        20. McCarthy B, Casey D, Devane D, et al. Pulmonary rehabilitation for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015(2):CD003793.

        21. Griffiths TL, Burr ML, Campbell IA, et al. Results at 1 year of outpatient multidisciplinary pulmonary rehabilitation: a randomised controlled trial. Lancet. 2000;355:362-368.

        22. Garcia-Aymerich J, Lange P, Benet M, et al. Regular physical activity reduces hospital admission and mortality in chronic obstructive pulmonary disease: a population based cohort study. Thorax. 2006;61:772-778.

        23. Global Initiative for Chronic Obstructive Lung Disease (GOLD): Global strategy for the diagnosis, management, and prevention of COPD 2017. www.goldcopd.org. Accessed July 9, 2019.

        24. Thabut G, Ravaud P, Christie JD, et al. Determinants of the survival benefit of lung transplantation in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;177:1156-1163.

        25. Lahzami S, Bridevaux PO, Soccal PM, et al. Survival impact of lung transplantation for COPD. Eur Respir J. 2010;36:74-80.

        26. Cerón Navarro J, de Aguiar Quevedo K, Ansótegui Barrera E, et al. Functional outcomes after lung transplant in chronic obstructive pulmonary disease. Arch Bronconeumol. 2015;51:109-114.

        27. Weill D, Benden C, Corris PA, et al. A consensus document for the selection of lung transplant candidates: 2014--an update from the Pulmonary Transplantation Council of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant. 2015;34:1-15.

        28. Minai OA, Shah S, Mazzone P, et al. Bronchogenic carcinoma after lung transplantation: characteristics and outcomes. J Thorac Oncol. 2008;3:1404-1409.

        29. Hajizadeh N, Goldfeld K, Crothers K. What happens to patients with COPD with long-term oxygen treatment who receive mechanical ventilation for COPD exacerbation? A 1-year retrospective follow- up study. Thorax. 2015;70:294-296.

        30. Siouta N, van Beek K, Preston N, et al. Towards integration of palliative care in patients with chronic heart failure and chronic obstructive pulmonary disease: a systematic literature review of European guidelines and pathways. BMC Palliat Care. 2016;15:18.

        31. Celli BR, MacNee W; ATS/ERS Task Force. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper. Eur Respir J. 2004;23:932-946.

        32. Szekendi MK, Vaughn J, Lal A, et al. The prevalence of inpatients at thirty-three U.S. hospitals appropriate for and receiving referral to palliative care. J Palliat Med. 2016;19:360-372.

        33. Rush B, Hertz P, Bond A, et al. Use of palliative care in patients with end-stage COPD and receiving home oxygen: national trends and barriers to care in the United States. Chest. 2017;151:41-46.

        References

        1. Sabanathan A, Sabanathan S, Shah R, Richardson J. Lung volume reduction surgery for emphysema: a review. J Cardiovasc Surg. 1998;39:237.

        2. Group NETTR. Patients at high risk of death after lung-volume–reduction surgery. N Engl J Med. 2001;345:1075-1083.

        3. Group NETTR. A randomized trial comparing lung-volume–reduction surgery with medical therapy for severe emphysema. N Engl J Med. 2003;348:2059-2073.

        4. Decker MR, Leverson GE, Jaoude WA, Maloney JD. Lung volume reduction surgery since the National Emphysema Treatment Trial: study of Society of Thoracic Surgeons database. J Thorac Cardiovasc Surg. 2014;148:2651-2658.

        5. Deslée G, Mal H, Dutau H, et al. Lung volume reduction coil treatment vs usual care in patients with severe emphysema: the REVOLENS randomized clinical trial. JAMA. 2016;315:175-184.

        6. Hartman JE, Klooster K, Gortzak K, et al. Long-term follow-up after bronchoscopic lung volume reduction treatment with coils in patients with severe emphysema. Respirology. 2015;20:319-326.

        7. Snell GI, Hopkins P, Westall G, et al. A feasibility and safety study of bronchoscopic thermal vapor ablation: a novel emphysema therapy. Ann Thorac Surg. 2009;88:1993-1998.

        8. Ingenito EP, Berger RL, Henderson AC, et al. Bronchoscopic lung volume reduction using tissue engineering principles. Am J Respir Crit Care Med. 2003;167:771-778.

        9. Ingenito EP, Loring SH, Moy ML, et al. Comparison of physiological and radiological screening for lung volume reduction surgery. Am J Respir Crit Care Med. 2001;163:1068-1073.

        10. Shah P, Slebos D, Cardoso P, et al. Bronchoscopic lung-volume reduction with Exhale airway stents for emphysema (EASE trial): randomised, sham-controlled, multicentre trial. Lancet. 2011;378:997-1005.

        11. Sciurba FC, Ernst A, Herth FJ, et al. A randomized study of endobronchial valves for advanced emphysema. N Engl J Med. 2010;363:1233-1244.

        12. Wan IY, Toma TP, Geddes DM, et al. Bronchoscopic lung volume reduction for end-stage emphysema: report on the first 98 patients. Chest. 2006;129:518-526.

        13. Gompelmann D, Eberhardt R, Schuhmann M, et al. Lung volume reduction with vapor ablation in the presence of incomplete fissures: 12-month results from the STEP-UP randomized controlled study. Respiration. 2016;92:397-403.

        14. Come CE, Kramer MR, Dransfield MT, et al. A randomised trial of lung sealant versus medical therapy for advanced emphysema. Eur Respir J. 2015;46:651-662.

        15. Group NOTT. Continuous or nocturnal oxygen therapy in hypoxemic chronic obstructive lung disease: a clinical trial. Ann Intern Med. 1980;93:391-398.

        16. Council M. Long term domiciliary oxygen therapy in chronic hypoxic cor pulmonale complicating chronic bronchitis and emphysema: Report of the Medical Research Council Working Party. Lancet. 1981;1:681-686.

        17. Qaseem A, Wilt TJ, Weinberger SE, et al. Diagnosis and management of stable chronic obstructive pulmonary disease: a clinical practice guideline update from the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society. Ann Intern Med. 2011;155:179-191.

        18. Vestbo J, Hurd SS, Agustí AG, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187:347-365.

        19. Group L-TOTTR. A randomized trial of long-term oxygen for COPD with moderate desaturation. N Engl J Med. 2016;375:1617-1627.

        20. McCarthy B, Casey D, Devane D, et al. Pulmonary rehabilitation for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2015(2):CD003793.

        21. Griffiths TL, Burr ML, Campbell IA, et al. Results at 1 year of outpatient multidisciplinary pulmonary rehabilitation: a randomised controlled trial. Lancet. 2000;355:362-368.

        22. Garcia-Aymerich J, Lange P, Benet M, et al. Regular physical activity reduces hospital admission and mortality in chronic obstructive pulmonary disease: a population based cohort study. Thorax. 2006;61:772-778.

        23. Global Initiative for Chronic Obstructive Lung Disease (GOLD): Global strategy for the diagnosis, management, and prevention of COPD 2017. www.goldcopd.org. Accessed July 9, 2019.

        24. Thabut G, Ravaud P, Christie JD, et al. Determinants of the survival benefit of lung transplantation in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;177:1156-1163.

        25. Lahzami S, Bridevaux PO, Soccal PM, et al. Survival impact of lung transplantation for COPD. Eur Respir J. 2010;36:74-80.

        26. Cerón Navarro J, de Aguiar Quevedo K, Ansótegui Barrera E, et al. Functional outcomes after lung transplant in chronic obstructive pulmonary disease. Arch Bronconeumol. 2015;51:109-114.

        27. Weill D, Benden C, Corris PA, et al. A consensus document for the selection of lung transplant candidates: 2014--an update from the Pulmonary Transplantation Council of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant. 2015;34:1-15.

        28. Minai OA, Shah S, Mazzone P, et al. Bronchogenic carcinoma after lung transplantation: characteristics and outcomes. J Thorac Oncol. 2008;3:1404-1409.

        29. Hajizadeh N, Goldfeld K, Crothers K. What happens to patients with COPD with long-term oxygen treatment who receive mechanical ventilation for COPD exacerbation? A 1-year retrospective follow- up study. Thorax. 2015;70:294-296.

        30. Siouta N, van Beek K, Preston N, et al. Towards integration of palliative care in patients with chronic heart failure and chronic obstructive pulmonary disease: a systematic literature review of European guidelines and pathways. BMC Palliat Care. 2016;15:18.

        31. Celli BR, MacNee W; ATS/ERS Task Force. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper. Eur Respir J. 2004;23:932-946.

        32. Szekendi MK, Vaughn J, Lal A, et al. The prevalence of inpatients at thirty-three U.S. hospitals appropriate for and receiving referral to palliative care. J Palliat Med. 2016;19:360-372.

        33. Rush B, Hertz P, Bond A, et al. Use of palliative care in patients with end-stage COPD and receiving home oxygen: national trends and barriers to care in the United States. Chest. 2017;151:41-46.

        Publications
        Publications
        Topics
        Article Type
        Display Headline
        Stable COPD: Managing Advanced Disease
        Display Headline
        Stable COPD: Managing Advanced Disease
        Sections
        Citation Override
        Pulmonary Disease Board Review. 2019 July;16(1)
        Disallow All Ads
        Content Gating
        No Gating (article Unlocked/Free)
        Alternative CME
        Disqus Comments
        Default
        Gate On Date
        Tue, 07/16/2019 - 11:30
        Un-Gate On Date
        Tue, 07/16/2019 - 11:30
        Use ProPublica
        CFC Schedule Remove Status
        Tue, 07/16/2019 - 11:30
        Hide sidebar & use full width
        render the right sidebar.