Pulmonary Complications of HIV Infection




Introduction


Since the recognition of the first cases of acquired immunodeficiency syndrome (AIDS) in 1981, our understanding of human immunodeficiency virus (HIV) and its myriad infectious and noninfectious complications has markedly increased. The prognosis for persons infected with HIV has undergone dramatic change for those with access to effective combination antiretroviral therapy (ART) to suppress HIV replication and medications for treatment and prevention of HIV-associated complications. ART in this chapter refers to the use of combination ART, often taken in a fixed-dose combination, that suppresses viral replication and prevents selection of drug-resistant HIV. These therapeutic advances have resulted in a dramatic decline in the incidence of new AIDS cases and in the number of AIDS-associated deaths in countries where this care is widely and consistently available. Unfortunately, access to ART and other medications is not universal, and an HIV vaccine remains elusive. Therefore, efforts to prevent the spread of new HIV infection, to identify and treat those persons who are already HIV infected, and to slow the progression from HIV to AIDS continue to be worldwide priorities.


The lungs are one of the chief target organs of HIV and, accordingly, a major source of morbidity and mortality. The spectrum of pulmonary manifestations is broad and includes many infectious and noninfectious complications ( Table 90-1 ). These complications include diseases that are AIDS-defining (e.g., Pneumocystis pneumonia) or HIV-associated (e.g., bacterial pneumonia), disorders that are not classified as HIV-associated but appear to be more common in those with HIV infection (e.g., lung cancer, pulmonary arterial hypertension [PAH], and chronic obstructive pulmonary disease [COPD]), and conditions whose association with HIV is inconclusive or purely coincidental (e.g., sarcoidosis). HIV-infected patients may also frequently experience critical illness either as a result of pulmonary complications or other diseases and, although uncommon, lung transplantation in HIV-infected patients has been performed. This chapter provides a brief review of the epidemiology of HIV infection and its immunologic abnormalities, followed by an approach to the evaluation of pulmonary disease in an HIV-infected patient. Then, the major HIV-associated pulmonary diseases are discussed, with an emphasis on clinical features, diagnosis, treatment, and prevention.



Table 90-1

Spectrum of Pulmonary Complications




















































































INFECTIONS
Bacteria *
Streptococcus pneumoniae
Haemophilus species
Staphylococcus aureus
Pseudomonas aeruginosa
Other bacteria
Mycobacteria
Mycobacterium tuberculosis
Mycobacterium avium complex
Mycobacterium kansasii
Other mycobacteria
Fungi
Pneumocystis jirovecii (formerly P. carinii ) §
Cryptococcus neoformans
Histoplasma capsulatum
Coccidioides immitis
Aspergillus species (most often A. fumigatus )
Blastomyces dermatitidis
Penicillium marneffei
Other fungi
Viruses
Cytomegalovirus §
Other viruses
Parasites
Toxoplasma gondii
Other parasites
MALIGNANCIES
Kaposi sarcoma §
Non-Hodgkin lymphoma §
Bronchogenic carcinoma
Interstitial Pneumonitides
Lymphocytic interstitial pneumonitis
Nonspecific interstitial pneumonitis
OTHER
Chronic obstructive pulmonary disease
Asthma
Pulmonary arterial hypertension
Sarcoidosis
Immune reconstitution inflammatory syndrome

Modified from Murray JF, Mills J: Pulmonary infectious complications of human immunodeficiency virus infection. Am Rev Respir Dis 141:1356–1372, 1582–1598, 1990.

* Acquired immunodeficiency syndrome (AIDS)–defining diagnosis in adults and adolescents (>13 years) if recurrent (≥2 episodes in 12 months). Not applicable as AIDS-defining diagnosis in children.


AIDS-defining diagnosis in adults and adolescents. AIDS-defining diagnosis in children if extrapulmonary or disseminated.


AIDS-defining diagnosis if extrapulmonary or disseminated.


§ AIDS-defining diagnosis.


AIDS-defining diagnosis if central nervous system is involved.


AIDS-defining diagnosis in children. Not applicable as AIDS-defining diagnosis in adults or adolescents.





Epidemiology


The AIDS epidemic has had a major impact throughout the world. A disproportionate number of all of the HIV-infected people in the world live in low- and middle-income countries, especially in sub-Saharan Africa, which has the world’s highest prevalence of HIV infection. Global estimates indicate that 35.3 million people were living with HIV in 2012. Overall, approximately 0.8% of adults aged 18 to 49 years are infected with HIV worldwide. In sub-Saharan Africa, 4.9% of adults are living with HIV. Following sub-Saharan Africa, the regions with the highest burden of HIV infection are the Caribbean, Eastern Europe, and Central Asia. The Centers for Disease Control and Prevention (CDC) estimates that 1.1 million people were living with HIV in the United States as of 2012.


Encouragingly, new HIV infections have declined over the past decade. Although 2.3 million people worldwide were newly infected with HIV in 2011, this number represents a 20% drop from the number in 2001. AIDS-related mortality has also declined: in 2012, 1.6 million people died of AIDS-related causes compared with 2.3 million in 2005. Because survival has improved for HIV-infected patients since the introduction of ART, more people are living with HIV in the United States than ever before. Furthermore, HIV-infected adults in clinical care in the United States are more likely in the recent era to be prescribed ART, to have a suppressed HIV viral load, and to have a higher median CD4 + T cell-count.


The gender and racial/ethnic demographics of HIV-infected populations vary worldwide. In sub-Saharan Africa, women account for approximately 58% of people estimated to be living with HIV. In the United States, women comprise approximately 21% of adult AIDS cases. Although early in the epidemic HIV was predominantly seen among non-Hispanic white men who have sex with other men (MSM), HIV is increasingly affecting minorities, with new infections disproportionately represented in minorities, especially blacks and Hispanic/Latin Americans.




Immunologic Abnormalities


After infection with HIV and without ART, HIV-infected persons experience a gradual but inexorable loss of host immunity that has been characterized as a syndrome of immune dysregulation, dysfunction, and deficiency, involving many components of the immune system. HIV is tropic for CD4 + lymphocytes and monocytes. After initial infection, HIV leads to massive depletion of CD4 + lymphocytes of the effector-memory type from mucosal associated lymphoid tissue. During the chronic phase of untreated HIV infection, there is a generalized immune activation and ultimately a progressive decline in the naive and memory T-cell pool that results in systemic CD4 + lymphocyte depletion. As well as being decreased in number, T cells are also dysfunctional and mount abnormal host responses to T-cell–dependent antigens. HIV infection is also associated with B-cell dysfunction. Abnormal polyclonal activation, hypergammaglobulinemia, and lack of specific antibody responses ensue. This combination of immune dysfunction, dysregulation, and depletion of CD4 + lymphocytes results in a substantially increased risk for infections and other complications. Effective ART substantially decreases opportunistic infections and mortality in HIV-infected patients. The chronic immune activation that takes place in the second phase of HIV infection can be decreased by ART. Nonetheless, immune activation and chronic inflammation can persist, particularly in those who initiated ART at lower CD4 + cell counts or who have treatment interrupted. Premature mortality and comorbidities such as cardiovascular, renal, and liver disease have been associated with residual inflammation and immunodeficiency despite treatment with ART.


HIV infection also causes alteration in several lines of host defense in the lung and respiratory tract that contribute to an increased risk for pulmonary infections. These include abnormalities in both mucociliary function and soluble defense molecules within respiratory secretions. Within the lung parenchyma, innate and adaptive immune responses to pathogens may be impaired. For example, alveolar macrophages from HIV-infected individuals have been shown to have deficiencies in pathogen recognition, including abnormalities in Toll-like receptor 4 signaling. Bronchoalveolar CD4 + T cells from HIV-infected individuals display impaired responses to important respiratory pathogens, including influenza and Mycobacterium tuberculosis . Evidence also suggests that HIV results in chronic stimulation and activation of inflammatory cells within the alveolar space. Furthermore, animal models suggest that HIV-related proteins may disrupt the barrier function of the alveolar epithelium.


The consequences of persistently increased inflammation and immune activation related to HIV on the pathogenesis of chronic lung complications are less clear. Within the alveolar space, HIV infection is associated with changes in the cellular profile ; however, the mucosal depletion of CD4 + T cells that takes place in the gastrointestinal tract does not appear to happen within the lung. Instead, bronchoalveolar lavage (BAL) fluid from HIV-infected individuals has more CD4 + T cells than does the terminal ileum. Lung cells that can be infected with HIV include alveolar macrophages, T cells, and fibroblasts. While alveolar macrophages have been viewed as a potential reservoir of HIV-infected cells within the lung, recent evidence suggests that T cells instead may be a long-lived reservoir of infection within the lung. The clinical significance of the lung as a protected compartment for HIV-infected cells is incompletely understood.




Diagnostic Approach


The evaluation of an HIV-infected patient with pulmonary disease is aimed at making a specific diagnosis. Definitive diagnosis is preferred over empirical therapy for a number of reasons. Although each of the HIV-associated pulmonary diseases has characteristic clinical and radiographic features, these features overlap. Occasionally, the presenting features are atypical or more than one pulmonary disease is present. Without appropriate therapy, HIV-infected patients with pneumonia can progress rapidly to respiratory failure and death. The therapies used to treat HIV-associated pulmonary diseases have substantial toxicities and interactions with other medications.


The evaluation should begin with the history and physical examination. Laboratory testing should be performed and a chest radiograph obtained in patients with suspected pulmonary disease. Frequently, the results of this evaluation suggest a specific differential diagnosis and management plan. Occasionally, additional studies such as pulmonary function testing, chest computed tomography (CT), or high-resolution CT are needed to narrow the differential diagnosis and to refine the management plan.


Owing to their particular importance in the diagnostic evaluation of many patients with known or suspected HIV-related pulmonary disease, the value of CD4 + lymphocyte counts and fiberoptic bronchoscopy is briefly reviewed here. The utility of other diagnostic tests is considered in the discussions of the specific complications and disorders that follow this section. Additional information on specific diagnostic tests is also available in Chapter 17 .


Cd4 + Lymphocyte Count


The CD4 + lymphocyte count is an indicator of an HIV-infected patient’s risk for development of a specific opportunistic infection or HIV-associated neoplasm. Many HIV-associated pulmonary diseases present at or below a characteristic CD4 + lymphocyte count and are seldom seen at higher counts. Exceptions are diseases that can present in persons without HIV infection, such as bacterial pneumonia, tuberculosis, and non-Hodgkin lymphoma (NHL). In HIV-infected persons, these diseases can present at any CD4 + lymphocyte count, although their incidence increases and their characteristic presentation changes as the CD4 + lymphocyte count declines (see later disease discussion).


At CD4 + lymphocyte counts less than 200 cells/µL, bacterial pneumonia is often accompanied by bacteremia, and M. tuberculosis disease is often extrapulmonary or disseminated. Moreover, below this CD4 + lymphocyte count, Pneumocystis and Cryptococcus pneumonia become important considerations, whereas neither typically presents in a patient with a count significantly higher than 200 cells/µL. At CD4 + lymphocyte counts less than 50 to 100 cells/µL, endemic fungi, certain viruses ( cytomegalovirus [CMV]), protozoa ( Toxoplasma gondii ), nontuberculous mycobacteria (Mycobacterium avium complex [MAC]), and Kaposi sarcoma (KS) become important considerations. Many of these diseases present with extrapulmonary or disseminated manifestations that may dominate the clinical presentation.


The risk of opportunistic infections is best reflected by the current CD4 + lymphocyte count. Primary and secondary Pneumocystis prophylaxis can be safely discontinued in patients whose CD4 + lymphocyte counts increase above 200 cells/µL for at least 3 months in response to ART; similar evidence has emerged indicating that it is safe to discontinue prophylaxis against MAC, Cryptococcus, Histoplasma capsulatum , Coccidioides immitis , and Toxoplasma when CD4 + lymphocyte counts have been restored to greater than certain CD4 + thresholds.


Fiberoptic Bronchoscopy


In general, bronchoscopy should be considered for any HIV-infected patient with pulmonary disease whose severity warrants a prompt and accurate diagnosis, for patients with suspected pulmonary KS, for patients with suspected NHL (if other diagnostic procedures are thought to be more hazardous or less likely to provide the diagnosis), for patients whose diagnosis remains unknown despite less invasive diagnostic studies (e.g., induced sputum), and for patients whose empirical therapy is failing. Conventional contraindications pertain to HIV-infected patients; HIV infection should not be considered a contraindication to bronchoscopy.


The decision to perform BAL with or without transbronchial biopsy (TBB) depends on the suspected diagnosis and on the sensitivities of these procedures for that diagnosis. At the beginning of the AIDS epidemic, both BAL and TBB were usually performed during the initial bronchoscopy. Studies demonstrated that the yield from these two procedures was complementary in identifying pathogens: the sensitivity of BAL was 86% and that of TBB 87%; when the two procedures were combined, the sensitivity was 98% for all pathogens and 100% for Pneumocystis. Currently, BAL alone is generally performed when Pneumocystis pneumonia (PCP) is the suspected pathogen. The sensitivity of BAL fluid examination for Pneumocystis is 97% or higher; therefore, a negative BAL fluid examination for Pneumocystis rules out the diagnosis of PCP in all but the rarest cases.


In contrast, TBB improves the sensitivity of bronchoscopy for diagnosing a number of other important pathogens, including tuberculosis and endemic fungal pneumonias. Furthermore, tissue confirmation from TBB or another biopsy specimen is required for a definitive diagnosis of invasive aspergillosis, CMV pneumonia, and NHL. Therefore, when the clinical and radiographic features suggest one of these diseases, both BAL and TBB are warranted during the initial bronchoscopy.




Infectious Complications


In this section, the major pulmonary infectious complications of HIV disease are reviewed according to category of infectious microorganism (see Table 90-1 ).


Bacteria


Early in the AIDS epidemic, bacterial pneumonia accounted for only a small proportion of reported pulmonary diseases. Subsequently, bacterial pneumonia became recognized as a frequent complication of HIV infection and one that often preceded other opportunistic infections. As a result, recurrent bacterial pneumonia (defined as two or more episodes within 12 months) was included as an AIDS-defining illness in the 1993 CDC Expanded Surveillance Case Definition for AIDS.


Prior to the ART era, Hirschtick and coworkers in the Pulmonary Complications of HIV Infection Study (PCHIS) confirmed the increased incidence of community-acquired bacterial pneumonia in HIV-infected persons. In this study, there were 5.5 episodes of bacterial pneumonia per 100 person-years in the HIV-infected cohort compared with 0.9 episodes per 100 person-years in the HIV-seronegative control cohort ( P < 0.001). Bacterial pneumonia was more common in HIV-infected patients than in the HIV-seronegative controls, even in the subset of HIV-infected patients with CD4 + lymphocyte counts greater than 500 cells/µL ( P < 0.004). Furthermore, the rate of bacterial pneumonia in the HIV-infected cohort increased as the CD4 + lymphocyte count declined. The rate of bacterial pneumonia was 2.3 episodes per 100 person-years in subjects with a CD4 + lymphocyte count greater than 500 cells/µL compared with 10.8 episodes per 100 person-years in persons with a CD4 + lymphocyte count less than 200 cells/µL. The incidence of bacterial pneumonia was significantly greater in intravenous drug users than in either MSM or female partners of HIV-infected men. Cigarette smoking was also associated with an increased incidence of bacterial pneumonia, especially in HIV-infected individuals with a CD4 + lymphocyte count less than 200 cells/µL.


During the ART era (1996 to present), the incidence of community-acquired bacterial pneumonia among HIV-infected persons has declined, although not to the same extent as opportunistic pneumonias such as PCP. In the CDC-sponsored Adult/Adolescent Spectrum of HIV Disease project, a prospective medical record review of HIV-infected persons aged 13 years and older in 11 U.S. cities, the incidence of recurrent pneumonia declined from 22 per 1000 person-years in 1992 to 10.7 per 1000 person-years in 1997. Other studies have reported similar decreases from the use of ART in both community-acquired pneumonia and hospital-acquired pneumonia. Thus, ART is clearly associated with a decreased risk for bacterial pneumonia. Another reason credited for decreased bacterial pneumonia among HIV-infected patients is the use of trimethoprim-sulfamethoxazole (TMP-SMX) as prophylaxis for PCP.


Yet despite the significant decrease in incidence, bacterial pneumonia remains more frequent in HIV-infected individuals than in noninfected individuals. Furthermore, bacterial pneumonia is a common cause of morbidity and is associated with increased short- and long-term mortality in HIV-infected patients in the ART era. One reason for the persistence of bacterial pneumonia despite effective ART is the high prevalence of cigarette smoking in HIV-infected populations. Cigarette smoking is associated with substantially increased rates of pneumonia in multiple studies of HIV-infected patients, at all CD4 + lymphocyte cell counts, whereas smoking cessation decreases the risk for pneumonia. Intravenous drug use remains a significant risk factor for bacterial pneumonia in the ART era. Additionally, neutrophil number and function can be impaired in HIV infection, which may be exacerbated by concomitant cigarette smoking and contribute to increased risk for bacterial pneumonia.


Several bacterial species can cause pneumonia in HIV-infected patients. Similar to community-acquired bacterial pneumonia in non–HIV-infected populations, Streptococcus pneumoniae and Haemophilus species are the most frequently identified pathogens in people with HIV infection. In several studies, the two organisms have accounted for approximately 50% to 60% of cases combined, and they remain the most commonly isolated bacterial organisms in the ART era. Staphylococcus aureus can be isolated as a cause of community-acquired pneumonia in patients both with and without a history of intravenous drug use. In addition to being a cause of nosocomial disease, Pseudomonas aeruginosa is well recognized as a cause of community-acquired bacterial pneumonia in HIV-infected persons, especially those with advanced AIDS. S. pneumoniae and P. aeruginosa ( eFig. 90-1 ) were the two most common bacteria seen in a series of 111 cases. Although this series included community-acquired as well as hospital-acquired pneumonias, most cases of pneumococcal pneumonia (91%) as well as pseudomonal pneumonia (63%) were community acquired. Other bacteria that are occasionally identified as causes of community-acquired bacterial pneumonia in HIV-infected persons include atypical organisms such as Legionella species, Rhodococcus equi, and Nocardia species; these are discussed in more detail later.


Of hospital-acquired pneumonias, the two most common bacterial causes in HIV-infected persons are S. aureus and P. aeruginosa. Both of these organisms are more likely to be isolated in patients with lower CD4 + lymphocyte counts. Staphylococcal pneumonia is frequently complicated by bacteremia, methicillin resistance, and high mortality. S. pneumoniae and Klebsiella pneumoniae are also relatively frequent causes of pneumonia in HIV-infected hospitalized patients.


Clinical Features.


In HIV-infected persons, the clinical and radiographic features of bacterial pneumonia are similar to those in immunocompetent persons, with a few important exceptions. Of note, HIV-infected individuals have a substantially increased risk for bacteremia and invasive disease due to S. pneumoniae . The rate of pneumococcal bacteremia in HIV-infected patients has been estimated to be almost 100 times greater than that in age-matched non–HIV-infected persons. Findings that confer greater risk for bacteremic pneumococcal pneumonia include alcohol abuse, cigarette smoking, recent hospitalization, and presence of comorbid illnesses. The risk of invasive pneumococcal disease is decreased in those on ART and in those who have received pneumococcal vaccine. However, while the incidence of invasive pneumococcal disease has decreased in the current era, the clinical presentation can be severe and may be complicated by respiratory failure. Providers should consider HIV testing for individuals with unknown serostatus presenting with such disease without other obvious risk factors for invasive pneumococcal disease.


CD4 + Lymphocyte Count.


Bacterial pneumonia can develop throughout the course of HIV infection and at any CD4 + lymphocyte count. As the CD4 + lymphocyte count declines, both the incidence of bacterial pneumonia and the frequency of bacteremia increase. Most cases of P. aeruginosa pneumonia are seen in HIV-infected patients with a CD4 + lymphocyte count less than 100 cells/µL, and usually less than 50 cells/µL.


Imaging.


As illustrated in Figure 90-1 , the majority of bacterial pneumonias due to S. pneumoniae in HIV-infected patients present with segmental, lobar, or multilobar consolidation on chest radiograph. Radiographic findings of pneumococcal pneumonia do not appear to be different among patients on ART than in those not on ART. Although less common, focal or diffuse alveolar patterns as well as interstitial opacities in association with S. pneumoniae or Haemophilus influenzae are also seen. Pneumonia due to P. aeruginosa may present with focal consolidation, similar to pneumococcal or Haemophilus pneumonias, although a significant proportion of radiographs demonstrate cavitary opacities.




Figure 90-1


Pneumococcal pneumonia.

Frontal chest radiograph in a 50-year-old HIV-infected man with positive urinary pneumococcal antigen shows homogeneous opacity with air bronchograms, extending to the pleural surface, representing a “lobar pneumonia” pattern, typical of pneumococcal pneumonia.

(Courtesy Stephen Aston, MBChB; Malawi-Liverpool-Wellcome Trust Clinical Research Programme.)


Diagnosis.


The diagnostic approach to bacterial pneumonia is the same for HIV-infected as for non–HIV-infected persons. HIV-infected patients continue to be at increased risk for invasive pneumococcal disease in the ART era. Blood cultures should be obtained, particularly in those with low CD4 + cell counts, given the increased risk for bacteremia in this group. Thoracentesis should be considered for patients with pleural effusion, especially if the effusion is large or if there is concern about a possible empyema. Efforts to culture the causative bacteria to enable drug susceptibility testing are especially important in communities where drug-resistant bacteria are prevalent.


Pneumococcal urinary antigen testing offers the potential for early, specific diagnosis. In a study of 70 adults with pneumococcal pneumonia, 47 of whom were HIV-infected, pneumococcal urinary antigen testing had a sensitivity of 81%, specificity of 98%, positive predictive value of 98%, and negative predictive value of 82%. The test appeared to perform equally well in HIV-infected and non–HIV-infected persons.


Serum procalcitonin and C-reactive protein levels may assist in the early differentiation of bacterial community-acquired pneumonia and pulmonary tuberculosis in HIV-infected patients; procalcitonin and C-reactive protein levels were higher in patients with bacterial pneumonia than in patients with pulmonary tuberculosis, although overlap exists between these causes of pneumonia, particularly in regions endemic for tuberculosis. Because procalcitonin increases in response to bacterial infections and decreases in the presence of viral infections, levels are sometimes used as a biologic marker of community-acquired bacterial pneumonia. Elevated procalcitonin levels can predict increased mortality in HIV-infected patients with lower respiratory tract infections, due either to tuberculosis or to bacterial causes. However, further studies of these biomarkers in larger cohorts of HIV-infected individuals with pneumonia are needed.


Treatment.


The treatment of patients with community-acquired pneumonia is similar for HIV-infected and non–HIV-infected persons ( Chapter 33 ). The choice of antimicrobial agent should be based on a number of factors, such as the results of a sputum Gram stain, the presence of comorbid conditions (e.g., COPD, congestive heart failure, alcohol use), the clinical and radiographic presentation, and the severity of the pneumonia. As in non–HIV-infected persons, treatment should be initiated promptly. Initial empirical therapy should include coverage against frequently identified organisms (e.g., S. pneumoniae and Haemophilus species). Local prevailing drug resistance patterns must be considered when the antibiotic is selected. Empirical monotherapy with a macrolide is not advised in HIV-infected patients, particularly when they are already on macrolide prophylaxis for MAC, because of increasing pneumococcal resistance to macrolides. Patients on TMP-SMX prophylaxis may be more likely to have penicillin- and TMP-SMX–resistant S. pneumoniae. For patients with CD4 + lymphocyte counts less than 100 cells/µL, especially if associated with recent hospitalization, neutropenia, or broad-spectrum antimicrobial use, consideration should be given to including coverage against P. aeruginosa.


Prevention.


Pneumococcal vaccine should be given to HIV-infected patients. For adult (age 19 and older) immunocompromised patients including those with HIV who have not been previously immunized, current recommendations from the Advisory Committee on Immunization Practices to the CDC are for the 13-valent pneumococcal conjugate vaccine to be given first followed by 23-valent pneumococcal polysaccharide vaccine at least 8 weeks or more later. For those who have already received the 23-valent pneumococcal polysaccharide vaccine, the 13-valent pneumococcal conjugate vaccine should be given at least 1 year later. A repeat dose of 23-valent pneumococcal polysaccharide vaccine should be given 5 years after the first, with another dose given after age 65 years. These recommendations may be revised based on ongoing clinical trials; refer to the Advisory Committee on Immunization Practices and CDC, National Institutes of Health (NIH), and the HIV Medicine Association of the Infectious Diseases Society of America (HIVMA/IDSA) guidelines for additional details. Because influenza is a major risk factor for bacterial pneumonia, prevention also includes yearly administration of inactivated influenza vaccine in all HIV-infected individuals.


Other Bacteria


Legionella pneumonia is an infrequent cause of bacterial pneumonia in HIV-infected patients and can be community acquired or nosocomial. Cases have been reported in persons with severe immunosuppression and in individuals with well-controlled HIV on ART. The chest radiograph typically reveals alveolar opacities; bilateral involvement is common. Extrapulmonary manifestations can also be seen, especially of the gastrointestinal tract and central nervous system. In one series, respiratory failure developed in 12 of 15 hospitalized, HIV-infected subjects (80%) with confirmed Legionella pneumonia and 3 died. Although Legionella pneumophila is the most common species identified, other Legionella species have been reported. Rapid diagnosis of Legionella pneumonia can be obtained with urinary antigen testing. However, cultures and/or serology should still be considered as part of the diagnostic evaluation, in that the urinary antigen primarily detects L. pneumophila serogroup 1 and can have a lower sensitivity in cases of mild disease. Its performance has not been prospectively evaluated in HIV-infected patients.


R. equi pulmonary disease has been reported in HIV-infected persons. A review of more than 100 reported cases of R. equi infection found that approximately two thirds were in HIV-infected persons, with pulmonary disease present in the majority. HIV-infected persons were more likely to have R. equi bacteremia or extrapulmonary manifestations, or both, than persons without HIV infection. Most cases present when the CD4 + lymphocyte count is less than 200 cells/µL and usually less than 100 cells/µL. In one multicenter series of 67 HIV-infected patients with R. equi infection, the mean CD4 + lymphocyte count was 35 cells/µL (range, 1 to 183 cells/µL). The chest radiograph often reveals cavitary lesions that may mimic tuberculosis or nocardiosis, or focal consolidation that may mimic bacterial pneumonia. Sputum, blood, bronchoscopy, and pleural fluid specimens can establish the diagnosis of R. equi pneumonia. The optimum treatment regimen and duration are not well defined. Combination antimicrobial therapy is generally recommended, and treatment for a minimum of 2 months (and frequently 6 months) is often required. Because relapses are common, chronic suppressive therapy is probably indicated.


Nocardia asteroides is the most common species identified in several reports of Nocardia infection in HIV-infected patients, and the lung is the most commonly affected site. Most cases present when the CD4 + lymphocyte count is less than 200 cells/µL. In one series of 30 HIV-infected patients with nocardiosis, the mean CD4 + lymphocyte count was 109 cells/µL (median 92 cells/µL; range, 12 to 266 cells/µL). The presenting symptoms often mimicked tuberculosis. Imaging ( eFig. 90-2 ) usually reveals cavitary lesions or lobar or multilobar opacities, especially in the upper lung zones, although reticulonodular opacities, solitary masses, and pleural effusions are seen also. Sputum or bronchoscopy can establish the diagnosis of pulmonary nocardiosis. A modified acid-fast bacilli (AFB) stain may provide an early presumptive diagnosis in the proper clinical setting. Long-term TMP-SMX is the treatment of choice.


Mycobacteria


Early in the AIDS epidemic, mycobacteria were recognized as major sources of HIV-associated morbidity and mortality. Initially, disseminated MAC was noted to develop in HIV-infected patients. Subsequently, the strong association between HIV and M. tuberculosis and nontuberculous mycobacteria, such as Mycobacterium kansasii, became clearer. Other nontuberculous mycobacteria are also occasionally identified as causes of pneumonia in HIV-infected persons.


Mycobacterium tuberculosis


Tuberculosis is the most prevalent opportunistic infection complicating the HIV epidemic worldwide. Although other pathogens may predominate in individual areas, no other pathogen poses as great a global threat to persons immunocompromised by HIV infection as M. tuberculosis. Moreover, unlike most HIV-associated infections, M. tuberculosis is transmissible from person to person, including to those without HIV infection. In fact, clusters of transmission of tuberculosis that include at least one HIV-infected person are larger, last longer, and have a shorter time period between successive cases than clusters of solely HIV-negative persons.


The AIDS epidemic contributed to a veritable explosion of tuberculosis throughout the world, especially in low-income countries, where it is often the first manifestation of HIV and a leading cause of mortality. The World Health Organization estimated that there were 1.1 million cases of tuberculosis in persons with HIV in 2011. Almost 80% of these cases were in residents of sub-Saharan Africa. There are also approximately 11 million HIV-infected individuals with latent tuberculosis. More than 400,000 HIV-infected persons died of tuberculosis in 2011, making it the leading cause of death in HIV-infected populations worldwide.


The AIDS epidemic has also affected tuberculosis in the United States. Before 1985, the incidence of tuberculosis in the United States regularly declined by 5% to 6% per year. From 1985 through 1992, tuberculosis cases increased by 20%, which resulted in 51,700 additional cases of tuberculosis being reported in excess of that predicted by the annual decline previously noted. Since 1992, tuberculosis cases have decreased again. Indeed, in 2012, fewer than 10,000 cases were reported, the lowest number since tuberculosis reporting began in 1953. The proportion of tuberculosis cases with known HIV infection has also decreased from 15% in 2003 to 7.7% in 2012.


It remains unclear whether HIV-infected persons are more likely to acquire tuberculous infection after exposure to M. tuberculosis than non–HIV-infected persons. Once an individual becomes infected, however, there is no doubt that HIV infection increases the risk for development of primary tuberculosis as well as progressing from latent tuberculosis infection (LTBI) to active tuberculous disease. Instead of a 5% lifetime risk of disease, the risk for tuberculosis among HIV-infected persons with LTBI in the pre-combination ART era has been estimated to be as high as 10% per year.


Multidrug-resistant tuberculosis (MDR-TB), defined as resistance to at least isoniazid and rifampin (also known as rifampicin in many countries), poses a particularly grim prospect for the HIV-infected person. The risk of drug-resistant tuberculosis has been reported to be greater among HIV-infected patients than among others, and drug-resistant tuberculosis appears to be associated with decreased survival, particularly in those with lower CD4 + cell counts or in individuals not receiving ART. In a multivariate analysis, HIV infection was found to be an independent risk factor for isoniazid resistance, for both isoniazid and rifampin resistance, and especially for rifampin monoresistance. A meta-analysis of risk factors for MDR-TB in Europe found that HIV was an independent risk factor (odds ratio, 3.52). In addition, HIV-infected patients treated with intermittent rifabutin-based therapy have been found to have a high risk for acquired rifamycin resistance, including monoresistance, particularly if their CD4 + lymphocyte counts are less than 100 cells/µL. Factors that lead to acquired drug resistance include an inadequate initial treatment regimen, patient nonadherence with the prescribed regimen, and the addition of a single drug to a failing regimen. Studies using restriction fragment length polymorphism analysis have shown that MDR-TB can also develop as a result of exogenous reinfection from a multidrug-resistant source.


Extensively drug-resistant tuberculosis (XDR-TB), defined as resistance to at least isoniazid and rifampin (i.e., MDR-TB), plus resistance to any fluoroquinolone and at least one second-line injectable drug, was first reported in 2006. It has been documented in all regions of the world and in HIV-infected persons. The true extent of XDR-TB is unknown because many laboratories worldwide are unable to perform susceptibility tests, particularly to second-line drugs, and rely only on smear results for diagnosing tuberculosis. A case series of 53 persons with XDR-TB in South Africa found that, of the 44 patients who underwent HIV testing, all were HIV infected; 52 of the 53 patients died, and the median survival was only 16 days. Epidemiologic evidence strongly suggested that the outbreak of XDR-TB was a result of person-to-person transmission, especially in a nosocomial setting.


Clinical Features.


Worldwide, tuberculosis is the most common initial manifestation of underlying HIV infection. The clinical and radiographic features of tuberculosis in both HIV-infected and non–HIV-infected persons vary and are fully discussed in Chapter 35 . HIV infection is independently associated with an increased risk for extrapulmonary tuberculosis. HIV infection is also associated with a lower frequency of cavitary tuberculosis, especially in patients with fewer than 200 CD4 + T lymphocytes/µL at the time of tuberculosis diagnosis.


CD4 + Lymphocyte Count.


Tuberculosis may develop throughout the course of HIV infection, regardless of the CD4 + lymphocyte count, but the incidence of tuberculosis increases as the count decreases. Tuberculosis is much less common in patients receiving ART, but can still happen. The clinical expression of tuberculosis in HIV-infected persons is largely dependent on the degree of host immunosuppression, as indicated by the CD4 + lymphocyte count ( Table 90-2 ). HIV-infected persons with early HIV disease typically present with a picture suggestive of reactivation tuberculosis, with disease usually limited to the lungs. In contrast, persons with more advanced HIV disease typically present with a picture reminiscent of primary pulmonary tuberculosis, often with disseminated or extrapulmonary tuberculosis. In a series of 97 HIV-infected patients with tuberculosis, Jones and colleagues reported that extrapulmonary tuberculosis was seen in 30 of 43 patients (70%) with a CD4 + lymphocyte count of 100 cells/µL or fewer, in 10 of 20 patients (50%) with a count between 101 and 200 cells/µL, in 7 of 16 patients (44%) with a count between 201 and 300 cells/µL, and in only 5 of 18 patients (28%) with a count greater than 300 cells/µL. Virtually any extrapulmonary site may be involved; common sites include lymph nodes (usually cervical, supraclavicular, and axillary), bone marrow, genitourinary tract, central nervous system, and liver.



Table 90-2

Manifestations of HIV-Related Tuberculosis in “Early” and “Late” HIV Infection




































Feature Early HIV Late HIV
Extrapulmonary 10-15% >50%
Radiographic distribution Upper zones Lower and middle zones
Radiographic findings
Cavitation Common Uncommon
Adenopathy Uncommon Common
Miliary Uncommon Common
Pleural effusion Uncommon Rare

Adapted from Murray JF: Cursed duet: HIV infection and tuberculosis. Respiration 57:210–220, 1990.


Imaging.


The prevalence of specific chest radiograph features of tuberculosis also depends on the degree of host immunosuppression. Early in the course of HIV infection, tuberculosis mirrors that encountered in immunocompetent persons: upper lung zone opacities, often with cavitation, on chest radiograph. However, later in the course of HIV infection, tuberculosis often presents with middle and lower lung zone opacities ( Fig. 90-2 , eFig. 90-3 ); with diffuse opacities, including a miliary pattern ( eFig. 90-4 ); or with a normal chest radiograph. As the CD4 + lymphocyte count declines, cavitation becomes less common and intrathoracic adenopathy ( Fig. 90-3 , eFig. 90-5 ) becomes more common. Jones and colleagues reported that adenopathy was seen on the radiographs of 20 of 58 HIV-infected patients (34%) with a CD4 + lymphocyte count of 200 cells/µL or fewer compared with 4 of 29 patients (14%) with a CD4 + lymphocyte count greater than 200 cells/µL ( P = 0.04). ART also affects the chest radiographic appearance in HIV-infected patients with tuberculosis; in one study of 209 patients, 82% of patients receiving ART presented with a radiographic pattern of tuberculosis resembling that in immunocompetent persons, compared with 44% of patients not receiving ART ( P < 0.001).




Figure 90-2


Tuberculosis.

Frontal chest radiograph of an HIV-infected patient showing focal right lung consolidation with air bronchograms. Culture of sputum revealed Mycobacterium tuberculosis that was mono-rifampin resistant. Knowledge of the patient’s CD4 + lymphocyte count (<50 cells/µL) and understanding that tuberculosis can present with middle to lower lung zone consolidation during “late” HIV infection were instrumental in making the diagnosis of tuberculosis. The key was in the patient’s pocket.

(Courtesy L. Huang.)



Figure 90-3


Tuberculosis.

Frontal chest radiograph of an HIV-infected patient showing mediastinal adenopathy. Culture of biopsy material revealed Mycobacterium tuberculosis. Among HIV-infected persons, the proportion of cases that present with intrathoracic adenopathy on chest radiograph increases as the CD4 + lymphocyte count decreases.

(From Murray JF, Mills J: Pulmonary complications of HIV infection. Am Rev Respir Dis 141:1356–1372, 1582–1598, 1990.)


Diagnosis.


The diagnostic approach to tuberculosis is the same for HIV-infected as for non–HIV-infected persons, with the “gold standard” diagnostic test being isolation and identification of M. tuberculosis by culture or by nucleic acid amplification ( Chapter 35 ). Three sputum specimens should be examined for AFB and cultured for mycobacteria. Current recommendations support the use of nucleic acid amplification testing of at least one sputum specimen because these tests can confirm the presence of tuberculosis or another mycobacteria in smear-positive patients and because nucleic acid amplification testing is more sensitive than AFB smear, allowing more rapid identification of smear-negative, culture-positive samples. In patients with a nonproductive cough or scant secretions, sputum induction should be performed. Virtually any specimen can be studied for mycobacteria, including sputum, pleural fluid, urine, cerebrospinal fluid, and BAL fluid; Wang needle, fine-needle, and bone marrow aspirates; and all biopsy and tissue specimens. The sensitivity of mycobacterial culture of induced sputum for HIV-infected patients with pleural effusions due to tuberculosis has been reported to be as high as 77%, and cultures of samples from extrapulmonary sites have a higher sensitivity in those with advanced immunodeficiency. Blood cultures are specific and should be obtained, especially from those persons with a CD4 + lymphocyte count less than 200 cells/µL.


All cultures positive for M. tuberculosis should be submitted for susceptibility testing, because the results are essential in identifying cases of drug resistance and in tailoring definitive treatment. More rapid identification of drug-resistance may be possible with genotypic testing to identify drug-resistance mutations in places where these tests are available.


Treatment.


The treatment of patients with tuberculosis is similar for HIV-infected and non–HIV-infected persons ( Chapter 35 ). Because tuberculosis is transmissible, persons with suspected tuberculosis should be started promptly on empirical antituberculosis treatment to reduce the risk of transmission. The detection of AFB on smear or in culture, regardless of the source, is an indication for empirical antituberculosis treatment until final identification is obtained. HIV-infected persons with suspected tuberculosis should be started on four antituberculous drugs: isoniazid, rifampin, pyrazinamide, and ethambutol along with pyridoxine. Directly observed therapy is recommended. If resistance to rifampin is suspected or confirmed, moxifloxacin or levofloxacin and an aminoglycoside or capreomycin should be included in the initial regimen and a tuberculosis expert consulted. When results of species identification and susceptibility testing become available, treatment can be tailored as needed. In the United States, the response to tuberculosis therapy and the time to convert sputum cultures from positive to negative appear to be similar in HIV-infected and non–HIV-infected patients. In sub-Saharan Africa, however, the likelihood of death, especially during the first 2 months of therapy, and of relapse following apparently successful treatment is considerably higher in HIV-infected than in non–HIV-infected patients.


HIV-infected patients with tuberculosis are more likely to experience toxicity from antituberculous drugs than are non–HIV-infected persons. One series found that 40% of HIV-infected persons suffered serious adverse events from antituberculous drugs compared with 26% of non–HIV-infected persons ( P = 0.008). HIV-infected patients are frequently receiving a number of additional medications, and it is often difficult to distinguish an adverse drug effect to antituberculous drugs from adverse effects to these other medications. As a result, the first-line antituberculous drugs (especially isoniazid and rifampin) should only be discontinued permanently if there is strong evidence that these medications were the cause.


Despite challenges in ART use during TB therapy, as described later, ART given concurrently with tuberculosis treatment rather than sequentially has been associated with decreased mortality, greater AIDS-free survival, and more rapid conversion of sputum smears and culture in randomized studies. Thus, all HIV-infected individuals with TB should receive ART. If the patient is naive to ART, it is recommended that ART be initiated within 2 weeks of tuberculosis treatment initiation in individuals with CD4 + cell counts less than 50 cells/µL and within 8 to 12 weeks in all others. Individuals receiving ART should continue its use during TB therapy.


Clinicians need to be aware of several challenges of ART use during TB treatment. HIV-infected patients with tuberculosis who begin therapy for both M. tuberculosis and HIV infection may develop immune reconstitution inflammatory syndrome (IRIS), a paradoxical reaction presenting as a temporary exacerbation of clinical and radiographic features, which is more common with ART started early during antituberculosis therapy and among those with disseminated tuberculosis. Furthermore, simultaneous treatment of HIV and M. tuberculosis poses additional challenges in terms of drug interactions. Both HIV protease inhibitors (PIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) have significant interactions with the rifamycins, principally related to the induction or inhibition (or both) of the hepatic cytochrome P-450 (CYP) enzyme system. Decreased drug levels are associated with development of drug resistance by both HIV and M. tuberculosis, whereas increased drug levels are correlated with toxicity. In persons receiving PIs, rifabutin is preferred over rifampin because it is a less potent CYP 3A4 inducer. Information on the interactions between specific antiretroviral drugs and different rifamycins is regularly updated and can be found on such websites as the CDC website http://www.cdc.gov/tb . Clinicians caring for HIV-infected patients with tuberculosis should consult experts for aid in management prior to initiating tuberculosis or HIV therapy.


Prevention of Exposure.


In hospitals and health care facilities, all persons with HIV infection and signs or symptoms compatible with tuberculosis should be placed in respiratory isolation until three sputum smears are negative. Prevention of transmission of drug-resistant tuberculosis is particularly important, for those with and without HIV infection. In a modeling study, measures such as improved ventilation, rapid drug sensitivity testing, HIV treatment, and tuberculosis isolation facilities were found to be effective preventive measures, but involuntary detention for XDR-TB treatment was thought to lead to an increase in transmission. This study also estimated that mask use and a shift to outpatient therapy could prevent about one third of XDR-TB cases in a highly prevalent area. A subsequent randomized study of patient mask use demonstrated a 56% decrease in TB transmission.


Prevention of Disease.


HIV-infected persons are at a high risk for progressing from LTBI to active tuberculosis. Therefore, HIV-infected persons should be tested for LTBI by either a tuberculin skin test or interferon-γ (IFN-γ) release assay when HIV infection is first identified. HIV-infected persons who are at risk for exposure to M. tuberculosis (either ongoing or repeated) should be tested for LTBI annually. In addition, persons who initially had a negative test, but subsequently experienced an increase in their CD4 + lymphocyte count to ≥200 cells/µL due to ART should be retested for LTBI. Any individual with a positive test for LTBI should be screened for symptoms and chest radiographic findings suggestive of active TB.


Several studies document that the risk of LTBI reactivation is virtually eliminated if an HIV-infected person who has been infected with a drug-susceptible organism takes adequate prophylaxis. HIV-infected persons with a positive tuberculin skin reaction (defined as 5-mm induration or greater), a positive IFN-γ release assay, or with a history of either of these and who have not received prior treatment for either LTBI or active tuberculosis should receive prophylaxis. HIV-infected individuals who are close contacts of anyone with active TB should also receive prophylactic therapy. The standard prophylaxis treatment is isoniazid (300 mg daily or 900 mg twice weekly) plus pyridoxine for 9 months. An alternative treatment for LTBI is rifampin daily for 4 months. The two-drug regimen of rifampin plus pyrazinamide for 2 months is no longer recommended due to an increased incidence of severe hepatotoxicity and death. Although a 12-week regimen of high-dose isoniazid and rifapentine by weekly directly observed therapy is another alternative, this regimen is not recommended for HIV-infected patients who are on ART due to drug interactions between certain antiretrovirals and rifapentine. Expert consultation should be sought for patients receiving ART or for individuals exposed to drug-resistant tuberculosis. Routine use of preventive therapy in anergic or purified protein derivative–negative HIV-infected persons is not generally recommended.


Mycobacterium avium Complex


As discussed in Chapter 36 , MAC consists of several related Mycobacterium species, including M. avium and M. intracellulare. Until 1980, only 24 cases of disseminated MAC had been reported. Coincident with the onset of the AIDS epidemic, the number of cases rose dramatically. With the use of ART, however, the incidence of disseminated MAC has declined since 1996.


The pathogenesis of disseminated MAC is incompletely understood, but it is believed to result from primary acquisition of the microorganism, which is ubiquitous in the environment, rather than reactivation of latent infection. These mycobacteria probably enter the body chiefly through the gastrointestinal tract and occasionally through the lungs. Person-to-person transmission is believed to be uncommon. Nearly all cases of disseminated MAC are caused by M. avium serotypes, which suggests that there are important differences in exposure to or virulence of these particular strains.


Clinical Features.


Although the lungs are an important potential portal of entry for MAC into the bloodstream, isolated MAC pulmonary disease is rare in the context of AIDS. The most common clinical presentation of disseminated MAC disease is a febrile wasting syndrome consisting of fever, night sweats, fatigue, anorexia, and weight loss; other manifestations include abdominal pain and chronic diarrhea, hepatosplenomegaly, lymphadenopathy, progressive anemia, and, rarely, extrabiliary obstructive jaundice. Laboratory abnormalities include anemia, which is often severe, and an elevated alkaline phosphatase level.


CD4 + Lymphocyte Count.


More than 95% of cases of disseminated MAC are seen in HIV-infected patients whose CD4 + lymphocyte count is 50 cells/µL or less. Other risk factors for disseminated MAC include a plasma HIV viral level greater than 100,000 copies/mL, history of previous opportunistic infection, and previous MAC colonization of the respiratory or gastrointestinal tract.


Imaging.


The chest radiograph is typically normal, even when the organism is cultured from respiratory tract secretions. Focal pneumonia has been reported ( eFig. 90-6 ) but is extremely rare, as is presentation as a solitary pulmonary nodule ( eFig. 90-7 ). More frequently seen—but still uncommon—are endobronchial lesions without pneumonia. These endobronchial lesions appear to be submucosal “pearls” ( eFig. 90-8 ) that are teeming with AFB on biopsy. Lymphadenopathy—with or without necrosis—is one of the more common imaging manifestations of thoracic MAC infection in HIV-infected patients ( eFig. 90-9 ). Many of the cases of isolated pulmonary MAC were reported in patients who received ART, thus raising the possibility that this particular manifestation is an example of immune reconstitution. Clinicians should be aware of this possibility when starting patients on ART because IRIS may be seen either in patients with diagnosed disease or in those with subclinical infection.


Diagnosis.


In HIV-infected patients with MAC, the organism can be cultured from numerous sites, but the most productive sources are blood, bone marrow, liver, or lymph nodes. The sensitivity of blood cultures for disseminated MAC has ranged from 86% to 98% of cases in which disseminated disease was confirmed by autopsy. The diagnosis of disseminated MAC can also be established by cultures from any normally sterile body site. Often, MAC is cultured from respiratory specimens such as sputum or BAL fluid but, depending on the clinical situation, this finding may not indicate either pulmonary or disseminated disease. If patients have abnormal chest radiographs with positive sputum cultures, the diagnostic criteria for nonimmunosuppressed hosts recommended by the American Thoracic Society and the Infectious Disease Society of America (ATS/IDSA), listed in Table 36-3 , should be applied. The pathologic findings of MAC infection are characteristic, but not definitive; AFBs are remarkably abundant and are often packed within foamy macrophages or histiocytes; granulomas are usually absent or poorly formed.


Treatment.


As emphasized in Chapter 36 , the key to any regimen is the use of at least two drugs including a macrolide: clarithromycin (500 mg twice daily) is preferred, but azithromycin (500 to 600 mg daily) can be substituted if needed. One of these should be combined with ethambutol (15 mg/kg/day). Use of a third or fourth agent such as rifabutin (300 mg daily), an aminoglycoside (i.e., amikacin 10 to 15 mg/kg intravenously daily), or a fluoroquinolone (i.e., levofloxacin 500 mg daily) can be considered in patients with advanced immunosuppression, high mycobacterial burden, or the inability to take ART. As with tuberculosis, the use of rifabutin must be carefully scrutinized with concurrent administration of an NNRTI or a PI.


Prevention of Exposure.


Although MAC can be found in environmental sources such as food and water, there are no specific recommendations regarding exposure avoidance as a prevention strategy. Similarly, although the presence of MAC in a stool or respiratory specimen is predictive of disseminated disease, routine screening of either is not recommended.


Prevention of Disease.


The CDC, NIH, and HIVMA/IDSA guidelines recommend that MAC prophylaxis be administered to HIV-infected patients with a CD4 + lymphocyte count less than 50 cells/µL and no clinical evidence of disseminated MAC. Preferred MAC prophylaxis regimens include azithromycin (1200 mg weekly or 600 mg twice weekly) or clarithromycin (500 mg twice daily). Rifabutin (300 mg daily) is an alternative regimen if patients are intolerant of azithromycin or clarithromycin. If rifabutin is to be used, tuberculosis must first be ruled out, because its use for MAC has been associated with rifampin monoresistance in M. tuberculosis . Primary MAC prophylaxis should be discontinued in persons who have experienced a significant response to ART with an increase in their CD4 + lymphocyte count to greater than 100 cells/µL for at least 3 months. Individuals with a history of disseminated MAC should receive secondary prophylaxis/chronic maintenance therapy with a MAC treatment regimen cited earlier. In the setting of ART with increases in the CD4 + lymphocyte count to more than 100 cells/µL for at least 6 months—and after a minimum of 12 months of a macrolide-based MAC treatment regimen—most patients can discontinue therapy without relapse.


Mycobacterium kansasii


As described in Chapter 36 , M. kansasii has a particular geographic distribution, predominantly in the southern and central United States. Clusters of disease have also been reported in Europe, Asia, and Africa. Before the AIDS epidemic, infection and disease due to M. kansasii were uncommon; thereafter, a dramatic increase in the incidence associated with HIV infection was observed, even in areas where M. kansasii was not thought to be endemic. Bloch and associates found a cumulative incidence of M. kansasii infection of 2.4 cases per 100,000 HIV-infected adults in three counties in northern California, a rate that is almost five times higher than the national rate. Although M. kansasii pulmonary disease can be clinically indistinguishable from tuberculosis, person-to-person transmission has not been documented, and infection is thought to arise from environmental sources.


CD4 + Lymphocyte Count.


Similar to tuberculosis in HIV-infected persons, M. kansasii pneumonia can develop at any CD4 + lymphocyte count. Most HIV-infected patients with M. kansasii, however, have evidence of severe immunosuppression with CD4 + lymphocyte counts typically less than 100 cells/µL. In a study from the ART era, Canueto-Quintero and colleagues reported a mean CD4 + lymphocyte count of 20 cells/µL among 25 HIV-infected patients with M. kansasii. Witzig and coworkers found that 32 of 49 HIV-infected patients (65%) with M. kansasii had isolated pulmonary disease (mean CD4 + lymphocyte count, 75 cells/µL), whereas the remainder had disseminated disease (mean CD4 + lymphocyte count, 28 cells/µL).


Imaging.


The chest radiographic findings of M. kansasii are varied. The most common radiographic findings re­­semble tuberculosis and include alveolar opacities, diffuse opacities, and cavities (see Fig. 36-5 ; eFig. 90-10 ); masses, intrathoracic adenopathy, and pleural effusions have also been reported. In a series of 83 HIV-infected patients with M. kansasii, the most frequent findings were consolidation (66%) and nodules (42%); abnormalities were most often located in the mid-lung and lower lung zones (89%), and in 10%, radiographs were normal.


Diagnosis.


The diagnosis of M. kansasii rests on its isolation and subsequent identification by culture. Pulmonary disease can be diagnosed by all of the techniques used for the diagnosis of tuberculosis. Unlike tuberculosis, however, in which identification of M. tuberculosis is diagnostic of disease, the identification of M. kansasii can occasionally represent colonization rather than disease. As previously mentioned, the ATS/IDSA guidelines have defined criteria for diagnosis of nontuberculous mycobacterial pulmonary disease that includes M. kansasii (see Table 36-3 ). In addition, the use of subtyping defined by PCR-restriction enzyme analysis of the hsp65 gene may help distinguish pathogenic from nonpathogenic isolates.


Treatment.


The recommended regimen for treatment of M. kansasii pulmonary disease consists of isoniazid (300 mg daily), rifampin (600 mg daily), and ethambutol (15 mg/kg daily). Patients should also receive pyridoxine (50 mg daily) during treatment. Patients who take PI or NNRTI therapy for HIV infection should receive rifabutin or clarithromycin in place of rifampin. Pyrazinamide is unac­ceptable as an alternative drug because all M. kansasii isolates are resistant. Patients should receive treatment for a minimum of 12 months after their cultures become negative.


Prevention.


There are no recommended prevention strategies for M. kansasii .


Other Mycobacteria


MAC and M. kansasii account for the great majority of nontuberculous mycobacterial infections complicating HIV disease. Other mycobacteria, however, have occasionally been identified. Increased frequency of exposure or specific defects in host defenses might account for the predominance of MAC and M. kansasii over other nontuberculous mycobacteria in AIDS patients; differences in the virulence of the various mycobacterial species are also likely to play an important role.


Fungi


Early in the AIDS epidemic, fungi were recognized as major sources of morbidity and mortality. Despite the overall decline in HIV-associated opportunistic infections in the United States, fungi remain important causes of disease. Several fungi can cause pulmonary disease in HIV-infected persons. Pneumocystis jirovecii, formerly Pneumocystis carinii, previously classified as a protozoan but currently considered a fungus, remains the most common AIDS-defining opportunistic infection in the United States and Western Europe and is a common cause of HIV-associated pneumonia. Cryptococcus neoformans, the most common cause of meningitis in HIV-infected persons, often pre­sents with an associated pneumonia. The endemic fungi H. capsulatum, C. immitis, Penicillium marneffei, and, to a lesser extent, Blastomyces dermatitidis are among the chief causes of HIV-associated disease seen in their particular geographic regions, and all have important pulmonary presentations. Finally, invasive aspergillosis is often a devastating pulmonary disease seen in HIV-infected persons with severe immunodeficiency. Further information about fungal pulmonary diseases is provided in Chapters 37 and 38 .


Pneumocystis jirovecii


PCP (shorthand for P neumo c ystis p neumonia) remains the most common AIDS-defining opportunistic infection in the United States, even though its overall incidence has declined substantially. Early in the AIDS epidemic, PCP accounted for nearly two thirds of AIDS-defining diagnoses, and PCP developed in an additional 15% to 20% of patients at some time during their HIV disease. Two factors—the use of ART and Pneumocystis prophylaxis—have dramatically reduced the overall number of cases. Nevertheless, PCP remains a risk, chiefly among persons who are unaware of their HIV infection, those who fail to seek medical care, and those who fail to adhere to or respond to ART or Pneumocystis prophylaxis. Recent reports of increased PCP cases in immunosuppressed non-HIV populations have unclear implications for HIV-infected populations.


History and Epidemiology.


First identified early in the 20th century by Chagas, Pneumocystis was initially recognized as a human pathogen in the 1940s and 1950s. Despite more than a century of experience, several gaps in our understanding of this omnipresent organism remain. One major obstacle that has impeded further advances has been the inability to culture Pneumocystis in vitro. Although mammals are the only known hosts of Pneumocystis and a number of mammals are susceptible to infection, Pneumocystis has been demonstrated to be species-specific. Although Pneumocystis can be easily transmitted between mammals of the same species, animal studies that have attempted to transmit Pneumocystis from one species to another have been unsuccessful. In recognition of the host-species specificity of Pneumocystis, the Pneumocystis that causes Pneumocystis pneumonia in humans is now referred to as P. jirovecii, in honor of Otto Jirovec, the parasitologist who is credited with first identifying Pneumocystis as the cause of pulmonary disease in humans.


The precise ecologic niche for P. jirovecii and its mode of transmission is unknown. The inability to maintain human Pneumocystis in culture suggests that P. jirovecii may be unable to grow outside of its human host. Given its species specificity, it is doubtful that other mammals are the reservoir for P. jirovecii. This host-species specificity also implies coevolution of humans and P. jirovecii that, in turn, implies long-term carriage of P. jirovecii in its human host. Infection with P. jirovecii is almost ubiquitous. Up to 85% to 100% of the U.S. population have specific antibodies directed against P. jirovecii by the age of 3 years. Clearly, PCP never develops in most of these individuals. In the setting of severe immunosuppression, however, PCP can affect significant proportions of people. These studies and others suggest that reactivation of latent infection is a cause of P. jirovecii disease among immunocompromised hosts. Outbreaks of newly acquired infection after de novo exposure to persons with PCP have also been documented among immunocompromised patients in pediatric wards, cancer clinics or wards, transplantation units, and other confined spaces, which suggests person-to-person transmission of P. jirovecii and the rapid development of disease. Results of studies that have used molecular tools to detect Pneumocystis are consistent with transmission of P. jirovecii from patients with PCP to their household and hospital contacts, probably via inhalation of a Pneumocystis -containing aerosol.


Risk Factors.


Traditional risk factors for the development of PCP among HIV-infected adults include a CD4 + lymphocyte count less than 200 cells/µL, a history of PCP, and oropharyngeal candidiasis. In the current ART era, HIV-infected persons with a CD4 + lymphocyte count between 101 and 200 cells/µL, but a suppressed HIV RNA level on ART appear to be at low risk for primary PCP.


CD4 + Lymphocyte Count.


Approximately 95% of adolescent and adult cases of PCP are seen in HIV-infected patients whose CD4 + lymphocyte count is less than 200 cells/µL. The Multicenter AIDS Cohort Study found a markedly increased risk for PCP among HIV-infected subjects with a CD4 + lymphocyte count of 200 cells/µL or fewer at study entry. These subjects had a nearly fivefold greater risk of developing PCP than did subjects who had a CD4 + lymphocyte count higher than 200 cells/µL at study entry. This study also demonstrated that the presence of fever for 2 weeks or more and the development of thrush were independent predictors for PCP. Stansell and colleagues found that the incidence of PCP increased as the CD4 + lymphocyte count declined. Subjects with a CD4 + lymphocyte count between 101 and 200 cells/µL had an incidence of 5.95 cases of PCP per 100 person-years, whereas those with a CD4 + lymphocyte count of 100 cells/µL or fewer had 11.13 cases per 100 person-years. In persons on ART, the most recent CD4 + lymphocyte count is generally a better indicator of an HIV-infected person’s risk for PCP than their nadir CD4 + lymphocyte count.


Clinical Features.


HIV-infected adults, unlike other immunocompromised persons, usually have a prolonged prodromal illness associated with PCP. Kovacs and coworkers found the median duration of symptoms in 40 HIV-infected patients was 28 days, which was significantly longer than the median duration of 5 days seen in 37 patients with other underlying conditions ( P < 0.0002). Kales and colleagues reported a median duration of symptoms of 3 weeks in 145 HIV-infected patients with PCP and found that 72 patients (50%) had a duration of symptoms of 2 weeks or longer. This duration of symptoms can often be used to distinguish PCP from pyogenic pneumonia, which typically presents with 3 to 5 days of symptoms.


Classically, PCP presents with fever, a nonproductive cough, and dyspnea on exertion. Fever was noted in 86%, cough in 91%, and dyspnea in 95% in one series of 145 HIV-infected patients with PCP. High temperature, rigors, purulent sputum, and pleuritic chest pain are uncommon and can be used to distinguish PCP from pyogenic pneumonia. In a multivariable analysis, Selwyn and associates found that the presence of purulent sputum was an independent predictor of bacterial pneumonia, rather than of PCP or tuberculosis.


Physical examination of the chest may be normal. In the series reported by Kales and colleagues, 78 patients (54%) with PCP had a normal lung examination. When abnormal, the most frequent findings on lung auscultation are inspiratory crackles, which have been reported to be associated with a greater disease severity and an increased mortality. Oxygen desaturation with exertion is a sensitive but nonspecific indicator of PCP.


Numerous studies have shown that the serum lactate dehydrogenase (LDH) level is increased in patients with PCP; however, an elevated serum LDH level does not establish the diagnosis of PCP, nor does a normal serum LDH value rule out the diagnosis. Serum S -adenosylmethionine concentration and β-D-glucan, have also been reported to be potential diagnostic tools for PCP.


Imaging.


Classically, PCP presents with bilateral, symmetrical reticular opacities, often manifesting as peribronchovascular indistinctness ( eFig. 90-11 ), or as granular or a ground-glass appearance ( eFig. 90-12A ); these opacities typically begin in the perihilar region and extend outward as the disease severity increases. The ground-glass opacity appearance is particularly associated with the chest CT manifestations of this disorder (see eFig. 90-12B ). Occasionally, the opacities are unilateral or asymmetrical ( eFig. 90-13 ); a lobar or focal radiographic pattern is uncommon. Thin-walled cysts, or pneumatoceles, are seen in 10% to 20% of cases ( Fig. 90-4 ). Pneumatoceles may be present at the time of diagnosis or may develop during PCP therapy. Pneumatoceles may be single ( eFig. 90-14A ) or multiple ( eFig. 90-14B-D ), and small or large, and predispose patients to pneumothorax ( eFig. 90-15 ), which is another radiographic presentation of PCP. Usually pneumatoceles resolve (see eFig. 90-14B-D ), but occasionally they persist despite successful therapy.




Figure 90-4


Pneumocystis pneumonia.

Frontal chest radiograph of an HIV-infected patient with Pneumocystis pneumonia showing bilateral, predominantly perihilar, granular opacities and three pneumatoceles ( arrows ). Pneumatoceles may be single or multiple in number, and small or large in size, and may predispose patients to pneumothorax.

(Courtesy L. Huang.)


Virtually every possible radiographic finding, including focal, lobar, or segmental consolidation ( eFigs. 90-16 through 90-18 ), nodules ( eFig. 90-19 ) with or without cavitation, and a miliary pattern, can be seen occasionally. Apical or upper lung zone disease that mimics tuberculosis ( Fig. 90-5 , eFig. 90-20 ) is typically associated with aerosolized pentamidine prophylaxis, although this presentation can be seen as well in patients taking oral prophylaxis or no preventive therapy. Intrathoracic adenopathy and pleural effusions are rarely due to PCP. These radiographic findings should prompt a search for an alternate or at least a coexisting process such as bacterial pneumonia, tuberculosis, fungal pneumonia, or pulmonary KS. PCP may also present with a normal chest radiograph. Published studies report the incidence of a normal radiograph to range from 0% to 39%. In persons with a high clinical suspicion for PCP but a normal chest radiograph, chest high resolution CT can be useful.




Figure 90-5


Pneumocystis pneumonia.

Frontal chest radiograph of an HIV-infected patient who had been receiving aerosolized pentamidine showing reticular infiltration, predominantly of the upper lung zones, secondary to relapsed Pneumocystis pneumonia (PCP). This radiographic presentation, however, may also be seen in patients on no PCP prophylaxis.

(From Murray JF, Mills J: Pulmonary complications of HIV infection. Am Rev Respir Dis 141:1356–1372, 1582–1598, 1990.)


Diagnosis.


The diagnosis of PCP rests on the microscopic visualization of the characteristic P. jirovecii cysts or trophic forms (or both) on stained respiratory specimens. The standard methods for detection of P. jirovecii have been with cyst wall stains such as methenamine silver and toluidine blue-O or with Giemsa and Diff-Quik, which stain both the cysts and trophic forms; monoclonal antibodies to P. jirovecii are also used. PCR-based techniques have also been employed and have been reported to be more sensitive but also less specific than these other methods.


Pneumocystis jirovecii can be detected in expectorated (infrequently) or induced sputum; in pulmonary secretions obtained by endotracheal suction, BAL, or percutaneous aspiration of lung parenchyma; and in pulmonary tissue obtained by transbronchial, thorascopic, or open-lung biopsy. Of these, sputum induction and BAL are the most widely used. Sputum induction is an appropriate initial diagnostic procedure for PCP, but the sensitivity of induced sputum is less than 100% and thus a negative result should be followed by a more definitive procedure, namely bronchoscopy with BAL. BAL is performed in the most affected lobe visualized on chest radiograph. For patients with diffuse radiographic disease, BAL in the right middle lobe is usually performed. For patients with an upper lung zone predominance on radiograph, lavage in both an upper lobe and the right middle lobe should be considered. Despite the widespread use of TMP-SMX and dapsone for PCP prophylaxis, no studies have evaluated whether the yield of diagnostic studies is changed in persons receiving one of these medications. Clinical experience suggests that, although the severity of chest radiograph abnormalities may be milder in patients receiving prophylaxis than in those receiving no prophylaxis, the diagnostic yields from both sputum induction and BAL remain the same.


Treatment.


The treatment of choice for patients with mild, moderate, and severe PCP remains TMP-SMX administered for 21 days. TMP-SMX possesses many benefits, including availability in both an intravenous and an oral form, excellent oral bioavailability, and activity against many community-acquired bacteria that may cause concomitant pyogenic infection. The usual dose of TMP is 15 mg/kg/day (range, 15 to 20 mg/kg/day) and that of SMX is 75 mg/kg/day (range, 75 to 100 mg/kg/day), divided into three or four daily doses ( Table 90-3 ). Dosing may be intravenous (recommended for patients with moderate or severe PCP) or oral. Unfortunately, adverse effects from TMP-SMX are frequent in HIV-infected patients and include rash, fever, gastrointestinal complaints (nausea, vomiting), elevated transaminases, hyperkalemia, and bone marrow suppression, especially anemia and neutropenia. These effects often develop during the second week of therapy. In a significant proportion of HIV-infected patients, the side effects of TMP-SMX are ultimately treatment-limiting. Rare adverse reactions include Stevens-Johnson syndrome, toxic epidermal necrolysis, and a clinical syndrome resembling septic shock with hypotension, fever, pulmonary opacities, and renal and hepatic dysfunction. Studies are conflicting regarding the presence of potential TMP-SMX drug resistance in Pneumocystis .



Table 90-3

Treatment Regimens for Pneumocystis Pneumonia *




















































Treatment Regimen Dose(s), Frequency Toxicities
MILD PCP (PaO 2 >70 mm Hg AND Alveolar-arterial O 2 difference <35 mm Hg)
Trimethoprim-sulfamethoxazole (TMP-SMX) 15-20 mg/kg (TMP component) daily (q 6-8 hours) Fever, dermatologic, gastrointestinal, hematologic
Trimethoprim plus 15-20 mg/kg daily (q 6-8 hours) Dermatologic, gastrointestinal, hematologic
Dapsone 100 mg once daily
Clindamycin plus 1800 mg daily (q 6-8 hours) Dermatologic, gastrointestinal, hematologic
Primaquine 30 mg (base) once daily
Atovaquone 750 mg thrice daily Dermatologic, gastrointestinal
MODERATE-SEVERE PCP (PaO 2 ≤70 mm Hg OR Alveolar-arterial O 2 difference ≥35 mm Hg)
Trimethoprim-sulfamethoxazole (TMP-SMX) 15-20 mg/kg (TMP component) daily (q 6-8 hours) Fever, dermatologic, gastrointestinal, hematologic
Pentamidine 3-4 mg/kg IV once daily Renal, pancreatic
Clindamycin plus 1800-2400 mg (q 6-8 hours) Dermatologic, gastrointestinal, hematologic
Primaquine 30 mg (base) once daily

* Recommended duration of therapy = 21 days.


Oral route is preferred for patients with mild Pneumocystis pneumonia (PCP) who are treated as outpatients.


Intravenous (IV) route is preferred (at least until clinical improvement) for patients with moderate-severe PCP. Adjunctive corticosteroids (prednisone 40 mg, PO, twice daily for 5 days, then 40 mg, PO, once daily for 5 days, then 20 mg, PO, once daily for 11 days or potency-equivalent Solu-Medrol IV) should also be administered.



For patients with an allergy to or intolerance of TMP-SMX, alternative treatment regimens (see Table 90-3 ) include intravenous pentamidine, clindamycin plus primaquine, trimethoprim plus dapsone, and atovaquone. Aerosolized pentamidine should not be used for PCP treatment. Small reports describe successful PCP treatment with echinocandins. Similar to tuberculosis, HIV-infected patients with PCP who begin therapy for Pneumocystis and initiate ART may develop paradoxical reactions, usually a temporary exacerbation of clinical and radiographic features but occasionally respiratory failure, due to immune reconstitution. The diagnosis of IRIS is one of exclusion. Patients with paradoxical reactions only rarely require discontinuation of antiretroviral therapies, and symptomatic therapy is recommended.


Corticosteroid Therapy.


In 1990, the NIH-University of California Expert Panel for Corticosteroids as Adjunctive Therapy for Pneumocystis Pneumonia concluded that adjunctive corticosteroid therapy “can clearly reduce the likelihood of death, respiratory failure, or deterioration of oxygenation in patients with moderate-to-severe Pneumocystis pneumonia.” The panel recommended that corticosteroids be given to adults or adolescents with documented or suspected PCP if they have an arterial P o 2 level less than 70 mm Hg or an alveolar-arterial P o 2 difference greater than 35 mm Hg. Adjunctive corticosteroids, either oral prednisone or intravenous methylprednisolone, should be started at the same time as specific anti -Pneumocystis treatment is begun regardless of whether the diagnosis has been confirmed.


Early in the AIDS epidemic, it was realized that acute respiratory failure secondary to PCP, if severe enough to warrant mechanical ventilation, had a mortality rate of 86% or greater. Subsequent reports from individual hospitals confirmed this gloomy prognosis; 39 of 45 patients who were intubated and ventilated for PCP at San Francisco General Hospital from 1981 to 1985 died, a mortality rate of 87%. Although the mortality associated with acute respiratory failure secondary to PCP remains significant (approximately 30% to 50%), encouraging progress has been made. HIV-infected patients with PCP and respiratory failure requiring mechanical ventilation should be managed with lung-protective strategies as for patients with acute respiratory distress syndrome. At least one retrospective study by Morris and colleagues suggests that ART may be beneficial in these cases, although prospective randomized clinical trials in the intensive care unit involving mechanically ventilated patients are lacking.


Prevention of Exposure.


The natural reservoir for human Pneumocystis remains unknown. Both an environmental and a human reservoir have been suggested. Whether PCP results from reactivation of latent infection or also from a recent infection is debated; reports of cluster outbreaks of PCP among different immunocompromised populations support the theory that PCP can result from a recent exposure and person-to-person transmission. Accordingly, some authorities have argued that HIV-infected and other immunocompromised persons who are at risk for PCP should avoid close contact with any individuals who have PCP. However, the current CDC, NIH, and HIVMA/IDSA guidelines state “data are insufficient to support isolation as standard practice.”


Prevention of Disease.


HIV-infected adults or adolescents (including those on ART) who have a CD4 + lymphocyte count less than 200 cells/µL or a history of oropharyngeal candidiasis should receive primary Pneumocystis prophylaxis, and persons with prior PCP should receive secondary prophylaxis ( Table 90-4 ). Once started, HIV-infected adults and adolescents should remain on PCP prophylaxis, unless their CD4 + lymphocyte counts increase from less than 200 cells/µL to greater than 200 cells/µL for at least 3 months as a result of ART; several studies have demonstrated that primary and secondary PCP prophylaxis can be safely discontinued in the vast majority of these persons. In rare cases, however, PCP has recurred after the discontinuation of secondary PCP prophylaxis, despite an apparent ART-associated immune reconstitution.



Table 90-4

Prevention of Pneumocystis Pneumonia
























Prevention Regimens Alternative Dosing Comments
Trimethoprim-sulfamethoxazole 1 double-strength (DS) tablet daily 1 single-strength tablet daily or 1 DS tablet thrice weekly Also effective prophylaxis against Toxoplasma gondii and many bacterial pathogens
Dapsone 100 mg daily Combine with pyrimethamine and leucovorin in persons who are T. gondii immunoglobulin G antibody positive. Consider combining with pyrimethamine and leucovorin when used for secondary prophylaxis
Atovaquone suspension 1500 mg daily, or 750 mg twice daily Improved bioavailability compared to tablets
Aerosolized pentamidine 300 mg monthly via RespirGard II nebulizer May be associated with increased risk of extrapulmonary disease


TMP-SMX, dapsone, atovaquone suspension, and aerosolized pentamidine are the standard options for PCP prophylaxis. TMP-SMX is the first-line choice for primary and secondary prophylaxis against Pneumocystis. For those patients intolerant of TMP-SMX, dapsone and atovaquone are both oral drugs that can be used. Many authorities would add pyrimethamine to these drugs for patients with a history of PCP, a CD4 + lymphocyte count less than 100 cells/µL, or both. For patients who are T. gondii immunoglobulin G antibody positive, pyrimethamine should be added. Aerosolized pentamidine remains an effective and well-tolerated prophylaxis option; however, caution must be exercised when using this drug for secondary prophylaxis or in patients with a CD4 + lymphocyte count less than 100 cells/µL because prophylaxis break-through may be more frequent in this population.


Cryptococcus Species


Cryptococcosis is a disease caused by C. neoformans or C. gattii. Cryptococcus is the only encapsulated fungus that infects humans, either healthy or immunocompromised, and India ink or mucicarmine staining can identify its polysaccharide capsule. C. neoformans and C. gattii can be subclassified into four serotypes, based upon capsular agglutination reactions, and two species with two varieties. Cryptococcus infection is transmitted after inhalation of a yeast-containing aerosol. C. neoformans is global in distribution, and is most commonly isolated from bird excrement, decaying fruit, and soil. Most cryptococcal disease in HIV-infected persons is due to C. neoformans . Cryptococcus gattii has been isolated from trees in Australia, as well as the Pacific Northwest of the United States and Southwestern Canada, where outbreaks of disease have been reported. C. gattii is more likely to infect immunocompetent hosts, although it also causes disease in HIV-infected individuals.


The incidence of cryptococcosis in HIV-infected persons has dramatically declined since the introduction of ART. In one population-based surveillance study, the annual incidence of cryptococcosis decreased from 66 per 1000 persons with AIDS in 1992 to 7 per 1000 persons in 2000 in the Atlanta area, and from 24 per 1000 persons with AIDS in 1993 to 2 per 1000 persons in 2000 in the Houston area. Using national surveillance data, a study from France found a 46% decrease in the incidence of cryptococcosis in HIV-infected patients from the pre–combination ART period (1985-1996) to the early combination ART period (1997-2001).


Despite a declining incidence, exposure to Cryptococcus and cryptococcal disease is increasingly recognized in Africa and Southeast Asia. In a study from Uganda, Cryptococcus was isolated from 11% of BAL specimens in patients who presented with cough of greater than 2 weeks duration. Although cryptococcal pneumonia can be a rare cause of mortality in this population, not all cases in which cryptococcus was isolated were treated with antifungal therapy, raising the possibility of colonization or isolated infection. Cryptococcal antigen (CRAG) testing suggests that cryptococcal infection may be more widespread than previously appreciated. In a study from Thailand of patients hospitalized with acute respiratory symptoms, 13% of HIV-infected patients compared to none of the HIV-uninfected patients had a positive CRAG test. The proportion of HIV-infected patients with a positive CRAG in other similar studies ranged from 5% to 11%; the likelihood of a positive test increases among patients with CD4 + cell counts less than 100 cells/µL.


Clinical Features.


Although the portal of entry is the lung, cryptococcal pulmonary infection is often asymptomatic or minimally symptomatic, and the most commonly encountered manifestation of cryptococcal disease is meningitis. In a large series of 106 HIV-infected patients with cryptococcal disease, 89 patients (84%) had meningitis, and only 4 patients (4%) had isolated pneumonia. In a population-based surveillance study that included 1322 HIV-infected patients with cryptococcal disease, only 45 patients (3%) had pulmonary disease in the absence of both fungemia and meningitis. Autopsy studies suggest that pulmonary disease is underdiagnosed, particularly in settings where diagnostic facilities are limited.


When present, the most frequent respiratory complaints are cough and dyspnea. Pleuritic chest pain and productive cough have also been reported, perhaps distinguishing pulmonary cryptococcosis from PCP. Although more rare, cases of acute respiratory failure have been reported.


CD4 + Lymphocyte Count.


In most cases, cryptococcosis infects patients with a CD4 + lymphocyte count less than 200 cells/µL, and usually less than 100 cells/µL. A study of 1644 patients reported a median CD4 + count of 24 cells/µL with a range of 0 to 480 cells/µL. A study from Uganda from the current ART era reported a median CD4 + count of 23 cells/µL.


Imaging.


Cryptococcal pneumonia most commonly presents with diffuse bilateral interstitial opacities. In one study, Meyohas and associates reported interstitial opacities in 60 of 92 radiographs (65%) ( eFig. 90-21 ); in addition, focal consolidation (13%) ( eFig. 90-22 ), nodular opacities (11%) ( eFig. 90-23 ), cavitation (11%) ( Fig. 90-6 ), pleural effusion (14%), and hilar adenopathy (27%) ( eFig. 90-24 ) were noted. More rarely, radiographic findings include a miliary pattern ( eFig. 90-25 ), solitary pulmonary nodules, pulmonary masses, isolated pleural effusion, and pneumothorax. Finally, cryptococcal pneumonia may also present with a normal chest radiograph; in the review by Meyohas and associates, for example, radiograph results were normal in 11% of the 92 cases.




Figure 90-6


Cryptococcal infection.

Frontal chest radiograph of an HIV-infected patient showing a solitary cavitary lesion in the right lower lung field. Culture of bronchoalveolar lavage fluid revealed Cryptococcus neoformans. In HIV-infected patients, cryptococcal disease can present with a wide range of chest radiographic findings, including a normal chest radiograph.

(From Stansell JD: Fungal disease in HIV-infected persons: cryptococcosis, histoplasmosis, and coccidioidomycosis. J Thorac Imaging 6:28–35, 1991.)


Diagnosis.


The diagnosis of cryptococcal infection begins with the CRAG test. The test can be performed on serum, cerebrospinal fluid, urine, BAL fluid, or pleural fluid. The serum CRAG test is sensitive and specific for cryptococcemia. A negative serum CRAG test virtually rules out the diagnosis of cryptococcal meningitis, but can be seen in some cases of isolated pulmonary cryptococcosis. A positive serum CRAG test should prompt an evaluation for disseminated disease, especially meningitis, but false-positive results can be seen in the presence of rheumatoid factor or infection with the fungus Trichosporon asahii (formerly T. beigelii ), or with bacteria from the Stomatococcus or Capnocytophaga genera . As noted, a positive CRAG can sometimes be found in individuals without clinical evidence of disease. Blood fungal cultures are specific and should be obtained as part of the diagnostic evaluation. New cutaneous lesions may be signs of dissemination, and their sudden appearance should prompt consideration for biopsy.


The diagnosis of pulmonary cryptococcosis is usually established by culture of sputum or BAL fluid and occasionally of pleural fluid. Specimens from TBB and pleural biopsy can also be diagnostic. Batungwanayo and coworkers found that BAL diagnosed 27 of 33 cases (sensitivity 82%) of cryptococcal pneumonia compared with TBB, which diagnosed 10 of 21 cases (sensitivity 48%). In some cases, cultures are negative, but a BAL or pleural fluid CRAG test can establish the diagnosis. Alternatively, because the treatment is identical, the diagnosis of pneumonia may be inferred in the presence of disseminated cryptococcal disease (e.g., meningitis) and a compatible radiographic presentation. However, caution must be exercised with this approach, because other opportunistic infections such as PCP may be present concurrently and can demonstrate identical radiographic findings.


Treatment.


In contrast to cryptococcal meningitis, there are no randomized controlled trials for HIV-infected patients presenting with isolated cryptococcal pneumonia or with concurrent cryptococcal pneumonia and meningitis. Some authorities would treat isolated cryptococcal pneumonia that is mild in severity with fluconazole alone (400 mg daily for 12 months) in combination with effective ART ; however, patients with clinically significant cryptococcal pneumonia should be considered at high risk for early deterioration and treated similarly to those with disseminated disease using the lipid formulation of amphotericin B (3 to 4 mg/kg/day) in combination with flucytosine for at least 2 weeks. Treatment should be continued until the patient is clinically improved, at which point the patient can be switched to fluconazole (400 mg daily) to complete at least an 8-week course. The patient should then be maintained on fluconazole (200 mg daily) to complete at least 12 months of azole therapy. As in patients with tuberculosis or PCP, IRIS may develop in HIV-infected patients with cryptococcosis who begin dual therapy for C. neoformans and HIV infection. Patients who had cryptococcal meningitis may present with aseptic meningitis and have elevated intracranial pressure. In addition, patients with cryptococcal pneumonia and nodules on chest radiograph may develop cavitation of their nodules or new intrathoracic adenopathy.


Prevention.


There are no specific recommendations regarding exposure avoidance or chemoprophylaxis for C. neoformans (e.g., fluconazole) . In addition, routine screening of asymptomatic persons with serum CRAG testing is not recommended.


Histoplasma capsulatum


Histoplasmosis is a disease caused by the dimorphic, soil-dwelling fungus H. capsulatum. The fungus is found on all continents except Antarctica, but it is most endemic to North America and the Caribbean basin. The heaviest concentration is found in the Mississippi, Ohio, and St. Lawrence River valleys. In these areas, the fungus exists in microenvironments related to the enrichment of the soil with bird or bat excrement, which helps promote sporulation. In the soil, the fungus exists in a mycelial form producing asexual spores, the characteristic tuberculate macroconidia and microconidia. Microconidia are easily aerosolized when disturbed and inhalation leads to primary pulmonary infection that is usually clinically silent.


Once deposited in the alveoli, H. capsulatum transforms into its yeast (or parasitic) form, and an area of pneumonitis develops. During this period before cell-mediated immunity develops, the organism spreads to regional lymph nodes and to reticuloendothelial organs. Two to 3 weeks after exposure, a flulike syndrome with fever, chills, myalgias, a nonproductive cough, and chest pain develops in about 40% of immunocompetent individuals. As emphasized in Chapter 37 , 99 % of these cases spontaneously resolve with the development of specific cell-mediated immunity. In contrast, progressive disseminated histoplasmosis develops in HIV-infected and other persons lacking cell-mediated immunity. Although most cases of HIV-associated histoplasmosis appear to result from de novo exposure, the disease can reactivate. These cases account for the histoplasmosis seen in nonendemic areas such as San Francisco or New York.


Clinical Features.


Although the portal of entry is the lung, in HIV-infected persons, histoplasmosis most often presents as a febrile wasting illness with disseminated infection. In a large series of 72 HIV-infected patients with disseminated histoplasmosis, 69 patients (96%) presented with fever and weight loss; in approximately 10% of cases, the presentation was dramatic with a sepsis-like syndrome associated with hypotension, respiratory failure, liver and renal failure, and coagulopathy. Prognosis in these patients is poor. Respiratory complaints at presentation, chiefly cough and dyspnea, are found in patients who are likely to have abnormal chest radiographs.


CD4 + Lymphocyte Count.


Most cases of disseminated histoplasmosis are seen in patients with a CD4 + lymphocyte count less than 100 cells/µL and often less than 50 cells/µL. Isolated respiratory disease is more likely in patients with CD4 + cell counts greater than 300 cells/µL. Frequent laboratory findings include anemia, leukopenia, thrombocytopenia, and liver function test elevations. Serum LDH and serum ferritin elevations, often pronounced, have also been reported.


Imaging.


Disseminated histoplasmosis presents with a normal chest radiograph in 35% to 55% of cases. In patients with abnormal findings, the most frequent are diffuse, coarse reticular or reticulonodular opacities as shown in Figure 90-7 . Occasionally, alveolar opacities are present ; focal opacities are less common and seen in 7% to 11% of cases. Hilar and mediastinal adenopathy and calcified granulomata are each found in less than 5% of patients, attesting to the low incidence of reactivation disease.




Figure 90-7


Disseminated fungal infection.

Frontal chest radiographic close-up of the left mid-lung of an HIV-infected patient showing a medium to coarse reticulonodular pattern characteristic of disseminated fungal disease.

(From Stansell JD: Fungal disease in HIV-infected persons: cryptococcosis, histoplasmosis, and coccidioidomycosis. J Thorac Imaging 6:28–35, 1991.)


Diagnosis.


The workup of histoplasmosis begins with the histoplasma antigen test, which is a sensitive method for rapid diagnosis of disease. The antigen test can be performed on urine, serum, cerebrospinal fluid, or BAL fluid. The performance of the newer quantitative histoplasma antigen test is excellent; in AIDS patients with disseminated histoplasmosis, the antigen was detected in the urine of 100% and in the serum of 92% of patients. In isolated, chronic pulmonary disease, serum or urine antigen testing is less sensitive. In patients with pulmonary involvement, antigen testing of BAL fluid combined with cytopathology evaluation can complement serum or urine testing and increase the sensitivity for diagnosis of disease. With successful therapy, antigen values fall and, during relapses, antigen values rise; thus changes in antigen values can be useful for assessing response to therapy and for evaluating possible relapse. A persistently positive histoplasma antigen test indicates continued disease and warrants continued therapy. False-positive histoplasma antigen test results have been seen in patients with other disseminated fungal diseases (blastomycosis, coccidioidomycosis, paracoccidioidomycosis, and penicilliosis, and rarely with aspergillosis), but such results have not been described in patients with cryptococcosis or candidiasis. Histoplasma can also be cultured from involved sites, although results may not be available for several weeks. Blood fungal cultures are specific and should be obtained as part of the diagnostic evaluation. Wheat and colleagues reported that fungal blood cultures were positive in 65 of their 72 cases (90%). Occasionally, the peripheral blood smear reveals intracellular yeast. Other potential diagnostic sources include bone marrow, lymph node, and skin. Serologic tests are generally less helpful in those with disseminated disease, but may be useful in patients who are less severely immunocompromised with isolated pulmonary disease.


Treatment.


A lipid formulation of amphotericin B is the treatment of choice for HIV-infected patients with moderate to severe disseminated histoplasmosis, whereas itraconazole is an alternative for patients with mild disease. Treatment with amphotericin B should be continued for at least 2 weeks or until the patient is clinically improved, at which point the patient can be switched to itraconazole to complete at least a 12-month course. Patients with isolated pulmonary disease and CD4 + cell counts greater than 300 cells/µL should be treated similarly as for non–HIV-infected patients ( Chapter 37 ).


Prevention of Exposure.


In areas endemic for H. capsulatum, HIV-infected patients, especially those with CD4 + lymphocyte counts less than 150 cells/µL, should avoid activities that would potentially increase their exposure, including cleaning chicken coops, disturbing native soil beneath bird-roosting sites, cleaning, remodeling, or demolishing old buildings, and exploring caves.


Prevention of Disease.


Routine serologic testing, even in histoplasmosis-endemic areas, is not recommended. Itraconazole for primary H. capsulatum prophylaxis can be considered in those who have a CD4 + lymphocyte count less than 150 cells/µL and who are at high-risk due to occupation or who live in hyperendemic regions.


Coccidioides immitis


Coccidioidomycosis is caused by the dimorphic, soil-dwelling fungi C. immitis and Coccidioides posadasii. As described in Chapter 37 , the chief manifestation of coccidioidomycosis is as a mild flulike, often undiagnosed illness, from which most previously healthy people spontaneously recover. Chronic pulmonary disease manifests in roughly 5% of infected persons and disseminated disease in even fewer. Manifestations of coccidioidomycosis in HIV-infected persons include focal or diffuse pneumonia, cutaneous disease, meningitis, liver or lymph node involvement, or disseminated disease that can be fatal. The fungus is endemic to the semiarid regions of North America, notably the southwestern United States (central California, southern Arizona, southern New Mexico, and west Texas) and also northern Mexico. California’s southern San Joaquin Valley and southern Arizona are hyperendemic areas. Coccidioides is also found in South America, especially central Argentina. In soil, the fungus exists in a mycelial form with characteristic arthrospores. When the soil is disrupted, the few micrometer–diameter arthrospores, which are just the right size for deposition in the distal airways and alveoli after inhalation, become airborne. After deposition in the lungs, the arthrospores transform into spherules that may develop several hundred endospores. Rupture of the spherules allows widespread dissemination of the endospores, which then form additional spherules, replicating the cycle. Although most cases of HIV-associated coccidioidomycosis appear to result from de novo inhalation, disease may also reactivate.


Clinical Features.


The clinical presentation is often nonspecific; fever and chills (68%), night sweats (36%), and weight loss (50%) are all common. Although the portal of entry is the lung, coccidioidomycosis can present with disseminated disease and meningitis. Other frequently involved sites include the skin, lymph nodes, liver, and skeletal system. In one study, 42% of HIV-infected patients with coccidioidomycosis presented with disseminated disease; overall, 25% of the patients in this series died. Focal pneumonia can present similarly to bacterial pneumonia, and diffuse pulmonary involvement can be difficult to distinguish from PCP.


CD4 + Lymphocyte Count.


Most cases of disseminated coccidioidomycosis are seen in patients with a CD4 + lymphocyte count less than 100 cells/µL and often fewer than 50 cells/µL. Focal pneumonia is more common when the CD4 + cell count is greater than 250 cells/µL. Cerebrospinal fluid examination is warranted in all patients with suspected disseminated coccidioidomycosis.


Imaging.


In a series of 91 HIV-infected patients with coccidioidomycosis, diffuse reticulonodular opacities (see Fig. 37-5 ) were seen in 65%; focal opacities were less common, in 14% of cases, and consisted of focal opacities, single or multiple nodules, and cavities. A miliary pattern may be seen ( eFig. 90-26 ). Pleural effusion and hilar adenopathy as well as normal chest radiographs have all been reported.


Diagnosis.


Serologic tests are useful in the evaluation of suspected coccidioidomycosis. Several studies have found an 80% to 90% sensitivity of complement fixation and tube precipitin tests. Sensitivity of enzyme-linked immunoassays may be higher, but may be less specific. False-negative titers usually arise in the most severely immunocompromised patients with diffuse pulmonary disease; in those with positive tests, the titer appeared to reflect the disease activity and proved useful for monitoring response to therapy. A urinary and serum antigen test specific to coccidioides has been developed and is useful for diagnosis of severe cases of coccidioidomycosis; however, other endemic fungi, including Histoplasma or Blastomyces , can cross-react.


A definitive diagnosis can be established by isolation and identification of the fungus by culture or identification of pathognomonic giant spherules in cytologic or histologic preparations. In cases of suspected coccidioidomycosis, it is critical to alert the microbiology laboratory so that proper precautions can be implemented to prevent laboratory transmission. Direct examination and culture of sputum, BAL fluid, or TBB can establish the diagnosis of pulmonary coccidioidomycosis in HIV-infected patients. Singh and associates found that sputum culture in 13 of 19 cases (68%) and cytology in 8 of 11 cases (73%) had a high yield. BAL fluid culture diagnosed 29 of 42 cases (69%), and BAL fluid cytology diagnosed 32 of 48 cases (67%); in addition, TBB culture diagnosed 8 of 10 cases (80%), and biopsy histology diagnosed all 14 cases (100%).


Treatment.


Either amphotericin B or a lipid formulation of amphotericin B is the treatment of choice for HIV-infected patients with severe (i.e., diffuse) pulmonary or disseminated coccidioidomycosis. Treatment with amphotericin B should be continued until the patient is clinically improved, at which point the patient can be switched to fluconazole or itraconazole. Monitoring complement-fixing antibody titers every 12 weeks can be useful to assess response to therapy. In patients with clinically mild infection, such as focal pneumonia, fluconazole or itraconazole may be used throughout. Although clinical experience is limited, voriconazole or posaconazole may be considered in refractory coccidioidomycosis. In patients with severe, diffuse disease, lifelong suppressive therapy with fluconazole or itraconazole should be continued after initial treatment is complete, particularly in those with meningeal involvement because the risk of relapse is high. In patients who have had focal coccidioidal pneumonia, and who have had a sustained response to ART with CD4 + lymphocyte counts greater than 250 cells/µL, therapy may be discontinued after 12 months, but periodic surveillance with chest radiograph and coccidioides serology is recommended.


Prevention of Exposure.


In areas endemic for C. immitis, HIV-infected persons should avoid activities that will potentially increase their exposure, such as visits to construction or other sites where soil is being disturbed.


Prevention of Disease.


Routine serologic testing is reasonable only in endemic areas. Pre-emptive treatment for persons living in or who have traveled to an endemic area is recommended only if the serology test result is newly positive and CD4 + lymphocyte count is less than 250 cells/µL.


Aspergillus Species


Species of Aspergillus are found worldwide, and exposure is universal. Nevertheless, disease is infrequent unless phagocyte number or function is reduced. Presently, greater than 180 species within the Aspergillus genus have been identified. Aspergillus fumigatus, however, is the most common disease-causing species, and it accounts for approximately 90% of cases of invasive aspergillosis. Although invasive aspergillosis is a well-documented complication of various immunosuppressive disorders, particularly in patients with hematologic malignancy or organ transplantation, it is an uncommon problem in patients with HIV disease. Risk factors for the development of aspergillosis other than HIV-induced immunosuppression included use of corticosteroids, neutropenia, marijuana use, and broad-spectrum antimicrobial drugs. In the ART era, the incidence of invasive aspergillosis has further declined. Holding and coworkers reported an aspergillosis incidence of 3.5 cases per 1000 person-years among HIV-infected individuals enrolled in the CDC-led Adult and Adolescent Spectrum of HIV Disease study.


Clinical Features.


The entire spectrum of Aspergillus -related lung disease ( Chapter 38 ) has been observed in HIV-infected persons, from colonization of the respiratory tract or a preexisting cavity, to tracheobronchitis or obstructing bronchial aspergillosis, to invasive aspergillosis, by far the most severe manifestation. Although most patients with invasive aspergillosis have a CD4 + lymphocyte count lower than 100 cells/µL, the classic risk factors for the disease relate more to phagocyte number and function (as determined by neutropenia and/or monocytopenia and use of corticosteroids or broad-spectrum antibiotics) than to absolute CD4 + lymphocyte counts. Patients with aspergillosis typically present with fever, cough, dyspnea, and occasionally pleuritic chest pain. Hemoptysis is another presenting feature. The imaging findings are variable and include unilateral or bilateral opacities, cavitary lesions ( eFigs. 90-27 and 90-28 ), nodular (see eFig. 90-28 ) and pleural-based opacities, and pleural effusions.


Diagnosis.


The definitive diagnosis of aspergillosis requires both demonstration of tissue invasion and isolation of the organism by culture. Neither sputum nor BAL is sufficient. Microscopy alone cannot distinguish Aspergillus species from Fusarium or Pseudallescheria species. The results of TBB are usually negative, but specimens from sputum, BAL fluid, or percutaneous aspirates are often positive on culture. The absence of histologic proof of tissue invasion is always somewhat disquieting, especially in attempting to distinguish invasive disease from possible colonization of damaged airways. Repeated isolation of the fungus in large numbers with a compatible clinical setting makes the diagnosis more tenable.


Galactomannan antigen testing on serum or BAL specimens is useful in the diagnosis of invasive pulmonary aspergillosis in other immunocompromised patients (see also Chapters 17 and 38 ), although its performance has not been prospectively evaluated in HIV-infected patients with suspected aspergillosis. Several other fungi, including Histoplasma and Blastomyces, can cross-react.


Treatment.


Compared with the fungi previously discussed, there is relatively little experience treating aspergillosis in HIV-infected patients. Voriconazole is the first-line recommended therapy. Alternative agents include amphotericin, caspofungin, and posaconazole. Interactions with ART drugs must be considered because the antifungal azoles inhibit the cytochrome P450 system. Even with the prompt institution of therapy, the prognosis is poor, largely because aspergillosis is nearly always a late complication of advanced HIV disease.


Prevention of Exposure.


Given the ubiquitous nature of Aspergillus species, it is impossible to prevent exposure to the organism. However, patients with advanced HIV disease should avoid activities that will potentially increase their exposure, including being near decaying vegetation (e.g., compost) and soil.


Prevention of Disease.


There are no specific recommendations regarding chemoprophylaxis for Aspergillus species.


Blastomyces dermatitidis


Blastomycosis is a disease caused by the endemic, dimorphic fungus Blastomyces dermatitidis. Blastomycosis is co-endemic with histoplasmosis throughout much of the central United States ( Chapter 37 ), but it is less common than histoplasmosis, and reports of HIV-associated blastomycosis are infrequent.


Clinical Features.


The largest case series reported 15 cases of HIV-associated blastomycosis; all but one patient had a CD4 + lymphocyte count less than 200 cells/µL. The authors noted two distinct patterns of disease: one group of patients had disease that was clinically limited to the respiratory system, whereas the other group had disseminated blastomycosis, commonly involving multiple organ systems, including the lungs. Eleven of 15 patients (73%) had abnormal chest radiographs, with diffuse interstitial or miliary disease (55%) as the most common radiographic finding. Definitive diagnosis requires the growth of B. dermatitidis, although visualization of the characteristic budding yeast form is strongly suggestive and therefore warrants antifungal therapy while awaiting the results of culture.


Treatment.


Intravenous amphotericin B is the treatment of choice for HIV-infected patients with severe disease. Treatment with amphotericin B should be continued until there is clinical improvement; patients can then be switched to oral itraconazole maintenance therapy for at least 12 months, with itraconazole continued indefinitely for AIDS patients without immune reconstitution. With prompt institution of therapy, most patients with disease limited to the lungs respond well; in contrast, patients with disseminated disease do poorly (40% mortality in 30 days).


Penicillium marneffei


Penicilliosis is a disease caused by P. marneffei, a dimorphic, soil-dwelling fungus. P. marneffei is endemic in southeastern Asian countries, and, in northern Thailand, it is the third most common opportunistic infection (after tuberculosis and cryptococcosis) in HIV-infected patients with AIDS, accounting for 15% to 20% of all AIDS-related illnesses. Disease is related to soil exposure, especially during the rainy season (May to October), and infection is probably acquired via inhalation.


Clinical Features.


Most cases of penicilliosis are seen in patients with a CD4 + lymphocyte count less than 100 cells/µL. The clinical presentation is often mistaken for tuberculosis, cryptococcosis, or histoplasmosis. The most common symptoms include fever, weight loss, cough, and generalized papular skin lesions, usually with central umbilication. Symptoms are often present for weeks. In addition to the cutaneous findings, physical examination often reveals peripheral lymphadenopathy and hepatomegaly. Anemia is a prominent laboratory finding.


Diagnosis.


P. marneffei is most commonly a disseminated disease in HIV-infected patients, and the diagnosis is usually made by fungal blood cultures. Other involved sites include the skin, lymph nodes, bone marrow, and lungs. In contrast to other Penicillium species that cause disease in humans, P. marneffei converts to a yeast form in its host, and yeast-laden macrophages can often be seen in peripheral blood, bone marrow aspirates, and touch preparations from tissue biopsies.


Treatment.


Amphotericin B, followed by itraconazole, is the standard treatment for P. marneffei . Mild forms of disease can be treated initially with itraconazole. The duration of amphotericin B treatment is 2 weeks, followed by an additional 10 weeks of itraconazole. This regimen is reported to have a greater than 97% response rate for disseminated P. marneffei infection. Voriconazole is an alternative drug for primary treatment. Without secondary prophylaxis, most patients will suffer a relapse within 6 to 12 months. Secondary prophylaxis can be discontinued in patients who are started on ART and have a sustained CD4 + lymphocyte count more than 100 cells/µL for more than 6 months.


Prevention of Exposure.


Given the strong association with soil exposure, especially during the rainy season, HIV-infected patients living in endemic areas should avoid activities that will potentially increase their exposure. Current CDC, NIH, and HIVMA/IDSA guidelines suggest that HIV-infected patients avoid visiting endemic areas if possible.


Prevention of Disease.


Itraconazole is recommended as primary prophylaxis against P. marneffei for subjects in endemic areas who have CD4 + cell counts less than 100 cells/µL. Fluconazole is a second-line alternative.


Candida Species


Despite the high frequency of mucocutaneous candidiasis in HIV-infected patients, pulmonary candidiasis is distinctly uncommon and is rarely diagnosed during life. Because of the small number of documented cases of pulmonary candidiasis, neither the clinical features nor the treatment is well established. Tissue invasion must be demonstrated by biopsy for a convincing diagnosis; the mere identification of the fungus in respiratory secretions by culture alone is insufficient.


Viruses


Many viruses are known to cause pulmonary disease in immunosuppressed persons; however, only CMV is regarded as a potentially important agent of pulmonary disease in HIV-infected persons.


Cytomegalovirus


CMV, a double-stranded DNA virus in the Herpesvirus family, is described in Chapter 32 . The risk of exposure to CMV increases with age, and evidence of CMV infection is extremely common in healthy persons. In HIV-infected patients, CMV disease is believed to result chiefly from reactivation of latent infection; however, disease has been documented from de novo infection in recipients of solid organs, bone marrow, and blood, which raises the possibility that new infection or superinfection from exogenous sources may also arise in HIV-infected persons. Although clearly the source of significant pathologic conditions in the retina, gastrointestinal tract, and nervous system, the role of CMV in producing pulmonary disease in HIV-infected persons is open to question. Many consider this ubiquitous virus a “passenger” rather than a pathogen in most instances. However, there are also clear instances in which documented pulmonary disease results from CMV.


Clinical Features.


Retinitis and gastrointestinal disease are the two most common forms of HIV-associated CMV disease. CMV is a frequent isolate from the BAL fluid of patients with advanced immunosuppression who undergo evaluation for opportunistic infections, notably Pneumocystis. Because CMV is shed in respiratory secretions, its mere presence in BAL fluid cannot be construed as diagnostic of CMV pulmonary disease. When dual pulmonary infection is discovered, treatment directed against the coexisting disease and not against CMV usually results in clinical resolution ; however, sometimes CMV causes pulmonary disease, and the challenge for clinicians is to recognize these instances.


The most common symptoms of CMV pneumonia are cough, dyspnea, and fever. In a study by Salomon and associates, these symptoms were seen in 94%, 94%, and 89%, respectively, of the 18 patients reported. Respiratory symptoms were present for up to 2 weeks in 50% and between 2 and 4 weeks in an additional 44%.


CD4 + Lymphocyte Count.


Most cases of CMV disease are seen in patients with a CD4 + lymphocyte count less than 50 cells/µL. In one study of 18 patients with biopsy-proven CMV pneumonia, the median CD4 + lymphocyte count was 4 cells/µL. The serum LDH has been reported to be elevated in CMV pneumonia.


Imaging.


The imaging findings of CMV pneumonia vary and include reticular or ground-glass, alveolar, and nodular opacities ( eFig. 90-29 ). Pleural effusions may be seen as well.


Diagnosis.


When CMV pulmonary disease is suspected in conjunction with other end-organ disease (e.g., retinitis), CMV therapy must be initiated immediately. CMV is usually a disseminated disease and commonly involves multiple organ systems simultaneously. Treatment of CMV disease in one end-organ treats all affected organs, although the length of therapy can differ by organ system. The therapeutic dilemma is much greater when only the lungs appear to be afflicted. The only precise criterion for diagnosis of CMV pulmonary disease is the demonstration of widespread specific cytopathic changes in the lungs. Neither culture of BAL fluid nor cytopathic inclusions on TBB specimens are sufficient to make the diagnosis of CMV pneumonitis. Patients suspected of having CMV pneumonitis should undergo a careful dilated retinal examination performed by an experienced ophthalmologist, even if there are no ocular complaints.


Treatment.


Data for treatment of CMV pneumonia in HIV-infected patients are limited. Intravenous ganciclovir or foscarnet is recommended for severe pneumonitis. Although oral valganciclovir has been suggested for less severe pneumonitis, there are no data regarding its use in this setting. With these drugs, an initial course of induction therapy should be continued until there is clinical improvement; the length of induction therapy for CMV pneumonia, however, is undetermined. A 21-day course for isolated CMV pneumonitis has been recommended. The usefulness of maintenance therapy in preventing a relapse of CMV pneumonitis is unclear.


Prevention of Exposure.


HIV-infected patients who are CMV immunoglobulin G negative should be given CMV-negative blood in the event that a transfusion is necessary.


Prevention of Disease.


CMV disease is best prevented by maintaining CD4 + lymphocyte counts greater than 100 cells/µL with ART.


Other Viruses


Symptomatic pulmonary viral infection other than CMV is uncommon. Herpes simplex virus, while frequently cultured from BAL fluid, is usually a contaminant from upper airway carriage. Lower respiratory tract involvement is rare in HIV-infected persons (0.2% to 4% of autopsy cases) and appears to be more frequent in other immunosuppressed hosts. Herpes simplex virus pulmonary disease causes a focal pneumonia or a diffuse interstitial pneumonitis. The focal pneumonias appear to result from contiguous spread of herpes simplex virus to the lung parenchyma and are often associated with necrotizing tracheobronchitis, whereas the diffuse interstitial pneumonias appear to be a manifestation of hematogenous herpes simplex virus dissemination. Varicella-zoster virus is a rare cause of pneumonia in HIV-infected adult patients. Epstein-Barr virus DNA has been identified in lung biopsy specimens of infants and children with lymphocytic interstitial pneumonitis, but the precise role of the virus in this disease is uncertain.


Influenza-related mortality is increased in adults with AIDS compared to the general population in studies using data from the United States as well as from sub-Saharan Africa. Influenza-related mortality is decreased in the ART era, but remains greater in those with AIDS compared to the general U.S. population. Influenza generally presents similarly and has a comparable clinical course in HIV-infected as in non–HIV-infected adults. Studies focused on H1N1 influenza generally confirmed these findings, although HIV-infected patients with more advanced HIV disease had a poorer prognosis than those with well-controlled HIV. Prompt initiation of antiviral therapy directed against influenza in HIV-infected patients with suspected or confirmed disease is recommended. All HIV-infected persons should receive the inactivated influenza vaccine annually.


Parasites


Of the classic unicellular and multicellular parasites that afflict otherwise healthy humans ( Chapter 39 ), several also cause pulmonary disease in HIV-infected hosts. Of these, T. gondii is the most frequent.


Toxoplasma gondii


Toxoplasmosis is a zoonosis caused by the intracellular protozoan T. gondii, with domestic cats as its definitive host, but with an infectious reservoir that encompasses all animals. Infection is transmitted to humans when raw or undercooked meat containing T. gondii is eaten. Domestic cat feces in litter boxes are an additional source of potential T. gondii infection. Infection can be transmitted vertically from mother to fetus. In HIV-infected patients, the majority of cases of toxoplasmosis result from reactivation of chronic, latent infection. Thus, the keys to preventing toxoplasmosis are prevention of exposure for those not yet infected and prophylaxis for those already seropositive. The seroprevalence of Toxoplasma antibodies varies. In the United States, the seroprevalence is 10% to 50%, whereas in Western Europe, the seroprevalence may be as high as 90%.


Clinical Features.


Central nervous system complications of T. gondii are well recognized in HIV disease and include encephalitis as well as focal brain abscess. Pulmonary involvement is uncommon, but is seen in patients with central nervous system or disseminated disease, can present as isolated pneumonia, and can rarely be associated with acute respiratory distress syndrome.


CD4 + Lymphocyte Count.


Toxoplasmosis presents at the lower range of CD4 + lymphocyte counts. In a large study of 64 patients with pulmonary toxoplasmosis conducted in France, the mean (± standard deviation [SD]) CD4 + lymphocyte count was 40 (±75) cells/µL.


Imaging.


The chest radiograph usually reveals bilateral opacities, either in a fine reticulonodular pattern indistinguishable from PCP or in a coarse nodular pattern similar to that seen with tuberculosis or fungal pneumonias. Pleural effusions can be seen, and a variety of other radiographic findings have also been described.


Diagnosis.


The diagnosis of pulmonary toxoplasmosis is usually established by bronchoscopy and study of BAL fluid. In one review, BAL fluid examination was diagnostic in 16 of 17 immunocompromised patients with pulmonary disease.


Treatment.


The treatment for pulmonary toxoplasmosis is identical to that for central nervous system toxoplasmosis. First-line treatment is sulfadiazine plus pyrimethamine with leucovorin to decrease the likelihood of hematologic toxicities associated with pyrimethamine. The preferred alternative regimen is clindamycin plus pyrimethamine with leucovorin.


Prevention of Exposure.


Persons who are Toxoplasma antibody negative should be instructed to avoid potential sources of infection.


Prevention of Disease.


HIV-infected persons should be tested for T. gondii antibodies; antibody seronegative patients should be re-tested if their CD4 + lymphocyte count falls below 100 cells/µL. Toxoplasma seropositive patients receive primary prophylaxis once their CD4 + lymphocyte count falls below 100 cells/µL. As a practical matter, this is often accomplished when the CD4 + lymphocyte count reaches 200 cells/µL or fewer because the prophylaxis of choice (TMP-SMX) is also the prophylaxis of choice for Pneumocystis. For patients who are intolerant of TMP-SMX, dapsone plus pyrimethamine/leucovorin or atovaquone with or without pyrimethamine/leucovorin) can also be used. Primary and secondary T. gondii prophylaxis can be discontinued in HIV-infected persons on ART who have experienced an increase in their CD4 + lymphocyte count to greater than 200 cells/µL for at least 3 to 6 months.


Other Parasites


In general, pulmonary helminth infections are uncommon in HIV-infected persons. Both pulmonary cryptosporidiosis and pulmonary microsporidiosis in HIV-infected patients with concurrent intestinal disease have been reported. In these cases, aspiration from the gastrointestinal tract appears to be the most probable route of pulmonary infection. Occasionally, patients have presented with disseminated disease, raising the likely possibility of hematogenous spread. The most common respiratory symptoms are cough, dyspnea, and pleuritic chest pain. One review found that these symptoms were present in 77%, 58%, and 33%, respectively, of patients with cryptosporidial pulmonary disease. The diagnosis of Strongyloides stercoralis pulmonary disease ( eFig. 90-30 ) can be made by either sputum study or bronchoscopy. Strongyloides hyperinfection syndrome has also been reported in HIV-infected patients, but does not appear to be an important HIV-associated complication, even in countries where the parasite is endemic.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 21, 2019 | Posted by in CARDIOLOGY | Comments Off on Pulmonary Complications of HIV Infection

Full access? Get Clinical Tree

Get Clinical Tree app for offline access