Pneumocystis (carinii) jirovecii

Authors: M. Patricia George, M.D., Matthew R. Gingo, M.D.Alison Morris, M.D.

Pneumocystis jirovecii is an important cause of pneumonia in immunocompromised hosts, especially those with human immunodeficiency virus (HIV), hematologic malignancies, organ transplants, congenital immunodeficiencies, and those receiving specific immunosuppressive drugs such as high dose corticosteroids and tumor-necrosis factor inhibitors. A dramatic increase in incidence of P. jirovecii pneumonia occurred as the acquired immunodeficiency syndrome (AIDS) epidemic grew. With the more widespread use of specific chemoprophylaxis for the appropriate immunosuppressed patient populations in general, and with the expanded availability of effective combination antiretroviral therapy (CART) for HIV-infected patients in particular, there has been a decline in cases (122); however,Pneumocystis continues to cause morbidity and mortality among non-HIV-infected patients who are immunosuppressed and remains a leading cause of AIDS-associated pneumonia.

MICROBIOLOGY

Pneumocystis was originally discovered by Carlos Chagas in 1906 and believed by Chagas and Antonio Carini to be a stage in the trypanosome life cycle. Inability to culture Pneumocystis has limited understanding of the biology of the organism. Pneumocystis was recognized by Pierre and Mme. Delanoe to be distinct from trypanosomes six years after Chagas first identified it and was subsequently classified as a separate protozoa. Recent application of molecular techniques demonstrated that the organism shares many characteristics such as cell wall and enzyme structure with fungi (241). Thus, the organism is now considered to be a fungus rather than a protozoan.

Based on morphology, Pneumocystis exists in two main forms: the trophic form (formerly called the trophozoite) and the cyst form (also known as the spore case) (46246). It has been hypothesized that trophic forms can conjugate and form cysts that mature to contain eight nuclei. These cysts rupture, freeing eight trophic forms from the intracystic nuclei. It has also been hypothesized that trophic forms can replicate asexually via binary fission (162246). Although the life-cycle has not been fully elucidated, the organism has been isolated in the alveolar spaces of the host organism, where the trophic form attaches to alveolar epithelial cells (148). This interaction is important for survival of the organism, and the organism has not been successfully maintained in vitro in a cell-free environment. Recent work suggests that formation of a biofilm is also crucial to the organism’s survival and may enable future in vitro experimentation (45).

The cell walls of the cysts are rich in carbohydrate, consisting primarily of ß-1,3-glucan, as well as chitins and proteins (246). The main cell surface antigen is known as the major surface glycoprotein (MSG) or glycoprotein A (gpA). It is highly variable and is distinct among different species of Pneumocystis that infect different mammalian hosts (79150192). MSG interacts with the host by binding to both surfactant protein D, a lung collagenous carbohydrate protein that accumulates in Pneumocystis pneumonia (Vuk-Pavlovic et al. Carbohydrate recognition domain of surfactant protein D mediates interactions with Pneumocystis carinii glycoprotein A. AJRCMB 2001; 24: 475-484), and host macrophages through mannose receptors (192). The ability of the MSG locus to undergo recombination helps the organism evade host defenses (239126). Pneumocystis also contains a kexin-like serine protease, encoded by the Kex1 locus. This protease has been found on the cell surface and is believed to be less antigenically variable than MSG (142200274); however, recent reports have found genetic polymorphisms in this gene as well (69).

Several enzymatic targets have been isolated from Pneumocystis including dihydropteroate synthase (DHPS) and dihydrofolate reductase (DHFR), the target enzymes for sulfonamides and trimethoprim, respectively. Genes encoding these enzymes, as well as the cytochrome B locus (the site of action of atovaquone), have been identified and sequenced. Mutations in these loci are postulated to be linked to drug resistance (62106115155183).

Pneumocystis exhibits host-specific speciation, and the nomenclature was revised to distinguish these different species (Frenkel JK.Pneumocystis jiroveci n. sp. from man: morphology, physiology, and immunology in relation to pathology. Natl Cancer Inst Monogr 1976; 43:13-30). The organism in rats is called P. carinii, but this species does not infect humans. The name of the Pneumocystis species that infects humans was changed from Pneumocystis carinii to Pneumocystis jirovecii after Otto Jirovec, one of the earliest scientists to recognize the organism in humans (80240). P. jirovecii pneumonia is still abbreviated PCP, with the abbreviation standing for Pneumocystis Pneumonia instead of Pneumocystis carinii Pneumonia.

back to top

EPIDEMIOLOGY

In 1981, cases of Pneumocystis pneumonia (PCP) were first reported in men who had sex with men and in intravenous drug users (1160). The disease became the first opportunistic infection associated with HIV/AIDS. The advent of effective CART and routine PCP prophylaxis have reduced the incidence of PCP, although it remains the most common serious opportunistic infection in HIV (43117172,245). The EuroSIDA study followed a cohort of over 8,500 HIV-infected patients in Europe and found that incidence of PCP fell from 4.9 cases per 100 person-years before the introduction of combination CART to 0.3 cases per 100 person-years after CART was widely available (266). The prevalence of PCP in the current era in the United States ranges between 6-19% (30). Mortality from PCP has also changed over the course of the AIDS epidemic with the latest mortality of hospitalized patients with PCP reported to be around 7-11% (215129). In one study of PCP in HIV-infected individuals hospitalized in London from 1985-2006, overall mortality was 13.5% (265). Mortality improved over time, from 10.1% from 1985 to 1989, to 16.9% from 1990 to June 1996, and 9.7% from July 1996 through 2006 (265). For patients requiring intensive care, studies performed shortly after the introduction of CART showed a mortality ranging from 53 to 62% (44131,177). Certain clinical factors such as patient age, low serum albumin, need for mechanical ventilation, and development of a pneumothorax have been associated with increased mortality (561652170177265). In a study of HIV-infected individuals admitted to the ICU between 1996 and 2004 with respiratory failure, lower tidal volume ventilation was also independently associated with improved survival (48).

The frequency of PCP among HIV-infected patients in tropical and developing countries has generally been thought to be much lower than in industrialized countries (175). The lower incidence of PCP in places like Africa may be secondary to a high mortality from diseases such as tuberculosis and bacterial pneumonia that occur at higher CD4 cell counts, thus preventing persons from ever becoming at risk for PCP. The apparently lower incidence might also result from geographic or climactic factors that influence PCP risk, or merely reflect an underreporting secondary to a failure to diagnose (1172175251).

PCP has also been reported in non-HIV-infected, immunocompromised patients. The incidence of PCP among patients with cancer varies by diagnosis, ranging from 22-45% for lymphomas, 25% for rhabdomyosarcomas, and 1.3% for solid tumors if patients are receiving at least 20 mg of prednisone or equivalent corticosteroid for over one month (111229230). Bone marrow transplant and solid organ recipients are also at increased risk of PCP, with reported incidences from 2.5% to >25% (5160639193102168272). The incidence of PCP in patients with collagen vascular disease has been reported at < 2%, although patients with Wegener’s granulomatosis have an incidence > 6% (84194). The clinical presentation of PCP differs in HIV-infected and non-HIV-infected individuals with HIV-infected individuals presenting with less severe hypoxemia and a more indolent course than other immunocompromised patients (68135147). Non-HIV-infected individuals have lower parasite numbers and greater BAL neutrophilia than AIDS patients (147).

While overt PCP in immunocompetent hosts is rare, there is a growing body of literature that colonization with Pneumocystis occurs in both immunocompetent and immunocompromised persons. Pneumocystis colonization is defined as detection of Pneumocystis without signs and symptoms of PCP, often requiring use of polymerase chain reaction (PCR) for detection of the organism’s DNA in respiratory secretions (175261). Pneumocystis colonization may be detected in infants and young children (178). Nevez and coworkers reported a 24% prevalence of Pneumocystis colonization in 178 infants with bronchiolitis (188). Similar levels of colonization (between 14% and 25%) have been noted by other investigators in the immunocompetent pediatric population with acute respiratory syndromes or chronic lung diseases (39123249).

As methods of detection improve, prevalence of colonization in normal, non-immunosuppressed adults is somewhat common, with reported colonization ranging from 20 to as high as 64.9% (163210253). In order to detect high prevalence of colonization in the normal host, however, large quantities of lung tissue need to be assayed, suggesting that the organism burden is quite low. The prevalence of Pneumocystis colonization among HIV-infected individuals is higher and has been reported between 10-68.8% (92107143161174185,213262). The wide prevalence range is likely due to differences in subject populations, the type of sample tested, and the PCR technique used. One study reported that the frequency of colonization increased with decreasing CD4 count, although colonization has been reported even in individuals with high CD4 cell counts (143174). The prevalence of colonization among non-HIV infected, immunocompromised individuals is also high, ranging from 15.5 to 58.5% (98158166187258270).

Pneumocystis colonization may be an important phenomenon for several reasons. The role of colonization in risk for PCP is not known, but it is possible that people who are colonized may have an increased risk of developing PCP or may act as a reservoir and transmit Pneumocystis to others. Colonization may also play a pathogenic role in other diseases. It has been linked to chronic obstructive pulmonary disease (COPD) in both HIV-infected and non-HIV-infected individuals (28176186). In one study, Pneumocystis colonization was detected in 36.7% of patients with very severe COPD compared to less than 10% of those with other end-stage lung diseases (176). In another study, Pneumocystis colonization in COPD subjects was associated with higher levels of serum proin flammatory cytokines, suggesting a role in disease pathogenesis (28). Animal models also demonstrate that Pneumocystis colonization can promote the development of airway obstruction and emphysema (34190234). In a non-human primate model of HIV, Pneumocystis-colonized animals develop airway obstruction as well as radiographic and anatomic emphysema (234). A rodent model demonstrated that cigarette exposure combined with Pneumocystis murina infection in immunocompetent mice resulted in pulmonary function and pathologic changes consistent with emphysema. In addition, these changes were associated with increased macrophages in the bronchoalveolar lavage fluid as well as alterations of CD4/CD8 ratios similar to humans who develop emphysema (34). Although the link of colonization to these diseases needs to be confirmed, clinicians and researchers should at least consider the possibility that Pneumocystis may cause more clinical manifestations than previously suspected.

back to top

CLINICAL MANIFESTATIONS 

Compared to other diffuse pulmonary processes, there is nothing unique about the presentation of PCP that would allow it to be diagnosed without a specific microbiologic test identifying the organism in pulmonary secretions or tissue. Clinicians must be alert to the possibility of PCP in at-risk patients and the importance of quickly establishing the diagnosis, so that appropriate therapy can be instituted early when the likelihood of response to therapy is greatest. As PCP is currently a common initial presentation of HIV infection, clinicians must consider the diagnosis in patients who are not previously known to be HIV-infected.

PCP typically presents as an acute or subacute pulmonary process with fever, nonproductive cough, dyspnea, and shortness of breath (67). AIDS patients tend to have a more indolent course with a longer duration of symptoms and less hypoxemia than patients immunosuppressed from cytotoxic chemotherapy or corticosteroids (135244). HIV-infected patients typically present after 1-4 weeks of symptoms, while other immunosuppressed patients present within a few days of symptom onset.

Physical examination is often unrevealing except for fever and tachypnea. Chest examination is commonly normal; however, diffuse rales, and eventually signs of consolidation, may be present as the disease progresses.

Routine laboratory testing is also unremarkable. Patients often have nonspecific elevations of serum lactate dehydrogenase (LDH) and hypoxemia. The serum LDH is a reflection of tissue damage, in this case to the lung, rather than a specific marker for PCP. Although LDH is generally elevated, it cannot be used to diagnose PCP, and a normal LDH does not rule out PCP. The peripheral white blood count may be modestly elevated. For patients with HIV infection, clinicians should recognize that a white blood count of 6-10,000 cells/mm3 may represent a marked elevation over baseline. Patients are generally hypoxemic with an elevated alveolar-arterial oxygen gradient, and the degree of hypoxemia is used to determine disease severity.

The chest radiographic manifestations depend on the severity of illness. Early in the course of PCP, especially for patients with HIV, the chest radiograph may be normal despite substantial hypoxemia (55135). In patients with a normal chest radiograph, a high resolution (thin section) computed tomography (CT) scan of the chest will usually demonstrate a characteristic diffuse ground glass appearance; however, this finding is not specific for PCP and further diagnostic testing should be pursued (90). A normal high resolution CT scan makes the diagnosis of PCP highly unlikely. As the disease worsens, diffuse interstitial infiltrates develop which then progress to dense alveolar filling.

A substantial number of patients have atypical chest radiographs. Almost all types of infiltrates have been described with PCP. Asymmetric patterns, upper lobe infiltrates, mediastinal adenopathy, nodules, cavities, and effusions have been attributed to PCP alone, without a concurrent process, but rare findings such as pleural effusions or adenopathy should prompt an evaluation for processes other than PCP. Pneumothorax in a patient with AIDS should raise the suspicion for PCP (167232). Patients may present primarily with a pneumothorax, although the contralateral lung usually has diffuse infiltrates. Images of the liver, spleen, kidneys, or even the brain may occasionally reveal inflammatory masses due to P. jirovecii. These extrapulmonary lesions rarely cause symptomatic disease and appear to resolve with therapy.

back to top

LABORATORY DIAGNOSIS 

A specific diagnosis of PCP should be sought because many infectious and non-infectious processes can present almost identically to PCP. Empiric treatment should be begun immediately when PCP is suspected because treatment does not decrease the ability to diagnose PCP, and treatment delay may worsen outcome. In HIV-infected patients, organisms can persist for weeks or months after initiation of effective therapy. For other immunosuppressed patients, there is less information regarding the persistence of organisms, but treatment during the several days that might be required for diagnosis is prudent (218).

The definitive diagnosis of P. jirovecii disease requires the demonstration of cysts or trophozoites in tissue or body fluids since human Pneumocystis cannot be cultured in laboratory animals or in vitro. Before the AIDS epidemic, the diagnosis of P. jirovecii pneumonia typically required an open lung biopsy. With the development of improved diagnostic techniques, diagnoses can now be established by less invasive methods. Virtually all HIV-infected patients can be diagnosed by careful analysis of bronchoalveolar lavage fluid (BAL), although BAL is less sensitive in non-HIV-infected patients (164193). Induced sputum has been shown to be a sensitive, simple and noninvasive means to diagnose P. jirovecii pneumonia in the HIV-infected population and often reduces the need for bronchoscopy (18132137189). Reported yields for recovery of the organism range from 70-95% at various institutions, and utility of the test depends on the laboratory’s familiarity with the technique (18137208). On rare occasions, tissue diagnosis will be necessary. Either transbronchial biopsy, video-assisted thorascopic surgery (VATS) biopsy, or open lung biopsy can be performed. Most often, when bronchoalveolar lavage has failed to reveal Pneumocystis, a biopsy will reveal a pathologic process other than PCP.

The identification of P. jirovecii in sputum or BAL fluid has typically been performed with colorimetric stains such as methenamine silver, toluidine blue-O, Geimsa, and Diff-Quik or by using immunofluorescent assays (direct fluorescent antigen testing). The development of monoclonal antibodies has resulted in a rapid, sensitive and easy-to-perform immunofluorescence assay, which is more efficient than conventional colorimetric stains for detecting Pneumocystis jirovecii in respiratory specimens (82134189). Many hospital laboratories use this immunofluorescent stain in preference to colorimetric stains.

More recently, molecular assays have been developed for detecting P. jirovecii in BAL fluid, induced sputum, or oral wash samples. PCR methods have used a variety of gene targets (196224), but those with the highest sensitivity use either a multicopy gene target, e.g., mitochondrial rRNA or major surface glycoprotein (109264), and/or a nested PCR assay in which two amplification rounds are used to increase sensitivity (719798140151153263). In one study, a quantitative touch-down PCR assay targeting the MSG gene tested had a sensitivity of 88% and a specificity of 85% for diagnosis of PCP in oral wash samples (140). Initiation of PCP treatment prior to oral wash collection decreased the sensitivity of the PCR assay, suggesting that specimens should be collected early if the assay is to be used in clinical settings (140). When PCR is used on induced sputum or BAL, the sensitivity increases, but the specificity decreases as more cases of colonization are detected. In a study of real-time PCR reaction against a single-copy gene (cdc2) rather than a multi-copy gene, detection of Pneumocytis pneumonia in immunosuppressed patients being evaluated for opportunistic respiratory infections was enhanced over direct fluorescent microscopy method, while detection of colonization in non-immunocompromised, asymptomatic individuals was reduced (268). These highly sensitive molecular techniques are not currently available in clinical practice, but they have been useful tools for research to examine drug resistance, to study Pneumocystis colonization, and to address questions regarding the transmission of the disease (108).

Other novel diagnostic techniques for PCP are being investigated. S-Adenosylmethionine (AdoMet) is a molecule that Pneumocystis requires for methylation reactions and polyamine synthesis and is scavenged by Pneumocystis from its hosts. Two studies showed that AdoMet levels are reduced in humans with PCP (235236). With a break point between 59 and 66 nM of AdoMet, the sensitivity and specificity were 0.88 and 1.0 respectively for PCP diagnosis. AdoMet levels rose with successful treatment suggesting that it could be a non-invasive, rapid assay for diagnosing PCP and following response to treatment (236). However, in a subsequent prospective study of 31 HIV-negative patients with and without PCP, AdoMet levels did not reliably distinguish between patients with and without PCP (de Boer MG, Gelinck LB, van Zeist BD, van de Sande WW, Willems LN, van Dissel JT, de Jonge R, Kroon FP. β -d-glucan and S-adenosylmethionine serum levels for the diagnosis of Pneumocystis pneumonia in HIV-negative patients: a prospective study. J Infect 2011; 62:93-100). Another rapid assay that has shown some usefulness is the serum β-D-glucan level. With a cut-off point of 31.1 pg/mL, the sensitivity and specificity of β-D-glucan were 92.3% and 86.1%, respectively (243). In a prospective study of HIV-negative patients, elevated serum β-D-glucan level with a cut-off point of 60 mg/mL had a sensitivity of 0.90 and specificity of 0.89 (50). This molecule is elevated in other fungal infections, and the specificity may be lower if applied more widely.

back to top

PATHOGENESIS 

Serologic and polymerase chain reaction data indicate that most humans become subclinically infected with P. jirovecii during childhood and that this infection is usually well-contained by an intact immune system (141188203207252). The mode of transmission in humans is likely respiratory (99197255). It was historically thought that PCP results solely from reactivation of latent infection; however, several lines of evidence suggest that de novo infection either from an environmental reservoir or from person-to-person transmission occurs. There have been clusters of PCP cases, and risk of PCP varies by location (4959179). Studies have found that Pneumocystis genotype reflects a patient’s city of residence rather than city of birth and that newly-diagnosed HIV patients with PCP have Pneumocystis containing mutations associated with prior use of PCP prophylaxis, in both cases suggesting that disease was recently acquired (1415105). Similarly, some patients have had recurrent episodes of PCP due to genotypically distinct organisms, indicating that their disease resulted from re-infection (1599127128214). In addition, animal models of PCP have shown that Pneumocystis can be transmitted between both immunosuppressed and immunocompetent animals (7314181). Thus, disease can occur after primary infection, re-infection, or after reactivation of latent infection acquired in the distant past.

Pneumocystis is presumably inhaled into the alveolar space. The organism has a unique tropism for the lung, where it exists primarily as an alveolar pathogen without invading the host. Following primary infection in an immunologically intact individual, the organism can be completely cleared or can colonize the airways, possibly allowing the person to act as a reservoir for transmission or increasing the risk of developing PCP in the future. Clinically apparent pneumonia occurs only if an individual develops specific types of immunosuppression. At that point, latent organisms reactivate, or an acute infection/re-infection produces acute disease. Organisms proliferate, evoking a mononuclear cell response. Alveoli become filled with proteinacous material and clinical pneumonia occurs. The disease is fatal if untreated.

CD4+ T lymphocytes are pivotal in the host’s defenses against Pneumocystis in both animals and humans. For patients with HIV infection, the degree of depletion of CD4+ T lymphocytes strongly correlates with the likelihood of developing PCP. About 95% of PCP cases in patients with HIV infection have been seen in HIV-infected patients with recent CD4+ T lymphocyte counts under 200 cells/μL (237). The majority of these cases occur at CD4+ T lymphocyte counts less than 100 cells/μL (237). About 10-15% of patients develop PCP at CD4+ T lymphocyte cell counts greater than 200 cells/mm3 (35). For HIV-infected patients who have benefited from therapy with antiretroviral drugs, considerable evidence supports the concept that CD4+ T lymphocyte cell counts continue to be an accurate indicator of susceptibility to PCP, and those who experience sustained increases of CD4+ T lymphocytes above 200 cells/μL are at much lower risk of PCP (6177121152). The nadir of the CD4+ T lymphocyte cell count fall prior to the institution of CART does not influence the predictive value of counts after response to CART (38128137). For patients with immunosuppressive disorders other than HIV, CD4+ T lymphocyte cell counts are less helpful (157). They should not be used to determine which non-HIV-infected patients are susceptible to PCP or which patients need prophylaxis.

PCP has also been recognized in patients with B cell defects, children with severe combined immunodeficiency disease, premature or debilitated infants, oncology patients receiving immunosuppressive drugs, and organ transplant recipients (2627111205231242,250). In one retrospective study, a corticosteroid dose equivalent to 16 mg of prednisone per day or more for a period of weeks was associated with a significant risk for PCP in patients who did not have AIDS (271). In a non-human primate model of Pneumocystis colonization in HIV, Kling demonstrated that low anti-Pneumocystis-kexin protein (KEX1) antibody titers were associated with increased risk of Pneumocystis colonization (133). A study in persons with HIV also found that low KEX1 titers were an early independent predictor of future PCP risk (Gingo in press), and vaccination against KEX1 in mice is protective against PCP (274). Thus, it appears that both humoral and cell-mediated immunity are important host defenses against this infection (47108219).

back to top

SUSCEPTIBILITY IN VIVO AND IN VITRO

Human P. jirovecii cannot be grown in vitro, so conventional susceptibility testing cannot be performed. Molecular techniques permit detection of mutations in the target enzymes for various drugs (130139), but the clinical significance of these mutations is currently unknown.

Because P. jirovecii has been widely exposed to sulfonamides, it is reasonable to expect that this organism might develop sulfonamide resistance. Both dapsone and TMP/SMX act by inhibiting the folate biosynthesis enzyme DHPS. Sulfonamide resistance could theoretically result from point mutations in the DHPS gene. DHPS mutations seem to be occurring more commonly in recent years than one or two decades ago and are more often detected among patients with heavy prior exposure to sulfonamides (4296124184). Interestingly, no trimethoprim resistance has been detected, implying that perhaps trimethoprim exerts no selective pressure because it has so little activity. In vivo rodent studies support this concept (155).

Whether the degree of sulfonamide resistance currently recognized is clinically sufficient to cause treatment or prophylaxis failure has not been conclusively determined (4296105124125165184). A Danish study reported that patients with mutant DHPS were less likely to survive PCP, although the response to TMP-SMX was not specifically addressed (96). A more recent study found that although the majority of patients with mutant genotype survived when given treatment doses of TMP-SMX, there was a trend for these patients to be more likely to require mechanical ventilation and to die (42). Moreover, two other trials reported that there was no effect on survival or response to therapy in those who had mutant DHPS (124184). There is concern that higher level resistance will occur in the future and diminish the effectiveness of sulfonamides and sulfones.

Recommendations for Treatment

Once the diagnosis of P. jirovecii is made, outpatient therapy with oral TMP-SMX for 21 days is recommended for mild to moderate disease (PaO2 >70 mm Hg, A-a gradient ≤35 mm Hg) (121149). Other alternatives for oral outpatient therapy include TMP-dapsone, clindamycin-primaquine, and atovaquone. Patients who are more severely ill with moderate to severe disease or who cannot tolerate oral medications should be hospitalized and given intravenous TMP-SMX. In sulfonamide-intolerant patients, intravenous pentamidine or clindamycin-primaquine should be administered.

As patients often worsen within the first several days of treatment, treatment failure is considered only if the patient has worsening clinical status after at least 4-8 days of therapy. Alternative treatment regimens include clindamycin-primaquine or IV pentamidine. Pentamidine has more evidence of efficacy than clindamycin-primaquine for first line therapy; however, a systematic review found that clindamycin-primaquine had a higher response rate than IV pentamidine for second-line salvage therapy for suspected treatment and can be considered for those failing TMP-SMX (17).

 back to top

ANTIMICROBIAL THERAPY 

General

The efficacy of chemotherapy for PCP depends on several factors: the degree of hypoxia at the time therapy is started, the degree of immunosuppression, co-morbid conditions, and the ability of the patient to tolerate the most effective agents (923). As with other infectious diseases, the earlier therapy is started, the better the prognosis is likely to be (220222247).

Trimethoprim-sulfamethoxazole (TMP-SMX) is the agent of choice for initial therapy of acute PCP regardless of severity (23112,121149159220222). TMP-SMX is as potent as intravenous pentamidine, and it is less toxic (226). If a patient has mild disease (PaO2 greater than 70 mm Hg and alveolar-arterial oxygen gradient less than 35 mmHg) and is able to tolerate oral medications, TMP-SMX may be given in the dosage of two oral double strength (DS) tablets (160 mg TMP and 800 mg SMX) every 8 hours. With more severe disease or if the patient is unable to tolerate oral medication reliably, intravenous TMP-SMX (15-20mg/kg TMP and 75-100 mg/kg SMX divided into three or four doses) should be given. Total duration of therapy is 21 days. Intravenous therapy can be switched to the oral dosing when the patient is clinically improved.

Toxicity associated with TMP-SMX continues to hinder its use in many patients and occurs more frequently in HIV-infected patients than in patients without HIV infection (86113). Toxicities include fever, rash, headache, nausea, vomiting, pancytopenia, hepatitis, aseptic meningitis, and renal dysfunction. Trimethoprim can also cause hyperkalemia. Patients with bone marrow suppression have predictable difficulty tolerating TMP-SMX, and anemia and neutropenia are common. Some toxicities of TMP-SMX can be life threatening, including Stevens-Johnson syndrome and a distributive shock syndrome that presents similarly to anaphylaxis. Treatment-limiting toxicities usually occur between day 6 and day 10 of therapy. For AIDS patients, trials suggest that approximately 25% of patients are unable to tolerate a full course of TMP-SMX (94212228). Minor laboratory abnormalities should not be an indication to switch to a less effective alternative therapy. Clinicians must recognize that providing optimal therapy for this life-threatening pneumonia should be the therapeutic priority compared to mild or moderate toxicities that are reversible.

Alternative Therapy

Intravenous pentamidine is the most potent alternative agent to TMP-SMX (226267). The standard dose of pentamidine is 4 mg/kg/day, given intravenously (IV) over at least one hour for a minimum of 14-21 days (121149). Small studies suggest that a lower dose of 3 mg/kg/day may be less toxic, but equally effective (36). Pentamidine can only be given parenterally because aerosolized pentamidine is not effective for acute therapy (149). Pentamidine is difficult to administer because of the incidence of substantial toxicities including renal dysfunction, dysglycemias, pancreatitis, and Torsades de pointes. An elevated creatinine commonly occurs and must be monitored closely, but dosages do not need to be adjusted for renal dysfunction. Because pentamidine can cause initial islet cell destruction that results in insulin release followed by insufficient insulin production, patients often develop both hypo- and hyperglycemia, and glucose levels should be monitored. Patients also need to be carefully assessed to determine if they are receiving other drugs that prolong the QT interval in order to reduce the likelihood that Torsades will occur. Although initially used before intravenous pentamidine was shown to be safe, intramuscular pentamidine is rarely, if ever, used because it causes large, painful sterile abscesses.

The combination of clindamycin and primaquine is another reasonable alternative for the treatment of PCP in both mild and more severe disease (21121149220247). A randomized trial assessing initial therapy found the combination to be comparable in efficacy to TMP-SMX or TMP-dapsone in mild to moderate disease (222). Clindamycin-primaquine has also been used as a salvage regimen in patients with Pneumocystis-induced respiratory failure (191), although many authorities are reluctant to use an oral agent (i.e. primaquine) in this setting. Investigators report success rates of 75-80% in open, noncomparative trials with patients who are intolerant to or have failed standard treatment (220247). Primaquine is given orally at a dose of 30 mg per day. Clindamycin is given either orally (300 mg to 450 mg every 6 to 8 hours) or intravenously (600 mg to 900 mg every 6 to 8 hours). Primaquine has been associated with methemoglobinemia and hemolytic anemia especially in patients with glucose-6-phosphate dehydrogenase deficiency (G-6-PD), and therefore patients should be checked for G-6-PD deficiency prior to being started on primaquine.

Dapsone, as a single agent, is not as effective as other alternatives in the treatment of P. jirovecii pneumonia. Failure rates are approximately 40% in patients with HIV infection (171223). In combination with trimethoprim (15 mg/kg/day), however, its efficacy is comparable to TMP-SMX (145222). TMP-dapsone is used as an alternative oral regimen in mild to moderate disease for patients intolerant of TMP-SMX. Approximately 50% of sulfonamide-intolerant patients tolerate dapsone. This regimen can only be given orally and is therefore not suitable for patients with severe disease or gastrointestinal dysfunction. In addition, the combination of trimethoprim and dapsone is not formulated into one pill and is not as convenient to use as trimethoprim-sulfamethoxazole. Dapsone has also been associated with methemoglobinemia and hemolytic anemia, especially in patients with G-6-PD deficiency, and patients should be checked for G-6-PD deficiency prior to initiation of dapsone.

Oral atovaquone, another approved agent for treating P. jirovecii pneumonia, is effective and well tolerated. Atovaquone has a role as therapy for patients with mild, stable, disease who have no evidence of gastrointestinal dysfunction (121). If neither TMP-SMX nor TMP-dapsone is tolerable for patients with mild disease, atovaquone is a reasonable option. Atovaquone is not as effective as TMP-SMX (110), but atovaquone and intravenous pentamidine were found to have similar success rates in mild and moderate P. jirovecii pneumonia in AIDS patients who were intolerant to TMP-SMX (58). Atovaquone was better tolerated, but patients receiving atovaquone more frequently failed to respond to therapy, and patients receiving pentamidine had more treatment-limiting adverse drug toxicities (58). Low plasma atovaquone levels are due in part to poor bioavailability of the drug and are associated with a poor response (110). Even with the liquid formulation of the drug, absorption can be unpredictable and steady state may not be reached for several days. Absorption can be improved with ingestion of a fatty meal. Atovaquone levels are also reduced when used concurrently with rifampin of rifabutin. The combination of these drugs should be avoided. The standard dose of atovaquone is 750 mg orally twice a day.

Trimetrexate in combination with leucovorin has been assessed in patients with moderate to severe disease (227). Although this drug is very effective, it is less effective and associated with more relapses than TMP-SMX (227). Trimetrexate was an alternative agent for patients who were intolerant of TMP-SMX and who could not tolerate or did not respond to intravenous pentamidine (227), but it is no longer commercially available.

Other agents under investigation include analogs of primaquine, analogs of pentamidine, albendazole, and echinocandins or pneumocandins. There is interest in the activity of caspofungin against the cyst form of Pneumocystis, but there is scant clinical evidence that this drug is useful for treating or preventing human disease (856).

back to top

ADJUNCTIVE THERAPY 

The use of corticosteroids in conjunction with anti-Pneumocystis agents has become the standard of care in the treatment of moderate-severe PCP in AIDS patients (2). Three randomized studies revealed that corticosteroids significantly decreased the frequency of early deterioration in oxygenation and improved survival in patients who had an initial room air PO2 <70mm Hg or an A-a gradient >35mm Hg (22278). Glucocorticoids should be given to patients with moderate to severe PCP at a dose of 40 mg of prednisone twice daily for 5 days, then 40 mg daily for 5 days and then 20 mg daily through day 21 (if the patient is unable to take medication by mouth, methylprednisolone can be given intravenously at 75% of the prednisone dose) (121149). Corticosteroids should be administered with initiation of anti-PCP therapy if possible, even if therapy is empiric, and at least within 72 hours of treatment initiation (121). Although corticosteroids hasten the symptomatic resolution of mild PCP and prevent early deterioration in oxygenation (often subclinical) in this population, they are rarely used in mild disease because of concern regarding the metabolic complications of corticosteroids and possible potentiation of osteonecrosis (169). Corticosteroid therapy is logical to use in patients with underlying immunodeficiency disorders other than HIV, although no prospective trials have documented this efficacy (54202). Most clinicians would introduce corticosteroid therapy, or augment the dose if their patient had severe PCP.

For patients with HIV-related PCP who are not receiving CART, clinicians often consider starting such therapy soon after the PCP is improving. A recent randomized, multicenter trial of initiating CART early during opportunistic infections including PCP found a benefit in the combined secondary endpoint of progression to AIDS and death (273). Although this study included patients hospitalized with PCP, it did not include any patients with PCP requiring intensive care. Some retrospective studies have found that that continuing or starting CART in PCP patients or general HIV-infected patients requiring intensive care has a survival benefit (40177), but some evidence suggests that improvements in mortality in the CART era have resulted from use of low tidal volume ventilation and improved supportive techniques rather than the actual use of antiviral therapies (48211). If antiretroviral therapy is initiated during PCP treatment, the immune reconstitution inflammatory syndrome (IRIS) may occur and may result in worsening hypoxemia, increased radiographic infiltrates, and, in some cases, respiratory failure (121138269). Low CD4 cell counts and higher pre-CART HIV viral levels are associated with an increased risk of IRIS (87).

 back to top

ENDPOINTS FOR MONITORING THERAPY 

Survival from an episode of PCP correlates most closely with the pretreatment arterial-alveolar gradient < 55 as well as with pre-existing co-morbidities (70). Other factors that influence outcome include the number of trophic forms in bronchoalveolar lavage, degree of chest radiograph abnormality, level of LDH elevation, and (for patients with HIV infection) low CD4+ T lymphocyte cell counts (23119,135). For a patient with an initial PaO2 of greater than 70 mm Hg while breathing room air, expected survival rates are 60 to 80% in non-AIDS patients and 90-95% in patients with AIDS (23226).

Respiratory rate, arterial oxygenation, temperature and chest imaging should be assessed to determine initial clinical status and then followed to assess response to therapy. Arterial blood gases provide more precise measurement of oxygenation than percutaneous oxygen saturation monitoring, but the latter can be used, especially with mild disease. The median time to clinical response to therapy is 4 to 10 days (135). During that time, the temperature should return to normal, and the respiratory rate and oxygen saturation should unequivocally improve. As with most infectious pulmonary processes, the chest radiograph or CT scan often do not improve for several weeks. The use of bronchoscopy to assess response to drug therapy is not helpful since cysts and trophozoites are difficult to quantitate, and P. jirovecii will be present in bronchoalveolar lavage specimens for many weeks after initiation of therapy, even in patients who rapidly improve (233).

Causes of Clinical Worsening After Initiation of Therapy

There are several reasons why patients with PCP may fail to improve (Table 1). During the initial two to five days of therapy, many HIV-infected patients worsen with a decline in partial pressure of oxygen by 10-30 mmHg (121). This decline has been attributed to dying organisms, which elicit an intense inflammatory response. The precise causes and mechanisms of this intensified inflammatory response have not been well-delineated. The benefit of corticosteroids appears to be the ability to blunt this “paradoxical” inflammatory response. After this initial worsening, patients should demonstrate clinical improvement.

 Another cause of worsening during PCP treatment is development of a pneumothorax. Patients with PCP, especially HIV-related PCP, develop pneumatocoeles that can rupture, resulting in a pneumothorax. If a patient with PCP, especially one receiving mechanical ventilation, experiences a sudden deterioration, clinical and radiographic assessment for a pneumothorax should be performed immediately.

The initial therapeutic regimen should be continued for at least 4 to 8 days before considering a change in therapy due to treatment failure (121). If patients do not improve or if they worsen, other causes of clinical deterioration should also be sought. Fluid status should be monitored carefully, because P. jirovecii pneumonia may cause increased permeability of alveolar capillary membranes, which can lead to accumulation of interstitial and alveolar fluid and respiratory failure. Intravenous TMP-SMX may be an unrecognized source of fluids. Concomitant congestive heart failure due to processes unrelated to Pneumocystis may also occur. Cardiovascular comorbidities are a particular consideration as patients with HIV infection live to an older age, and certain antiretroviral regimens have been association with accelerated atherosclerosis (20747576). In addition, HIV-related or chemotherapy-related cardiomyopathy may not be evident until the patient is challenged with large volumes of fluid.

Clinicians also need to assess for other concurrent pulmonary processes in patients who appear to be failing PCP treatment. Cytomegalovirus (CMV), fungi, mycobacteria, respiratory viruses, or agents of atypical pneumonia may be present. Most clinicians would perform bronchoalveolar lavage if patients are not responding promptly, especially if the initial diagnosis was established by sputum examination. In desperate situations, an open or video-assisted thoracoscopic biopsy would be warranted to determine if a treatable cause of deterioration had been missed by bronchoalveolar lavage. Tissue is especially helpful to determine if CMV is present and if it should be treated. Non-infectious causes such as congestive heart failure, embolic disease or alveolar hemorrhage should also be considered. Evaluation may thus require CT scan for pulmonary emboli, an echocardiogram, or placement of a pulmonary artery catheter.

Patients started on CART shortly after PCP treatment may also worsen due to IRIS (1253101138216269). The exact incidence of IRIS in PCP is unknown (101269). In this syndrome, as viral load declines and CD4+ T lymphocyte count increases, a more robust inflammatory response can occur at sites of recent infection or may be seen in response to latent or subclinical infection. IRIS is more common in patients with very low CD4+ T cells (< 50 cells/μL) and high serum HIV viral levels (>100,000 copies/μL) (216). IRIS can occur within days or weeks of instituting therapy. In patients with PCP, the syndrome manifests as deteriorating oxygenation, worsening cough, fever, and shortness of breath, and worsening chest imaging. The diagnosis of IRIS is one of exclusion. Patients must be reevaluated to determine which process in causing their deterioration, i.e. IRIS, another infection, or another non-infectious syndrome (101). Management of IRIS is not well-defined, but CART should be continued whenever possible. Non-steroidal agents and corticosteroids have been used (66,269). Decisions regarding treatment or withdrawal of CART should be influenced by the severity of the syndrome, the robustness of the virologic and CD4+ T lymphocyte response to CART and the specific pathogen involved.

Management of the PCP Patient Who Fails to Improve

When patients deteriorate, possible causes discussed above should be evaluated and empiric treatment of immediately life-threatening diagnoses initiated. Echocardiography, CT scanning, or placement of a pulmonary artery catheter may help evaluate for other causes of worsening. Empiric therapy for community-acquired or hospital-acquired pneumonia with consideration of bronchoscopy may also be a reasonable strategy. Prednisone should be added to the regimen if not already initiated. A change in therapy to a different anti-PCP agent is usually reserved until the patient has had 4 to 8 days of first-line therapy and after other pulmonary processes have been investigated. Most recommend a switch first from TMP-SMX to clindamycin-primaquine or intravenous pentamidine (121). If a patient is deteriorating or failing to improve on a regimen other than TMP-SMX, strong consideration should be given to using TMP-SMX, even if desensitization is required in the intensive care setting.

 back to top

VACCINES 

There are no vaccines available.

PREVENTION 

General

TMP-SMX is always the drug of choice for PCP prophylaxis for patients who can tolerate this agent (121). This drug has a long history of consistent efficacy in a wide variety of patient populations. Hughes demonstrated that TMP-SMX was almost completely protective against PCP in a randomized, placebo-controlled trial of children with acute lymphocytic leukemia in the mid 1970s (112). Smaller trials, especially in patients with HIV (64114136), and non-randomized studies have demonstrated that TMP-SMX is safe and highly effective in other patient populations when compared to patients receiving no prophylaxis. True breakthroughs of PCP are unusual for patients who are adherent to the recommended regimens of TMP-SMX. There are data in patients with HIV infection to suggest that intermittent regimens (one DS tablet thrice weekly) may be less effective than daily regimens, though lower doses of TMP-SMX are better tolerated than higher doses (64). Such differences were not observed in children with malignant neoplasms (101). Reported side effects of prophylactic doses include cytopenias, skin rashes, hepatitis, pancreatitis, and nephritis.

TMP-SMX has advantages not provided by alternative drugs such as aerosolized pentamidine including low cost, oral preparation and probable protective effect against disseminated Pneumocystis. In addition, because of its broad spectrum of antimicrobial activity, it offers protection against toxoplasmosis and enteric pathogens (29). It is also apparent that TMP-SMX confers protection against Haemophilus influenzae and Streptococcus pneumoniae, although it is not indicated for bacterial prophylaxis alone.

Alternative Therapy

Aerosolized pentamidine, dapsone, dapsone-trimethoprim, dapsone-pyrimethamine, and atovaquone also have a high degree of efficacy. Each of these regimens has some disadvantages, and none is likely to be as effective as TMP-SMX. In European and American trials evaluating primary and secondary prophylaxis for patients with HIV infection, either high dose (1 DS per day) or low dose (1 single strength [SS] per day) TMP-SMX was found to be significantly more effective in preventing PCP than aerosolized pentamidine (2195116,228). The rate of discontinuation of study drug because of toxicity was higher in the TMP-SMX groups than in the aerosolized pentamidine group. The incidence and types of adverse reactions were similar in both TMP-SMX groups, but the toxic effects occurred significantly sooner in the group receiving the higher dose (228). Thus, many clinicians prefer lower dose regimens. However, for patients with the most severe immunosuppression, higher doses may be somewhat more effective, leaving clinicians to use their best judgment when choosing between high dose and low dose regimens.

In a large trial (ACTG 081), 843 patients with HIV infection were randomized to TMP-SMX (1 DS tablet bid), dapsone (50 mg bid) or aerosolized pentamidine (300 mg once monthly). Fewer episodes of PCP occurred among patients receiving TMP-SMX than in the other two arms when patients with CD4+ T lymphocyte counts less than 100 cells/μL were considered, but not when patients with higher CD4+ T lymphocyte counts were assessed. In this trial, the efficacy of dapsone appeared to be better than aerosolized pentamidine. Dapsone given at doses of 50 mg per day or less was not as effective as 50 mg bid (21).

A secondary prophylaxis study in the United States involving 310 patients with HIV infection randomly assigned administration of aerosolized pentamidine by a Respirgard II nebulizer or one oral double-strength tablet of TMP-SMX daily (94). When analyzed by intent-to-treat method, the recurrence rate of PCP was significantly higher among the patients assigned to aerosolized pentamidine (18%) than among those who received TMP-SMX (4%). As expected, patients who received TMP-SMX experienced frequent toxicity resulting in discontinuation of the agent.

 Aerosolized pentamidine is usually well-tolerated when delivered by the Respirgard II nebulizer in the indicated dosing regimens. Coughing or wheezing occurs in 30-40% of patients, but this reaction may be diminished or prevented by the administration of beta-adrenergic agonist such as albuterol (144). Bronchospasm rarely necessitates discontinuation of prophylaxis with aerosolized pentamidine treatment. Patients with reactive airway disease or bullous lung disease may not distribute aerosolized pentamidine effectively and thus may not obtain maximum protection. There have been reports of disseminated pneumocystosis in patients receiving aerosolized pentamidine for prophylaxis (95).

There is concern about transmission of Mycobacterium tuberculosis between health-care workers and HIV-infected patients associated with the coughing induced by aerosol pentamidine. Before administering aerosolized pentamidine, all patients should be screened for tuberculosis, and health-care workers should follow guidelines provided by the Centers for Disease Control and Prevention to minimize the risk of spread of tuberculosis to other patients and health-care workers (19118). Ideally, aerosolized pentamidine should be administered in individual booths or rooms with negative pressure ventilation and direct exhaust to the outside. After the administration of aerosolized pentamidine, patients should not return to common waiting areas until coughing has subsided.

Dapsone is an attractive alternative to aerosolized pentamidine because it is oral, convenient, and inexpensive. It is considered by some experts to be the best alternative for patients who cannot tolerate TMP-SMX (21116). It is estimated that 50-80% of patients who are TMP-SMX intolerant will be able to tolerate dapsone (104). Dose reduction to improve tolerability is not recommended, because doses less than 100 mg daily are considerably less effective than the full dose regimen. A case-control study in bone marrow transplants showed the efficacy of daily dapsone was similar to TMP-SMX. Dapsone did not seem to cause hematologic toxicity among TMP-SMX allergic patients (225).

Weekly doses of dapsone (200 mg) and pyrimethamine (75 mg) are well-tolerated, but less effective than TMP-SMX. Dapsone-pyrimethamine has efficacy as a prophylactic regimen against P. jirovecii pneumonia that is similar to aerosol pentamidine, but less effective than TMP-SMX. This combination has been assessed as a daily regimen (dapsone 50 mg po qd plus pyrimethamine 75 mg weekly) or as a weekly regimen (dapsone 200 mg plus pyrimethamine 75 mg) (83199). It is not clear if pyrimethamine truly adds potency against PCP (21). Dapsone alone has no antibacterial activity, and it is not clear if it has adequate anti-toxoplasma activity when used without pyrimethamine.

Atovaquone is another option for prophylaxis. Two trials in patients with HIV infection demonstrated that a daily atovaqone dose of 1500 mg of the liquid suspension has comparable efficacy to aerosolized pentamidine or oral dapsone (3265). Atovaquone does have activity against toxoplasma, but the relative efficacy of this regimen for preventing toxoplasmosis has not been adequately studied. Atovaqone has no antibacterial activity. This regimen is also much more expensive than other drug regimens. Atovaquone has not been extensively evaluated in patients with immunosuppressive disease other than HIV, but there is no reason to believe it would be less effective as long as it is absorbed and there are no substantial drug interactions.

Other potential prophylactic agents that have been used empirically or evaluated in small clinical trials include pyrimethamine-sulfadoxine (Fansidar), trimethoprim-dapsone, parenteral pentamidine, and primaquine-clindamycin. Pyrimethamine-sulfadoxine is effective, but its high rate of adverse reactions and long half-life make this an unattractive option, and one that has little benefit over TMP-SMX. Small trials with parenteral pentamidine and clindamycin-primaquine have been surprisingly disappointing in terms of efficacy.

back to top

Recommendations for Prophylaxis

For patients with HIV infection, primary prophylaxis is indicated when the patient's absolute CD4+ T lymphocyte cell count falls below 200 cells/μL (121) and for patients with oropharyngeal candidiasis regardless of CD4+ T lymphocyte cell count. Other indications include a CD4+% <14% or a history of an opportunistic infection (121). It may also be indicated to give PCP prophylaxis to HIV-infected patients whose CD4 count is between 200 and 250 cells/µl when frequent laboratory evaluation is not possible (121). Secondary prophylaxis is indicated in any patient with a history of PCP.

The recommended agent for prophylaxis is TMP-SMX, ideally one double strength tablet daily (121). In patients who have had an adverse reaction to TMP-SMX, the preferred option is desensitization or lower dose TMP-SMX (121146201). In those who cannot tolerate TMP-SMX, alternative regimens include: dapsone or monthly aerosolized pentamidine (228) or daily atovaquone (3265). In patients who are seropositive for Toxoplasma gondii, the preferred regimen is TMP-SMX (121). If TMP-SMX is not tolerated, then alternatives include daily dapsone combined with weekly pyrimethamine and leucovorin (83199209), or atovaquone with or without pyrimethamine and leucovorin (121).

Prior to the era of CART, prophylaxis was recommended to be life-long (3136159206). However, there are now convincing data that chemoprophylaxis can be stopped if the CD4+ T lymphocyte count rises above 200 cells/μL for at least 3 months because of CART (88152). A recent study showed the incidence of primary PCP in patients with CD4+ T lymphocyte count > 100 cells/μL was low regardless of prophylaxis use (Opportunistic Infections Project Team of the Collaboration of Observational HIV Epidemiological Research in Europe (COHERE) et al. Is it safe to discontinue primary Pneumocystis jiroveci pneumonia prophylaxis in patients with virologically suppressed HIV infection and a CD4 cell count <200 cells/microL. Clin Infect Dis. 2010 Sep 1;51(5):611-9). It is currently recommended to discontinue primary prophylaxis if patients have responded to CART with a CD4 count greater than 200 cells/μL (121), as there is no known added benefit after immune restoration (88152182). Discontinuing primary prophylaxis lowers pill burden and reduces risk of adverse drug reactions. Even with CD4+ T lymphocyte counts > 200 cells/μL, it may be prudent to continue prophylaxis in some high-risk patients. Those with high viral loads (i.e. > 50,00-100,000 copies/μL), rapidly declining CD4+ T lymphocyte cell counts (especially if frequent monitoring is not possible), wasting, oral candidiasis or a prior episode of PCP may still benefit from prophylaxis.

For non-HIV-infected patients, most authorities believe that it is reasonable to institute primary prophylaxis for groups of patients felt to be at high risk (8991100217). Most clinicians would provide chemoprophylaxis for allogeneic bone marrow transplant recipients, human stem cell transplant recipients, solid organ transplant recipients, and patients receiving antineoplastic chemotherapy, high dose corticosteroids, or certain other immunosuppressive agents (89103111156168180195). Fludarabine and 2-chlorodeoxyadenosine have an especially strong association with PCP (2627). Recent reports link development of PCP with the use of anti-tumor necrosis factor therapy, and prophylaxis should be considered in these patients (120173).

It is generally recommended that prophylaxis should be continued for as long as the immunosuppressive condition continues. Defining this period of immunosuppression may be challenging in non-HIV-infected patients. CD4+ T cell counts are not reliable, nor are any other clinical or laboratory markers (3351). Thus, factors such as the temporal relationship to immunosuppressive drugs, time since transplantation, induction therapy used at transplant, or time since the occurrence of graft versus host disease are used to estimate the period of susceptibility. For recipients of allogeneic bone marrow transplants and human stem cell transplants, it is routine to administer prophylaxis from the time of engraftment to at least 6 months after transplant for all recipients (224). Prophylaxis should be continued for more than 6 months after transplant for all persons who are receiving increased immunosuppression for graft versus host disease or for rejection. Decisions about duration of prophylaxis are made on the basis of estimates of susceptibility rather than objective clinical data. Cases of PCP in bone marrow transplant recipients usually occurred when prophylaxis was stopped too early (51). As transplant regimens change in terms of drugs, intensity, and underlying disease, the indications for PCP prophylaxis need to be readdressed.

back to top

REFERENCES

1Pneumocystis pneumonia--Los Angeles. Morb Mortal Wkly Rep 1981;30:250-2. [PubMed]

2. Consensus statement on the use of corticosteroids as adjunctive therapy for Pneumocystis pneumonia in the acquired immunodeficiency syndrome. The National Institutes of Health-University of California Expert Panel for Corticosteroids as Adjunctive Therapy forPneumocystis Pneumonia. N Engl J Med 1990;323:1500-4. [PubMed]

3. Guidelines for Prevention and Treatment of Opportunistic Infections among HIV-Exposed and HIV-Infected Children. Guideline: Recommendations from Centers for Disease Control and Prevention, the National Institutes of Health, the HIV Medicine Association of the Infectious Diseases Society of America, and the Pediatric Infectious Diseases Society; 2008. June 20, 2008. [PubMed]

4. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services November 3, 2008:1-139. Available athttp://www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf.  Accessed April 24, 2009.

5. Afessa B, Green B. Clinical course, prognostic factors, and outcome prediction for HIV patients in the ICU. The PIP (Pulmonary complications, ICU support, and prognostic factors in hospitalized patients with HIV) study. Chest 2000;118:138-45. [PubMed]

6. Alves C, Nicolas JM, Miro JM, Torres A, Agusti C, Gonzalez J, Rano A, Benito N, Moreno A, Garcia F, Milla J, Gatell JM. Reappraisal of the aetiology and prognostic factors of severe acute respiratory failure in HIV patients. Eur Respir J 2001;17:87-93. [PubMed]

7. An CL, Gigliotti F, Harmsen AG. Exposure of immunocompetent adult mice to Pneumocystis carinii f. sp. muris by cohousing: growth ofP. carinii f. sp. muris and host immune response. Infect Immun 2003;71:2065-70. [PubMed]

8. Annaloro C, Della Volpe A, Usardi P, Lambertenghi Deliliers G. Caspofungin treatment of Pneumocystis pneumonia during conditioning for bone marrow transplantation. Eur J Clin Microbiol Infect Dis 2006;25:52-4. [PubMed]

9. Antinori A, Maiuro G, Pallavicini F, Valente F, Ventura G, Marasca G, Murri R, Pizzigallo E, Camilli G, Tamburrini E. Prognostic factors of early fatal outcome and long-term survival in patients with Pneumocystis carinii pneumonia and acquired immunodeficiency syndrome. Eur J Epidemiol 1993;9:183-9. [PubMed]

10. Arozullah AM, Yarnold PR, Weinstein RA, Nwadiaro N, McIlraith TB, Chmiel JS, Sipler AM, Chan C, Goetz MB, Schwartz DN, Bennett CL. A new preadmission staging system for predicting inpatient mortality from HIV-associated Pneumocystis carinii pneumonia in the early highly active antiretroviral therapy (HAART) era. Am J Respir Crit Care Med 2000;161:1081-86. [PubMed]

11. Bakeera-Kitaka S, Musoke P, Downing R, Tumwine JK. Pneumocystis carinii in children with severe pneumonia at Mulago Hospital, Uganda. Ann Trop Paediatr 2004;24:227-35. [PubMed]

12. Barry SM, Lipman MC, Deery AR, Johnson MA, Janossy G. Immune reconstitution pneumonitis following Pneumocystis cariniipneumonia in HIV-infected subjects. HIV Med 2002;3:207-11. [PubMed]

13. Basselin M, Hunt SM, Abdala-Valencia H, Kaneshiro ES. Ubiquinone synthesis in mitochondrial and microsomal subcellular fractions of Pneumocystis spp.: differential sensitivities to atovaquone. Eukaryot Cell 2005;4:1483-92. [PubMed]

14. Beard CB, Carter JL, Keely SP, Huang L, Pieniazek NJ, Moura IN, Roberts JM, Hightower AW, Bens MS, Freeman AR, Lee S, Stringer JR, Duchin JS, del Rio C, Rimland D, Baughman RP, Levy DA, Dietz VJ, Simon P, Navin TR. Genetic variation in Pneumocystis carinii isolates from different geographic regions: implications for transmission. Emerg Infect Dis 2000;6:265-72. [PubMed]

15. Beard CB, Fox MR, Lawrence GG, Guarner J, Hanzlick RL, Huang L, del Rio C, Rimland D, Duchin JS, Colley DG. Genetic differences in Pneumocystis isolates recovered from immunocompetent infants and from adults with AIDS: Epidemiological implications. J Infect Dis 2005;192:1815-8. [PubMed]

16. Bedos JP, Dumoulin JL, Gachot B, Veber B, Wolff M, Regnier B, Chevret S. Pneumocystis carinii pneumonia requiring intensive care management: survival and prognostic study in 110 patients with human immunodeficiency virus. Crit Care Med 1999;27:1109-15.[PubMed]

17. Benfield T, Atzori C, Miller RF, Helweg-Larsen J. Second-line salvage treatment of AIDS-associated Pneumocystis jirovecii pneumonia: a case series and systematic review. J Acquir Immune Defic Syndr 2008;48:63-7. [PubMed]

18. Bigby TD, Margolskee D, Curtis JL, Michael PF, Sheppard D, Hadley WK, Hopewell PC. The usefulness of induced sputum in the diagnosis of Pneumocystis carinii pneumonia in patients with the acquired immunodeficiency syndrome. Am Rev Respir Dis 1986;133:515-8. [PubMed]

19. Blumberg HM, Watkins DL, Berschling JD, Antle A, Moore P, White N, Hunter M, Green B, Ray SM, McGowan JE, Jr. Preventing the nosocomial transmission of tuberculosis. Ann Intern Med 1995;122:658-63. [PubMed]

20. Bozkurt B. Cardiovascular toxicity with highly active antiretroviral therapy: review of clinical studies. Cardiovasc Toxicol 2004;4:243-60. [PubMed]

21. Bozzette SA, Finkelstein DM, Spector SA, Frame P, Powderly WG, He W, Phillips L, Craven D, van der Horst C, Feinberg J. A randomized trial of three antipneumocystis agents in patients with advanced human immunodeficiency virus infection. NIAID AIDS Clinical Trials Group. N Engl J Med 1995;332:693-9. [PubMed]

22. Bozzette SA, Sattler FR, Chiu J, Wu AW, Gluckstein D, Kemper C, Bartok A, Niosi J, Abramson I, Coffman J, et al. A controlled trial of early adjunctive treatment with corticosteroids for Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. California Collaborative Treatment Group. N Engl J Med 1990;323:1451-7. [PubMed]

23. Brenner M, Ognibene FP, Lack EE, Simmons JT, Suffredini AF, Lane HC, Fauci AS, Parrillo JE, Shelhamer JH, Masur H. Prognostic factors and life expectancy of patients with acquired immunodeficiency syndrome and Pneumocystis carinii pneumonia. Am Rev Respir Dis 1987;136:1199-206. [PubMed]

24. Briel M, Boscacci R, Furrer H, Bucher HC. Adjunctive corticosteroids for Pneumocystis jiroveci pneumonia in patients with HIV infection: a meta-analysis of randomised controlled trials. BMC Infect Dis 2005;5:101. [PubMed]

25. Briel M, Bucher HC, Boscacci R, Furrer H, Briel M, Bucher HC, Boscacci R, Furrer H. Adjunctive corticosteroids for Pneumocystis jiroveci pneumonia in patients with HIV-infection. Cochrane Database of Systematic Reviews 2006;3:CD006150. [PubMed]

26. Browne MJ, Hubbard SM, Longo DL, Fisher R, Wesley R, Ihde DC, Young RC, Pizzo PA. Excess prevalence of Pneumocystis cariniipneumonia in patients treated for lymphoma with combination chemotherapy. Ann Intern Med 1986;104:338-44. [PubMed]

27. Byrd JC, Hargis JB, Kester KE, Hospenthal DR, Knutson SW, Diehl LF. Opportunistic pulmonary infections with fludarabine in previously treated patients with low-grade lymphoid malignancies: a role for Pneumocystis carinii pneumonia prophylaxis. Am J Hematol 1995;49:135-42. [PubMed]

28. Calderon EJ, Rivero L, Respaldiza N, Morilla R, Montes-Cano MA, Friaza V, Munoz-Lobato F, Varela JM, Medrano FJ, Horra Cde L. Systemic inflammation in patients with chronic obstructive pulmonary disease who are colonized with Pneumocystis jiroveci. Clin Infect Dis 2007;45:e17-9.[PubMed]

29. Carr A, Tindall B, Brew BJ, Marriott DJ, Harkness JL, Penny R, Cooper DA. Low-dose trimethoprim-sulfamethoxazole prophylaxis for toxoplasmic encephalitis in patients with AIDS. Ann Intern Med 1992;117:106-11. [PubMed]

30. Centers for Disease Control and Prevention. HIV/AIDS Surveillance Supplemental Report; 2003; 9(No 3). [PubMed]

31. Chabe M, Dei-Cas E, Creusy C, Fleurisse L, Respaldiza N, Camus D, Durand-Joly I. Immunocompetent hosts as a reservoir ofPneumocystis organisms: histological and rt-PCR data demonstrate active replication. Eur J Clin Microbiol Infect Dis 2004;23:89-97.[PubMed]

32. Chan C, Montaner J, Lefebvre EA, Morey G, Dohn M, McIvor RA, Scott J, Marina R, Caldwell P. Atovaquone suspension compared with aerosolized pentamidine for prevention of Pneumocystis carinii pneumonia in human immunodeficiency virus-infected subjects intolerant of trimethoprim or sulfonamides. J Infect Dis 1999;180:369-76. [PubMed]

33. Chen CS, Boeckh M, Seidel K, Clark JG, Kansu E, Madtes DK, Wagner JL, Witherspoon RP, Anasetti C, Appelbaum FR, Bensinger WI, Deeg HJ, Martin PJ, Sanders JE, Storb R, Storek J, Wade J, Siadak M, Flowers ME, Sullivan KM. Incidence, risk factors, and mortality from pneumonia developing late after hematopoietic stem cell transplantation. Bone Marrow Transplant 2003;32:515-22.[PubMed]

34. Christensen PJ, Preston AM, Ling T, Du M, Fields WB, Curtis JL, Beck JM. Pneumocystis murina infection and cigarette smoke exposure interact to cause increased organism burden, development of airspace enlargement, and pulmonary inflammation in mice. Infect Immun 2008;76:3481-90. [PubMed]

35. Chu SY, Hanson DL, Ciesielski C, Ward JW. Prophylaxis against Pneumocystis carinii pneumonia at higher CD4+ T-cell counts. JAMA 1995;273:848.[PubMed]

36. Conte JE, Jr., Chernoff D, Feigal DW, Jr., Joseph P, McDonald C, Golden JA. Intravenous or inhaled pentamidine for treatingPneumocystis carinii pneumonia in AIDS. A randomized trial. Ann Intern Med 1990;113:203-9. [PubMed]

37. Conte JE, Jr., Hollander H, Golden JA. Inhaled or reduced-dose intravenous pentamidine for Pneumocystis carinii pneumonia. A pilot study. Ann Intern Med 1987;107:495-8. [PubMed]

38. Conte JE, Jr., Upton RA, Phelps RT, Wofsy CB, Zurlinden E, Lin ET. Use of a specific and sensitive assay to determine pentamidine pharmacokinetics in patients with AIDS. J Infect Dis 1986;154:923-9. [PubMed]

39. Contini C, Romani R, Cultrera R, Angelici E, Villa MP, Ronchetti R. Carriage of Pneumocystis carinii in children with chronic lung diseases. J Eukaryot Microbiol 1997;44:15S. [PubMed]

40. Croda J, Croda MG, Neves A, De Sousa dos Santos S, Benefit of antiretroviral therapy on survival of human immunodeficiency virus-infected patients admitted to an intensive care unit.  Crit Care Med. 2009 May;37(5):1605-11)  [PubMed] 

41. Croix DA, Board K, Capuano S, 3rd, Murphey-Corb M, Haidaris CG, Flynn JL, Reinhart T, Norris KA. Alterations in T lymphocyte profiles of bronchoalveolar lavage fluid from SIV- and Pneumocystis carinii-coinfected rhesus macaques. AIDS Res Hum Retroviruses 2002;18:391-401. [PubMed]

42. Crothers K, Beard CB, Turner J, Groner G, Fox M, Morris A, Eiser S, Huang L. Severity and outcome of HIV-associated Pneumocystispneumonia containing Pneumocystis jirovecii dihydropteroate synthase gene mutations. AIDS 2005;19:801-5. [PubMed]

43. Crum NF, Riffenburgh RH, Wegner S, Agan BK, Tasker SA, Spooner KM, Armstrong AW, Fraser S, Wallace MR. Comparisons of causes of death and mortality rates among HIV-infected persons: analysis of the pre-, early, and late HAART (highly active antiretroviral therapy) eras. J Acquir Immune Defic Syndr 2006;41:194-200. [PubMed]

44. Curtis JR, Yarnold PR, Schwartz DN, Weinstein RA, Bennett CL. Improvements in outcomes of acute respiratory failure for patients with human immunodeficiency virus-related Pneumocystis carinii pneumonia. Am J Respir Crit Care Med 2000;162:393-8. [PubMed]

45. Cushion MT, Collins MS, Linke MJ. Biofilm formation by Pneumocystis spp. Eukaryot Cell 2009;8:197-206. [PubMed]

46. Cushion MT, Walzer PD, Smulian AG, Linke MJ, Ruffolo JJ, Kaneshiro ES, Stringer JR. Terminology for the life cycle ofPneumocystis carinii. Infect Immun 1997;65:4365. [PubMed]

47. Daly KR, Koch J, Levin L, Walzer PD. Enzyme-linked immunosorbent assay and serologic responses to Pneumocystis jiroveci. Emerg Infect Dis 2004;10:848-54. [PubMed]

48. Davis JL, Morris A, Kallet RH, Powell K, Chi AS, Bensley M, Luce JM, Huang L. Low tidal volume ventilation is associated with reduced mortality in HIV-infected patients with acute lung injury. Thorax 2008;63:988-93. [PubMed]

49. de Boer MG, Bruijnesteijn van Coppenraet LE, Gaasbeek A, Berger SP, Gelinck LB, van Houwelingen HC, van den Broek P, Kuijper EJ, Kroon FP, Vandenbroucke JP. An outbreak of Pneumocystis jiroveci pneumonia with 1 predominant genotype among renal transplant recipients: interhuman transmission or a common environmental source? Clin Infect Dis 2007;44:1143-9. [PubMed]

50. de Boer MG, Gelinck LB, van Zeist BD, van de Sande WW, Willems LN, van Dissel JT, de Jonge R, Kroon FP. β -d-glucan and S-adenosylmethionine serum levels for the diagnosis of Pneumocystis pneumonia in HIV-negative patients: a prospective study. J Infect 2011; 62:93-100 [PubMed]

51. De Castro N, Neuville S, Sarfati C, Ribaud P, Derouin F, Gluckman E, Socie G, Molina JM. Occurrence of Pneumocystis jirovecipneumonia after allogeneic stem cell transplantation: a 6-year retrospective study. Bone Marrow Transplant 2005;36:879-83. [PubMed]

52. De Palo VA, Millstein BH, Mayo PH, Salzman SH, Rosen MJ. Outcome of intensive care in patients with HIV infection. Chest 1995;107:506-10. [PubMed]

53. Dean GL, Williams DI, Churchill DR, Fisher MJ. Transient clinical deterioration in HIV patients with Pneumocystis carinii pneumonia after starting highly active antiretroviral therapy: another case of immune restoration inflammatory syndrome. Am J Respir Crit Care Med 2002;165:1670; author reply 70. [PubMed]

54. Delclaux C, Zahar JR, Amraoui G, Leleu G, Lebargy F, Brochard L, Schlemmer B, Brun-Buisson C. Corticosteroids as adjunctive therapy for severe Pneumocystis carinii pneumonia in non-human immunodeficiency virus-infected patients: retrospective study of 31 patients. Clin Infect Dis 1999;29:670-2. [PubMed]

55. DeLorenzo LJ, Huang CT, Maguire GP, Stone DJ. Roentgenographic patterns of Pneumocystis carinii pneumonia in 104 patients with AIDS. Chest 1987;91:323-7. [PubMed]

56. Deresinski SC, Stevens DA. Caspofungin. Clin Infect Dis 2003;36:1445-57. [PubMed]

57. Dohn MN, Baughman RP, Vigdorth EM, Frame DL. Equal survival rates for first, second, and third episodes of Pneumocystis cariniipneumonia in patients with acquired immunodeficiency syndrome. Arch Intern Med 1992;152:2465-70. [PubMed]

58. Dohn MN, Weinberg WG, Torres RA, Follansbee SE, Caldwell PT, Scott JD, Gathe JC, Jr., Haghighat DP, Sampson JH, Spotkov J, Deresinski SC, Meyer RD, Lancaster DJ. Oral atovaquone compared with intravenous pentamidine for Pneumocystis carinii pneumonia in patients with AIDS. Atovaquone Study Group. Ann Intern Med 1994;121:174-80. [PubMed]

59. Dohn MN, White ML, Vigdorth EM, Ralph Buncher C, Hertzberg VS, Baughman RP, George Smulian A, Walzer PD. Geographic clustering of Pneumocystis carinii pneumonia in patients with HIV infection. Am J Respir Crit Care Med 2000;162:1617-21. [PubMed]

60. Dummer JS, Montero CG, Griffith BP, Hardesty RL, Paradis IL, Ho M. Infections in heart-lung transplant recipients. Transplantation 1986;41:725-9. [PubMed]

61. Dworkin MS, Hanson DL, Navin TR. Survival of patients with AIDS, after diagnosis of Pneumocystis carinii pneumonia, in the United States. J Infect Dis 2001;183:1409-12. [PubMed]

62. Edman JC, Edman U, Cao M, Lundgren B, Kovacs JA, Santi DV. Isolation and expression of the Pneumocystis carinii dihydrofolate reductase gene. Proc Natl Acad Sci U S A 1989;86:8625-9. [PubMed]

63. Eitner F, Hauser IA, Reiikowski O, Rath T, Lopau K, Pliquett RU, Fieldler R, Guba M, Hilgers RD, Floege J, Fischereder M. Risk factors for Pneumocystis jiroveci pneumonia (PcP) in renal transplant recipients . Nephrol Dial Transplant 2010. Epub ahead of print.[PubMed]

64. El-Sadr WM, Luskin-Hawk R, Yurik TM, Walker J, Abrams D, John SL, Sherer R, Crane L, Labriola A, Caras S, Pulling C, Hafner R. A randomized trial of daily and thrice-weekly trimethoprim-sulfamethoxazole for the prevention of Pneumocystis carinii pneumonia in human immunodeficiency virus-infected persons. Terry Beirn Community Programs for Clinical Research on AIDS (CPCRA). Clin Infect Dis 1999;29:775-83. [PubMed]

65. El-Sadr WM, Murphy RL, Yurik TM, Luskin-Hawk R, Cheung TW, Balfour HH, Jr., Eng R, Hooton TM, Kerkering TM, Schutz M, van der Horst C, Hafner R. Atovaquone compared with dapsone for the prevention of Pneumocystis carinii pneumonia in patients with HIV infection who cannot tolerate trimethoprim, sulfonamides, or both. Community Program for Clinical Research on AIDS and the AIDS Clinical Trials Group. N Engl J Med 1998;339:1889-95. [PubMed]

66. Elston JW, Thaker H. Immune reconstitution inflammatory syndrome. Int J STD AIDS 2009;20:221-4. [PubMed]

67. Engelberg LA, Lerner CW, Tapper ML. Clinical features of Pneumocystis pneumonia in the acquired immune deficiency syndrome. Am Rev Respir Dis 1984;130:689-94. [PubMed]

68. Enomoto T, Azuma A, Kohno A, Kaneko K, Saito H, Kametaka M, Usuki J, Gemma A, Kudoh S, Nakamura S. Differences in the clinical characteristics of Pneumocystis jirovecii pneumonia in immunocompromised patients with and without HIV infection. Respirology 2010; 15: 126-31. [PubMed]

69. Esteves F, Tavares A, Costa MC, Gaspar J, Antunes F, Matos O. Genetic characterization of the UCS and Kex1 loci of Pneumocystis jirovecii. Eur J Clin Microbiol Infect Dis 2009;28:175-8. [PubMed]

70. Ewig S et al. Clinical characteristics and outcome of Pneumocystis carinii pneumonia in HIV-infected and otherwise immunosuppressed patients. Eur Respir J 1995; 8:1548-1553. [PubMed]

71. Fischer S, Gill VJ, Kovacs J, Miele P, Keary J, Silcott V, Huang S, Borio L, Stock F, Fahle G, Brown D, Hahn B, Townley E, Lucey D, Masur H. The use of oral washes to diagnose Pneumocystis carinii pneumonia: a blinded prospective study using a polymerase chain reaction-based detection system. J Infect Dis 2001;184:1485-8. [PubMed]

72. Fisk DT, Meshnick S, Kazanjian PH. Pneumocystis carinii pneumonia in patients in the developing world who have acquired immunodeficiency syndrome. Clin Infect Dis 2003;36:70-8. [PubMed]

73. Frenkel JK. Pneumocystis jiroveci n. sp. from man: morphology, physiology, and immunology in relation to pathology. Natl Cancer Inst Monogr 1976; 43:13-30. [PubMed]

74. Friis-Moller N, Reiss P, Sabin CA, Weber R, Monforte A, El-Sadr W, Thiebaut R, De Wit S, Kirk O, Fontas E, Law MG, Phillips A, Lundgren JD. Class of antiretroviral drugs and the risk of myocardial infarction. N Engl J Med 2007;356:1723-35. [PubMed]

75. Friis-Moller N, Sabin CA, Weber R, d'Arminio Monforte A, El-Sadr WM, Reiss P, Thiebaut R, Morfeldt L, De Wit S, Pradier C, Calvo G, Law MG, Kirk O, Phillips AN, Lundgren JD. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med 2003;349:1993-2003. [PubMed]

76. Friis-Moller N, Weber R, Reiss P, Thiebaut R, Kirk O, d'Arminio Monforte A, Pradier C, Morfeldt L, Mateu S, Law M, El-Sadr W, De Wit S, Sabin CA, Phillips AN, Lundgren JD. Cardiovascular disease risk factors in HIV patients--association with antiretroviral therapy. Results from the DAD study. AIDS 2003;17:1179-93. [PubMed]

77. Furrer H, Egger M, Opravil M, Bernasconi E, Hirschel B, Battegay M, Telenti A, Vernazza PL, Rickenbach M, Flepp M, Malinverni R. Discontinuation of primary prophylaxis against Pneumocystis carinii pneumonia in HIV-1-infected adults treated with combination antiretroviral therapy. Swiss HIV Cohort Study. N Engl J Med 1999;340:1301-6. [PubMed]

78. Gagnon S, Boota AM, Fischl MA, Baier H, Kirksey OW, La Voie L. Corticosteroids as adjunctive therapy for severe Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. A double-blind, placebo-controlled trial. N Engl J Med 1990;323:1444-50.[PubMed]

79. Gigliotti F. Host species-specific antigenic variation of a mannosylated surface glycoprotein of Pneumocystis carinii. J Infect Dis 1992;165:329-36. [PubMed]

80. Gigliotti F. Pneumocystis carinii: has the name really been changed? Clin Infect Dis 2005;41:1752-5. [PubMed]

81. Gigliotti F, Harmsen AG, Wright TW. Characterization of transmission of Pneumocystis carinii f. sp. muris through immunocompetent BALB/c mice. Infect Immun 2003;71:3852-6. [PubMed]

82. Gill VJ, Evans G, Stock F, Parrillo JE, Masur H, Kovacs JA. Detection of Pneumocystis carinii by fluorescent-antibody stain using a combination of three monoclonal antibodies. J Clin Microbiol 1987;25:1837-40. [PubMed]

83. Girard PM, Landman R, Gaudebout C, Olivares R, Saimot AG, Jelazko P, Certain A, Boue F, Bouvet E, et al. Dapsone-pyrimethamine compared with aerosolized pentamidine as primary prophylaxis against Pneumocystis carinii pneumonia and toxoplasmosis in HIV infection. The PRIO Study Group. N Engl J Med 1993;328:1514-20. [PubMed]

84. Godeau B, Coutant-Perronne V, Le Thi Huong D, Guillevin L, Magadur G, De Bandt M, Dellion S, Rossert J, Rostoker G, Piette JC, et al. Pneumocystis carinii pneumonia in the course of connective tissue disease: report of 34 cases. J Rheumatol 1994;21:246-51. [PubMed]

85. Godoy MC, Silva CI, Ellis J, Phillips P, Muller NL. Organizing pneumonia as a manifestation of Pneumocystis jiroveci immune reconstitution syndrome in HIV-positive patients: report of 2 cases. J Thorac Imaging 2008;23:39-43. [PubMed]

86. Gordin FM, Simon GL, Wofsy CB, Mills J. Adverse reactions to trimethoprim-sulfamethoxazole in patients with the acquired immunodeficiency syndrome. Ann Intern Med 1984;100:495-9. [PubMed]

87Grant PMKomarow LAndersen JSereti IPahwa SLederman MMEron JSanne IPowderly WHogg ESuckow CZolopa A. Risk factor analyses for immune reconstitution inflammatory syndrome in a randomized study of early vs. deferred ART during an opportunistic infection. PLoS One. 2010 Jul 1;5(7):e11416. [PubMed]

88. Green H, Hay P, Dunn DT, McCormack S. A prospective multicentre study of discontinuing prophylaxis for opportunistic infections after effective antiretroviral therapy. HIV Med 2004;5:278-83. [PubMed]

89. Green H, M. P, L. V, L. L. Prophylaxis for Pneumocystis pneumonia (PCP) in non-HIV immunocompromised patients. Cochrane Database of Systematic Reviews 2007. [PubMed]

90. Gruden JF, Huang L, Turner J, Webb WR, Merrifield C, Stansell JD, Gamsu G, Hopewell PC. High-resolution CT in the evaluation of clinically suspected Pneumocystis carinii pneumonia in AIDS patients with normal, equivocal, or nonspecific radiographic findings. Am J Roentgenol 1997;169:967-75. [PubMed]

91. Gryzan S, Paradis IL, Zeevi A, Duquesnoy RJ, Dummer JS, Griffith BP, Hardesty RL, Trento A, Nalesnik MA, Dauber JH. Unexpectedly high incidence of Pneumocystis carinii infection after lung-heart transplantation. Implications for lung defense and allograft survival. Am Rev Respir Dis 1988;137:1268-74. [PubMed]

92. Guiterrez S, Morilla R, Leon JA, Martin-Garrido I, Rivero L, Friaza V, Respaldiza N, Montes-Cano MA, Teran R, de la Horra C. High prevalence of Pneumocystis jiroveci colonization among young HIV-infected patients. J Adolesc Health 2011; 48:103-5.[PubMed]

93. Hardy AM, Wajszczuk CP, Suffredini AF, Hakala TR, Ho M. Pneumocystis carinii pneumonia in renal-transplant recipients treated with cyclosporine and steroids. J Infect Dis 1984;149:143-7. [PubMed]

94. Hardy WD, Feinberg J, Finkelstein DM, Power ME, He W, Kaczka C, Frame PT, Holmes M, Waskin H, Fass RJ, et al. A controlled trial of trimethoprim-sulfamethoxazole or aerosolized pentamidine for secondary prophylaxis of Pneumocystis carinii pneumonia in patients with the acquired immunodeficiency syndrome. AIDS Clinical Trials Group Protocol 021. N Engl J Med 1992;327:1842-8. [PubMed]

95. Hardy WD, Northfelt DW, Drake TA. Fatal, disseminated pneumocystosis in a patient with acquired immunodeficiency syndrome receiving prophylactic aerosolized pentamidine. Am J Med 1989;87:329-31. [PubMed]

96. Helweg-Larsen J, Benfield TL, Eugen-Olsen J, Lundgren JD, Lundgren B. Effects of mutations in Pneumocystis carinii dihydropteroate synthase gene on outcome of AIDS-associated P. carinii pneumonia. Lancet 1999;354:1347-51. [PubMed]

97. Helweg-Larsen J, Jensen JS, Benfield T, Svendsen UG, Lundgren JD, Lundgren B. Diagnostic use of PCR for detection of Pneumocystis carinii in oral wash samples. J Clin Microbiol 1998;36:2068-72. [PubMed]

98. Helweg-Larsen J, Jensen JS, Dohn B, Benfield TL, Lundgren B. Detection of Pneumocystis DNA in samples from patients suspected of bacterial pneumonia--a case-control study. BMC Infect Dis 2002;2:28. [PubMed]

99. Helweg-Larsen J, Tsolaki AG, Miller RF, Lundgren B, Wakefield AE. Clusters of Pneumocystis carinii pneumonia: analysis of person-to-person transmission by genotyping. QJM 1998;91:813-20. [PubMed]

100. Henson JW, Jalaj JK, Walker RW, Stover DE, Fels AO. Pneumocystis carinii pneumonia in patients with primary brain tumors. Arch Neurol 1991;48:406-9. [PubMed]

101. Hirsch HH, Kaufmann G, Sendi P, Battegay M. Immune reconstitution in HIV-infected patients. Clin Infect Dis 2004;38:1159-66.[PubMed]

102. Hofflin JM, Potasman I, Baldwin JC, Oyer PE, Stinson EB, Remington JS. Infectious complications in heart transplant recipients receiving cyclosporine and corticosteroids. Ann Intern Med 1987;106:209-16. [PubMed]

103. Hoffman GS, Kerr GS, Leavitt RY, Hallahan CW, Lebovics RS, Travis WD, Rottem M, Fauci AS. Wegener granulomatosis: an analysis of 158 patients. Ann Intern Med 1992;116:488-98. [PubMed]

104. Holtzer CD, Flaherty JF, Jr., Coleman RL. Cross-reactivity in HIV-infected patients switched from trimethoprim-sulfamethoxazole to dapsone. Pharmacotherapy 1998;18:831-5. [PubMed]

105. Huang L, Beard CB, Creasman J, Levy D, Duchin JS, Lee S, Pieniazek N, Carter JL, del Rio C, Rimland D, Navin TR. Sulfa or sulfone prophylaxis and geographic region predict mutations in the Pneumocystis carinii dihydropteroate synthase gene. J Infect Dis 2000;182:1192-8. [PubMed]

106. Huang L, Crothers K, Atzori C, Benfield T, Miller R, Rabodonirina M, Helweg-Larsen J. Dihydropteroate synthase gene mutations inPneumocystis and sulfa resistance. Emerg Infect Dis 2004;10:1721-8. [PubMed]

107. Huang L, Crothers K, Morris A, Groner G, Fox M, Turner JR, Merrifield C, Eiser S, Zucchi P, Beard CB. Pneumocystis colonization in HIV-infected patients. J Eukaryot Microbiol 2003;50 Suppl:616-7. [PubMed]

108. Huang L, Morris A, Limper AH, Beck JM. An Official ATS Workshop Summary: Recent advances and future directions inPneumocystis pneumonia (PCP). Proceedings of the American Thoracic Society 2006;3:655-64. [PubMed]

109. Huang SN, Fischer SH, O'Shaughnessy E, Gill VJ, Masur H, Kovacs JA. Development of a PCR assay for diagnosis of Pneumocystis carinii pneumonia based on amplification of the multicopy major surface glycoprotein gene family. Diagn Microbiol Infect Dis 1999;35:27-32. [PubMed]

110. Hughes W, Leoung G, Kramer F, Bozzette SA, Safrin S, Frame P, Clumeck N, Masur H, Lancaster D, Chan C, et al. Comparison of atovaquone (566C80) with trimethoprim-sulfamethoxazole to treat Pneumocystis carinii pneumonia in patients with AIDS. N Engl J Med 1993;328:1521-7. [PubMed]

111. Hughes WT, Feldman S, Aur RJ, Verzosa MS, Hustu HO, Simone JV. Intensity of immunosuppressive therapy and the incidence ofPneumocystis carinii pneumonitis. Cancer 1975;36:2004-9. [PubMed]

112. Hughes WT, Kuhn S, Chaudhary S, Feldman S, Verzosa M, Aur RJ, Pratt C, George SL. Successful chemoprophylaxis forPneumocystis carinii pneumonitis. N Engl J Med 1977;297:1419-26. [PubMed]

113. Hughes WT, LaFon SW, Scott JD, Masur H. Adverse events associated with trimethoprim-sulfamethoxazole and atovaquone during the treatment of AIDS-related Pneumocystis carinii pneumonia. J Infect Dis 1995;171:1295-301. [PubMed]

114. Hughes WT, Rivera GK, Schell MJ, Thornton D, Lott L. Successful intermittent chemoprophylaxis for Pneumocystis cariniipneumonitis. N Engl J Med 1987;316:1627-32. [PubMed]

115. Iliades P, Meshnick SR, Macreadie IG. Mutations in the Pneumocystis jirovecii DHPS gene confer cross-resistance to sulfa drugs. Antimicrob Agents Chemother 2005;49:741-8. [PubMed]

116. Ioannidis JP, Cappelleri JC, Skolnik PR, Lau J, Sacks HS. A meta-analysis of the relative efficacy and toxicity of Pneumocystis cariniiprophylactic regimens. Arch Intern Med 1996;156:177-88. [PubMed]

117. Ives NJ, Gazzard BG, Easterbrook PJ. The changing pattern of AIDS-defining illnesses with the introduction of highly active antiretroviral therapy (HAART) in a London clinic. J Infect 2001;42:134-9. [PubMed]

118. Jensen PA, Lambert LA, Iademarco MF, Ridzon R. Guidelines for preventing the transmission of Mycobacterium tuberculosis in health-care settings, 2005. MMWR Recomm Rep 2005;54:1-141. [PubMed]

119. Kales CP, Murren JR, Torres RA, Crocco JA. Early predictors of in-hospital mortality for Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. Arch Intern Med 1987;147:1413-7. [PubMed]

120. Kalyoncu U, Karadag O, Akdogan A, Kisacik B, Erman M, Erguven S, Ertenli AI. Pneumocystis carinii pneumonia in a rheumatoid arthritis patient treated with adalimumab. Scand J Infect Dis 2007;39:475-8. [PubMed]

121. Kaplan JE, Benson C, Holmes KH, Brooks JT, Pau A, Masur H. Guidelines for prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: recommendations from CDC, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America. MMWR Recomm Rep 2009;58:1-207. [PubMed]

122. Kaplan JE, Hanson D, Dworkin MS, Frederick T, Bertolli J, Lindegren ML, Holmberg S, Jones JL. Epidemiology of human immunodeficiency virus-associated opportunistic infections in the United States in the era of highly active antiretroviral therapy. Clin Infect Dis 2000;30 Suppl 1:S5-14. [PubMed]

123. Kasolo F, Lishimpi K, Chintu C, Mwaba P, Mudenda V, Maswahu D, Terunuma H, Fletcher H, Nunn A, Lucas S, Zumla A. Identification of Pneumocystis carinii DNA by polymerase chain reaction in necropsy lung samples from children dying of respiratory tract illnesses. J Pediatr 2002;140:367-9. [PubMed]

124. Kazanjian P, Armstrong W, Hossler PA, Burman W, Richardson J, Lee CH, Crane L, Katz J, Meshnick SR. Pneumocystis cariniimutations are associated with duration of sulfa or sulfone prophylaxis exposure in AIDS patients. J Infect Dis 2000;182:551-7. [PubMed]

125. Kazanjian P, Locke AB, Hossler PA, Lane BR, Bartlett MS, Smith JW, Cannon M, Meshnick SR. Pneumocystis carinii mutations associated with sulfa and sulfone prophylaxis failures in AIDS patients. AIDS 1998;12:873-8. [PubMed]

126. Keely SP, Stringer JR. Complexity of the MSG gene family of Pneumocystis carinii. BMC Genomics 2009; 10:367. [PubMed]

127. Keely SP, Stringer JR. Sequences of Pneumocystis carinii f. sp. hominis strains associated with recurrent pneumonia vary at multiple loci. J Clin Microbiol 1997;35:2745-7. [PubMed]

128. Keely SP, Stringer JR, Baughman RP, Linke MJ, Walzer PD, Smulian AG. Genetic variation among Pneumocystis carinii hominisisolates in recurrent pneumocystosis. J Infect Dis 1995;172:595-8. [PubMed]

129. Kelley CF, Checkley W, Mannino DM, Franco-Paredes C, Del Rio C, Holguin F. Trends in hospitalizations for AIDS-associated Pneumocystis jirovecii pneumonia in the United States (1986 to 2005). Chest: 2009; 136: 190-197. [PubMed] 

130. Kessl JJ, Hill P, Lange BB, Meshnick SR, Meunier B, Trumpower BL. Molecular basis for atovaquone resistance in Pneumocystis jirovecii modeled in the cytochrome bc(1) complex of Saccharomyces cerevisiae. J Biol Chem 2004;279:2817-24. [PubMed]

131. Khouli H, Afrasiabi A, Shibli M, Hajal R, Barrett CR, Homel P. Outcome of critically ill human immunodeficiency virus-infected patients in the era of highly active antiretroviral therapy. J Intensive Care Med 2005;20:327-33. [PubMed]

132. Kirsch CM, Jensen WA, Kagawa FT, Azzi RL. Analysis of induced sputum for the diagnosis of recurrent Pneumocystis cariniipneumonia. Chest 1992;102:1152-4. [PubMed]

133. Kling HM, Shipley TW, Patil SP, Kristoff J, Bryan M, Montelaro RC, Morris A, Norris KA. Relationship of Pneumocystis jiroveci humoral immunity to prevention of colonization and chronic obstructive pulmonary disease in a primate model of HIV infection. Infect Immun 2010; 78: 4320-30. [PubMed]

134. Kovacs JA, Halpern JL, Swan JC, Moss J, Parrillo JE, Masur H. Identification of antigens and antibodies specific for Pneumocystis carinii. J Immunol 1988;140:2023-31. [PubMed]

135. Kovacs JA, Hiemenz JW, Macher AM, Stover D, Murray HW, Shelhamer J, Lane HC, Urmacher C, Honig C, Longo DL, et al.Pneumocystis carinii pneumonia: a comparison between patients with the acquired immunodeficiency syndrome and patients with other immunodeficiencies. Ann Intern Med 1984;100:663-71. [PubMed]

136. Kovacs JA, Masur H. Prophylaxis for Pneumocystis carinii pneumonia in patients infected with human immunodeficiency virus. Clin Infect Dis 1992;14:1005-9. [PubMed]

137. Kovacs JA, Ng VL, Masur H, Leoung G, Hadley WK, Evans G, Lane HC, Ognibene FP, Shelhamer J, Parrillo JE, et al. Diagnosis ofPneumocystis carinii pneumonia: improved detection in sputum with use of monoclonal antibodies. N Engl J Med 1988;318:589-93.[PubMed]

138. Koval CE, Gigliotti F, Nevins D, Demeter LM. Immune reconstitution syndrome after successful treatment of Pneumocystis cariniipneumonia in a man with human immunodeficiency virus type 1 infection. Clin Infect Dis 2002;35:491-3. [PubMed]

139. Lane BR, Ast JC, Hossler PA, Mindell DP, Bartlett MS, Smith JW, Meshnick SR. Dihydropteroate synthase polymorphisms inPneumocystis carinii. J Infect Dis 1997;175:482-5. [PubMed]

140. Larsen HH, Huang L, Kovacs JA, Crothers K, Silcott VA, Morris A, Turner JR, Beard CB, Masur H, Fischer SH. A prospective, blinded study of quantitative touch-down polymerase chain reaction using oral-wash samples for diagnosis of Pneumocystis pneumonia in HIV-infected patients. J Infect Dis 2004;189:1679-83. [PubMed]

141. Larsen HH, von Linstow ML, Lundgren B, Hogh B, Westh H, Lundgren JD. Primary Pneumocystis infection in infants hospitalized with acute respiratory tract infection. Emerg Infect Dis 2007;13:66-72. [PubMed]

142. Lee LH, Gigliotti F, Wright TW, Simpson-Haidaris PJ, Weinberg GA, Haidaris CG. Molecular characterization of KEX1, a kexin-like protease in mouse Pneumocystis carinii. Gene 2000;242:141-50. [PubMed]

143. Leigh TR, Kangro HO, Gazzard BG, Jeffries DJ, Collins JV. DNA amplification by the polymerase chain reaction to detect sub-clinical Pneumocystis carinii colonization in HIV-positive and HIV-negative male homosexuals with and without respiratory symptoms. Respir Med 1993;87:525-9. [PubMed]

144. Leoung GS, Feigal DW, Jr., Montgomery AB, Corkery K, Wardlaw L, Adams M, Busch D, Gordon S, Jacobson MA, Volberding PA, et al. Aerosolized pentamidine for prophylaxis against Pneumocystis carinii pneumonia. The San Francisco community prophylaxis trial. N Engl J Med 1990;323:769-75. [PubMed]

145. Leoung GS, Mills J, Hopewell PC, Hughes W, Wofsy C. Dapsone-trimethoprim for Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. Ann Intern Med 1986;105:45-8. [PubMed]

146. Leoung GS, Stanford JF, Giordano MF, Stein A, Torres RA, Giffen CA, Wesley M, Sarracco T, Cooper EC, Dratter V, Smith JJ, Frost KR. Trimethoprim-sulfamethoxazole (TMP-SMZ) dose escalation versus direct rechallenge for Pneumocystis carinii pneumonia prophylaxis in human immunodeficiency virus-infected patients with previous adverse reaction to TMP-SMZ. J Infect Dis 2001;184:992-7. [PubMed]

147. Limper AH, Offord KP, Smith TF, Martin WJ, 2nd. Pneumocystis carinii pneumonia. Differences in lung parasite number and inflammation in patients with and without AIDS. Am Rev Respir Dis 1989;140:1204-9. [PubMed]

148. Limper AH, Thomas CF, Jr., Anders RA, Leof EB. Interactions of parasite and host epithelial cell cycle regulation during Pneumocystis carinii pneumonia. J Lab Clin Med 1997;130:132-8. [PubMed]

149. Limper AH, Know KS, Sarosi GA, Ampel NM, Bennett JE, Catanzaro A, Davies SF, Dismukes WE, Hage CA, Marr KA, Mody CH, Perfect JR, Stevens DA. American Thoracic Society Working Group. An official American Thoracic Society statement: Treatment of fungal infections in adult pulmonary and critical care patients. AJRCCM 2011; 183: 96-128. [PubMed]

150. Linke MJ, Cushion MT, Walzer PD. Properties of the major antigens of rat and human Pneumocystis carinii. Infect Immun 1989;57:1547-55. [PubMed]

151. Lipschik GY, Gill VJ, Lundgren JD, Andrawis VA, Nelson NA, Nielsen JO, Ognibene FP, Kovacs JA. Improved diagnosis ofPneumocystis carinii infection by polymerase chain reaction on induced sputum and blood. Lancet 1992;340:203-6. [PubMed]

152. Lopez Bernaldo de Quiros JC, Miro JM, Pena JM, Podzamczer D, Alberdi JC, Martinez E, Cosin J, Claramonte X, Gonzalez J, Domingo P, Casado JL, Ribera E. A randomized trial of the discontinuation of primary and secondary prophylaxis against Pneumocystis carinii pneumonia after highly active antiretroviral therapy in patients with HIV infection. Group de Estudio del SIDA 04/98. N Engl J Med 2001;344:159-67. [PubMed]

153. Lu JJ, Chen CH, Bartlett MS, Smith JW, Lee CH. Comparison of six different PCR methods for detection of Pneumocystis carinii. J Clin Microbiol 1995;33:2785-8. [PubMed]

154. Lundgren JD, Barton SE, Katlama C, Ledergerber B, Gonzalez-Lahoz J, Pinching AJ, Proenca R, Hemmer R, Pedersen C, Phillips AN. Changes in survival over time after a first episode of Pneumocystis carinii pneumonia for European patients with acquired immunodeficiency syndrome. Multicentre Study Group on AIDS in Europe. Arch Intern Med 1995;155:822-8. [PubMed]

155. Ma L, Borio L, Masur H, Kovacs JA. Pneumocystis carinii dihydropteroate synthase but not dihydrofolate reductase gene mutations correlate with prior trimethoprim-sulfamethoxazole or dapsone use. J Infect Dis 1999;180:1969-78. [PubMed]

156. Machado CM, Macedo MC, Medeiros RS, Massumoto C, Silva AC, Castelli JB, Silva RL, Ostronoff M, Dulley FL. PrimaryPneumocystis carinii prophylaxis with aerosolized pentamidine after bone marrow transplantation. Acta haematologica 1998;99:54-6.[PubMed]

157. Mansharamani NG, Balachandran D, Vernovsky I, Garland R, Koziel H. Peripheral blood CD4 + T-lymphocyte counts duringPneumocystis carinii pneumonia in immunocompromised patients without HIV infection. Chest 2000;118:712-20. [PubMed]

158. Maskell NA, Waine DJ, Lindley A, Pepperell JC, Wakefield AE, Miller RF, Davies RJ. Asymptomatic carriage of Pneumocystis jiroveci in subjects undergoing bronchoscopy: a prospective study. Thorax 2003;58:594-7. [PubMed]

159. Masur H. Prevention and treatment of Pneumocystis pneumonia. N Engl J Med 1992;327:1853-60. [PubMed]

160. Masur H, Michelis MA, Greene JB, Onorato I, Stouwe RA, Holzman RS, Wormser G, Brettman L, Lange M, Murray HW, Cunningham-Rundles S. An outbreak of community-acquired Pneumocystis carinii pneumonia: initial manifestation of cellular immune dysfunction. N Engl J Med 1981;305:1431-8. [PubMed]

161. Matos O, Costa MC, Lundgren B, Caldeira L, Aguiar P, Antunes F. Effect of oral washes on the diagnosis of Pneumocystis cariniipneumonia with a low parasite burden and on detection of organisms in subclinical infections. Eur J Clin Microbiol Infect Dis 2001;20:573-5.[PubMed]

162. Matsumoto Y, Yoshida Y. Sporogony in Pneumocystis carinii: synaptonemal complexes and meiotic nuclear divisions observed in precysts. J Protozool 1984;31:420-8. [PubMed]

163. Medrano FJ, Montes-Cano M, Conde M, de la Horra C, Respaldiza N, Gasch A, Perez-Lozano MJ, Varela JM, Calderon EJ.Pneumocystis jirovecii in general population. Emerg Infect Dis 2005;11:245-50. [PubMed]

164. Meduri GU, Stover DE, Greeno RA, Nash T, Zaman MB. Bilateral bronchoalveolar lavage in the diagnosis of opportunistic pulmonary infections. Chest 1991;100:1272-6. [PubMed]

165. Mei Q, Gurunathan S, Masur H, Kovacs JA. Failure of co-trimoxazole in Pneumocystis carinii infection and mutations in dihydropteroate synthase gene. Lancet 1998;351:1631-2. [PubMed]

166. Mekinian A, Durand-Joly I, Hatron PY, Moranne O, Denis G, Dei-Cas E, Morell-Dubois S, Lambert M, Launay D, Delhaes L, Hachulla E, Queyrel V. Pneumocystis jirovecii colonization in patients with systemic autoimmune diseases: prevalence, risk factors, and outcome. Rheumatology (Oxford) 2010 50: 569-77. [PubMed]

167. Metersky ML, Colt HG, Olson LK, Shanks TG. AIDS-related spontaneous pneumothorax. Risk factors and treatment. Chest 1995;108:946-51. [PubMed]

168. Meyers JD, Flournoy N, Thomas ED. Nonbacterial pneumonia after allogeneic marrow transplantation: a review of ten years' experience. Rev Infect Dis 1982;4:1119-32. [PubMed]

169. Miller KD, Masur H, Jones EC, Joe GO, Rick ME, Kelly GG, Mican JM, Liu S, Gerber LH, Blackwelder WC, Falloon J, Davey RT, Polis MA, Walker RE, Lane HC, Kovacs JA. High prevalence of osteonecrosis of the femoral head in HIV-infected adults. Ann Intern Med 2002;137:17-25. [PubMed]

170. Miller RF, Allen E, Copas A, Singer M, Edwards SG. Improved survival for HIV-infected patients with severe Pneumocystis jiroveciipneumonia is independent of highly active antiretroviral therapy. Thorax 2006;61:716-21. [PubMed]

171. Mills J, Leoung G, Medina I, Hopewell PC, Hughes WT, Wofsy C. Dapsone treatment of Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. Antimicrob Agents Chemother 1988;32:1057-60. [PubMed]

172. Mocroft A, Katlama C, Johnson AM, Pradier C, Antunes F, Mulcahy F, Chiesi A, Phillips AN, Kirk O, Lundgren JD. AIDS across Europe, 1994-98: the EuroSIDA study. Lancet 2000;356:291-6. [PubMed]

173. Mori S, Imamura F, Kiyofuji C, Ito K, Koga Y, Honda I, Sugimoto M. Pneumocystis jiroveci pneumonia in a patient with rheumatoid arthritis as a complication of treatment with infliximab, anti-tumor necrosis factor alpha neutralizing antibody. Mod Rheumatol 2006;16:58-62. [PubMed]

174. Morris A, Kingsley LA, Groner G, Lebedeva IP, Beard CB, Norris KA. Prevalence and clinical predictors of Pneumocystiscolonization among HIV-infected men. AIDS 2004;18:793-8. [PubMed]

175. Morris A, Lundgren JD, Masur H, Walzer PD, Hanson DL, Frederick T, Huang L, Beard CB, Kaplan JE. Current epidemiology ofPneumocystis pneumonia. Emerg Infect Dis 2004;10:1713-20. [PubMed]

176. Morris A, Sciurba FC, Lebedeva IP, Githaiga A, Elliott WM, Hogg JC, Huang L, Norris KA. Association of chronic obstructive pulmonary disease severity and Pneumocystis colonization. Am J Respir Crit Care Med 2004;170:408-13. [PubMed]

177. Morris A, Wachter RM, Luce J, Turner J, Huang L. Improved survival with highly active antiretroviral therapy in HIV-infected patients with severe Pneumocystis carinii pneumonia. AIDS 2003;17:73-80. [PubMed]

178. Morris A, Wei K, Afshar K, Huang L. Epidemiology and clinical significance of Pneumocystis colonization. J Infect Dis 2008;197:10-7. [PubMed]

179. Morris AM, Swanson M, Ha H, Huang L. Geographic distribution of human immunodeficiency virus-associated Pneumocystis cariniipneumonia in San Francisco. Am J Respir Crit Care Med 2000;162:1622-6. [PubMed]

180. Munoz P, Munoz RM, Palomo J, Rodriguez-Creixems M, Munoz R, Bouza E. Pneumocystis carinii infection in heart transplant recipients. Efficacy of a weekend prophylaxis schedule. Medicine (Baltimore) 1997;76:415-22. [PubMed]

181. Murphy RL, Lavelle JP, Allan JD, Gordin FM, Dupliss R, Boswell SL, Waskin HA, Davies SF, Graziano FM, Saag MS, et al. Aerosol pentamidine prophylaxis following Pneumocystis carinii pneumonia in AIDS patients: results of a blinded dose-comparison study using an ultrasonic nebulizer. Am J Med 1991;90:418-26. [PubMed]

182. Mussini C, Pezzotti P, Govoni A, Borghi V, Antinori A, d'Arminio Monforte A, De Luca A, Mongiardo N, Cerri MC, Chiodo F, Concia E, Bonazzi L, Moroni M, Ortona L, Esposito R, Cossarizza A, De Rienzo B. Discontinuation of primary prophylaxis forPneumocystis carinii pneumonia and toxoplasmic encephalitis in human immunodeficiency virus type I-infected patients: the changes in opportunistic prophylaxis study. J Infect Dis 2000;181:1635-42. [PubMed]

183. Nahimana A, Rabodonirina M, Bille J, Francioli P, Hauser PM. Mutations of Pneumocystis jirovecii dihydrofolate reductase associated with failure of prophylaxis. Antimicrob Agents Chemother 2004;48:4301-5. [PubMed]

184. Navin TR, Beard CB, Huang L, del Rio C, Lee S, Pieniazek NJ, Carter JL, Le T, Hightower A, Rimland D. Effect of mutations inPneumocystis carinii dihydropteroate synthase gene on outcome of P carinii pneumonia in patients with HIV-1: a prospective study. Lancet 2001;358:545-9. [PubMed]

185. Nevez G, Jounieaux V, Linas MD, Guyot K, Leophonte P, Massip P, Schmit JL, Seguela JP, Camus D, Dei-Cas E, Raccurt C, Mazars E. High frequency of Pneumocystis carinii sp.f. hominis colonization in HIV-negative patients. J Eukaryot Microbiol 1997;44:36S.[PubMed]

186. Nevez G, Magois E, Duwat H, Gouilleux V, Jounieaux V, Totet A. Apparent absence of Pneumocystis jirovecii in healthy subjects. Clin Infect Dis 2006;42:e99-101. [PubMed]

187. Nevez G, Raccurt C, Vincent P, Jounieaux V, Dei-Cas E. Pulmonary colonization with Pneumocystis carinii in human immunodeficiency virus-negative patients: assessing risk with blood CD4+ T cell counts. Clin Infect Dis 1999;29:1331-2. [PubMed]

188. Nevez G, Totet A, Pautard JC, Raccurt C. Pneumocystis carinii detection using nested-PCR in nasopharyngeal aspirates of immunocompetent infants with bronchiolitis. J Eukaryot Microbiol 2001;Suppl:122S-23S. [PubMed]

189. Ng VL, Virani NA, Chaisson RE, Yajko DM, Sphar HT, Cabrian K, Rollins N, Charache P, Krieger M, Hadley WK, et al. Rapid detection of Pneumocystis carinii using a direct fluorescent monoclonal antibody stain. J Clin Microbiol 1990;28:2228-33. [PubMed]

190. Norris KA, Morris A, Patil S, Fernandes E. Pneumocystis colonization, airway inflammation, and pulmonary function decline in acquired immunodeficiency syndrome. Immunol Res 2006;36:175-87. [PubMed]

191. Noskin GA, Murphy RL, Black JR, Phair JP. Salvage therapy with clindamycin/primaquine for Pneumocystis carinii pneumonia. Clin Infect Dis 1992;14:183-8. [PubMed]

192. O'Riordan DM, Standing JE, Limper AH. Pneumocystis carinii glycoprotein A binds macrophage mannose receptors. Infect Immun 1995;63:779-84. [PubMed]

193. Ognibene FP, Shelhamer J, Gill V, Macher AM, Loew D, Parker MM, Gelmann E, Fauci AS, Parrillo JE, Masur H. The diagnosis ofPneumocystis carinii pneumonia in patients with the acquired immunodeficiency syndrome using subsegmental bronchoalveolar lavage. Am Rev Respir Dis 1984;129:929-32. [PubMed]

194. Ognibene FP, Shelhamer JH, Hoffman GS, Kerr GS, Reda D, Fauci AS, Leavitt RY. Pneumocystis carinii pneumonia: a major complication of immunosuppressive therapy in patients with Wegener's granulomatosis. Am J Respir Crit Care Med 1995;151:795-9.[PubMed]

195. Olsen SL, Renlund DG, O'Connell JB, Taylor DO, Lassetter JE, Eastburn TE, Hammond EH, Bristow MR. Prevention ofPneumocystis carinii pneumonia in cardiac transplant recipients by trimethoprim sulfamethoxazole. Transplantation 1993;56:359-62.[PubMed]

196. Olsson M, Elvin K, Lofdahl S, Linder E. Detection of Pneumocystis carinii DNA in sputum and bronchoalveolar lavage samples by polymerase chain reaction. J Clin Microbiol 1993;31:221-6. [PubMed]

197. Olsson M, Lidman C, Latouche S, Bjorkman A, Roux P, Linder E, Wahlgren M. Identification of Pneumocystis carinii f. sp. hominisgene sequences in filtered air in hospital environments. J Clin Microbiol 1998;36:1737-40. [PubMed]

198Opportunistic Infections Project Team of the Collaboration of Observational HIV Epidemiological Research in Europe (COHERE) et al. Is it safe to discontinue primary Pneumocystis jiroveci pneumonia prophylaxis in patients with virologically suppressed HIV infection and a CD4 cell count <200 cells/microL. Clin Infect Dis. 2010 Sep 1;51(5):611-9. [PubMed]

199. Opravil M, Hirschel B, Lazzarin A, Heald A, Pechere M, Ruttimann S, Iten A, von Overbeck J, Oertle D, Praz G, et al. Once-weekly administration of dapsone/pyrimethamine vs. aerosolized pentamidine as combined prophylaxis for Pneumocystis carinii pneumonia and toxoplasmic encephalitis in human immunodeficiency virus-infected patients. Clin Infect Dis 1995;20:531-41. [PubMed]

200. Palmer RJ, Wakefield AE. Functional glycosylphosphatidylinositol anchor signal sequences in the Pneumocystis carinii PRT1 protease family. Am J Respir Cell Mol Biol 2001;25:466-73. [PubMed]

201. Para MF, Finkelstein D, Becker S, Dohn M, Walawander A, Black JR. Reduced toxicity with gradual initiation of trimethoprim-sulfamethoxazole as primary prophylaxis for Pneumocystis carinii pneumonia: AIDS Clinical Trials Group 268. J Acquir Immune Defic Syndr 2000;24:337-43. [PubMed]

202. Pareja JG, Garland R, Koziel H. Use of adjunctive corticosteroids in severe adult non-HIV Pneumocystis carinii pneumonia. Chest 1998;113:1215-24. [PubMed]

203. Peglow SL, Smulian AG, Linke MJ, Pogue CL, Nurre S, Crisler J, Phair J, Gold JW, Armstrong D, Walzer PD. Serologic responses toPneumocystis carinii antigens in health and disease. J Infect Dis 1990;161:296-306.  [PubMed]

204. Peters SE, Wakefield AE, Sinclair K, Millard PR, Hopkin JM. A search for Pneumocystis carinii in post-mortem lungs by DNA amplification. J Pathol 1992;166:195-8. [PubMed]

205. Peters SG, Prakash UB. Pneumocystis carinii pneumonia. Review of 53 cases. Am J Med 1987;82:73-8. [PubMed]

206. Phair J, Munoz A, Detels R, Kaslow R, Rinaldo C, Saah A. The risk of Pneumocystis carinii pneumonia among men infected with human immunodeficiency virus type 1. Multicenter AIDS Cohort Study Group. N Engl J Med 1990;322:161-5. [PubMed]

207. Pifer LL, Hughes WT, Stagno S, Woods D. Pneumocystis carinii infection: evidence for high prevalence in normal and immunosuppressed children. Pediatrics 1978;61:35-41. [PubMed]

208. Pitchenik AE, Ganjei P, Torres A, Evans DA, Rubin E, Baier H. Sputum examination for the diagnosis of Pneumocystis cariniipneumonia in the acquired immunodeficiency syndrome. Am Rev Respir Dis 1986;133:226-9. [PubMed]

209. Podzamczer D, Salazar A, Jimenez J, Consiglio E, Santin M, Casanova A, Rufi G, Gudiol F. Intermittent trimethoprim-sulfamethoxazole compared with dapsone-pyrimethamine for the simultaneous primary prophylaxis of Pneumocystis pneumonia and toxoplasmosis in patients infected with HIV. Ann Intern Med 1995;122:755-61. [PubMed]

210. Ponce CA, Gallo M, Bustamante R, Vargas SL. Pneumocystis colonization is highly prevalent in the autopsied lungs of the general population. Clin Infect Dis 2010; 40: 347-53. [PubMed]

211. Powell K, Davis JL, Morris AM, Chi A, Bensley MR, Huang L. Survival for patients with Human Immunodeficiency Virus admitted to the intensive care unit continues to improve in the current era of highly active antiretroviral therapy. Chest 2009;135:11-17. [PubMed]

212. Rabaud C, Charreau I, Izard S, Raffi F, Meiffredy V, Leport C, Guillemin F, Yeni P, Aboulker JP. Adverse reactions to cotrimoxazole in HIV-infected patients: predictive factors and subsequent HIV disease progression. Scand J Infect Dis 2001;33:759-64. [PubMed]

213. Rabodonirina M, Raffenot D, Cotte L, Boibieux A, Mayencon M, Bayle G, Persat F, Rabatel F, Trepo C, Peyramond D, Piens MA. Rapid detection of Pneumocystis carinii in bronchoalveolar lavage specimens from human immunodeficiency virus-infected patients: use of a simple DNA extraction procedure and nested PCR. J Clin Microbiol 1997;35:2748-51. [PubMed]

214. Rabodonirina M, Vanhems P, Couray-Targe S, Gillibert RP, Ganne C, Nizard N, Colin C, Fabry J, Touraine JL, van Melle G, Nahimana A, Francioli P, Hauser PM. Molecular evidence of interhuman transmission of Pneumocystis pneumonia among renal transplant recipients hospitalized with HIV-infected patients. Emerg Infect Dis 2004;10:1766-73. [PubMed]

215. Radhi S, Alexander T, Ukwu M, Saleh S, Morris A. Outcome of HIV-associated Pneumocystis pneumonia in hospitalized patients from 2000 through 2003. BMC Infect Dis 2008;8:118. [PubMed]

216. Ratnam I, Chiu C, Kandala NB, Easterbrook PJ. Incidence and risk factors for immune reconstitution inflammatory syndrome in an ethnically diverse HIV type 1-infected cohort. Clin Infect Dis 2006;42:418-27. [PubMed]

217. Rodriguez M, Fishman JA. Prevention of infection due to Pneumocystis spp. in human immunodeficiency virus-negative immunocompromised patients. Clin Microbiol Rev 2004;17:770-82, table of contents. [PubMed]

218. Roger PM, Vandenbos F, Pugliese P, De Salvador F, Durant J, Le Fichoux Y, Dellamonica P. Persistence of Pneumocystis carinii after effective treatment of P. carinii pneumonia is not related to relapse or survival among patients infected with human immunodeficiency virus. Clin Infect Dis 1998;26:509-10. [PubMed]

219. Roths JB, Sidman CL. Single and combined humoral and cell-mediated immunotherapy of Pneumocystis carinii pneumonia in immunodeficient SCID mice. Infect Immun 1993;61:1641-9. [PubMed]

220. Ruf B, Rohde I, Pohle HD. Efficacy of clindamycin/primaquine versus trimethoprim/sulfamethoxazole in primary treatment ofPneumocystis carinii pneumonia. Eur J Clin Microbiol Infect Dis 1991;10:207-10. [PubMed]

221. Saah AJ, Hoover DR, Peng Y, Phair JP, Visscher B, Kingsley LA, Schrager LK. Predictors for failure of Pneumocystis cariniipneumonia prophylaxis. Multicenter AIDS Cohort Study. JAMA 1995;273:1197-202. [PubMed]

222. Safrin S, Finkelstein DM, Feinberg J, Frame P, Simpson G, Wu A, Cheung T, Soeiro R, Hojczyk P, Black JR. Comparison of three regimens for treatment of mild to moderate Pneumocystis carinii pneumonia in patients with AIDS. A double-blind, randomized, trial of oral trimethoprim-sulfamethoxazole, dapsone-trimethoprim, and clindamycin-primaquine. ACTG 108 Study Group. Ann Intern Med 1996;124:792-802. [PubMed]

223. Safrin S, Sattler FR, Lee BL, Young T, Bill R, Boylan CT, Mills J. Dapsone as a single agent is suboptimal therapy for Pneumocystis carinii pneumonia. J Acquir Immune Defic Syndr 1991;4:244-9. [PubMed]

224. Sandhu GS, Kline BC, Espy MJ, Stockman L, Smith TF, Limper AH. Laboratory diagnosis of Pneumocystis carinii infections by PCR directed to genes encoding for mitochondrial 5S and 28S ribosomal RNA. Diagn Microbiol Infect Dis 1999;33:157-62. [PubMed]

225. Sangiolo D, Storer B, Nash R, Corey L, Davis C, Flowers M, Hackman RC, Boeckh M. Toxicity and efficacy of daily dapsone asPneumocystis jiroveci prophylaxis after hematopoietic stem cell transplantation: a case-control study. Biol Blood Marrow Transplant 2005;11:521-9. [PubMed]

226. Sattler FR, Cowan R, Nielsen DM, Ruskin J. Trimethoprim-sulfamethoxazole compared with pentamidine for treatment ofPneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. A prospective, noncrossover study. Ann Intern Med 1988;109:280-7. [PubMed]

227. Sattler FR, Frame P, Davis R, Nichols L, Shelton B, Akil B, Baughman R, Hughlett C, Weiss W, Boylen CT, et al. Trimetrexate with leucovorin versus trimethoprim-sulfamethoxazole for moderate to severe episodes of Pneumocystis carinii pneumonia in patients with AIDS: a prospective, controlled multicenter investigation of the AIDS Clinical Trials Group Protocol 029/031. J Infect Dis 1994;170:165-72.[PubMed]

228. Schneider MM, Hoepelman AI, Eeftinck Schattenkerk JK, Nielsen TL, van der Graaf Y, Frissen JP, van der Ende IM, Kolsters AF, Borleffs JC. A controlled trial of aerosolized pentamidine or trimethoprim-sulfamethoxazole as primary prophylaxis against Pneumocystis carinii pneumonia in patients with human immunodeficiency virus infection. The Dutch AIDS Treatment Group. N Engl J Med 1992;327:1836-41. [PubMed]

229. Sepkowitz KA. Pneumocystis carinii pneumonia among patients with neoplastic disease. Semin Respir Infect 1992;7:114-21.[PubMed]

230. Sepkowitz KA, Brown AE, Armstrong D. Pneumocystis carinii pneumonia without acquired immunodeficiency syndrome. More patients, same risk. Arch Intern Med 1995;155:1125-8. [PubMed]

231. Sepkowitz KA, Brown AE, Telzak EE, Gottlieb S, Armstrong D. Pneumocystis carinii pneumonia among patients without AIDS at a cancer hospital. JAMA 1992;267:832-7. [PubMed]

232. Sepkowitz KA, Telzak EE, Gold JW, Bernard EM, Blum S, Carrow M, Dickmeyer M, Armstrong D. Pneumothorax in AIDS. Ann Intern Med 1991;114:455-9. [PubMed]

233. Shelhamer JH, Ognibene FP, Macher AM, Tuazon C, Steiss R, Longo D, Kovacs JA, Parker MM, Natanson C, Lane HC, et al. Persistence of Pneumocystis carinii in lung tissue of acquired immunodeficiency syndrome patients treated for Pneumocystis pneumonia. Am Rev Respir Dis 1984;130:1161-5. [PubMed]

234. Shipley TW , Kling HM, Morris A, Patil S, Kristoff J, Guyach SE, Murphy JE, Shao X, Sciurba FC, Rogers RM, Richards T, Thompson P, Montelaro RC, Coxson HO, Hogg, JC, Norris KA. Persistent Pneumocystis colonization leads to the developments of chronic obstructive pulmonary disease in a nonhuman primate model of AIDS. J Infect Dis 2010; 202: 302-12. [PubMed]

235. Skelly M, Hoffman J, Fabbri M, Holzman RS, Clarkson AB, Jr., Merali S. S-adenosylmethionine concentrations in diagnosis ofPneumocystis carinii pneumonia. Lancet 2003;361:1267-8. [PubMed]

236. Skelly MJ, Holzman RS, Merali S. S-adenosylmethionine levels in the diagnosis of Pneumocystis carinii pneumonia in patients with HIV infection. Clin Infect Dis 2008;46:467-71. [PubMed]

237. Stansell JD, Osmond DH, Charlebois E, LaVange L, Wallace JM, Alexander BV, Glassroth J, Kvale PA, Rosen MJ, Reichman LB, Turner JR, Hopewell PC. Predictors of Pneumocystis carinii pneumonia in HIV-infected persons. Pulmonary Complications of HIV Infection Study Group. Am J Respir Crit Care Med 1997;155:60-6. [PubMed]

238. Stein CR, Poole C, Kazanjian P, Meshnick SR. Sulfa use, dihydropteroate synthase mutations, and Pneumocystis jirovecii pneumonia. Emerg Infect Dis 2004;10:1760-5. [PubMed]

239. Stringer JR. Antigenic variation in Pneumocystis. J Eukaryot Microbiol 2007;54:8-13. [PubMed]

240. Stringer JR, Beard CB, Miller RF, Wakefield AE. A new name (Pneumocystis jiroveci) for Pneumocystis from humans. Emerg Infect Dis 2002;8:891-6. [PubMed]

241. Stringer SL, Hudson K, Blase MA, Walzer PD, Cushion MT, Stringer JR. Sequence from ribosomal RNA of Pneumocystis cariniicompared to those of four fungi suggests an ascomycetous affinity. J Protozool 1989;36:14S-16S. [PubMed]

242. Sugimoto H, Uchida H, Akiyama N, Nagao T, Tomikawa S, Mita K, Beck Y, Inoue S, Watanabe K, Nakayama Y, et al. Improved survival of renal allograft recipients with Pneumocystis carinii pneumonia by early diagnosis and treatment. Transplant Proc 1992;24:1556-8.[PubMed]

243. Tasaka S, Hasegawa N, Kobayashi S, Yamada W, Nishimura T, Takeuchi T, Ishizaka A. Serum indicators for the diagnosis ofPneumocystis pneumonia. Chest 2007;131:1173-80. [PubMed]

244. Thomas CF, Limper AH. Pneumocystis pneumonia: clinical presentation and diagnosis in patients with and without acquired immune deficiency syndrome. Semin Respir Infect 1998;13:289-95. [PubMed]

245. Thomas CF, Jr., Limper AH. Pneumocystis pneumonia. N Engl J Med 2004;350:2487-98. [PubMed]

246. Thomas CF, Jr., Limper AH. Current insights into the biology and pathogenesis of Pneumocystis pneumonia. Nat Rev Microbiol 2007;5:298-308. [PubMed]

247. Toma E. Clindamycin/primaquine for treatment of Pneumocystis carinii pneumonia in AIDS. Eur J Clin Microbiol Infect Dis 1991;10:210-3. [PubMed]

248. Toma E, Fournier S, Poisson M, Morisset R, Phaneuf D, Vega C. Clindamycin with primaquine for Pneumocystis carinii pneumonia. Lancet 1989;1:1046-8. [PubMed]

249. Totet A, Meliani L, Lacube P, Pautard JC, Raccurt C, Roux P, Nevez G. Immunocompetent infants as a human reservoir forPneumocystis jirovecii: rapid screening by non-invasive sampling and real-time PCR at the mitochondrial large subunit rRNA gene. J Eukaryot Microbiol 2003;50 Suppl:668-9. [PubMed]

250. Tuan IZ, Dennison D, Weisdorf DJ. Pneumocystis carinii pneumonitis following bone marrow transplantation. Bone Marrow Transplant 1992;10:267-72. [PubMed]

251. Tudor-Williams G. HIV infection in children in developing countries. Trans R Soc Trop Med Hyg 2000;94:3-4. [PubMed]

252. Vargas SL, Hughes WT, Santolaya ME, Ulloa AV, Ponce CA, Cabrera CE, Cumsille F, Gigliotti F. Search for primary infection byPneumocystis carinii in a cohort of normal, healthy infants. Clin Infect Dis 2001;32:855-61. [PubMed]

253Vargas SL, Pizarro P, Lopez-Vieyra M, Neira-Aviles P, Bustamante R, Ponce CA. Pneumocystis colonization in older adults and diagnostic yield of single versus paired noninvasive sampling. Clin Infect Dis. 2010 50(3):e19-21. [PubMed]

254. Vargas SL, Ponce CA, Galvez P, Ibarra C, Haas EA, Chadwick AE, Krous HF. Pneumocystis is not a direct cause of sudden infant death syndrome. Pediatr Infect Dis J 2007;26:81-3. [PubMed]

255. Vargas SL, Ponce CA, Gigliotti F, Ulloa AV, Prieto S, Munoz MP, Hughes WT. Transmission of Pneumocystis carinii DNA from a patient with P. carinii pneumonia to immunocompetent contact health care workers. J Clin Microbiol 2000;38:1536-8. [PubMed]

256. Vargas SL, Ponce CA, Hughes WT, Wakefield AE, Weitz JC, Donoso S, Ulloa AV, Madrid P, Gould S, Latorre JJ, Avila R, Benveniste S, Gallo M, Belletti J, Lopez R. Association of primary Pneumocystis carinii infection and sudden infant death syndrome. Clin Infect Dis 1999;29:1489-93. [PubMed]

257. Vargas SL, Ponce CA, Luchsinger V, Silva C, Gallo M, Lopez R, Belletti J, Velozo L, Avila R, Palomino MA, Benveniste S, Avendano LF. Detection of Pneumocystis carinii f. sp. hominis and viruses in presumably immunocompetent infants who died in the hospital or in the community. J Infect Dis 2005;191:122-6. [PubMed]

258. Vargas SL, Ponce CA, Sanchez CA, Ulloa AV, Bustamante R, Juarez G. Pregnancy and asymptomatic carriage of Pneumocystis jiroveci. Emerg Infect Dis 2003;9:605-6. [PubMed]

259. Volpe F, Ballantine SP, Delves CJ. The multifunctional folic acid synthesis fas gene of Pneumocystis carinii encodes dihydroneopterin aldolase, hydroxymethyldihydropterin pyrophosphokinase and dihydropteroate synthase. Eur J Biochem 1993;216:449-58. [PubMed]

260. Vuk-Pavlovic et al. Carbohydrate recognition domain of surfactant protein D mediates interactions with Pneumocystis cariniiglycoprotein A. AJRCMB 2001; 24: 475-484. [PubMed]

261. Wakefield AE, Banerji S, Pixley FJ, Hopkin JM. Molecular probes for the detection of Pneumocystis carinii. Trans R Soc Trop Med Hyg 1990;84 Suppl 1:17-8. [PubMed]

262. Wakefield AE, Lindley AR, Ambrose HE, Denis CM, Miller RF. Limited asymptomatic carriage of Pneumocystis jiroveci in human immunodeficiency virus-infected patients. J Infect Dis 2003;187:901-8. [PubMed]

263. Wakefield AE, Pixley FJ, Banerji S, Sinclair K, Miller RF, Moxon ER, Hopkin JM. Amplification of mitochondrial ribosomal RNA sequences from Pneumocystis carinii DNA of rat and human origin. Mol Biochem Parasitol 1990;43:69-76. [PubMed]

264. Wakefield AE, Pixley FJ, Banerji S, Sinclair K, Miller RF, Moxon ER, Hopkin JM. Detection of Pneumocystis carinii with DNA amplification. Lancet 1990;336:451-3. [PubMed]

265. Walzer PD, Evans HE, Copas AJ, Edwards SG, Grant AD, Miller RF. Early predictors of mortality from Pneumocystis jiroveciipneumonia in HIV-infected patients: 1985-2006. Clin Infect Dis 2008;46:625-33. [PubMed]

266. Weverling GJ, Mocroft A, Ledergerber B, Kirk O, Gonzales-Lahoz J, d'Arminio Monforte A, Proenca R, Phillips AN, Lundgren JD, Reiss P. Discontinuation of Pneumocystis carinii pneumonia prophylaxis after start of highly active antiretroviral therapy in HIV-1 infection. EuroSIDA Study Group. Lancet 1999;353:1293-8. [PubMed]

267. Wharton JM, Coleman DL, Wofsy CB, Luce JM, Blumenfeld W, Hadley WK, Ingram-Drake L, Volberding PA, Hopewell PC. Trimethoprim-sulfamethoxazole or pentamidine for Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. A prospective randomized trial. Ann Intern Med 1986;105:37-44. [PubMed]

268. Wilson JW, Limper AH, Grys TE, Karre T, Wengenack NL, Binnicker MJ. Pneumocystis jirovecii testing by real-time polymerase chain reaction and direct examination among immunocompetent and immunosuppressed patient groups and correlation to disease severity (Diagn Microbiol Infect Dis 2011; 69: 145-52. [PubMed]

269. Wislez M, Bergot E, Antoine M, Parrot A, Carette MF, Mayaud C, Cadranel J. Acute respiratory failure following HAART introduction in patients treated for Pneumocystis carinii pneumonia. Am J Respir Crit Care Med 2001;164:847-51. [PubMed]

270. Wissman G, Morilla R, Martin-Garrido I, Friaza V, Respaldiza N, Povedano J, Praena-Fernandez JM, Montes-Cano MA, Medrano FJ, Goldani LZ, de la Horra C, Varela JM, Calderon EJ. Pneumocystis jirovecii colonization in patients treated with infliximab. Eur J Clin Invest 2011; 41: 343-8. [PubMed]

271. Yale SH, Limper AH. Pneumocystis carinii pneumonia in patients without acquired immunodeficiency syndrome: associated illness and prior corticosteroid therapy. Mayo Clin Proc 1996;71:5-13. [PubMed]

272. Yazaki H, Goto N, Uchida K, Kobayashi T, Gatanaga H, Oka S. Outbreak of Pneumocystis jiroveci pneumonia in renal transplant patients: P. jiroveci is contagious to the susceptible host. Transplantation 2009; 88: 380-5. [PubMed]

273. Zolopa A, Andersen J, Powderly W, Sanchez A, Sanne I, Suckow C, Hogg E, Komarow L. Early antiretroviral therapy reduces AIDS progression/death in individuals with acute opportunistic infections: a multicenter randomized strategy trial. PLoS One. 2009;4(5):e5575. Epub 2009 May 18. [PubMed]

274. Zheng M, Ramsay AJ, Robichaux MB, Norris KA, Kliment C, Crowe C, Rapaka RR, Steele C, McAllister F, Shellito JE, Marrero L, Schwarzenberger P, Zhong Q, Kolls JK. CD4+ T cell-independent DNA vaccination against opportunistic infections. J Clin Invest 2005;115:3536-44. [PubMed]

back to top

 

Table 1. Reasons for Failure to Improve with PCP Therapy

 Paradoxical response (initial worsening with effective treatment)
 Pneumothorax

 Pulmonary edema

     Non-cardiogenic

     Cardiogenic

 Undiagnosed or new infection
  Pulmonary embolism
  Alveolar hemorrhage
  Immune reconstitution inflammatory syndrome
 PCP treatment failure

 

What's New

Choukri F, Menotti J, Sarfati C, Lucet JC, Nevez G, Garin YJ, Derouin F, Totet A. Spread of Pneumocystis jirovecii in the surrounding air of patients with Pneumocystis pneumonia. Clin Infect Dis. 2010 Aug 1;51(3):259-65.

Guided Medline Search for

Epidemiology

Clinical Manifestations

Laboratory Diagnosis

Pathogenesis

Therapy

Prevention