ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Fungal infections following lung transplantation

Fungal infections following lung transplantation
Literature review current through: Jan 2024.
This topic last updated: Aug 23, 2022.

INTRODUCTION — Lung transplantation is an effective treatment for a wide range of advanced lung diseases. While the survival of lung transplant recipients continues to improve [1], outcomes after lung transplantation remain inferior to other types of solid organ transplantation. Infectious complications contribute substantially to morbidity and mortality following lung transplantation.

Fungal infections are a frequent complication in lung transplant recipients, with one-year cumulative incidence of 8.6 percent [2]. Aspergillus and Candida spp cause the majority of fungal infections in lung transplant recipients; Cryptococcus spp, the agents of mucormycosis, endemic fungi (Histoplasma, Coccidioides, and Blastomyces spp), Scedosporium spp, Fusarium spp, and dematiaceous molds are other important causes (table 1) [3,4].

This topic reviews fungal infections in lung transplant recipients. Bacterial, viral, and mycobacterial infections in lung transplant recipients, as well as the evaluation, treatment, and prophylaxis of infection in solid organ transplant recipients, are reviewed separately. (See "Bacterial infections following lung transplantation" and "Prevention of cytomegalovirus infection in lung transplant recipients" and "Tuberculosis in solid organ transplant candidates and recipients" and "Nontuberculous mycobacterial infections in solid organ transplant candidates and recipients" and "Evaluation for infection before solid organ transplantation" and "Infection in the solid organ transplant recipient" and "Prophylaxis of infections in solid organ transplantation".)

RISK OF INFECTION — Lung transplant recipients are at high risk of infectious complications due to the following factors:

The high level of immunosuppression required to prevent rejection

Adverse effects of transplantation on local pulmonary host defenses (loss of lymphatics, reduced mucociliary clearance, decreased cough)

Constant environmental contact allowing pathogens direct access into the allograft

The likelihood and type of infection varies with the degree of host immunosuppression, timing since transplantation, nature and duration of antimicrobial prophylaxis, local hospital and regional microbiology, and active or latent infections in the donor. Pneumonia is the most common type of infection in lung transplant recipients. Bacterial, viral, fungal, and mycobacterial infections all occur at an increased frequency after lung transplantation. (See "Bacterial infections following lung transplantation" and "Prevention of cytomegalovirus infection in lung transplant recipients" and "Tuberculosis in solid organ transplant candidates and recipients" and "Nontuberculous mycobacterial infections in solid organ transplant candidates and recipients".)

The majority of non-Candida fungal infections are acquired through inhalation. Pretransplant colonization, particularly with filamentous molds (eg, Aspergillus spp), also increases the risk of post-transplant pulmonary fungal infection. (See 'Risk factors' below.)

In addition to the direct morbidity and mortality caused by infectious complications, they may also lead to loss of allograft function and contribute to the development of bronchiolitis obliterans syndrome (BOS) [5]. As an example, in a cohort of 201 lung transplant recipients in which 54 (27 percent) were colonized with Aspergillus spp, such colonization was found to be an independent risk factor for BOS on multivariate analysis (hazard ratio 1.81, 95% CI 1.03-3.19) [6]. In another study, airway colonization with Aspergillus spp, especially those that have smaller conidia, was found to be a risk factor for BOS following lung transplantation [7]. (See "Infection in the solid organ transplant recipient" and "Chronic lung allograft dysfunction: Bronchiolitis obliterans syndrome".)

TIMELINE OF INFECTION — The epidemiology of fungal infections after lung transplant varies by the nature and duration of post-transplant prophylaxis. Candidemia usually occurs during the first month following lung transplantation as a result of intensive care unit exposure, increased antibiotic use, and recent surgery. Aspergillosis occurs a median of 3.2 months following lung transplantation, with 72 percent of cases occurring within the first six months [8].

Non-Aspergillus molds (eg, Scedosporium, Purpureocillium, Scopulariopsis) are frequently isolated from respiratory samples from lung transplant recipients receiving inhaled liposomal amphotericin B prophylaxis (pretransplant 11.9 percent, post-transplant 16.9 percent) but only a minority (2.4 percent) developed an invasive fungal infection [9]. Non-Aspergillus molds may present later than Aspergillus (median 419 days post-transplant compared with 363 days post-transplant) and are associated with significantly higher mortality (60.5 versus 39.5 percent) [10]. The hospital environment itself can also be considered a risk factor for the development of fungal infection after lung transplantation, and center-specific outbreaks have been observed [11].

A general discussion of the pattern of infections seen following solid organ transplantation is discussed in detail separately (figure 1). (See "Infection in the solid organ transplant recipient".)

EVALUATION FOR INFECTION — Evaluation for infection in lung transplant recipients should follow the general guidelines used in other solid organ transplant recipients and consider the diversity of pulmonary infectious diseases including tracheobronchitis, anastomotic infection, cavitary disease, and pleural space infection. (See "Infection in the solid organ transplant recipient".)

Since pneumonia is the most common type of infection in lung transplant recipients, the diagnostic evaluation of lung transplant patients generally includes early and aggressive evaluation of the lungs as a potential source. (See "Bacterial infections following lung transplantation" and 'Diagnosis' below.)

Pleural effusions diagnosed within the first three months of transplant also warrant further investigation, particularly in patients with systemic signs of infection. Although often presumed to be benign, some pleural effusions in the early post-transplant period represent infection [12].

ASPERGILLOSIS — Aspergillus spp are the most common cause of invasive fungal infection following lung transplantation and occur more often among lung transplant recipients compared with recipients of other organs (40.5 versus 1.2 cases per 1000 patient-years among lung transplant recipients and renal transplant recipients, respectively; 4.8 cases per 1000 patient-years among all solid organ transplant recipients) [13]. Approximately 6 percent (range 3 to 15 percent) of lung transplant recipients develop aspergillosis [8,13-15].

Aspergillus fumigatus is the most common species implicated in invasive aspergillosis, but other species, such as Aspergillus terreus, Aspergillus flavus, and Aspergillus niger, also cause disease [16]. A. terreus is of particular importance because it is resistant to amphotericin B in vitro [17,18]. (See "Epidemiology and clinical manifestations of invasive aspergillosis", section on 'Microbial epidemiology'.)

Risk factors — Lung transplant recipients are at increased risk for invasive aspergillosis and other infections compared with other solid organ transplant recipients because of the higher doses of immunosuppression that are required, as well as other factors that are described above. (See 'Risk of infection' above.)

Specific risk factors for invasive aspergillosis in lung transplant recipients include [8,14,16,19,20]:

Airway colonization with Aspergillus spp

Airway ischemia

Bronchiolitis obliterans (rejection)

Airway colonization with Aspergillus spp occurs in 25 to 30 percent of lung transplant recipients [14] but in approximately 50 percent among those with cystic fibrosis [21]. Cystic fibrosis patients colonized with Aspergillus pretransplant appear to have an elevated risk of Aspergillus tracheobronchitis but not of invasive pulmonary aspergillosis [21-23].

Recipients of single-lung transplants are at risk for aspergillosis in the native lung [8,24]. In a retrospective study, aspergillosis in single-lung transplant recipients was associated with significantly higher mortality than in double-lung and heart-lung transplant recipients [8]. Furthermore, single-lung transplant recipients were more likely to develop invasive aspergillosis late in the post-transplant course. Those who developed invasive aspergillosis were more likely to have chronic obstructive pulmonary disease, which predisposes to colonization with Aspergillus spp.

Risk factors for invasive aspergillosis include rejection, cytomegalovirus disease, high doses of glucocorticoids, renal failure, use of anti-T cell agents (eg, antithymocyte globulin), single-lung transplant, and colonization with Aspergillus at one year post-transplant [24,25]. (See "Infection in the solid organ transplant recipient" and "Epidemiology and clinical manifestations of invasive aspergillosis".)

Clinical manifestations — The most common acute manifestations of invasive aspergillosis in lung transplant recipients are tracheobronchitis, pneumonia, and disseminated disease. In a literature review of 78 cases of aspergillosis in lung transplant recipients, tracheobronchitis was responsible for 37 percent of cases, followed by invasive pulmonary aspergillosis (32 percent), bronchial anastomosis infections (20 percent), and disseminated infection (10 percent) [8]. Other sites of involvement may include the sinuses and/or central nervous system (CNS), and, less commonly, the vertebrae, intervertebral discs, eyes, pleural space, or skin [16]. (See "Epidemiology and clinical manifestations of invasive aspergillosis".)

Tracheobronchial aspergillosis — Tracheobronchial aspergillosis occurs most commonly in lung transplant recipients, typically within three months of transplantation [8,14,26,27].

Tracheobronchial aspergillosis can be asymptomatic and detected only by surveillance bronchoscopy [28]. Reported clinical findings have included fever, cough, wheezing, and/or hemoptysis.

The range of findings in tracheobronchial aspergillosis is varied and includes simple tracheobronchitis, obstructing bronchial aspergillosis, ulcerative tracheobronchitis, necrotizing pseudomembranous tracheobronchitis, and tracheobronchial aspergillosis with bronchopleural fistulae. Tracheobronchial aspergillosis may also progress to invasive and disseminated disease [8]. Tracheobronchial disease can occur early post-transplant at the vulnerable anastomotic site, can be a complication from the use of airway stents, and can lead to airway dehiscence [29,30].

Pulmonary aspergillosis — Pulmonary aspergillosis generally occurs later than tracheobronchial aspergillosis, with median time to diagnosis of six months post-transplant [31]. Symptoms include fever, dry cough, and dyspnea with or without hemoptysis [16]. Physical exam findings are consistent with pneumonia.

Other sites — After the lungs and airways, other common sites of invasive aspergillosis in lung transplant recipients include the sinuses, orbits, and CNS [16]. Rarer manifestations include vertebral osteomyelitis, discitis, endophthalmitis, empyema, retroperitoneal or intra-abdominal abscess, pericarditis, and skin lesions. In a literature review that included 78 lung transplant recipients with aspergillosis, disseminated infection occurred in 10 percent of patients [8].

Diagnosis — Given the increased risk of pulmonary fungal infections in lung transplant recipients, many centers employ frequent early surveillance bronchoscopies to examine the anastomosis as well as fungal staining and culture of bronchoalveolar lavage (BAL) fluid (table 2). Practices for surveillance bronchoscopy vary widely by transplant center. At centers that routinely perform bronchoscopy, the primary purpose is to evaluate for allograft rejection although BAL provides an opportunity for extensive microbiologic sampling of the allograft. In addition, diagnostic bronchoscopies performed based upon clinical need (respiratory symptoms, radiographic abnormalities) are useful for the diagnosis of fungal infection.

The diagnosis of tracheobronchial aspergillosis is made by visualization of erythema, pseudomembranes, or ulcerations on bronchoscopy with confirmation by histopathology and/or fungal culture (picture 1) [32]. Bronchoscopy may also identify airway dehiscence, which can be a complication of tracheobronchial aspergillosis. Chest computed tomography (CT) can help define the extent of lung involvement.  

Although definitive diagnosis of invasive pulmonary aspergillosis requires a biopsy demonstrating tissue invasion, evidence of a mold in the airway is suggestive of invasive infection in the right clinical context, such as when there are chest CT findings that are consistent with invasive infection. In lung transplant recipients, BAL and/or transbronchial lung biopsy is helpful in proving the diagnosis but cultures and/or fungal stains are positive in only 50 to 70 percent of cases [33]. CT scan findings range from consolidation (40 percent) to cavitary lesions (30 percent) to nodular or mass-like lesions (30 percent) (picture 2) [8]. Detection of fungal antigens such as galactomannan has been used to increase the sensitivity of detecting Aspergillus from BAL fluid. However, a positive BAL galactomannan is not specific for invasive disease because poor airway clearance can be associated with colonization alone. (See 'Galactomannan, beta-D-glucan, and PCR' below.)

CT or magnetic resonance imaging of the CNS is advised in lung transplant recipients with invasive pulmonary aspergillosis to evaluate for dissemination among patients where suspicion for CNS disease exists based on a careful neurologic exam or in patients where a neurologic exam may be unreliable. Tracheobronchitis, however, would not warrant CNS imaging unless CNS symptoms or signs were present.

Galactomannan, beta-D-glucan, and PCR — Data are limited regarding the utility of the galactomannan antigenemia assay, the beta-D-glucan assay, or the polymerase chain reaction (PCR) for the diagnosis of invasive aspergillosis in lung transplant recipients. One study evaluated the serum galactomannan assay in 70 lung transplant recipients through prospective monitoring with biweekly monitoring. Using an index cut-off value of ≥0.66, the assay had a sensitivity of 30 percent and a specificity of 95 percent, with false positives typically occurring in the first week post-transplant [34]. None of the four patients with tracheobronchial aspergillosis had a positive serum galactomannan assay.

Another prospective study evaluated the galactomannan assay in 333 bronchoalveolar lavage samples from 116 lung transplant recipients, two of whom had proven invasive aspergillosis, and four of whom had probable invasive aspergillosis [35]. Using an index cut-off value of ≥1, the assay had a sensitivity of 60 percent and a specificity of 98 percent [35].

A retrospective study compared the performance of an Aspergillus real-time PCR assay with the galactomannan assay in 150 BAL specimens from lung transplant recipients who underwent bronchoscopy for surveillance or diagnostic evaluation [36]. Of these, 16 patients had proven or probable invasive pulmonary aspergillosis, 26 had Aspergillus colonization, 11 had non-Aspergillus mold colonization, and 97 were negative controls. The sensitivity and specificity of a pan-Aspergillus spp PCR for diagnosing invasive pulmonary aspergillosis were 100 and 88 percent, respectively. In comparison, the sensitivity and specificity of an A. fumigatus-specific PCR were 85 and 96 percent, respectively, and the sensitivity and specificity of the galactomannan assay (using a cut-off value ≥0.5) were 93 and 89 percent, respectively.

Beta-D-glucan is a cell wall component of many fungi. A serum assay for beta-D-glucan is available and can be used to screen for invasive fungal infections, including those caused by Aspergillus spp, Candida spp, Pneumocystis, and other fungi. However, it has not been adequately studied in lung transplant recipients. While we do not routinely use or recommend using beta-D-glucan testing to diagnose aspergillosis, an elevated level corroborates the diagnosis.

These assays are discussed in greater detail separately. (See "Diagnosis of invasive aspergillosis".)

Treatment — Treatment of invasive aspergillosis involves antifungal therapy, most often with voriconazole, although other antifungal agents can be used [37,38]. In addition, immunosuppression should be reduced whenever possible. (See "Treatment and prevention of invasive aspergillosis".)

Tracheobronchial aspergillosis — Treatment of tracheobronchial aspergillosis involves systemic therapy with voriconazole in combination with nebulized amphotericin B; in addition, debridement of the bronchial anastomosis is often necessary when a substantial amount of necrotic tissue is present [29,37]. The use of voriconazole is discussed in the following section. (See 'Pulmonary and disseminated aspergillosis' below.)

Bronchoscopic debridement is an important component of therapy for tracheobronchial aspergillosis when a substantial amount of necrotic debris is present at the bronchial anastomosis, particularly when there is risk of airway obstruction. When there are ulcerations or bronchitis in the absence of devitalized tissue, there is no role for debridement.

Nebulized amphotericin B products may help treat the devascularized anastomotic site when this is the site of infection, although its therapeutic efficacy has not been proven. Anecdotal reports have described the use of combined systemic and inhaled antifungal agents in the treatment of anastomotic infection resulting in resolution of disease [39]. Occasionally, stenting is required to maintain airway patency.

Duration of therapy for tracheobronchial aspergillosis has not been adequately studied and is based on the extent and invasiveness of the infection, the host immune status, and the response to therapy. In uncomplicated cases, at least three months of voriconazole and nebulized amphotericin B or amphotericin B lipid complex are typically given, with longer durations reserved for nonresolving disease [37]. Complicated cases necessitate longer therapy, with consideration of lifelong suppressive therapy, particularly when the bronchial anastomosis is involved. Patients should continue antifungal therapy until tracheobronchial aspergillosis has completely resolved [37].

Pulmonary and disseminated aspergillosis — Voriconazole is the treatment of choice for invasive aspergillosis and is recommended for this indication in the 2016 Infectious Diseases Society of America (IDSA) guidelines on the treatment of aspergillosis [37]. We typically use combination therapy with voriconazole plus an echinocandin in lung transplant recipients with severe invasive aspergillosis. Similarly, the IDSA guidelines recommend consideration of such combination therapy in the setting of severe disease. Data supporting this practice are limited; combination therapy has been best studied in those with hematologic malignancy or profound and persistent neutropenia (not specifically in lung transplant recipients) [37]. Some experts prefer monotherapy with voriconazole. (See "Treatment and prevention of invasive aspergillosis", section on 'Dosing and drug effects'.)

An important consideration when giving voriconazole (or other azoles) to solid organ transplant recipients is its significant interactions with calcineurin inhibitors (eg, tacrolimus, cyclosporine) and mTOR inhibitors (eg, sirolimus, everolimus). Azoles increase levels of these immunosuppressants. Thus, dose reduction of these immunosuppressants and careful monitoring of serum concentrations of both azoles and immunosuppressants are needed when azoles are used. Because interaction between voriconazole and sirolimus can be extreme, this combination is usually avoided. We also suggest checking a voriconazole trough concentration during the first week of therapy (eg, between days 4 and 7) in all patients receiving treatment for invasive aspergillosis; the goal concentration is >1 mcg/mL and <5.5 mcg/mL. (See "Pharmacology of azoles", section on 'Drug interactions' and "Pharmacology of azoles", section on 'Voriconazole'.)

The treatment of invasive aspergillosis is discussed in greater detail separately. (See "Treatment and prevention of invasive aspergillosis".)

Outcomes — Historically, there was a high mortality with invasive aspergillosis (over 50 percent) in lung transplant recipients, but this varies depending upon the site of infection and has likely been reduced with the availability of newer antifungal azoles [8,28]. The mortality is lower in those with tracheobronchitis compared with invasive pulmonary infections [14].

OTHER FUNGAL INFECTIONS — In addition to Aspergillus spp, other important causes of invasive fungal infection in lung transplant recipients include Candida spp, Cryptococcus spp, endemic fungi, and the agents of mucormycosis; rarer causes are Fusarium spp, Scedosporium spp, and dematiaceous molds (table 1) [3,4,40].

Candidiasis — Candidiasis usually manifests as candidemia during the first month following transplantation as a result of intensive care unit exposure and the recent transplant surgery. In addition to candidemia, other manifestations include invasive disease, pleural space infections, incision site infections, and local anastomotic site infections [39,41]. In a survey of bloodstream infections following lung transplantation, Candida was the second most common isolate [41]. Despite frequent oropharyngeal colonization and isolation from sputum or bronchoalveolar lavage specimens, invasive pulmonary disease caused by Candida spp is exceedingly rare. (See "Clinical manifestations and diagnosis of candidemia and invasive candidiasis in adults" and "Candida infections of the abdomen and thorax".)

Cryptococcosis — Both pulmonary, central nervous system, and disseminated cryptococcal infection may occur in lung transplant recipients (image 1). The 12-month cumulative incidence of cryptococcosis following solid organ transplantation is estimated at 0.2 percent [2]. The risk of cryptococcosis is estimated to be highest in lung transplantation recipients (0.66 percent) compared with liver, heart, and kidney transplantation (0.44, 0.42, and 0.32 percent, respectively; hazard ratio 2.10, 95% CI 1.21-3.60) [42]. The median time to onset of disease after lung transplantation was approximately 190 days (range 7.5 to 1816).

Immune reconstitution inflammatory syndrome (IRIS) following initiation of treatment of cryptococcal disease in solid organ transplant recipients has been reported and should be considered as a potential complication of cryptococcosis [43]. Reduction of immunosuppression may have contributed to the development of IRIS in some cases. Cryptococcal cellulitis has been reported in lung transplant recipients [44]. (See "Cryptococcus neoformans infection outside the central nervous system" and "Epidemiology of pulmonary infections in immunocompromised patients" and "Cryptococcus neoformans: Treatment of meningoencephalitis and disseminated infection in patients without HIV", section on 'Immune reconstitution inflammatory syndrome'.)

Mucormycosis — Mucormycosis is another cause of invasive fungal infections in immunocompromised hosts. Presentation in lung transplant recipients typically consists of pulmonary disease, although bronchial anastomosis infection and gastrointestinal disease have also been reported (image 2) [3,45,46].

Mucormycosis is characterized by infarction and necrosis of host tissues that results from invasion of the vasculature by hyphae. The progression is usually fast and the outcome in patients with pulmonary mucormycosis is poor, with mortality rates as high as 87 percent. Treatment of mucormycosis involves a combination of surgical debridement of involved tissues and antifungal therapy. (See "Mucormycosis (zygomycosis)", section on 'Treatment'.)

Endemic fungi — Endemic fungi that cause infection following solid organ transplantation include Histoplasma capsulatum, Coccidioides immitis, and, rarely, Blastomyces dermatitidis [47]. Infections with endemic fungi may occur as a result of reactivation of dormant infection, new acquisition from the environment, or transmission from donors from endemic areas. (See "Infection in the solid organ transplant recipient".)

When endemic fungal infection occurs in solid organ transplant recipients, severe pneumonia and disseminated disease are more likely and the mortality is higher compared with immunocompetent hosts [48]. (See "Pathogenesis and clinical features of pulmonary histoplasmosis" and "Clinical manifestations and diagnosis of blastomycosis" and "Management considerations, screening, and prevention of coccidioidomycosis in immunocompromised individuals and pregnant patients" and "Primary pulmonary coccidioidal infection".)

Pneumocystis jirovecii — Approximately 5 to 15 percent of patients who undergo solid organ transplantation develop Pneumocystis jirovecii (formerly P. carinii) pneumonia (PCP) in the absence of prophylaxis [49-52]. The rates are highest among lung and heart-lung transplant recipients [49,50,53,54].

The period of highest risk for PCP following solid organ transplantation is from one to six months following transplantation if prophylaxis is not given. The incidence of PCP has decreased dramatically due to nearly universal prophylaxis, which is typically given indefinitely. (See "Epidemiology, clinical manifestations, and diagnosis of Pneumocystis pneumonia in patients without HIV".)

PROPHYLAXIS

Invasive fungal infections

Approach to prophylaxis — We provide antifungal prophylaxis for most patients in the early post-transplant period because the risk of fungal infection during this period and its associated morbidity is high [55]. However, providing antifungal prophylaxis to all patients is not universal practice [56-60]. The American Society of Transplantation recommends either a universal prophylaxis or pre-emptive monitoring strategy for invasive aspergillosis in lung transplant recipients. Although data to guide the best approach are limited [38,61], most centers in the United States use universal prophylaxis [62].

Our approach to prophylaxis is as follows:

We use inhaled liposomal amphotericin B for all patients because the potential benefit of decreased fungal infection appears to outweigh the risks. Typically, we use inhaled amphotericin B lipid complex in extubated patients, dosed at 100 mg nebulized daily for four days post-transplant and then 50 mg nebulized weekly until hospital discharge [63,64].

We add an azole for most patients to prevent infection with Candida spp, particularly surgical site infections (eg, empyema); use of inhaled amphotericin alone is rare.

Azole selection varies based on risk factors for mold infection (eg, a history of mold infection or current of past colonization with mold). (See 'Risk factors for mold infection' below and 'Systemic azoles' below.)

For patients without risk factors for mold infection, we generally use fluconazole for three months following transplantation. However, if the chest is left open, we use an echinocandin (micafungin, caspofungin, anidulafungin) or fluconazole until the chest is closed.

For patients with risk factors for mold infection, we use a broader spectrum azole (eg, voriconazole, posaconazole, isavuconazole). While voriconazole is most often used, selection varies based on patient comorbidities.

For patients with a history of mold colonization or infection, we begin prophylaxis when the patient is listed for transplant and continue for at least three months post-transplant. Longer or shorter durations might be appropriate, depending on the clinical context and burden of disease.

We reinitiate antifungal prophylaxis for patients receiving immunosuppression augmentation with thymoglobulin, alemtuzumab, or high-dose glucocorticoids [37].

In addition to nebulized or systemic antifungal prophylaxis, nystatin suspension (5 cc swish and swallow four times daily for six months following transplantation) is often used to reduce gastrointestinal colonization with Candida spp. We use nystatin in patients who are not receiving a systemic antifungal agent.

The approach to universal prophylaxis varies among centers and not all use inhaled amphotericin in addition to systemic antifungal agents. Transplant centers that use a pre-emptive approach typically perform fungal culture and galactomannan testing on bronchoalveolar lavage fluid obtained on routine bronchoscopy for the first three to four months following transplantation.

Risk factors for mold infection — The most important risk factors for mold infection in the early post-transplant period are:

A history of mold infection or colonization

Detection of mold in the airway at the time of transplantation

Detection of mold in the airway on post-transplant surveillance bronchoscopy

Several other patient groups may be at higher risk for fungal infections after lung transplantation [61]. These include recipients of a single-lung transplant, patients with early airway ischemia, patients whose sinuses are colonized with mold, patients with concurrent cytomegalovirus infection, patients with rejection requiring increased immunosuppression (eg, antithymocyte globulin), patients with delayed sternal closure, patients requiring renal replacement therapy, patients on extracorporeal membrane oxygenation, and patients with acquired hypogammaglobulinemia (immunoglobulin [Ig]G <400 mg/dL.) In such individuals, systemic antifungal prophylaxis is likely warranted. Further study is necessary to better define the benefits of antifungal prophylaxis in these patient groups.

Antifungal agents and their efficacy

Inhaled amphotericin B — We prefer inhaled lipid complex amphotericin B (eg, liposomal amphotericin B, amphotericin B lipid complex) to inhaled amphotericin B deoxycholate since the former agent appears to be associated with a lower rate of adverse effects and also has a longer half-life and improved lung deposition. Because inhaled therapy has minimal side effects and coverage against a broad range of fungal pathogens, we use this agent in nearly all patients in the early post-transplant period.

Efficacy data are limited in observational studies, which show reduced rates of invasive fungal infection with nebulized amphotericin use [19,63,65-67]. However, reduced infection rates were not observed in all studies [68] and there are disadvantages to utilizing inhaled therapy alone. At least one center that practices this approach reported a relatively high incidence of fungal pleural space infections in the early post-transplant period, suggesting that a disadvantage of inhaled therapy is the failure to prevent systemic fungal infections, including pleural space infections [12,57].

Systemic azoles — Fluconazole is active against many Candida spp but has limited to no activity against molds. Thus, we use fluconazole for patients without risk factors for mold.

For patients who have respiratory tract colonization with mold (especially Aspergillus spp), we use an oral azole that has activity against molds, such as voriconazole, isavuconazole, or posaconazole.

Voriconazole is given as a loading dose of 6 mg/kg intravenously (IV; or 400 mg orally) every 12 hours for two doses, then 4 mg/kg IV (or 200 mg orally) every 12 hours.

Isavuconazole is given as a loading dose of 372 mg by mouth (isavuconazonium sulfate 372 mg IV) every eight hours for six doses, then 372 mg once daily.

Posaconazole is given as a loading dose of 300 mg (by mouth/IV) twice daily for two doses, then 300 mg once daily. When administering posaconazole orally, we use the delayed-release formulation because it is better absorbed than other formulations. However, the extended-release form cannot be given to intubated patients; while the liquid form can be given via an endotracheal tube, absorption is erratic. In these cases, at our center, we either use the IV formulation of posaconazole or switch to another azole.

When azoles are used, the clinician must be aware of their significant interactions with calcineurin inhibitors (tacrolimus, cyclosporine) and mTor inhibitors (eg, everolimus, sirolimus) and reduce the dose of the immunosuppressant agent accordingly. We also perform therapeutic drug monitoring to ensure adequate absorption.

A small retrospective study evaluated the use of antifungal prophylaxis in lung transplant recipients [69]. The 65 patients who received universal voriconazole prophylaxis had significantly lower rates of invasive aspergillosis at one year post-transplant than the 30 patients who received targeted itraconazole prophylaxis (with or without inhaled amphotericin) for pre- or post-transplant Aspergillus colonization except A. niger (1.5 versus 23 percent). One downside of long-term voriconazole prophylaxis is that can increase the risk of skin cancer in lung transplant recipients [70].

Posaconazole and isavuconazole are two additional azoles with activity against Aspergillus spp and other molds. In a cohort study evaluating 300 lung transplant recipients, isavuconazole appeared to be as effective as voriconazole for prevention of invasive fungal infections but was better tolerated. The breakthrough infection rate was 8 percent for both azoles; 11 percent of patients receiving isavuconazole stopped prophylaxis prematurely due to adverse effects compared with 35 percent of patients receiving voriconazole [71].

At our center, we tend to use isavuconazole in lung transplant recipient who are risk for QTc prolongation and recipients with renal impairment. Most clinical trials of isavuconazole have not shown QTc prolongation unlike other azoles [72,73]. Compared with IV voriconazole and posaconazole, isavuconazole’s prodrug, isavuconazonium sulfate, does not contain cyclodextrin, which can accumulate with renal impairment and cause toxicity [74]. A potential downside is that isavuconazole may be less effective against Candida spp when compared with other azoles [75].

Posaconazole has not been directly compared with voriconazole or isavuconazole in lung transplant recipients, but observational data suggest it is also effective and well tolerated [71,76].

Pneumocystis pneumonia — Because the risk of P. jirovecii (formerly P. carinii) pneumonia (PCP) in the allografted lung is high, indefinite prophylaxis is recommended for all recipients.

A retrospective study evaluated 25 cases of PCP among 1299 solid organ transplant recipients at a single center between 1987 and 1996 [77]. The incidence of PCP was highest among lung transplant recipients compared with other organ recipients (22 versus 4.8 cases per 1000 person-transplant years) and did not decline after the first year following transplantation, in contrast with recipients of other organs. No patients developed PCP while receiving prophylaxis but the duration of prophylaxis was shorter for the recipients of certain organs than is standard (eg, lung transplant recipients received prophylaxis for one year; liver transplant recipients did not routinely receive prophylaxis until five years into the study). A subsequent multicenter study evaluating PCP episodes between 2010 and 2017 found that the annual incidence was 2.7 cases per 1000 person-transplant years [78]. Sixty-five percent of the patients were not on PCP prophylaxis when they developed PCP infection.

The incidence of PCP among solid organ transplant recipients has decreased dramatically due to nearly universal prophylaxis. In a meta-analysis of 12 randomized controlled trials that included 1245 immunocompromised patients without HIV infection who had undergone autologous hematopoietic stem cell or solid organ transplantation or had a hematologic malignancy, trimethoprim-sulfamethoxazole (TMP-SMX) prophylaxis was associated with a 91 percent reduction in the occurrence of PCP (relative risk [RR] 0.09, 95% CI 0.02-0.32) [79]. Mortality due to PCP was also significantly reduced (RR 0.17, 95% CI 0.03-0.94), although all-cause mortality was not. No differences in efficacy were found between once-daily and thrice-weekly administration schedules for TMP-SMX. (See "Treatment and prevention of Pneumocystis pneumonia in patients without HIV", section on 'Prophylaxis'.)

Although PCP prophylaxis is highly effective, some cases have still been reported in the setting of prophylaxis. For example, in a series of 609 bronchoscopies performed (surveillance and clinically indicated) in lung and heart-lung transplant recipients at a single center from 1994 to 2002, 23 samples were positive for P. jirovecii, 15 of which were from patients receiving prophylaxis [80]. Seven of these 15 patients were taking inhaled pentamidine, which is less effective than TMP-SMX. Furthermore, two samples were positive in samples obtained from surveillance bronchoscopies, which could have represented colonization rather than infection.

TMP-SMX is recommended for all patients who can tolerate it, given its excellent efficacy and its broad spectrum against a range of organisms in addition to P. jirovecii (eg, Toxoplasma gondii, Listeria spp, Nocardia spp, Streptococcus pneumoniae) [81]. The usual dosing of TMP-SMX for lung transplant recipients is one double-strength tablet daily or three times per week, continued indefinitely. Daily dosing is needed for patients at increased risk of toxoplasmosis (donor seropositive/recipient seronegative). For those who are allergic to TMP-SMX, desensitization should be attempted when possible. Alternative regimens are discussed separately. (See "Treatment and prevention of Pneumocystis pneumonia in patients without HIV" and "Prophylaxis of infections in solid organ transplantation".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Infections in solid organ transplant recipients".)

SUMMARY AND RECOMMENDATIONS

Important pathogensAspergillus and Candida spp cause the majority of fungal infections in lung transplant recipients. Cryptococcus spp, the agents of mucormycosis, Scedosporium spp, and endemic fungi (Histoplasma, Blastomyces, and Coccidioides spp) are other important causes (table 1). (See 'Introduction' above.)

Risk of infection − Lung transplant recipients are at high risk for infectious complications due to the high level of immunosuppression required to prevent rejection, the adverse effects of transplantation on local pulmonary host defenses (eg, reduced mucociliary clearance), and constant environmental contact allowing pathogens direct access into the allograft. (See 'Risk of infection' above.)

Clinical features and diagnosis of aspergillosis

Manifestations of invasive aspergillosis − The most common acute manifestations of invasive aspergillosis in lung transplant recipients are tracheobronchitis, pneumonia, and disseminated disease. (See 'Clinical manifestations' above.)

Diagnosis of tracheobronchial aspergillosis − The diagnosis of tracheobronchial aspergillosis is made by visualization of erythema, pseudomembranes, or ulcerations on bronchoscopy with confirmation by histopathology and/or fungal culture. (See 'Diagnosis' above.)

Diagnosis of pulmonary aspergillosis − Although definitive diagnosis of invasive pulmonary aspergillosis requires a biopsy demonstrating tissue invasion, this is not always necessary or practical. More commonly, the diagnosis of pulmonary aspergillosis is made presumptively by chest computed tomography in combination with bronchoscopy with fungal stains and cultures performed on bronchoalveolar lavage fluid. (See 'Diagnosis' above.)

Treatment of aspergillosis

Tracheobronchial aspergillosis − For patients with tracheobronchial aspergillosis, we recommend systemic therapy with voriconazole (Grade 1B). In addition, we suggest nebulized amphotericin B (Grade 2C). Bronchoscopic debridement is an important component of therapy in patients with a substantial amount of necrotic debris at the bronchial anastomosis, particularly when there is risk of airway obstruction. (See 'Tracheobronchial aspergillosis' above.)

Invasive aspergillosisVoriconazole is the treatment of choice for invasive aspergillosis. We typically use combination therapy with voriconazole plus an echinocandin in lung transplant recipients with severe invasive aspergillosis. (See 'Treatment' above and "Treatment and prevention of invasive aspergillosis".)

Important drug interactions − When voriconazole (or another azole) is used, the clinician must be aware of its significant interactions with calcineurin inhibitors (eg, tacrolimus, cyclosporine) and mTor inhibitors (eg, sirolimus, everolimus) and reduce the dose of the immunosuppressant agent or change therapy accordingly. (See "Pharmacology of azoles", section on 'Drug interactions' and "Pharmacology of azoles", section on 'Voriconazole'.)

Antifungal prophylaxis in the early post-transplant period

Our approach − We provide antifungal prophylaxis for most patients in the early post-transplant period because the risk of fungal infection during this period and its associated morbidity is high. However, providing antifungal prophylaxis to all patients is not universal practice. The American Society of Transplantation recommends either a universal prophylaxis or pre-emptive monitoring strategy for invasive aspergillosis in lung transplant recipients; data to guide the best approach is limited. (See 'Invasive fungal infections' above and 'Approach to prophylaxis' above.)

Antifungal selection − We use inhaled liposomal amphotericin B for all patients because the potential benefit of decreased fungal infection appears to outweigh the risks. We add an additional systemic agent depending on the patient’s operative risk factors (eg, whether the chest was open) and risk factors for specific mold infections. (See 'Approach to prophylaxis' above and 'Risk factors for mold infection' above.)

Pneumocystis prophylaxis − We recommend Pneumocystis jirovecii (formerly P. carinii) pneumonia prophylaxis for all lung transplant recipients (Grade 1A). The duration of prophylaxis is typically lifelong. We recommend trimethoprim-sulfamethoxazole (Grade 1A). The usual dosing is one double-strength tablet daily or one double-strength tablet three times per week, continued indefinitely. (See 'Pneumocystis pneumonia' above.)

ACKNOWLEDGMENTS — The UpToDate editorial staff acknowledges David Zaas, MD, Kieren A Marr, MD, and Cameron Wolfe, MBBS (Hons), MPH, who contributed to earlier versions of this topic review.

  1. Yusen RD, Edwards LB, Dipchand AI, et al. The Registry of the International Society for Heart and Lung Transplantation: Thirty-third Adult Lung and Heart-Lung Transplant Report-2016; Focus Theme: Primary Diagnostic Indications for Transplant. J Heart Lung Transplant 2016; 35:1170.
  2. Pappas PG, Alexander BD, Andes DR, et al. Invasive fungal infections among organ transplant recipients: results of the Transplant-Associated Infection Surveillance Network (TRANSNET). Clin Infect Dis 2010; 50:1101.
  3. Silveira FP, Husain S. Fungal infections in lung transplant recipients. Curr Opin Pulm Med 2008; 14:211.
  4. Shoham S. Emerging fungal infections in solid organ transplant recipients. Infect Dis Clin North Am 2013; 27:305.
  5. Husain S, Singh N. Bronchiolitis obliterans and lung transplantation: evidence for an infectious etiology. Semin Respir Infect 2002; 17:310.
  6. Weigt SS, Elashoff RM, Huang C, et al. Aspergillus colonization of the lung allograft is a risk factor for bronchiolitis obliterans syndrome. Am J Transplant 2009; 9:1903.
  7. Weigt SS, Copeland CA, Derhovanessian A, et al. Colonization with small conidia Aspergillus species is associated with bronchiolitis obliterans syndrome: a two-center validation study. Am J Transplant 2013; 13:919.
  8. Singh N, Husain S. Aspergillus infections after lung transplantation: clinical differences in type of transplant and implications for management. J Heart Lung Transplant 2003; 22:258.
  9. Peghin M, Monforte V, Martin-Gomez MT, et al. Epidemiology of invasive respiratory disease caused by emerging non-Aspergillus molds in lung transplant recipients. Transpl Infect Dis 2016; 18:70.
  10. Vazquez R, Vazquez-Guillamet MC, Suarez J, et al. Invasive mold infections in lung and heart-lung transplant recipients: Stanford University experience. Transpl Infect Dis 2015; 17:259.
  11. Bonnal C, Leleu C, Brugière O, et al. Relationship between Fungal Colonisation of the Respiratory Tract in Lung Transplant Recipients and Fungal Contamination of the Hospital Environment. PLoS One 2015; 10:e0144044.
  12. Wahidi MM, Willner DA, Snyder LD, et al. Diagnosis and outcome of early pleural space infection following lung transplantation. Chest 2009; 135:484.
  13. Minari A, Husni R, Avery RK, et al. The incidence of invasive aspergillosis among solid organ transplant recipients and implications for prophylaxis in lung transplants. Transpl Infect Dis 2002; 4:195.
  14. Singh N, Paterson DL. Aspergillus infections in transplant recipients. Clin Microbiol Rev 2005; 18:44.
  15. Herrera S, Davoudi S, Farooq A, et al. Late Onset Invasive Pulmonary Aspergillosis in Lung Transplant Recipients in the Setting of a Targeted Prophylaxis/Preemptive Antifungal Therapy Strategy. Transplantation 2020; 104:2575.
  16. Gordon SM, Avery RK. Aspergillosis in lung transplantation: incidence, risk factors, and prophylactic strategies. Transpl Infect Dis 2001; 3:161.
  17. Sutton DA, Sanche SE, Revankar SG, et al. In vitro amphotericin B resistance in clinical isolates of Aspergillus terreus, with a head-to-head comparison to voriconazole. J Clin Microbiol 1999; 37:2343.
  18. Baddley JW, Pappas PG, Smith AC, Moser SA. Epidemiology of Aspergillus terreus at a university hospital. J Clin Microbiol 2003; 41:5525.
  19. Monforte V, Roman A, Gavalda J, et al. Nebulized amphotericin B prophylaxis for Aspergillus infection in lung transplantation: study of risk factors. J Heart Lung Transplant 2001; 20:1274.
  20. Cahill BC, Hibbs JR, Savik K, et al. Aspergillus airway colonization and invasive disease after lung transplantation. Chest 1997; 112:1160.
  21. Helmi M, Love RB, Welter D, et al. Aspergillus infection in lung transplant recipients with cystic fibrosis: risk factors and outcomes comparison to other types of transplant recipients. Chest 2003; 123:800.
  22. Avery RK. Prophylactic strategies before solid-organ transplantation. Curr Opin Infect Dis 2004; 17:353.
  23. Nunley DR, Ohori P, Grgurich WF, et al. Pulmonary aspergillosis in cystic fibrosis lung transplant recipients. Chest 1998; 114:1321.
  24. Aguilar CA, Hamandi B, Fegbeutel C, et al. Clinical risk factors for invasive aspergillosis in lung transplant recipients: Results of an international cohort study. J Heart Lung Transplant 2018; 37:1226.
  25. Patterson TF, Thompson GR 3rd, Denning DW, et al. Executive Summary: Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis 2016; 63:433.
  26. Kramer MR, Denning DW, Marshall SE, et al. Ulcerative tracheobronchitis after lung transplantation. A new form of invasive aspergillosis. Am Rev Respir Dis 1991; 144:552.
  27. Fernández-Ruiz M, Silva JT, San-Juan R, et al. Aspergillus tracheobronchitis: report of 8 cases and review of the literature. Medicine (Baltimore) 2012; 91:261.
  28. Mehrad B, Paciocco G, Martinez FJ, et al. Spectrum of Aspergillus infection in lung transplant recipients: case series and review of the literature. Chest 2001; 119:169.
  29. Hadjiliadis D, Howell DN, Davis RD, et al. Anastomotic infections in lung transplant recipients. Ann Transplant 2000; 5:13.
  30. Kennedy CC, Razonable RR. Fungal Infections After Lung Transplantation. Clin Chest Med 2017; 38:511.
  31. Morgan J, Wannemuehler KA, Marr KA, et al. Incidence of invasive aspergillosis following hematopoietic stem cell and solid organ transplantation: interim results of a prospective multicenter surveillance program. Med Mycol 2005; 43 Suppl 1:S49.
  32. Paterson DL, Singh N. Invasive aspergillosis in transplant recipients. Medicine (Baltimore) 1999; 78:123.
  33. Geltner C, Lass-Flörl C. Invasive pulmonary Aspergillosis in organ transplants--Focus on lung transplants. Respir Investig 2016; 54:76.
  34. Husain S, Kwak EJ, Obman A, et al. Prospective assessment of Platelia Aspergillus galactomannan antigen for the diagnosis of invasive aspergillosis in lung transplant recipients. Am J Transplant 2004; 4:796.
  35. Husain S, Paterson DL, Studer SM, et al. Aspergillus galactomannan antigen in the bronchoalveolar lavage fluid for the diagnosis of invasive aspergillosis in lung transplant recipients. Transplantation 2007; 83:1330.
  36. Luong ML, Clancy CJ, Vadnerkar A, et al. Comparison of an Aspergillus real-time polymerase chain reaction assay with galactomannan testing of bronchoalvelolar lavage fluid for the diagnosis of invasive pulmonary aspergillosis in lung transplant recipients. Clin Infect Dis 2011; 52:1218.
  37. Patterson TF, Thompson GR 3rd, Denning DW, et al. Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis 2016; 63:e1.
  38. Husain S, Sole A, Alexander BD, et al. The 2015 International Society for Heart and Lung Transplantation Guidelines for the management of fungal infections in mechanical circulatory support and cardiothoracic organ transplant recipients: Executive summary. J Heart Lung Transplant 2016; 35:261.
  39. Palmer SM, Perfect JR, Howell DN, et al. Candidal anastomotic infection in lung transplant recipients: successful treatment with a combination of systemic and inhaled antifungal agents. J Heart Lung Transplant 1998; 17:1029.
  40. Abela IA, Murer C, Schuurmans MM, et al. A cluster of scedosporiosis in lung transplant candidates and recipients: The Zurich experience and review of the literature. Transpl Infect Dis 2018; 20.
  41. Palmer SM, Alexander BD, Sanders LL, et al. Significance of blood stream infection after lung transplantation: analysis in 176 consecutive patients. Transplantation 2000; 69:2360.
  42. George IA, Santos CAQ, Olsen MA, Powderly WG. Epidemiology of Cryptococcosis and Cryptococcal Meningitis in a Large Retrospective Cohort of Patients After Solid Organ Transplantation. Open Forum Infect Dis 2017; 4:ofx004.
  43. Singh N, Lortholary O, Alexander BD, et al. An immune reconstitution syndrome-like illness associated with Cryptococcus neoformans infection in organ transplant recipients. Clin Infect Dis 2005; 40:1756.
  44. Rakvit A, Meyerrose G, Vidal AM, et al. Cellulitis caused by Cryptococcus neoformans in a lung transplant recipient. J Heart Lung Transplant 2005; 24:642.
  45. McGuire FR, Grinnan DC, Robbins M. Mucormycosis of the bronchial anastomosis: a case of successful medical treatment and historic review. J Heart Lung Transplant 2007; 26:857.
  46. Manchikalapati P, Canon CL, Jhala N, Eloubeidi MA. Gastrointestinal zygomycosis complicating heart and lung transplantation in a patient with Eisenmenger's syndrome. Dig Dis Sci 2005; 50:1181.
  47. Miller R, Assi M, AST Infectious Diseases Community of Practice. Endemic fungal infections in solid organ transplant recipients-Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019; 33:e13553.
  48. Kauffman CA, Freifeld AG, Andes DR, et al. Endemic fungal infections in solid organ and hematopoietic cell transplant recipients enrolled in the Transplant-Associated Infection Surveillance Network (TRANSNET). Transpl Infect Dis 2014; 16:213.
  49. Sepkowitz KA. Opportunistic infections in patients with and patients without Acquired Immunodeficiency Syndrome. Clin Infect Dis 2002; 34:1098.
  50. Sepkowitz KA, Brown AE, Armstrong D. Pneumocystis carinii pneumonia without acquired immunodeficiency syndrome. More patients, same risk. Arch Intern Med 1995; 155:1125.
  51. Gerrard JG. Pneumocystis carinii pneumonia in HIV-negative immunocompromised adults. Med J Aust 1995; 162:233.
  52. Radisic M, Lattes R, Chapman JF, et al. Risk factors for Pneumocystis carinii pneumonia in kidney transplant recipients: a case-control study. Transpl Infect Dis 2003; 5:84.
  53. Dummer JS, Montero CG, Griffith BP, et al. Infections in heart-lung transplant recipients. Transplantation 1986; 41:725.
  54. Gryzan S, Paradis IL, Zeevi A, et al. 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.
  55. Neoh CF, Snell GI, Kotsimbos T, et al. Antifungal prophylaxis in lung transplantation--a world-wide survey. Am J Transplant 2011; 11:361.
  56. Schaenman JM. Is universal antifungal prophylaxis mandatory in lung transplant patients? Curr Opin Infect Dis 2013; 26:317.
  57. Baker AW, Maziarz EK, Arnold CJ, et al. Invasive Fungal Infection After Lung Transplantation: Epidemiology in the Setting of Antifungal Prophylaxis. Clin Infect Dis 2020; 70:30.
  58. Pilarczyk K, Haake N, Heckmann J, et al. Is universal antifungal prophylaxis mandatory in adults after lung transplantation? A review and meta-analysis of observational studies. Clin Transplant 2016; 30:1522.
  59. Linder KA, Kauffman CA, Patel TS, et al. Evaluation of targeted versus universal prophylaxis for the prevention of invasive fungal infections following lung transplantation. Transpl Infect Dis 2021; 23:e13448.
  60. Pennington KM, Baqir M, Erwin PJ, et al. Antifungal prophylaxis in lung transplant recipients: A systematic review and meta-analysis. Transpl Infect Dis 2020; 22:e13333.
  61. Husain S, Camargo JF. Invasive Aspergillosis in solid-organ transplant recipients: Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019; 33:e13544.
  62. Pennington KM, Yost KJ, Escalante P, et al. Antifungal prophylaxis in lung transplant: A survey of United States' transplant centers. Clin Transplant 2019; 33:e13630.
  63. Drew RH, Dodds Ashley E, Benjamin DK Jr, et al. Comparative safety of amphotericin B lipid complex and amphotericin B deoxycholate as aerosolized antifungal prophylaxis in lung-transplant recipients. Transplantation 2004; 77:232.
  64. Palmer SM, Drew RH, Whitehouse JD, et al. Safety of aerosolized amphotericin B lipid complex in lung transplant recipients. Transplantation 2001; 72:545.
  65. Peghin M, Monforte V, Martin-Gomez MT, et al. 10 years of prophylaxis with nebulized liposomal amphotericin B and the changing epidemiology of Aspergillus spp. infection in lung transplantation. Transpl Int 2016; 29:51.
  66. Reichenspurner H, Gamberg P, Nitschke M, et al. Significant reduction in the number of fungal infections after lung-, heart-lung, and heart transplantation using aerosolized amphotericin B prophylaxis. Transplant Proc 1997; 29:627.
  67. Borro JM, Solé A, de la Torre M, et al. Efficiency and safety of inhaled amphotericin B lipid complex (Abelcet) in the prophylaxis of invasive fungal infections following lung transplantation. Transplant Proc 2008; 40:3090.
  68. Monforte V, Ussetti P, Gavaldà J, et al. Feasibility, tolerability, and outcomes of nebulized liposomal amphotericin B for Aspergillus infection prevention in lung transplantation. J Heart Lung Transplant 2010; 29:523.
  69. Husain S, Paterson DL, Studer S, et al. Voriconazole prophylaxis in lung transplant recipients. Am J Transplant 2006; 6:3008.
  70. Kolaitis NA, Duffy E, Zhang A, et al. Voriconazole increases the risk for cutaneous squamous cell carcinoma after lung transplantation. Transpl Int 2017; 30:41.
  71. Samanta P, Clancy CJ, Marini RV, et al. Isavuconazole Is as Effective as and Better Tolerated Than Voriconazole for Antifungal Prophylaxis in Lung Transplant Recipients. Clin Infect Dis 2021; 73:416.
  72. Schmitt-Hoffmann A, Roos B, Maares J, et al. Multiple-dose pharmacokinetics and safety of the new antifungal triazole BAL4815 after intravenous infusion and oral administration of its prodrug, BAL8557, in healthy volunteers. Antimicrob Agents Chemother 2006; 50:286.
  73. Wilson DT, Dimondi VP, Johnson SW, et al. Role of isavuconazole in the treatment of invasive fungal infections. Ther Clin Risk Manag 2016; 12:1197.
  74. von Mach MA, Burhenne J, Weilemann LS. Accumulation of the solvent vehicle sulphobutylether beta cyclodextrin sodium in critically ill patients treated with intravenous voriconazole under renal replacement therapy. BMC Clin Pharmacol 2006; 6:6.
  75. Zhou J, Wang Y, Xu G, et al. Structural Insight into Enantioselective Inversion of an Alcohol Dehydrogenase Reveals a "Polar Gate" in Stereorecognition of Diaryl Ketones. J Am Chem Soc 2018; 140:12645.
  76. Jeong W, Snell GI, Levvey BJ, et al. Clinical effectiveness of early posaconazole suspension pre-emptive therapy in lung transplant recipients: The Alfred's experience. J Antimicrob Chemother 2017; 72:2089.
  77. Gordon SM, LaRosa SP, Kalmadi S, et al. Should prophylaxis for Pneumocystis carinii pneumonia in solid organ transplant recipients ever be discontinued? Clin Infect Dis 1999; 28:240.
  78. Delbove A, Alami H, Tissot A, et al. Pneumocystis pneumonia after lung transplantation: A retrospective multicenter study. Respir Med 2020; 169:106019.
  79. Green H, Paul M, Vidal L, Leibovici L. Prophylaxis of Pneumocystis pneumonia in immunocompromised non-HIV-infected patients: systematic review and meta-analysis of randomized controlled trials. Mayo Clin Proc 2007; 82:1052.
  80. Lehto JT, Koskinen PK, Anttila VJ, et al. Bronchoscopy in the diagnosis and surveillance of respiratory infections in lung and heart-lung transplant recipients. Transpl Int 2005; 18:562.
  81. Fishman JA, Gans H, AST Infectious Diseases Community of Practice. Pneumocystis jiroveci in solid organ transplantation: Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019; 33:e13587.
Topic 1394 Version 39.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟