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

Nontuberculous mycobacterial pulmonary infections in children

Nontuberculous mycobacterial pulmonary infections in children
Author:
Andrea T Cruz, MD, MPH
Section Editor:
Sheldon L Kaplan, MD
Deputy Editor:
Diane Blake, MD
Literature review current through: Jan 2024.
This topic last updated: Dec 01, 2022.

INTRODUCTION — Nontuberculous mycobacteria (NTM) are a miscellaneous collection of acid-fast bacteria that are widespread in the environment [1]. They have been isolated from numerous environmental sources including water, soil, food products, and domestic and wild animals [2]. Health care-associated transmission has occurred with medical equipment [3-5].

This topic will provide an overview of NTM pulmonary infections in children. NTM lymphadenitis, skin and soft tissue infection, disseminated infection, and bacteremia in children are discussed separately.

(See "Nontuberculous mycobacterial lymphadenitis in children".)

(See "Nontuberculous mycobacterial skin and soft tissue infections in children".)

(See "Disseminated nontuberculous mycobacterial (NTM) infections and NTM bacteremia in children".)

MICROBIOLOGY — More than 170 species of NTM have been identified, not all of which have been documented to cause disease in humans [6-9].

Classification – NTM pathogens are classified as rapidly growing or slowly growing (table 1). Rapidly growing species grow within seven days and include Mycobacterium fortuitum, Mycobacterium abscessus, and Mycobacterium chelonae. Slowly growing species require several weeks to grow and include Mycobacterium avium complex (MAC), Mycobacterium marinum, and Mycobacterium kansasii. (See "Microbiology of nontuberculous mycobacteria", section on 'Classification'.)

Disease associations – In children, NTM cause four main clinical syndromes: lymphadenopathy, skin and soft tissue infection, pulmonary disease (predominantly in children with underlying pulmonary conditions), and disseminated disease (predominantly in immune-compromised children). The type of infection varies with the species of NTM and host characteristics.

There is little information about which species of NTM cause pulmonary disease in children. In one series of 17 children with NTM pulmonary disease, MAC was more common in previously healthy children (n = 5) and M. abscessus or M. chelonae in children with cystic fibrosis (n = 8) [10]. MAC (M. avium and M. intracellulare), M. abscessus, and M. kansasii are the most common NTM species that cause pulmonary disease in adults in the United States [11]. In countries with a high burden of tuberculosis, up to 40 percent of mycobacterial isolates are NTM species, most commonly MAC, M. abscessus, and M. fortuitum [12,13].

In a registry of over 16,000 pediatric and adult patients with cystic fibrosis, 20 percent had an NTM species isolated at least once during 2010 to 2014, with the prevalence increasing each year [14]. MAC (M. avium and Mycobacterium intracellulare) and M. abscessus are the species most frequently isolated from children with cystic fibrosis [15-20]. In a retrospective cohort study that was limited to children slow-growing NTM species (eg, MAC) were detected more often in patients who were younger at the time of cystic fibrosis diagnosis (median age 1.2 months, range 0.1 to 2.8 months), and rapidly growing NTM species (eg, M. abscessus) were detected more often in patients diagnosed with cystic fibrosis at an older age (median age 4.5 months, range 1.5 to 84.3 months) [21].

EPIDEMIOLOGY — Estimates of the true burden of NTM infections in children are unavailable, in part because NTM infections may be asymptomatic and because NTM infections are not communicable; reporting of NTM infections is not required in the United States or many other countries [22]. Cystic fibrosis registries provide some of the more robust pediatric data. The overall prevalence of NTM disease appears to be increasing with time (possibly as a result of enhanced detection) [14,23-25]. (See "Epidemiology of nontuberculous mycobacterial infections".)

In case series, the prevalence of NTM among respiratory specimens from patients with cystic fibrosis ranges from 4 to 20 percent [15,17,22,26]. However, isolation of NTM from a respiratory specimen does not necessarily indicate NTM disease. In a prospective study of 682 sputum specimens from 106 children with cystic fibrosis, NTM were isolated from 6.6 percent, but only 1.9 percent had NTM disease (ie, clinical and functional decline in addition to isolation) [26]. Among patients with cystic fibrosis, NTM isolation is more common in teenagers and young adults than in younger patients [15,16,18,26-28]. There is geographic variation in species detection. In one large cystic fibrosis registry, M. abscessus was most prevalent in Hawaii, Florida, and Louisiana, whereas MAC was most prevalent in Nevada, Kansas, Hawaii, and Arizona [14].

NTM is transmitted through environmental sources. They are ubiquitous in the environment and have been isolated from water, soil, food products, and domestic and wild animals [22]. An observational study found near-identical isolates of M. abscessus in cystic fibrosis clinics, suggesting the possibility of person-to-person transmission [19]. However, this is controversial. In a multicenter cohort, when isolates were analyzed with whole genome sequencing, unique strains were detected, even after intensive contact with other patients [29]. The respiratory or gastrointestinal tract is the usual portal of entry for NTM pulmonary disease. The incubation periods are variable [9].

RISK FACTORS — Cystic fibrosis is the most common risk factor for pulmonary NTM disease in children in industrialized nations [18,30]. In adults with cystic fibrosis, highly effective modulator therapy, which improves the function of the cystic fibrosis transmembrane conductance regulator protein, may decrease the risk of pulmonary NTM infection [31]; the impact in children is less clear. Screening patients with cystic fibrosis to make sure that NTM cultures are negative before initiation of anti-inflammatory therapy with a macrolide antibiotic is discussed separately. (See "Cystic fibrosis: Overview of the treatment of lung disease", section on 'Azithromycin'.)

Other risk factors for NTM pulmonary disease include polymorphisms in the cystic fibrosis transmembrane conductance regulatory genes without classic cystic fibrosis, polymorphisms in the natural resistance-associated macrophage protein 1 (NRAMP1) gene, hematopoietic stem cell transplantation, and congenital defects in interferon gamma and interleukin (IL)-12 synthesis and response pathways [32-39]. While children with severe combined immunodeficiency, DiGeorge syndrome, chronic granulomatous disease, and hyperimmunoglobulin M syndrome also have defects in portions of the IL-12 pathway, they are not prone to disseminated infection with NTM species [40]. (See "Mendelian susceptibility to mycobacterial diseases: Specific defects".)

Although the mechanism is unclear, vaping-related lung injury also may be a risk factor for NTM pulmonary infection. There is a case report of three patients who developed NTM pulmonary infections in association with vaping-related lung injury [41].

CLINICAL FEATURES — Clinically significant NTM pulmonary disease is most often described in children with preexisting lung disease (eg, cystic fibrosis) and in children with non-human immunodeficiency virus (HIV) immune deficiency who have disseminated NTM disease; it is less common in patients with HIV infection who have disseminated NTM disease [42,43]. (See "Disseminated nontuberculous mycobacterial (NTM) infections and NTM bacteremia in children", section on 'Risk factors'.)

The clinical features of NTM pulmonary disease are variable and nonspecific [22]. Most patients have chronic or recurrent cough [44]. Other pulmonary symptoms and signs include increased sputum production, dyspnea, hemoptysis, chest pain, rhonchi, crackles, wheezing, and stridor. In patients with cystic fibrosis, use of transmembrane conductance regulator modulator therapy may alter the clinical manifestations of pulmonary NTM [45].

The clinical features of NTM pulmonary disease may depend, to some extent, upon the underlying medical problem(s).

Constitutional symptoms (eg, fever, weight loss, fatigue, malaise) are common in children with NTM pulmonary disease, whether or not they have medical comorbidities or underlying lung disease [46,47]. However, they are more prevalent in immunodeficient children and children with advanced disease [44,48].

Pulmonary symptoms of NTM lung disease in children without underlying lung disease include wheezing or stridor that does not respond to bronchodilator therapy; these symptoms may be related to endobronchial granulomas and/or hilar lymphadenopathy [49,50].

In children with underlying lung disease, NTM pulmonary disease is associated with worsening of existing pulmonary symptoms (eg, cough, increased sputum production, exercise intolerance, decline in pulmonary status) [47].

The clinical manifestations of NTM pulmonary disease in patients with cystic fibrosis (including disease due to infection) are discussed separately. (See "Cystic fibrosis: Clinical manifestations of pulmonary disease".)

EVALUATION AND DIAGNOSIS

Diagnostic criteria — The diagnosis of NTM pulmonary disease is established based on the combination of compatible clinical and radiographic findings, the isolation of an NTM species in culture, and exclusion of other conditions in the differential diagnosis (table 2) [2,22]. The clinical, radiographic, and microbiologic criteria must be fulfilled. (See 'Differential diagnosis' below.)

When NTM is isolated from a patient with underlying lung disease, it can be difficult to determine whether NTM represents a true pathogen, colonization, or a contaminant. Consultation with an expert in infectious diseases and/or pulmonary medicine may be necessary. (See "Diagnosis of nontuberculous mycobacterial infections of the lungs".)

Clinical suspicion — NTM pulmonary disease may be suspected in [22,30,49] (see 'Clinical features' above):

Children with underlying pulmonary disease with worsening of pulmonary symptoms/function, weight loss, or low-grade fever, in whom other causes have been excluded

Children with cystic fibrosis and exacerbation of pulmonary symptoms that does not respond to routine treatment and antipseudomonal antibiotics (see "Cystic fibrosis: Overview of the treatment of lung disease" and "Cystic fibrosis: Antibiotic therapy for chronic pulmonary infection")

Children with HIV immune deficiency and disseminated NTM disease

Imaging studies — Imaging of the chest is necessary to diagnose NTM pulmonary disease [22]. In children with underlying pulmonary disease, it is important to compare radiographs and computed tomographic (CT) findings with previous studies to determine if there has been a change. It can be difficult to differentiate between worsening of the primary pulmonary disorder and NTM pulmonary disease. In addition, in patients with cystic fibrosis, use of transmembrane conductance regulator modulator therapy may alter the radiographic manifestations of pulmonary NTM [45].

Radiographic findings in NTM pulmonary disease include:

Nodular or cavitary opacities on chest radiograph (image 1), or

Multifocal bronchiectasis with multiple small nodules on high-resolution CT (image 2)

Other radiographic features of NTM pulmonary disease may vary with the clinical scenario. The most common radiographic findings in otherwise healthy children include hilar lymphadenopathy and a tree-in-bud appearance associated with endobronchial spread [49]. Infiltrates are more likely in children with constitutional symptoms [46]. Pleural effusions are uncommon, and cavitary lesions in young children are rare.

Radiographic features of NTM pulmonary disease also may vary depending on the causative species [22]:

M. avium complex (MAC) – Apical fibrocavitary disease or nodular and interstitial nodular infiltrates often involving the right middle lobe or lingula

M. kansasii – Cavitary infiltrates in the upper lobes

M. abscessus – Multilobar, patchy, reticulonodular or mixed interstitial-alveolar opacities with an upper lobe predominance; cavitation may occur

Microbiologic testing — When NTM pulmonary disease is suspected, pulmonary specimens should be sent for histopathology, acid-fast bacillus (AFB) staining, and mycobacterial culture and susceptibility testing (as well as bacterial and fungal stains and cultures as clinically indicated). A positive NTM culture is necessary for diagnosis. Compatible histopathology, positive AFB staining, rapid diagnostic tests, and tests that exclude Mycobacterium tuberculosis support the diagnosis.

It is helpful to discuss specimen collection, transport, and culture processing with the microbiology laboratory before sending tissue or fluid to the laboratory. Most NTM species will not grow in traditional bacterial culture media; rapidly growing NTM species (eg, M. abscessus and M. fortuitum) are an exception [51]. Techniques for maximizing the yield of cultures are discussed separately. (See "Microbiology of nontuberculous mycobacteria", section on 'Culture'.)

AFB staining – AFB staining provides supportive information but is neither necessary nor sufficient to make the diagnosis of NTM disease. A positive AFB stain increases the likelihood that an isolate is clinically significant but cannot distinguish among NTM species or between NTM species and M. tuberculosis [22]. Negative AFB stains do not exclude a diagnosis of mycobacterial disease. In a prospective series of 104 positive NTM cultures from patients with cystic fibrosis, the AFB smear was positive only in one-third [15]. (See "Microbiology of nontuberculous mycobacteria", section on 'Microscopy' and 'Clinical significance' below.)

Culture – A positive NTM culture (from sputum, bronchial wash or lavage, transbronchial or other lung biopsy) in a patient with compatible clinical and radiographic findings establishes the diagnosis of NTM pulmonary disease (table 2). In addition, the culture identifies the NTM species and the presence or absence of M. tuberculosis; these results are necessary to determine the clinical significance of the infection and appropriate antimycobacterial regimen.

Mycobacterial cultures may take days to several weeks to grow. The time (in days) to detection of mycobacterial growth may help to distinguish between NTM species.

The following species typically grow within seven days:

-M. abscessus

-M. fortuitum

-M. chelonae

-Mycobacterium smegmatis

-Mycobacterium mucogenicum

-Mycobacterium peregrinum

The following species require several weeks for growth:

-MAC

-M. kansasii

Obtaining adequate specimens for culture and proper processing are essential. The yield of cultures may be reduced by insufficient quality (eg, nasopharyngeal swab rather than expectorated sputum) or quantity of a respiratory specimen [52]. (See "Microbiology of nontuberculous mycobacteria", section on 'Culture'.)

Rapid diagnostic tests – Rapid diagnostic tests confirm the presence of NTM but do not distinguish between colonization and NTM-related disease. They do not satisfy the microbiologic criteria for diagnosis (table 2) [22]. Rapid methods for identification of NTM species include polymerase chain reaction (PCR) and high-pressure liquid chromatography (HPLC).

PCR – PCR may identify NTM species before identification in culture results are available. Nucleic acid probes for MAC and M. kansasii are commercially available. However, there is little experience with PCR and other molecular modalities for the diagnosis of NTM pulmonary disease in children. In adult patients, real-time multiplex PCR (eg, Xpert MTB/RIF) is increasingly used to differentiate between M. tuberculosis and NTM species such as MAC and M. abscessus [53]. Newer multiplex PCRs can also detect mutations conferring macrolide and aminoglycoside resistance [54,55].

HPLC – HPLC examines the mycolic acid fingerprint patterns that differ among most species or complexes of mycobacteria and can be used to speciate NTM, including rapidly growing mycobacteria [56]. However, HPLC cannot reliably differentiate between M. abscessus and M. chelonae.

Excluding M. tuberculosis – Although culture is necessary for definitive diagnosis, interferon gamma release assays (IGRAs) and tuberculin skin testing (TST) may be helpful in differentiating NTM from M. tuberculosis.

Pulmonary NTM and M. tuberculosis coinfection may occur in immunocompromised children. In observational studies in settings where M. tuberculosis is hyperendemic, NTM species were isolated in 3 to 15 percent of positive mycobacterial cultures. In another observational study in a tuberculosis-endemic setting, 2.8 percent of adults with confirmed pulmonary M. tuberculosis had coinfection with NTM [57].

IGRAs – IGRAs are the optimal test to exclude M. tuberculosis in children with NTM pulmonary disease. They are far more specific than TST. IGRAs measure interferon production by sensitized lymphocytes after exposure to antigens that are predominantly, but not exclusively, found in M. tuberculosis. (See "Tuberculosis infection (latent tuberculosis) in children", section on 'Interferon-gamma release assays'.)

Among the NTM species that most commonly cause pulmonary disease, the IGRAs do not include antigens shared by MAC or M. abscessus, but do include antigens shared by M. kansasii [58,59]. Thus, in a child with clinical and radiographic findings of pulmonary disease, a positive IGRA supports a diagnosis of M. tuberculosis or M. kansasii, a negative IGRA supports a diagnosis of NTM (other than M. kansasii) or another condition in the differential diagnosis, and an indeterminate IGRA is not helpful.

TST – The TST contains numerous antigens shared by both M. tuberculosis and NTM species. NTM infections may account for many positive PPD reactions [58].

Interpretation of TST depends upon risk factors for M. tuberculosis and receipt of bacille Calmette-Guérin (BCG) vaccination.

-In children without risk factors for M. tuberculosis and who have not received BCG vaccination, TST with ≥5 mm induration at 48 hours supports a diagnosis of NTM infection.

-In children with risk factors for M. tuberculosis or who have received BCG vaccination, TST ≥5 mm does not distinguish between NTM, M. tuberculosis, or reaction to BCG vaccination [22,60-62]. TST ≥15 mm may be more suggestive of M. tuberculosis than NTM but does not exclude NTM. (See "Tuberculosis disease in children: Epidemiology, clinical manifestations, and diagnosis".)

-Independent of risk factors for M. tuberculosis, TST with <5 mm of induration at 48 hours is not helpful in excluding mycobacterial disease. If NTM continues to be suspected in a child with negative TST, specimens should be obtained and sent for microbiologic and histopathologic studies.

Clinical significance — When NTM is isolated from a child with pulmonary symptoms and signs, the clinical significance of the isolate must be determined. The pathogenicity of the various NTM species varies substantially. The possibility that NTM was isolated as a result of contamination or represents colonization of diseased lung also must be considered [22]. Consultation with a specialist in infectious diseases may be helpful in determining the clinical significance of the isolate.

Factors that increase the likelihood of clinical significance include recovery from multiple specimens, recovery in large quantities (ie, AFB-positive specimens), and recovery from a normally sterile site [22].

DIFFERENTIAL DIAGNOSIS — Important considerations in the differential diagnosis of NTM pulmonary disease in children include:

Progression of primary pulmonary disease (eg, worsening bronchiectasis in patients with cystic fibrosis) – In children with underlying lung disease, it can be difficult to determine whether pulmonary symptoms are related to the progression of the underlying lung disease or NTM pulmonary infection. Mycobacterial cultures of respiratory specimens (eg, expectorated sputum, bronchial wash/lavage, lung biopsy) are necessary.

M. tuberculosis – The cavitary appearance and endobronchial spread of NTM can mimic radiographic findings of M. tuberculosis. It is important to distinguish tuberculosis from NTM pulmonary disease because they require different treatments; in addition, M. tuberculosis requires contact tracing and airborne isolation precautions, whereas NTM does not.

In a child with risk factors for M. tuberculosis and acid-fast bacillus-positive sputum, NTM and M. tuberculosis must be differentiated by mycobacterial culture and speciation; interferon gamma release assays and tuberculin skin testing cannot reliably differentiate NTM from M. tuberculosis. In a series of respiratory specimens from 17 children in Texas who met criteria for diagnosis of NTM pulmonary disease (table 2), M. tuberculosis was isolated more frequently than all other mycobacterial species (23 versus 17 specimens) [10]. (See 'Microbiologic testing' above.)

It may be necessary to initiate empiric therapy before the distinction between NTM and M. tuberculosis is made. In such cases, coverage for both pathogens is provided in a multidrug regimen. (See 'NTM and M. tuberculosis' below.)

Other forms of nodular lung disease, including disseminated fungal disease (table 3) – Clinical features (eg, exposures) may help to differentiate NTM from other nodular lung diseases, but microbiologic studies are necessary for definitive diagnosis. (See "Diagnosis and treatment of pulmonary histoplasmosis" and "Primary pulmonary coccidioidal infection" and "Clinical manifestations and diagnosis of blastomycosis".)

Hypersensitivity reactions, as seen in vaping (ie, use of electronic cigarette devices to inhale aerosolized substances) [63]. (See "E-cigarette or vaping product use-associated lung injury (EVALI)".)

Pneumocystis jirovecii pneumonia – The ground-glass appearance occasionally seen in NTM pulmonary disease can mimic P. jirovecii pneumonia. Microbiologic studies can be helpful in differentiating between NTM and P. jirovecii pneumonia. (See "Epidemiology, clinical manifestations, and diagnosis of Pneumocystis pneumonia in patients without HIV", section on 'Diagnosis'.)

TREATMENT

Decision to treat — Antimycobacterial treatment generally is warranted for children who meet clinical and microbiologic criteria for NTM pulmonary disease (table 2). However, the potential risks and benefits of antimycobacterial therapy disease (table 4) must be considered before initiating treatment.

As an example, in an adolescent with cystic fibrosis who is awaiting lung transplantation in whom M. abscessus is isolated, the benefits of decreased burden of M. abscessus and potential decreased risk of recurrence posttransplant may outweigh the adverse effects of parenteral multidrug therapy with ototoxic and nephrotoxic drugs [64]. On the other hand, the risks of therapy may outweigh the benefits in a young child in whom a potentially nonpathogenic species or contaminant (eg, M. mucogenicum) is isolated from a respiratory specimen. Similarly, an asymptomatic child with cystic fibrosis with stable pulmonary function testing and chest imaging in whom sputum samples demonstrate an NTM species may not benefit from treatment at that time. (See 'M. abscessus' below.)

Susceptibility testing — Susceptibility testing is recommended for clinically significant NTM pulmonary isolates [22]. The antibiotic susceptibilities of NTM species vary substantially. However, with the exception of macrolides in the treatment of M. avium complex (MAC), drug susceptibility testing for NTM has less correlation with in vivo response than for tuberculosis or other bacterial pathogens.

Given that the diagnostic criteria for NTM pulmonary disease require sequential sputum samples (table 2), speciation and drug susceptibility results often are available before initiation of antimicrobial therapy [22]. However, it may be necessary to initiate empiric therapy before speciation is complete, particularly when M. tuberculosis cannot be excluded. (See 'Diagnostic criteria' above and 'Differential diagnosis' above.)

Antimycobacterial therapy — Suggested antimycobacterial regimens for NTM pulmonary infections are summarized in the table (table 5).

NTM and M. tuberculosis — If the possibility of M. tuberculosis cannot be excluded after clinical, radiographic, and initial laboratory evaluation (eg, children with risk factors for M. tuberculosis and positive smears for acid-fast bacilli [AFB]), we suggest empiric treatment with the following regimen until speciation is available (table 4 and table 5) [65]:

Isoniazid, plus

Rifampin (rifampicin), plus

Ethambutol, plus

Azithromycin (preferred) or clarithromycin, plus

Pyrazinamide

Based on in vitro susceptibilities, this regimen provides initial empiric coverage for M. tuberculosis and the two NTM species most frequently isolated from children with NTM pulmonary disease (MAC and M. kansasii). It is consistent with the recommendations of the American Thoracic Society/Infectious Diseases Society of America (ATS/IDSA) and the American Academy of Pediatrics (AAP) Committee on Infectious Diseases [9,11]. This initial regimen can be tailored according to speciation and susceptibility testing when the results are available. (See 'Microbiologic testing' above.)

For children in whom the high pill burden of this five-drug regimen is too difficult, it is reasonable to exclude pyrazinamide. However, if M. tuberculosis is confirmed, the course of therapy should be nine months (rather than the six-month course that can be used for isolated noncavitary pulmonary disease in most immunocompetent children). (See "Tuberculosis disease in children: Treatment and prevention".)

We generally prefer azithromycin to clarithromycin for pulmonary NTM infection. Azithromycin can be administered once daily, and azithromycin suspension is more palatable than clarithromycin suspension [66], which may increase adherence. In addition, there is the potential for drug interaction between clarithromycin and rifamycins – rifamycins can decrease the serum concentration of clarithromycin [67,68]. It is unclear the degree to which the benefit of combination therapy is outweighed by the risk of selecting for clarithromycin resistance. However, use of azithromycin rather than clarithromycin permits combination therapy without potentially sacrificing the minimal inhibitory concentration for the macrolide. A systematic review found no evidence of a difference in the effectiveness of different antimicrobial regimens for NTM pulmonary infection in patients with cystic fibrosis [69].

M. avium complex

Children without HIV

Nodular or noncavitating bronchiectasis – We suggest that children without HIV infection children with nodular or noncavitating bronchiectatic MAC lung disease be treated with a three-drug regimen that includes (table 5 and table 4) [9,22]:

-A rifamycin (rifampin [rifampicin] or rifabutin), plus

-Ethambutol, plus

-Azithromycin (preferred) or clarithromycin

We generally prefer azithromycin to clarithromycin for pulmonary NTM infection. (See 'NTM and M. tuberculosis' above.)

The medications are administered three times per week until the sputum cultures have been negative for at least one year [22]. Three-times-weekly dosing for patients with nodular or nonsevere bronchiectatic MAC disease is supported by several prospective studies in adult patients demonstrating effectiveness [70-72] and a systematic review demonstrating increased adherence [73].

Fibrocavitary or extensive bronchiectasis – We suggest that children without HIV infection children with extensive bronchiectasis or fibrocavitary MAC lung disease be treated with a regimen that includes (table 5 and table 4) [9,22]:

-A rifamycin (rifampin [rifampicin] or rifabutin), plus

-Ethambutol, plus

-Azithromycin (preferred) or clarithromycin), plus

-An aminoglycoside (amikacin or streptomycin)

For the macrolide, we generally prefer azithromycin to clarithromycin. The first three drugs should be administered daily to avoid the interval development of macrolide resistance. The aminoglycoside is also administered daily in children; however, in older adolescents, amikacin or streptomycin may be administered three times per week. In a randomized study in adult patients with tuberculous or nontuberculous mycobacterial infections, the administration of amikacin 25 mg/kg three times per week was not associated with increased toxicity compared with daily dosing [74].

The aminoglycoside should be discontinued after eight weeks; the remainder of the drugs should be continued until the child's sputum cultures have been negative for at least one year [22].

The recommendations for treatment of MAC pulmonary disease in children without HIV infection are based upon in vitro susceptibilities and observational studies in HIV-negative adult patients, in which multidrug treatment with a macrolide-containing regimen was associated with clearance of MAC from sputum in >60 percent of patients [70,75,76]. They are consistent with the recommendations of the ATS/IDSA and the AAP Committee on Infectious Diseases [9,11].

Children with HIV infection Respiratory symptoms are uncommon among children with HIV infection with disseminated MAC, and isolated pulmonary disease is rare [43]. We suggest that children with HIV infections and MAC who meet the diagnostic criteria for pulmonary disease (table 2) be treated in the same manner as those with disseminated infection. (See "Disseminated nontuberculous mycobacterial (NTM) infections and NTM bacteremia in children", section on 'Antimycobacterial therapy'.)

M. abscessus — For children with M. abscessus pulmonary disease, we suggest initial treatment with (table 5 and table 4) [9,11]:

Azithromycin (preferred) or clarithromycin, plus

Amikacin, plus

Cefoxitin or a carbapenem (eg, imipenem, meropenem)

Another drug (eg, linezolid, tigecycline) should be added if M. abscessus subspecies abscessus or M. abscessus subspecies bolletii is isolated [11]. These subspecies have inducible macrolide resistance although resistance to other antibiotics remains constant [77]. Although the macrolide may be continued for its immunomodulatory effects, it should not count as one of the active drugs in the regimen.

Although there is limited evidence to determine the optimal combination, the initial regimen should include at least three drugs to which there is documented susceptibility, bearing in mind that in vitro susceptibilities may not correlate with clinical response [22,78]. For children with substantial antibiotic exposures, it may not be possible to find three antibiotics to which the isolate has documented susceptibility. When this occurs, although there is little evidence for guidance, we start with the agent(s) to which M. abscessus is apparently susceptible and then add one or more bactericidal agents, trying to choose those with the fewest adverse effects and the most convenient dosing regimens (eg, oral rather than parenteral medications).

While both azithromycin and clarithromycin have been used to treat M. abscessus lung disease, we prefer azithromycin for several reasons: decreased drug-drug interactions, increased palatability of the liquid formulation, and an observational study suggesting that sustained culture conversion was more common in adults with M. abscessus lung disease who received azithromycin than in those who received clarithromycin [79].

The goal of M. abscessus therapy is not microbiologic eradication. Instead, symptomatic and radiographic improvements should be the endpoints of therapy [80]. There is limited evidence to determine the optimal duration of treatment. In addition to the difficulty in eradicating M. abscessus, it is possible for patients to be reinfected by different M. abscessus genotypes during or after completion of antimicrobial therapy [81]. In a retrospective review of adult patients with M. abscessus lung disease, >40 percent relapsed within a year of treatment cessation; AFB sputum smear positivity was the only consistent predictor of treatment outcome [81]. In another study, lower body mass index, bilateral lung involvement, and fibrocavitary disease were associated with poor prognoses [79].

After four to eight weeks of intensive therapy with at least three drugs with documented susceptibility, two drugs may be used to complete therapy (usually for a total duration of 6 to 12 months). The duration of therapy may be limited by the patient's ability to tolerate the medications [22,78]. M. abscessus has a predilection for infecting diseased lungs and is difficult to eradicate [82]; cure is generally possible only with surgical resection when lung disease is limited [44]. (See 'Surgical therapy' below.)

Parenteral agents with in vitro activity against M. abscessus include amikacin, cefoxitin, tigecycline, and imipenem. Oral drugs with some in vitro activity include macrolides, clofazimine, linezolid, and fluoroquinolones. Linezolid and fluoroquinolones can be used for patients with refractory M. abscessus pulmonary disease. However, the utility of linezolid in children may be limited by bone marrow suppression or optic neuritis, and high rates of fluoroquinolone resistance have been reported in children with no previous exposure to fluoroquinolones [10].

Bedaquiline and delamanid are available for the treatment of drug-resistant M. tuberculosis in adults and may have potential in the treatment of M. abscessus infections [83-86]. However, there is little experience with these agents for the treatment of M. tuberculosis or NTM pulmonary disease in children [87,88]. Consultation with an expert in mycobacterial disease is warranted. (See "Tuberculosis disease in children: Treatment and prevention".)

M. kansasii

Rifampin (rifampicin)-susceptible M. kansasii – We suggest that children with rifampin (rifampicin)-susceptible M. kansasii pulmonary disease be treated with (table 5 and table 4):

Rifampin (rifampicin), plus

Ethambutol, plus

A macrolide (azithromycin [preferred] or clarithromycin) or isoniazid

If isoniazid is included in the initial regimen, children should receive therapy daily; if a macrolide is used, therapy can be given three times a week. All children with cavitary M. kansasii pulmonary disease should receive daily therapy.

We generally prefer azithromycin to clarithromycin for pulmonary NTM infections. (See 'NTM and M. tuberculosis' above.)

Treatment for rifampin (rifampicin)-susceptible M. kansasii should continue until sputum cultures have been negative for at least one year [22].

Rifampin (rifampicin)-resistant M. kansasii or intolerance to a first-line medication – We suggest that children with rifampin (rifampicin)-resistant isolates or intolerance to any of the first-line medications be treated with three drugs based upon in vitro susceptibilities. Potential agents include macrolides, fluoroquinolones, aminoglycosides, and trimethoprim-sulfamethoxazole (TMP-SMX) (table 5 and table 4). Treatment for rifampin (rifampicin)-resistant M. kansasii should continue until sputum cultures have been negative for 12 to 18 months [22].

There have been no randomized trials of treatment for M. kansasii. In in vitro studies, M. kansasii is susceptible to macrolides, TMP-SMX, amikacin, fluoroquinolones, rifamycins, bedaquiline, and delamanid [89-93]. In observational studies, multidrug regimens that included rifampin (rifampicin) were associated with clearance of M. kansasii within four months in all patients [94-97]. Relapse, which was rare, was associated with development of rifampin (rifampicin) resistance [94].

Surgical therapy — Surgical resection may be required as an adjunct to antimycobacterial therapy for children with MAC with isolated cavities, M. abscessus, failure to convert sputum cultures after six months, or patients who cannot tolerate antimycobacterial therapy [22]. (See 'Monitoring response to therapy' below.)

In patients with limited pulmonary disease, surgical resection can be curative. In patients with extensive pulmonary disease, surgical resection may be performed in an attempt to decrease organism burden.

Monitoring response to therapy — Children who are being treated for pulmonary NTM disease should be monitored by monthly symptom screening, sputum analyses, and pulmonary function tests. Monitoring for adverse effects of therapy is discussed below. (See 'Monitoring adverse effects' below.)

We do not routinely monitor response to therapy for pulmonary NTM disease with serial CT of the chest, although repeat imaging may be warranted one to two months after initiation of therapy and for children who have clinical deterioration during therapy. Children with pulmonary NTM disease, by virtue of their underlying medical conditions, are likely to have higher cumulative radiation exposure at baseline than children without such conditions [98].

Successful treatment of patients with MAC pulmonary disease is indicated by clinical improvement within three to six months and negative sputum cultures within 12 months of initiation of therapy [75]. Possible reasons for failure to respond within these timeframes include poor adherence/drug intolerance, macrolide resistance, or anatomic limitations (eg, focal cystic or cavitary disease) [22]. An alternative medication regimen or surgery may be warranted. (See 'M. avium complex' above.)

The endpoint of therapy for M. abscessus is symptomatic improvement and prevention of disease progression rather than microbiologic eradication. Microbiologic eradication is potentially possible only with surgical resection of limited lung disease [44]. (See 'M. abscessus' above and 'Surgical therapy' above.)

Although not yet available for clinical use, studies suggest that certain biomarkers may be used to monitor response to therapy and distinguish exacerbations from disease. Levels of antiglycopeptidolipid core immunoglobulin A antibody (anti-GPL-core IgA) fall during treatment and are correlated with symptomatic improvement; anti-GPL-core IgA levels are also higher in patients with MAC and bronchiectasis than those with MAC without bronchiectasis [99]. Urine lipoarabinomannan (LAM) is a point-of-care assay best used to detect tuberculosis in persons living with HIV, but urine LAM may be a marker of progression from pulmonary colonization to disease in patients with MAC [100].

Monitoring adverse effects — The antimycobacterial agents that are used to treat NTM pulmonary infections often are difficult to tolerate and some have important toxicities (table 4). It may be challenging to determine which of the multiple medications required for treatment of NTM pulmonary infections is responsible for a given reaction. In addition, children with comorbid medical conditions may require additional medications that can interact with antimycobacterial therapy, particularly with rifamycins [101]. They also may have baseline hepatic or renal dysfunction, which may decrease medication tolerance.

Although most adverse effects are monitored clinically by assessing the patient for symptoms (eg, dizziness, vomiting), some specific parameters should be monitored at baseline or periodically as indicated below, even in children without comorbid medical conditions [22].

For children receiving combination rifampin (rifampicin), ethambutol, and macrolide therapy (with or without other agents), we suggest baseline complete blood count (CBC), electrocardiogram (ECG), alkaline phosphatase, aspartate aminotransferase (AST), and alanine aminotransferase (ALT). Thereafter, the need for subsequent laboratory evaluation should be driven by symptoms and/or examination findings; scheduled laboratory evaluation may not be needed unless patients are receiving other marrow-suppressive or potentially hepatotoxic medications. If the regimen includes other agent(s), the specific parameters for those agents should be monitored as indicated below. Ethambutol is rapidly metabolized in children and ocular toxicity is extremely rare in children with normal renal function [102,103]. For the treatment of M. tuberculosis in children, the World Health Organization recommends vision screening if possible, but unavailability of vision screening is not a contraindication [104].

For children receiving other regimens, we suggest baseline CBC, alkaline phosphatase, AST, and ALT followed by periodic monitoring of specific parameters for particular agents as follows:

Rifamycins (rifampin, rifabutin, rifapentine) – Periodic CBC to evaluate granulocytopenia and thrombocytopenia (see "Rifamycins (rifampin, rifabutin, rifapentine)")

Macrolides (eg, azithromycin, clarithromycin) – Baseline ECG before initiation of therapy to evaluate prolongation of the QT interval; periodic alkaline phosphatase, AST, and ALT during the first three months of therapy to evaluate hepatotoxicity (see "Azithromycin and clarithromycin", section on 'Adverse reactions')

Isoniazid – Periodic monitoring of AST and ALT may be warranted in children with pulmonary NTM who are at increased risk for liver disease (eg, those with cystic fibrosis) or are receiving other medications that are metabolized in the liver (see "Isoniazid hepatotoxicity")

Aminoglycosides (including amikacin and streptomycin) – Periodic hearing evaluations and renal function tests to monitor ototoxicity (both sensorineural hearing loss and vestibulitis) and nephrotoxicity; for children receiving amikacin, periodic amikacin levels

Cefoxitin – Periodic CBC to monitor bone marrow suppression

TMP-SMX – Periodic CBC to monitor bone marrow suppression

Linezolid – Periodic CBC to monitor bone marrow suppression; periodic eye examinations by an ophthalmologist to evaluate optic neuritis

Bedaquiline or delamanid may be used in certain children with M. abscessus or M. kansasii pulmonary disease. Adverse effects of bedaquiline include nausea, hepatitis, prolongation of the QT interval, and pancreatitis; obtain baseline alkaline phosphatase, AST, ALT, ECG, and lipase before initiation and periodically during treatment. Adverse effects of delamanid include nausea, vomiting, hepatitis, anemia, eosinophilia, dizziness, peripheral neuropathy, and prolongation of the QT interval; obtain baseline CBC, alkaline phosphatase, AST, ALT, and ECG before initiation.

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: Nontuberculous mycobacteria".)

SUMMARY AND RECOMMENDATIONS

Risk factors and microbiology – Nontuberculous mycobacterial (NTM) pulmonary disease in children usually affects patients with underlying lung disease, particularly cystic fibrosis. Mycobacterium avium complex (MAC) and Mycobacterium abscessus are the species most frequently isolated from children with NTM pulmonary disease. (See 'Risk factors' above and 'Microbiology' above.)

Clinical features – Clinical features of NTM pulmonary disease are variable and nonspecific. Most patients have chronic or recurrent cough. Other findings include increased sputum, dyspnea, hemoptysis, chest pain, abnormal auscultatory findings (eg, crackles, wheezing, stridor), fever, fatigue, and weight loss. (See 'Clinical features' above.)

Diagnosis and differential diagnosis – Diagnosis of NTM pulmonary disease requires a combination of clinical symptoms, compatible radiographic findings (eg, nodular or cavitary opacities on chest radiograph (image 1), multifocal bronchiectasis with multiple small nodules on high-resolution CT (image 2)), isolation of NTM in culture, and exclusion of other conditions in the differential diagnosis (table 2). (See 'Evaluation and diagnosis' above.)

It is particularly important to exclude Mycobacterium tuberculosis because its treatment and public health implications differ from those of NTM. Additional considerations in the differential diagnosis include progression of primary pulmonary disease, other forms of nodular lung disease (table 3), and Pneumocystis jirovecii pneumonia. Microbiologic studies are necessary to distinguish among the possibilities. (See 'Differential diagnosis' above.)

Indications for treatment – Although antimycobacterial treatment generally is warranted for children who meet clinical and microbiologic criteria for NTM pulmonary disease (table 2), the risks and benefits of treatment and clinical significance of the isolate must be considered. (See 'Decision to treat' above and 'Clinical significance' above.)

Treatment regimens – Treatment regimens for NTM pulmonary infections in children are summarized in the tables (table 5 and table 4).

NTM and M. tuberculosis – For children in whom the distinction between NTM and M. tuberculosis remains uncertain, we suggest empiric treatment with isoniazid, rifampin (rifampicin), ethambutol, pyrazinamide, and a macrolide (azithromycin or clarithromycin) (table 4) rather than other combinations pending speciation and results of antimycobacterial susceptibility testing (Grade 2C). It is reasonable to exclude pyrazinamide if the five-drug regimen is too difficult. (See 'NTM and M. tuberculosis' above.)

MAC – Treatment of MAC pulmonary disease in HIV-negative children depends upon the severity (see 'M. avium complex' above):

-For children with nodular or noncavitating bronchiectatic disease, we suggest treatment with a rifamycin, ethambutol, and a macrolide (azithromycin preferred) (table 4) rather than other combinations (Grade 2C). Therapy is administered three times per week and continued until sputum cultures have been negative for at least one year.

-For children with cavitary or extensive bronchiectatic disease, we suggest treatment with a rifamycin, ethambutol, a macrolide (azithromycin preferred), and an aminoglycoside (table 4) rather than other combinations (Grade 2C). Therapy is administered daily; thrice-weekly therapy has been used in adolescents and adults. The aminoglycoside is discontinued after eight weeks; the other drugs are continued until the child's sputum cultures have been negative for at least one year.

Treatment of MAC pulmonary disease in HIV-positive children is the same as that for disseminated NTM infections and is discussed separately. (See "Disseminated nontuberculous mycobacterial (NTM) infections and NTM bacteremia in children", section on 'Antimycobacterial therapy'.)

M. abscessus – For children with M. abscessus pulmonary disease, we suggest initial treatment with a macrolide (azithromycin preferred), amikacin, and either cefoxitin or a carbapenem (table 4) rather than other combinations (Grade 2C). The endpoint of therapy for M. abscessus is symptomatic improvement and prevention of disease progression rather than microbiologic eradication. (See 'M. abscessus' above.)

M. kansasii

-Rifampin-susceptible – For children with rifampin-susceptible isolates, we suggest combination therapy with rifampin plus ethambutol plus either a macrolide (azithromycin preferred) or isoniazid (table 4) (Grade 2C). Treatment is continued until sputum cultures have been negative for at least one year.

-Rifampin-resistant or intolerance to a first-line agent – For children with rifampin-resistant isolates or resistance to a first-line agent, we suggest treatment with three drugs based on in vitro susceptibilities (Grade 2C). Potential agents include macrolides, fluoroquinolones, aminoglycosides, and trimethoprim-sulfamethoxazole (table 4). Treatment is continued until sputum cultures have been negative for 12 to 18 months.

Adjunctive surgery – Adjunctive surgical resection may be necessary for children with MAC with isolated cavities, M. abscessus, failure to convert sputum cultures after six months, or patients who cannot tolerate antimycobacterial therapy. (See 'Surgical therapy' above.)

Monitoring response and adverse events – Children who are being treated for pulmonary NTM disease should be monitored by monthly symptom screening, evaluation for medication-associated adverse events (table 4), sputum analyses, and pulmonary function tests. (See 'Monitoring response to therapy' above and 'Monitoring adverse effects' above.)

  1. Fraser L, Moore P, Kubba H. Atypical mycobacterial infection of the head and neck in children: a 5-year retrospective review. Otolaryngol Head Neck Surg 2008; 138:311.
  2. Daley CL, Iaccarino JM, Lange C, et al. Treatment of Nontuberculous Mycobacterial Pulmonary Disease: An Official ATS/ERS/ESCMID/IDSA Clinical Practice Guideline. Clin Infect Dis 2020; 71:905.
  3. Celdrán A, Esteban J, Mañas J, Granizo JJ. Wound infections due to Mycobacterium fortuitum after polypropylene mesh inguinal hernia repair. J Hosp Infect 2007; 66:374.
  4. Ferguson DD, Gershman K, Jensen B, et al. Mycobacterium goodii infections associated with surgical implants at Colorado hospital. Emerg Infect Dis 2004; 10:1868.
  5. Al Soub H, Al Maslamani M, Al Khuwaiter J, et al. Myocardial abscess and bacteremia complicating Mycobacterium fortuitum pacemaker infection: case report and review of the literature. Pediatr Infect Dis J 2009; 28:1032.
  6. McNabb A, Eisler D, Adie K, et al. Assessment of partial sequencing of the 65-kilodalton heat shock protein gene (hsp65) for routine identification of Mycobacterium species isolated from clinical sources. J Clin Microbiol 2004; 42:3000.
  7. Tortoli E. Impact of genotypic studies on mycobacterial taxonomy: the new mycobacteria of the 1990s. Clin Microbiol Rev 2003; 16:319.
  8. Parte AC. List of prokaryotic names with standing in nomenclature. Genus Mycobacterium. Available at: http://www.bacterio.net/mycobacterium.html (Accessed on October 04, 2019).
  9. American Academy of Pediatrics. Nontuberculous mycobacteria (environmental mycobacteria, mycobacteria other than Mycobacterium tuberculosis). In: Red Book: 2021-2024 Report of the Committee on Infectious Diseases, 32nd ed, Kimberlin DW, Barnett ED, Lynfield R, Sawyer MH (Eds), American Academy of Pediatrics, Itasca, IL 2021. p.814.
  10. Cruz AT, Ong LT, Starke JR. Mycobacterial infections in Texas children: a 5-year case series. Pediatr Infect Dis J 2010; 29:772.
  11. Daley CL, Iaccarino JM, Lange C, et al. Treatment of Nontuberculous Mycobacterial Pulmonary Disease: An Official ATS/ERS/ESCMID/IDSA Clinical Practice Guideline. Clin Infect Dis 2020; 71:e1.
  12. Suresh P, Kumar A, Biswas R, et al. Epidemiology of Nontuberculous Mycobacterial Infection in Tuberculosis Suspects. Am J Trop Med Hyg 2021; 105:1335.
  13. Okoi C, Anderson ST, Mulwa S, et al. Pulmonary non-tuberculous mycobacteria in colonisation and disease in The Gambia. Sci Rep 2022; 12:19523.
  14. Adjemian J, Olivier KN, Prevots DR. Epidemiology of Pulmonary Nontuberculous Mycobacterial Sputum Positivity in Patients with Cystic Fibrosis in the United States, 2010-2014. Ann Am Thorac Soc 2018; 15:817.
  15. Roux AL, Catherinot E, Ripoll F, et al. Multicenter study of prevalence of nontuberculous mycobacteria in patients with cystic fibrosis in france. J Clin Microbiol 2009; 47:4124.
  16. Levy I, Grisaru-Soen G, Lerner-Geva L, et al. Multicenter cross-sectional study of nontuberculous mycobacterial infections among cystic fibrosis patients, Israel. Emerg Infect Dis 2008; 14:378.
  17. Olivier KN, Weber DJ, Wallace RJ Jr, et al. Nontuberculous mycobacteria. I: multicenter prevalence study in cystic fibrosis. Am J Respir Crit Care Med 2003; 167:828.
  18. Pierre-Audigier C, Ferroni A, Sermet-Gaudelus I, et al. Age-related prevalence and distribution of nontuberculous mycobacterial species among patients with cystic fibrosis. J Clin Microbiol 2005; 43:3467.
  19. Bryant JM, Grogono DM, Rodriguez-Rincon D, et al. Emergence and spread of a human-transmissible multidrug-resistant nontuberculous mycobacterium. Science 2016; 354:751.
  20. Gardner AI, McClenaghan E, Saint G, et al. Epidemiology of Nontuberculous Mycobacteria Infection in Children and Young People With Cystic Fibrosis: Analysis of UK Cystic Fibrosis Registry. Clin Infect Dis 2019; 68:731.
  21. Eikani MS, Nugent M, Poursina A, et al. Clinical course and significance of nontuberculous mycobacteria and its subtypes in cystic fibrosis. BMC Infect Dis 2018; 18:311.
  22. Griffith DE, Aksamit T, Brown-Elliott BA, et al. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med 2007; 175:367.
  23. Pham-Huy A, Robinson JL, Tapiéro B, et al. Current trends in nontuberculous mycobacteria infections in Canadian children: A pediatric investigators collaborative network on infections in Canada (PICNIC) study. Paediatr Child Health 2010; 15:276.
  24. Vu TT, Daniel SJ, Quach C. Nontuberculous mycobacteria in children: a changing pattern. J Otolaryngol 2005; 34 Suppl 1:S40.
  25. Hatziagorou E, Orenti A, Drevinek P, et al. Changing epidemiology of the respiratory bacteriology of patients with cystic fibrosis-data from the European cystic fibrosis society patient registry. J Cyst Fibros 2020; 19:376.
  26. Fauroux B, Delaisi B, Clément A, et al. Mycobacterial lung disease in cystic fibrosis: a prospective study. Pediatr Infect Dis J 1997; 16:354.
  27. Esther CR Jr, Henry MM, Molina PL, Leigh MW. Nontuberculous mycobacterial infection in young children with cystic fibrosis. Pediatr Pulmonol 2005; 40:39.
  28. Cavalli Z, Reynaud Q, Bricca R, et al. High incidence of non-tuberculous mycobacteria-positive cultures among adolescent with cystic fibrosis. J Cyst Fibros 2017; 16:579.
  29. Doyle RM, Rubio M, Dixon G, et al. Cross-transmission Is Not the Source of New Mycobacterium abscessus Infections in a Multicenter Cohort of Cystic Fibrosis Patients. Clin Infect Dis 2020; 70:1855.
  30. Whittaker LA, Teneback C. Atypical mycobacterial and fungal infections in cystic fibrosis. Semin Respir Crit Care Med 2009; 30:539.
  31. Ricotta EE, Prevots DR, Olivier KN. CFTR modulator use and risk of nontuberculous mycobacteria positivity in cystic fibrosis, 2011-2018. ERJ Open Res 2022; 8.
  32. Holland SM. Interferon gamma, IL-12, IL-12R and STAT-1 immunodeficiency diseases: disorders of the interface of innate and adaptive immunity. Immunol Res 2007; 38:342.
  33. Marazzi MG, Chapgier A, Defilippi AC, et al. Disseminated Mycobacterium scrofulaceum infection in a child with interferon-gamma receptor 1 deficiency. Int J Infect Dis 2010; 14:e167.
  34. Tsolia MN, Chapgier A, Taprantzi P, et al. Disseminated nontuberculous mycobacterial infection in a child with interferon-gamma receptor 1 deficiency. Eur J Pediatr 2006; 165:458.
  35. Koscielniak E, de Boer T, Dupuis S, et al. Disseminated Mycobacterium peregrinum infection in a child with complete interferon-gamma receptor-1 deficiency. Pediatr Infect Dis J 2003; 22:378.
  36. Vesterhus P, Holland SM, Abrahamsen TG, Bjerknes R. Familial disseminated infection due to atypical mycobacteria with childhood onset. Clin Infect Dis 1998; 27:822.
  37. Colombo RE, Hill SC, Claypool RJ, et al. Familial clustering of pulmonary nontuberculous mycobacterial disease. Chest 2010; 137:629.
  38. Koh WJ, Kwon OJ, Kim EJ, et al. NRAMP1 gene polymorphism and susceptibility to nontuberculous mycobacterial lung diseases. Chest 2005; 128:94.
  39. Unal E, Yen C, Saiman L, et al. A low incidence of nontuberculous mycobacterial infections in pediatric hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant 2006; 12:1188.
  40. Wu UI, Holland SM. Host susceptibility to non-tuberculous mycobacterial infections. Lancet Infect Dis 2015; 15:968.
  41. Chen L, Arens R, Chidambaram AG, et al. Vaping Associated Pulmonary Nontuberculous Mycobacteria. Lung 2021; 199:21.
  42. Aronchick JM, Miller WT Jr. Disseminated nontuberculous mycobacterial infections in immunosuppressed patients. Semin Roentgenol 1993; 28:150.
  43. Panel on Opportunistic Infections in HIV-Exposed and HIV-Infected Children. Guidelines for the prevention and treatment of opportunistic infections in HIV-exposed and HIV-infected children. Department of Health and Human Services. Available at: https://aidsinfo.nih.gov/guidelines/html/5/pediatric-opportunistic-infection/0 (Accessed on March 25, 2019).
  44. Griffith DE, Girard WM, Wallace RJ Jr. Clinical features of pulmonary disease caused by rapidly growing mycobacteria. An analysis of 154 patients. Am Rev Respir Dis 1993; 147:1271.
  45. Martiniano SL, Nick JA, Daley CL. Nontuberculous Mycobacterial Infections in Cystic Fibrosis. Clin Chest Med 2022; 43:697.
  46. Nolt D, Michaels MG, Wald ER. Intrathoracic disease from nontuberculous mycobacteria in children: two cases and a review of the literature. Pediatrics 2003; 112:e434.
  47. Esther CR Jr, Esserman DA, Gilligan P, et al. Chronic Mycobacterium abscessus infection and lung function decline in cystic fibrosis. J Cyst Fibros 2010; 9:117.
  48. Tebruegge M, Curtis N. Mycobacterium nontuberculosis species. In: Principles and Practice of Pediatric Infectious Diseases, 5th ed, Long SS, Prober CG, Fischer M (Eds), Elsevier, Philadelphia 2018. p.806.
  49. Freeman AF, Olivier KN, Rubio TT, et al. Intrathoracic nontuberculous mycobacterial infections in otherwise healthy children. Pediatr Pulmonol 2009; 44:1051.
  50. Kröner C, Griese M, Kappler M, et al. Endobronchial lesions caused by nontuberculous mycobacteria in apparently healthy pediatric patients. Pediatr Infect Dis J 2015; 34:532.
  51. Murillo J, Torres J, Bofill L, et al. Skin and wound infection by rapidly growing mycobacteria: an unexpected complication of liposuction and liposculpture. The Venezuelan Collaborative Infectious and Tropical Diseases Study Group. Arch Dermatol 2000; 136:1347.
  52. Somoskovi A, Mester J, Hale YM, et al. Laboratory diagnosis of nontuberculous mycobacteria. Clin Chest Med 2002; 23:585.
  53. Keerthirathne TP, Magana-Arachchi DN, Madegedara D, Sooriyapathirana SS. Real time PCR for the rapid identification and drug susceptibility of Mycobacteria present in Bronchial washings. BMC Infect Dis 2016; 16:607.
  54. Marras SAE, Chen L, Shashkina E, et al. A Molecular-Beacon-Based Multiplex Real-Time PCR Assay To Distinguish Mycobacterium abscessus Subspecies and Determine Macrolide Susceptibility. J Clin Microbiol 2021; 59:e0045521.
  55. Bouzinbi N, Marcy O, Bertolotti T, et al. Evaluation of the GenoType NTM-DR assay performance for the identification and molecular detection of antibiotic resistance in Mycobacterium abscessus complex. PLoS One 2020; 15:e0239146.
  56. Butler WR, Ahearn DG, Kilburn JO. High-performance liquid chromatography of mycolic acids as a tool in the identification of Corynebacterium, Nocardia, Rhodococcus, and Mycobacterium species. J Clin Microbiol 1986; 23:182.
  57. Lin CK, Yang YH, Lu ML, et al. Incidence of nontuberculous mycobacterial disease and coinfection with tuberculosis in a tuberculosis-endemic region: A population-based retrospective cohort study. Medicine (Baltimore) 2020; 99:e23775.
  58. Mazurek GH, Jereb J, Vernon A, et al. Updated guidelines for using Interferon Gamma Release Assays to detect Mycobacterium tuberculosis infection - United States, 2010. MMWR Recomm Rep 2010; 59:1.
  59. Pong A, Moser KS, Park SM, et al. Evaluation of an Interferon Gamma Release Assay to Detect Tuberculosis Infection in Children in San Diego, California. J Pediatric Infect Dis Soc 2012; 1:74.
  60. Lindeboom JA, Kuijper EJ, Prins JM, et al. Tuberculin skin testing is useful in the screening for nontuberculous mycobacterial cervicofacial lymphadenitis in children. Clin Infect Dis 2006; 43:1547.
  61. Haimi-Cohen Y, Zeharia A, Mimouni M, et al. Skin indurations in response to tuberculin testing in patients with nontuberculous mycobacterial lymphadenitis. Clin Infect Dis 2001; 33:1786.
  62. Staufner C, Sommerburg O, Holland-Cunz S. Algorithm for early diagnosis in nontuberculous mycobacterial lymphadenitis. Acta Paediatr 2012; 101:e382.
  63. Schier JG, Meiman JG, Layden J, et al. Severe Pulmonary Disease Associated with Electronic-Cigarette-Product Use - Interim Guidance. MMWR Morb Mortal Wkly Rep 2019; 68:787.
  64. Kavaliunaite E, Harris KA, Aurora P, et al. Outcome according to subspecies following lung transplantation in cystic fibrosis pediatric patients infected with Mycobacterium abscessus. Transpl Infect Dis 2020; 22:e13274.
  65. Nahid P, Dorman SE, Alipanah N, et al. Official American Thoracic Society/Centers for Disease Control and Prevention/Infectious Diseases Society of America Clinical Practice Guidelines: Treatment of Drug-Susceptible Tuberculosis. Clin Infect Dis 2016; 63:e147.
  66. Steele RW, Thomas MP, Bégué RE. Compliance issues related to the selection of antibiotic suspensions for children. Pediatr Infect Dis J 2001; 20:1.
  67. Magis-Escurra C, Alffenaar JW, Hoefnagels I, et al. Pharmacokinetic studies in patients with nontuberculous mycobacterial lung infections. Int J Antimicrob Agents 2013; 42:256.
  68. Hafner R, Bethel J, Power M, et al. Tolerance and pharmacokinetic interactions of rifabutin and clarithromycin in human immunodeficiency virus-infected volunteers. Antimicrob Agents Chemother 1998; 42:631.
  69. Waters V, Ratjen F. Antibiotic treatment for nontuberculous mycobacteria lung infection in people with cystic fibrosis. Cochrane Database Syst Rev 2020; 6:CD010004.
  70. Griffith DE, Brown BA, Girard WM, et al. Azithromycin-containing regimens for treatment of Mycobacterium avium complex lung disease. Clin Infect Dis 2001; 32:1547.
  71. Griffith DE, Brown BA, Murphy DT, et al. Initial (6-month) results of three-times-weekly azithromycin in treatment regimens for Mycobacterium avium complex lung disease in human immunodeficiency virus-negative patients. J Infect Dis 1998; 178:121.
  72. Griffith DE, Brown BA, Cegielski P, et al. Early results (at 6 months) with intermittent clarithromycin-including regimens for lung disease due to Mycobacterium avium complex. Clin Infect Dis 2000; 30:288.
  73. Kwak N, Park J, Kim E, et al. Treatment Outcomes of Mycobacterium avium Complex Lung Disease: A Systematic Review and Meta-analysis. Clin Infect Dis 2017; 65:1077.
  74. Peloquin CA, Berning SE, Nitta AT, et al. Aminoglycoside toxicity: daily versus thrice-weekly dosing for treatment of mycobacterial diseases. Clin Infect Dis 2004; 38:1538.
  75. Wallace RJ Jr, Brown BA, Griffith DE, et al. Clarithromycin regimens for pulmonary Mycobacterium avium complex. The first 50 patients. Am J Respir Crit Care Med 1996; 153:1766.
  76. Tanaka E, Kimoto T, Tsuyuguchi K, et al. Effect of clarithromycin regimen for Mycobacterium avium complex pulmonary disease. Am J Respir Crit Care Med 1999; 160:866.
  77. Mougari F, Bouziane F, Crockett F, et al. Selection of Resistance to Clarithromycin in Mycobacterium abscessus Subspecies. Antimicrob Agents Chemother 2017; 61.
  78. Haworth CS, Banks J, Capstick T, et al. British Thoracic Society guidelines for the management of non-tuberculous mycobacterial pulmonary disease (NTM-PD). Thorax 2017; 72:ii1.
  79. Park J, Cho J, Lee CH, et al. Progression and Treatment Outcomes of Lung Disease Caused by Mycobacterium abscessus and Mycobacterium massiliense. Clin Infect Dis 2017; 64:301.
  80. Lamb GS, Starke JR. Mycobacterium abscessus Infections in Children: A Review of Current Literature. J Pediatric Infect Dis Soc 2018; 7:e131.
  81. Koh WJ, Jeong BH, Kim SY, et al. Mycobacterial Characteristics and Treatment Outcomes in Mycobacterium abscessus Lung Disease. Clin Infect Dis 2017; 64:309.
  82. Chacko A, Wen SCH, Hartel G, et al. Improved Clinical Outcome After Treatment of Mycobacterium abscessus Complex Pulmonary Disease in Children With Cystic Fibrosis. Pediatr Infect Dis J 2019; 38:660.
  83. Kurosawa K, Rossenu S, Biewenga J, et al. Population Pharmacokinetic Analysis of Bedaquiline-Clarithromycin for Dose Selection Against Pulmonary Nontuberculous Mycobacteria Based on a Phase 1, Randomized, Pharmacokinetic Study. J Clin Pharmacol 2021; 61:1344.
  84. Gil E, Sweeney N, Barrett V, et al. Bedaquiline as Treatment for Disseminated Nontuberculous Mycobacteria Infection in 2 Patients Co-Infected with HIV. Emerg Infect Dis 2021; 27:944.
  85. Lee J, Ammerman N, Agarwal A, et al. Differential In Vitro Activities of Individual Drugs and Bedaquiline-Rifabutin Combinations against Actively Multiplying and Nutrient-Starved Mycobacterium abscessus. Antimicrob Agents Chemother 2021; 65.
  86. Chew KL, Octavia S, Go J, et al. In vitro susceptibility of Mycobacterium abscessus complex and feasibility of standardizing treatment regimens. J Antimicrob Chemother 2021; 76:973.
  87. D'Ambrosio L, Centis R, Tiberi S, et al. Delamanid and bedaquiline to treat multidrug-resistant and extensively drug-resistant tuberculosis in children: a systematic review. J Thorac Dis 2017; 9:2093.
  88. The Use of Delamanid in the Treatment of Multidrug-Resistant Tuberculosis in Children and Adolescents: Interim Policy Guidance, World Health Organization, Geneva 2016.
  89. Shitrit D, Baum GL, Priess R, et al. Pulmonary Mycobacterium kansasii infection in Israel, 1999-2004: clinical features, drug susceptibility, and outcome. Chest 2006; 129:771.
  90. da Silva Telles MA, Chimara E, Ferrazoli L, Riley LW. Mycobacterium kansasii: antibiotic susceptibility and PCR-restriction analysis of clinical isolates. J Med Microbiol 2005; 54:975.
  91. Guna R, Muñoz C, Domínguez V, et al. In vitro activity of linezolid, clarithromycin and moxifloxacin against clinical isolates of Mycobacterium kansasii. J Antimicrob Chemother 2005; 55:950.
  92. Alcaide F, Calatayud L, Santín M, Martín R. Comparative in vitro activities of linezolid, telithromycin, clarithromycin, levofloxacin, moxifloxacin, and four conventional antimycobacterial drugs against Mycobacterium kansasii. Antimicrob Agents Chemother 2004; 48:4562.
  93. Kim DH, Jhun BW, Moon SM, et al. In Vitro Activity of Bedaquiline and Delamanid against Nontuberculous Mycobacteria, Including Macrolide-Resistant Clinical Isolates. Antimicrob Agents Chemother 2019; 63.
  94. Pezzia W, Raleigh JW, Bailey MC, et al. Treatment of pulmonary disease due to Mycobacterium kansasii: recent experience with rifampin. Rev Infect Dis 1981; 3:1035.
  95. Ahn CH, Lowell JR, Ahn SS, et al. Chemotherapy for pulmonary disease due to Mycobacterium kansasii: efficacies of some individual drugs. Rev Infect Dis 1981; 3:1028.
  96. Ahn CH, Lowell JR, Ahn SS, et al. Short-course chemotherapy for pulmonary disease caused by Mycobacterium kansasii. Am Rev Respir Dis 1983; 128:1048.
  97. Banks J, Hunter AM, Campbell IA, et al. Pulmonary infection with Mycobacterium kansasii in Wales, 1970-9: review of treatment and response. Thorax 1983; 38:271.
  98. Brenner DJ, Hall EJ. Computed tomography--an increasing source of radiation exposure. N Engl J Med 2007; 357:2277.
  99. Hernandez AG, Brunton AE, Ato M, et al. Use of Anti-Glycopeptidolipid-Core Antibodies Serology for Diagnosis and Monitoring of Mycobacterium avium Complex Pulmonary Disease in the United States. Open Forum Infect Dis 2022; 9:ofac528.
  100. De P, Amin AG, Graham B, et al. Urine lipoarabinomannan as a marker for low-risk of NTM infection in the CF airway. J Cyst Fibros 2020; 19:801.
  101. Chesney PJ. Nontuberculous mycobacteria. Pediatr Rev 2002; 23:300.
  102. Bekker A, Schaaf HS, Draper HR, et al. Pharmacokinetics of Rifampin, Isoniazid, Pyrazinamide, and Ethambutol in Infants Dosed According to Revised WHO-Recommended Treatment Guidelines. Antimicrob Agents Chemother 2016; 60:2171.
  103. Ethambutol efficacy and toxicity: Literature review and recommendations for daily and intermittent dosage in children. World Health Organization, 2006. Available at: https://apps.who.int/iris/bitstream/handle/10665/69366/WHO_HTM_TB_2006.365_eng.pdf?sequence=1&isAllowed=y.
  104. World Health Organization. Guidance for national tuberculosis programmes on the management of tuberculosis in children (2014) https://www.who.int/tb/publications/childtb_guidelines/en/ (Accessed on May 29, 2021).
Topic 85894 Version 31.0

References

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