Next Article in Journal
Effect of pH and Carbon Source on Phosphate Solubilization by Bacterial Strains in Pikovskaya Medium
Next Article in Special Issue
Multiomic Investigations into Lung Health and Disease
Previous Article in Journal
Effect of Trehalose and Lactose Treatments on the Freeze-Drying Resistance of Lactic Acid Bacteria in High-Density Culture
Previous Article in Special Issue
Gut Dysbiosis in Children with Cystic Fibrosis: Development, Features and the Role of Gut–Lung Axis on Disease Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnosis and Management of Pulmonary NTM with a Focus on Mycobacterium avium Complex and Mycobacterium abscessus: Challenges and Prospects

Department of Internal Medicine, Saint Louis University, St. Louis, MO 63104, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(1), 47; https://doi.org/10.3390/microorganisms11010047
Submission received: 3 October 2022 / Revised: 15 December 2022 / Accepted: 20 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Role of Microbiome in Lung Health)

Abstract

:
Background: Nontuberculous mycobacteria (NTM) are ubiquitous. NTM can affect different organs and may cause disseminated diseases, but the pulmonary form is the most common form. Pulmonary NTM is commonly seen in patients with underlying diseases. Pulmonary Mycobacterium avium complex (MAC) is the most common NTM disease and M. abscessus (MAB) is the most challenging to treat. This review is prepared with the following objectives: (a) to evaluate new methods available for the diagnosis of pulmonary MAC or MAB, (b) to assess advances in developing new therapeutics and their impact on treatment of pulmonary MAC or MAB, and (c) to evaluate the prospects of preventive strategies including vaccines against pulmonary MAC or MAB. Methods: A literature search was conducted using PubMed/MEDLINE and multiple search terms. The search was restricted to the English language and human studies. The database query resulted in a total of 197 publications. After the title and abstract review, 64 articles were included in this analysis. Results: The guidelines by the American Thoracic Society (ATS), European Respiratory Society (ERS), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), and Infectious Diseases Society of America (IDSA) are widely applicable. The guidelines are based on expert opinion and there may be a need to broaden criteria to include those with underlying lung diseases who may not fulfill some of the criteria as ‘probable cases’ for better follow up and management. Some cases with only one culture-positive sputum sample or suggestive histology without a positive culture may benefit from new methods of confirming NTM infection. Amikacin liposomal inhalation suspension (ALIS), gallium containing compounds and immunotherapies will have potential in the management of pulmonary MAC and MAB. Conclusions: the prevalence of pulmonary NTM is increasing. The efforts to optimize diagnosis and treatment of pulmonary NTM are encouraging. There is still a need to develop new diagnostics and therapeutics.
Keywords:
NTM; MAC; MAB; Mycobacterium

1. Background

Nontuberculous mycobacteria (NTM) are vast and include all mycobacteria except M. tuberculosis (Mtb) and M. leprae. Nontuberculous mycobacteria (NTM) have a lipid-rich outer membrane generating hydrophobicity and can produce biofilms, thus allowing a wide variety of hosts within nature [1].
NTM are ubiquitous in nature. Currently, over 190 species of nontuberculous mycobacteria (NTM) have been identified [2]. It is believed that NTM infections are acquired from the environment via inhalation, ingestion and skin contact, which may result in pulmonary disease, lymphadenitis, skin and soft tissue infections, or disseminated disease. Pulmonary disease is the most common form of NTM disease in patients who are negative for human immunodeficiency virus (HIV) [3]. Pulmonary NTM commonly occurs in patients with underlying lung diseases [4,5]. Disseminated diseases are commonly seen in patients who are immunocompromised including HIV, immunosuppressive medications, and genetic defects in Th1 immune responses [6].
In developed countries where tuberculosis (TB) is well controlled, the prevalence of pulmonary NTM and the mortality rate associated with NTM infections are increasing [3,7,8,9,10,11]. A US study of Medicare Part B beneficiaries showed that the prevalence of NTM increased from 20 to 47 per 100,000 persons between 1997 and 2007, an 8.2% increase per year [7]. A more recent report estimated that the number of pulmonary NTM cases in the US increased by at least two-fold between 2010 and 2014 [12]. Data on NTM isolates from pulmonary samples obtained from 30 countries across six continents in 2008 showed that M. avium complex (MAC) constituted 37% of isolates in Europe, 52% of in North America and 71% in Australia [13]. In North America, recent data suggest that MAC and M. abscessus (MAB) are common causes of pulmonary NTM [14]. Pulmonary MAB is associated with treatment failure rates exceeding 50%, a rapid decline in lung function, significant morbidity and mortality [15,16]. Therefore, this review focuses on pulmonary MAC and MAB, and is prepared with the following objectives: (a) evaluate new methods available for the diagnosis of pulmonary MAC or MAB, (b) assess advances in developing new therapeutics and their impact on treatment of pulmonary MAC or MAB, and (c) evaluate the prospects of preventive strategies including vaccines against pulmonary MAC or MAB.

2. Methods

A literature search was conducted using PubMed/MEDLINE and multiple search terms. The search included publication dates 1 January 1980–16 December 2022 and was restricted to the English language and human studies. A search term ‘pulmonary NTM and new methods’ revealed 116 publications, pulmonary NTM and new drug 136, pulmonary NTM and immunotherapy 13, pulmonary NTM and vaccine 46 publications. The database query resulted in a total of 197 publications. After the title and abstract review, 64 articles were included in this analysis.

2.1. Risk Factors of Pulmonary NTM

Patients with no identifiable structural lung diseases may develop pulmonary NTM but pulmonary NTM usually occurs in patients with underlying diseases [17,18,19,20]. Cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) are common risk factors for pulmonary NTM. Other risk factors include old age, non-CF bronchiectasis, interstitial lung disease (ILD), and immunodeficiency [6,21,22].
CF is the most common genetic disorder in the US and Europe with incidence of about 1 in 3200 in Caucasians [23]. The incidence is much lower in other races. In the US, CF occurs in approximately 1:15,000 blacks, 1:35,000 individuals of Asian descent, and 1:10,900 Native Americans [24,25]. In US, there are more than 30,000 individuals with CF [26]. The number of adults with CF continues to increase, while the number of children remains relatively stable [26], because advances in medical care have prolonged life in these patients. With increases in survival the spectrum of life-threatening pulmonary infections in CF has changed. An extensive literature indicates that pulmonary NTM infections are becoming common in CF, with prevalence increasing from 10% in children aged 10 years to more than 30% in adults over 40 years [5]. Pulmonary NTM increases the rate of exacerbations in CF threefold [27], and accelerates deterioration of lung function [28,29,30] more than other serious CF pulmonary infections such as Pseudomonas and Burkholderia [29]. In addition, pre-transplant pulmonary NTM in CF patients makes post-lung transplant recovery more challenging [31]. It is likely that susceptibility to pulmonary NTM in CF is at least partly due to viscous secretions, deceased mucocilliary clearance, mucus tethering, and impaired innate immunity [32,33,34,35]. CF transmembrane conductance regulator (CFTR) gene modulator therapies have decreased rates of new or recurrent Pseudomonas lung infection and improved quality of life [36]. To our knowledge, there are no studies on the impact of CFTR modulator on pulmonary NTM. In fact, the prevalence of pulmonary NTM has continued to increase even after the wider use of CFTR modulators in the CF population [36]. CFTR modulators are drugs that act by improving production, intracellular processing, and/or function of the defective CFTR protein. The management may include two correctors and a potentiator (e.g., elexacaftor-tezacaftor-ivacaftor) to restore the function of the mutant CFTR protein more fully [37]. CFTR modulators may prevent lung damage or improve lung function but there is no evidence supporting reversal of already damaged bronchi, suggesting that CF patients with structural damage will continue to be high risk for pulmonary NTM. NTM infection significantly decreases lung function [30].
Chronic obstructive pulmonary disease (COPD) is another common risk factor for pulmonary NTM. The prevalence of (COPD) in the US adult population ranges from 5.1 to 14% [38,39,40,41]. Similar prevalence rates were reported from Canada and Europe with estimated global prevalence of 13% [42,43]. The prevalence of pNTM in COPD patients is about 0.7% with a hazard ratio of 15.5 for patients older than 35 years, with a 10× higher hazard ratio for patients ≥ 65 years old [4]. Pulmonary NTM doubles the rate of severe exacerbations requiring hospital admissions and leads to a significant deterioration in lung function [44]. In a population-based study of more than 6 million people, the fully adjusted hazard ratio for pulmonary NTM was significantly high in COPD (8.7, 95% CI 8.3–9.2) with incidence rate of 143 per 100,000 person-years compared to 6.6 per 100,000 person-years in a group without COPD [4].

2.2. Diagnosis of Pulmonary NTM

The diagnosis of pulmonary NTM relies on clinical, microbiologic, and radiologic criteria. Based on the guidelines by American Thoracic Society (ATS), European Respiratory Society (ERS), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), and Infectious Diseases Society of America (IDSA), all of the following criteria have to be met for the diagnosis of pulmonary NTM [45,46]:
  • New or worsening pulmonary symptoms with or without systemic symptoms.
  • New or worsening radiologic findings suggestive of nodular, cavitary opacities on chest X-ray or bronchiectasis with nodules on computed tomography (CT).
  • Exclusion of other diagnosis.
  • Supportive microbiologic findings including (i) cultures of at least two separate sputum samples positive for NTM, (ii) culture of bronchial wash or lavage positive for NTM or (iii) lung histology showing granulomatous inflammation or acid-fast bacilli (AFB) and at least one positive NTM culture from biopsy or another respiratory specimen.
Productive cough and shortness of breath are the main presenting symptoms of pulmonary NTM, and some patients may have fatigue, fever, chest pain and weight loss [47,48]. Figure 1 shows typical radiologic findings of nodular and fibrocavitary pulmonary MAC
The ATS/ERS/ESCMID/IDSA criteria are based on expert opinion, not on high quality studies. Therefore, it may not be surprising to see some patients with underlying lung disease treated for pulmonary NTM even when they do not fulfill microbiologic (e.g., having only one culture positive sputum culture) or radiologic criteria (e.g., abnormal imaging study findings but not nodular, fibrocavitary or bronchiectasis) [48,49,50,51]. This may suggest the need to broaden the criteria for pulmonary disease caused by some of the NTMs. A retrospective study on 53 patients with and 19 without underlying lung diseases recommended the definition of definite pulmonary MAC for those who fulfill the ATS/ERS/ESCMID/IDSA criteria and probable pulmonary MAC for those with a single culture positive sputum or radiologic abnormalities other than nodular bronchiectasis or fibrocavitation [49]. The suggestion for classifying patients with pulmonary MAC into definite and probable groups must be tested on a large number of MAC patients with known treatment outcomes.

2.3. Clinical Relevance of Drug Susceptibility Testing

Macrolides, rifampin, ethambutol, and aminoglycosides are drugs used for treatment of pulmonary MAC. The first three drugs are used for all patients and an aminoglycoside is added for patients with fibrocavitary lesions [46]. In vitro susceptibility is not commonly done for new patients and only in vitro susceptibility for clarithromycin result has a good correlation with clinical efficacy [52]. Despite lack of correlation between in vitro susceptibility to some of the MAC drugs and clinical efficacy, it has been shown that high MIC particularly ≥8 mg/L for rifampin and ethambutol or ≥64 mg/L for aminoglycosides is associated with treatment failure [53,54]. In vitro testing using macrophages infected with MAC isolates from patients with disseminated MAC showed some promising results [55]. A decrease in colony forming units by at least one log was predictive of clinical efficacy of some of the drugs used for MAC [55].
The optimal drugs for treatment of pulmonary MAB are not known [46]. However, in one study that used combination regimen, treatment success was associated with in vitro susceptibility to clarithromycin but not with ciprofloxacin, doxycycline, cefoxitin or amikacin [56].

2.4. Treatment of Pulmonary MAC and MAB

In patients with pulmonary MAC, a susceptibility-based treatment for macrolides and amikacin is recommended by the ATS/ERS/ESCMID/IDSA guidelines, and it is suggested that patients with macrolide-susceptible MAC receive a treatment regimen with at least three drugs (including a macrolide and ethambutol) for at least 12 months after culture conversion [46]. For patients with cavitary, advanced/severe bronchiectasis or macrolide-resistant pulmonary MAC addition of amikacin or streptomycin is suggested [46]. The cure rates of treatment for pulmonary MAC ranges from 55% to 66% [48,57,58].
The optimal drugs, regimens, and duration of therapy for pulmonary MAB are not well defined. However, a susceptibility-based treatment for macrolides and amikacin is recommended [46]. Studies, mainly from South Korea, have shown that clarithromycin-containing oral regimen with 4–16 weeks of one or two intravenous drugs showed clinical improvement in 50–97% of patients [56,59,60]. In a retrospective study done on 65 patients in South Korea, efficacy of a combination regimen including clarithromycin, ciprofloxacin and doxycycline three-drug treatment with an initial 4-week course of intravenous cefoxitin and amikacin resulted in clinical improvement in 83% of patients, radiologic improvement in 74% and microbiologic cure in 58% of patients [56]. In this study, treatment for pulmonary MAB continued for at least 12 months after sputum culture conversion or a total duration of 24 months.

2.5. Prospects in Improving Diagnosis and Treatment

Microbiologic criteria for the diagnosis of pulmonary MAC and MAB include culture of respiratory specimen and histology [46]. In patients with only a single positive sputum culture or histology showing granuloma with neg cultures, new methods of detecting NTM infection may help in diagnosis. A dual skin test with MAC sensitin (MAS) and with Mtb purified protein derivative (PPD) was used for identification of patients with pulmonary MAC. MAS skin test with induration size of ≥5 mm larger than induration from PPD had a specificity of 97% for discriminating pulmonary MAC and TB [61].
Use of a simple and stable antigen for use in serodiagnosis has been explored. Glycopeptidolipids (GPL) have been the main focus because of their abundance in NTM isolates [62]. MAC strains produce highly antigenic and typeable serovar-specific GPLs. Observational study of anti-GPL on samples from 369 patients with rheumatoid arthritis, 10 with and 359 without pulmonary MAC showed a positive predictive value of 67% and a negative predictive value of 97%. Pulmonary MAC patients in this study fulfilled the criteria [63]. Using GPL from 11 reference strains of MAC in pulmonary MAC patients without rheumatoid arthritis, positive predictive values of 80% for IgG and 89% for IgA with corresponding negative predictive values of 89% and 97% [64].
Patients with cavitary, advanced/severe bronchiectasis or macrolide-resistant pulmonary MAC benefit from adding aminoglycosides as part of a treatment regimen [46]. In addition, aminoglycosdies are among the drugs to be considered in other pulmonary MAC patients who fail treatment. Unfortunately, use of intravenous or intramuscular aminoglycosides for extended duration is limited by serious side effects [43]. In the last few years, amikacin liposomal inhalation suspension (ALIS) has been shown to improve culture conversion in treatment refractory cases of pulmonary MAC [54,65,66]. The new guidelines now recommend the addition of ALIS in patients with pulmonary MAC pulmonary who have failed therapy after at least six months of guideline-based therapy [46].
Another approach to improve treatment outcome is finding ways to optimize the use of existing drugs. A multicenter study on efficacy of a single tablet with fixed dose combinations (FDC) of clarithromycin, rifabutin and clofazimine for treatment of pulmonary MAC with nodular bronchiectasis is underway [67]. FDC may prevent emergence of drug resistance but may not prevent treatment default associated with drug side effects. In a recent multicenter study of 297 pulmonary NTM patients, 90 (30.3%) required change in treatment because of drug side effects [48]. Therefore, simplifying treatment regimens to minimize the number of drugs helps improve treatment compliance. In a preliminary open-label study, a two-drug combination with clarithromycin and ethambutol was shown to be noninferior to a three-drug regimen with addition of rifampin [68]. This has the potential to decrease drug side effects and challenges arising from drug interactions. Currently a large randomized controlled trial is undergoing recruitment of 500 participants to study the benefit of two versus three-antibiotic therapy for MAC disease [69].
There are efforts to develop new drugs for MAC and MAB [70,71]. Part of this effort relies on testing new drugs developed for TB treatment [70,71]. There are encouraging results from studies that use cell cultures and small animal but not many have made it to human trials [72]. Potential candidates include gallium containing compounds. In mice, compounds containing gallium which interferes with iron metabolism or uptake by mycobacteria have been shown to have activity against pulmonary MAB [73]. A phase 1b study on intravenous gallium nitrate in CF colonized with NTM is underway [74].
A unique approach that showed some promising results is immunotherapy. In a small placebo-controlled intramuscular IFN-γ daily for one month and then three times per week for up to 6 months as adjunct to guideline-based therapy, a more rapid clinical and radiological responses were seen in patients who received IFN-γ [75]. Nitric oxide (NO), an important part of the innate immune system with bactericidal activities, has been tried for treatment of pulmonary MAB [76,77]. The use of inhaled NO by Yaacoby-Bianu et al. in 2018 in two cystic fibrosis patients with pulmonary MAB resulted in a reduction in bacterial load as measured by quantitative polymerase chain [78]. The use of inhaled NO by Goldbart et al. in a CF patient with pulmonary MAB showed bacterial growth inhibition as well as improvement of lung pathology on CT [79]. The observations from these case reports show that NO is safe and well tolerated and could play a crucial role in the treatment of NTM pulmonary disease [71,78,79]. Additional trials are investigating treatment outcomes with intermittent inhaled nitric oxide (NO) in cystic fibrosis versus non-cystic fibrosis patients, both with pulmonary MAB [80]. Another important immunomodulating therapy that attracted some interest is granulocyte-macrophage colony-stimulating factor (GM-CSF). GM-CSF is produced by different types of cells including T cells, macrophages and alveolar epithelial cells, and is known to increase the ability of macrophages to control mycobacterial infection [81]. Inhaled GM-CSF was tested in patients who failed MAC or MAB antibiotic treatment and helped achieve culture conversion in 5/32 (15.6%) but the culture conversion was durable after end of guideline-based therapy only in two patients [82]. There is still an interest to carefully examine the benefits of GM-CSF in treatment refractory pulmonary NTM cases, particularly MAB [83].
Immunotherapy with BCG has been considered. BCG, the only licensed mycobacterial vaccine, live attenuated Mycobacterium bovis is used globally for the prevention of pulmonary TB. BCG, is known to stimulate innate immunity [84,85,86] and induces adaptive immune responses [87]. Long term follow-up studies have demonstrated that BCG provides durable protection against pulmonary TB as summarized in two recent reviews [88,89]. BCG has also been shown to be effective in preventing infections due to other mycobacteria including M. leprae, M ulcerans, and strains of M. avium causing NTM lymphadenitis in children, indicating cross-protective immunity within the genus [90,91,92]. Our recent study showed that BCG induces cross-protective immunity to NTM [93]. The use of BCG as adjunct immunotherapy or prevention of pulmonary MAC and MAB is attractive. One potential advantage of whole cell vaccines over protein-adjuvant formulations and viral-vectored constructs is their broad antigen composition, which includes the complete protein repertoire, lipids, carbohydrates, and other moieties that may be antigenic and induce donor unrestricted T-cell responses, B-cell responses, and possibly also natural killer and innate lymphoid cell responses [94]. However, most patients with pulmonary MAC and MAB have underlying lung diseases and therefore, the use of BCG has a potential of causing serious BCG lung disease.

3. Conclusions

The prevalence of pulmonary NTM is increasing. The exact reasons for this increase in prevalence are not known, but medical advances to increase the lifespan of patients with underlying lung diseases could be one factor. Underlying lung diseases such as CF and COPD are major risk factors for pulmonary MAC and MAB. ATS/ERS/ESCMID/IDSA guidelines include recommendations for diagnosis and treatment of pulmonary MAC and MAB. Despite the guidelines, diagnosis of pulmonary MAC and MAB is challenging. Similarly, treatment of pulmonary MAC and MAB is complex with high failure rates. There is a need to improve the diagnosis and treatment of patients with pulmonary MAC and MAB. Efforts to simplify the treatment regimen for most cases of pulmonary MAC and findings new therapeutics are encouraging. Two-drug antibiotic regimens, inhaled amikacin, and immunotherapies have potential for the management of pulmonary NTM.

Author Contributions

C.H. and M.M. contributed to the preparation of most sections of the draft manuscript. R.H. provided typical imaging findings after consent was obtained from 2 patients and revised manuscripts. G.A. prepared an outline for manuscript preparation, performed a critical review and provided significant revisions to all sections of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Data Availability Statement

This review did not include data.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jeon, D. Infection Source and Epidemiology of Nontuberculous Mycobacterial Lung Disease. Tuberc. Respir. Dis. (Seoul) 2019, 82, 94–101. [Google Scholar] [CrossRef] [PubMed]
  2. Forbes, B.A. Mycobacterial Taxonomy. J. Clin. Microbiol. 2017, 55, 380–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Cassidy, P.M.; Hedberg, K.; Saulson, A.; McNelly, E.; Winthrop, K.L. Nontuberculous mycobacterial disease prevalence and risk factors: A changing epidemiology. Clin. Infect. Dis. 2009, 49, e124–e129. [Google Scholar] [CrossRef] [PubMed]
  4. Marras, T.K.; Campitelli, M.A.; Kwong, J.C.; Lu, H.; Brode, S.K.; Marchand-Austin, A.; Gershon, A.S.; Jamieson, F.B. Risk of nontuberculous mycobacterial pulmonary disease with obstructive lung disease. Eur. Respir. J. 2016, 48, 928–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rodman, D.M.; Polis, J.M.; Heltshe, S.L.; Sontag, M.K.; Chacon, C.; Rodman, R.V.; Brayshaw, S.J.; Huitt, G.A.; Iseman, M.D.; Saavedra, M.T.; et al. Late diagnosis defines a unique population of long-term survivors of cystic fibrosis. Am. J. Respir. Crit. Care Med. 2005, 171, 621–626. [Google Scholar] [CrossRef]
  6. Honda, J.R.; Knight, V.; Chan, E.D. Pathogenesis and risk factors for nontuberculous mycobacterial lung disease. Clin. Chest Med. 2015, 36, 1–11. [Google Scholar] [CrossRef]
  7. Adjemian, J.; Olivier, K.N.; Seitz, A.E.; Holland, S.M.; Prevots, D.R. Prevalence of nontuberculous mycobacterial lung disease in U.S. Medicare beneficiaries. Am. J. Respir. Crit. Care Med. 2012, 185, 881–886. [Google Scholar] [CrossRef] [Green Version]
  8. Mirsaeidi, M.; Machado, R.F.; Garcia, J.G.; Schraufnagel, D.E. Nontuberculous mycobacterial disease mortality in the United States, 1999–2010: A population-based comparative study. PLoS ONE 2014, 9, e91879. [Google Scholar] [CrossRef]
  9. Ringshausen, F.C.; Wagner, D.; de Roux, A.; Diel, R.; Hohmann, D.; Hickstein, L.; Welte, T.; Rademacher, J. Prevalence of Nontuberculous Mycobacterial Pulmonary Disease, Germany, 2009–2014. Emerg. Infect. Dis. 2016, 22, 1102–1105. [Google Scholar] [CrossRef]
  10. Marras, T.K.; Mendelson, D.; Marchand-Austin, A.; May, K.; Jamieson, F.B. Pulmonary nontuberculous mycobacterial disease, Ontario, Canada, 1998-2010. Emerg. Infect. Dis. 2013, 19, 1889–1891. [Google Scholar] [CrossRef]
  11. Brode, S.K.; Daley, C.L.; Marras, T.K. The epidemiologic relationship between tuberculosis and non-tuberculous mycobacterial disease: A systematic review. Int. J. Tuberc. Lung Dis. 2014, 18, 1370–1377. [Google Scholar] [CrossRef]
  12. Strollo, S.E.; Adjemian, J.; Adjemian, M.K.; Prevots, D.R. The Burden of Pulmonary Nontuberculous Mycobacterial Disease in the United States. Ann. Am. Thorac. Soc. 2015, 12, 1458–1464. [Google Scholar] [CrossRef] [Green Version]
  13. Hoefsloot, W.; van Ingen, J.; Andrejak, C.; Angeby, K.; Bauriaud, R.; Bemer, P.; Beylis, N.; Boeree, M.J.; Cacho, J.; Chihota, V.; et al. The geographic diversity of nontuberculous mycobacteria isolated from pulmonary samples: An NTM-NET collaborative study. Eur. Respir. J. 2013, 42, 1604–1613. [Google Scholar] [CrossRef]
  14. Johnson, M.M.; Odell, J.A. Nontuberculous mycobacterial pulmonary infections. J. Thorac. Dis. 2014, 6, 210–220. [Google Scholar] [CrossRef]
  15. Diel, R.; Ringshausen, F.; Richter, E.; Welker, L.; Schmitz, J.; Nienhaus, A. Microbiological and Clinical Outcomes of Treating Non-Mycobacterium Avium Complex Nontuberculous Mycobacterial Pulmonary Disease: A Systematic Review and Meta-Analysis. Chest 2017, 152, 120–142. [Google Scholar] [CrossRef]
  16. Pasipanodya, J.G.; Ogbonna, D.; Ferro, B.E.; Magombedze, G.; Srivastava, S.; Deshpande, D.; Gumbo, T. Systematic Review and Meta-analyses of the Effect of Chemotherapy on Pulmonary Mycobacterium abscessus Outcomes and Disease Recurrence. Antimicrob. Agents Chemother. 2017, 61, e01206-17. [Google Scholar] [CrossRef] [Green Version]
  17. Reich, J.M.; Johnson, R.E. Mycobacterium avium complex pulmonary disease presenting as an isolated lingular or middle lobe pattern. The Lady Windermere syndrome. Chest 1992, 101, 1605–1609. [Google Scholar] [CrossRef] [Green Version]
  18. Prince, D.S.; Peterson, D.D.; Steiner, R.M.; Gottlieb, J.E.; Scott, R.; Israel, H.L.; Figueroa, W.G.; Fish, J.E. Infection with Mycobacterium avium complex in patients without predisposing conditions. N. Engl. J. Med. 1989, 321, 863–868. [Google Scholar] [CrossRef]
  19. Kim, R.D.; Greenberg, D.E.; Ehrmantraut, M.E.; Guide, S.V.; Ding, L.; Shea, Y.; Brown, M.R.; Chernick, M.; Steagall, W.K.; Glasgow, C.G.; et al. Pulmonary nontuberculous mycobacterial disease: Prospective study of a distinct preexisting syndrome. Am. J. Respir. Crit. Care Med. 2008, 178, 1066–1074. [Google Scholar] [CrossRef] [Green Version]
  20. Iseman, M.D.; Buschman, D.L.; Ackerson, L.M. Pectus excavatum and scoliosis. Thoracic anomalies associated with pulmonary disease caused by Mycobacterium avium complex. Am. Rev. Respir. Dis. 1991, 144, 914–916. [Google Scholar] [CrossRef]
  21. Donohue, M.J.; Wymer, L. Increasing Prevalence Rate of Nontuberculous Mycobacteria Infections in Five States, 2008–2013. Ann. Am. Thorac. Soc. 2016, 13, 2143–2150. [Google Scholar] [CrossRef] [PubMed]
  22. Chan, E.D.; Iseman, M.D. Underlying host risk factors for nontuberculous mycobacterial lung disease. Semin. Respir. Crit. Care Med. 2013, 34, 110–123. [Google Scholar] [CrossRef] [PubMed]
  23. Sontag, M.K.; Hammond, K.B.; Zielenski, J.; Wagener, J.S.; Accurso, F.J. Two-tiered immunoreactive trypsinogen-based newborn screening for cystic fibrosis in Colorado: Screening efficacy and diagnostic outcomes. J. Pediatr. 2005, 147, S83–S88. [Google Scholar] [CrossRef] [PubMed]
  24. Palomaki, G.E.; FitzSimmons, S.C.; Haddow, J.E. Clinical sensitivity of prenatal screening for cystic fibrosis via CFTR carrier testing in a United States panethnic population. Genet. Med. 2004, 6, 405–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Hamosh, A.; FitzSimmons, S.C.; Macek, M., Jr.; Knowles, M.R.; Rosenstein, B.J.; Cutting, G.R. Comparison of the clinical manifestations of cystic fibrosis in black and white patients. J. Pediatr. 1998, 132, 255–259. [Google Scholar] [CrossRef]
  26. Cystic Fibrosis Foundation, Bethesda, MA, USA. 2016 Patient Registry: Annual Data Report. 2017. Available online: chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/https://www.cff.org/sites/default/files/2021-11/Patient-Registry-Annual-Data-Report.pdf (accessed on 3 October 2022).
  27. Low, D.; Wilson, D.A.; Flume, P.A. Screening practices for nontuberculous mycobacteria at us cystic fibrosis centers. J. Cyst. Fibros. 2020, 19, 59–574. [Google Scholar] [CrossRef] [Green Version]
  28. Esther, C.R., Jr.; Esserman, D.A.; Gilligan, P.; Kerr, A.; Noone, P.G. Chronic Mycobacterium abscessus infection and lung function decline in cystic fibrosis. J. Cyst. Fibros. 2010, 9, 117–123. [Google Scholar] [CrossRef] [Green Version]
  29. Qvist, T.; Taylor-Robinson, D.; Waldmann, E.; Olesen, H.V.; Hansen, C.R.; Mathiesen, I.H.; Hoiby, N.; Katzenstein, T.L.; Smyth, R.L.; Diggle, P.J.; et al. Comparing the harmful effects of nontuberculous mycobacteria and Gram negative bacteria on lung function in patients with cystic fibrosis. J. Cyst. Fibros. 2016, 15, 380–385. [Google Scholar] [CrossRef] [Green Version]
  30. Eikani, M.S.; Nugent, M.; Poursina, A.; Simpson, P.; Levy, H. Clinical course and significance of nontuberculous mycobacteria and its subtypes in cystic fibrosis. BMC Infect. Dis. 2018, 18, 311. [Google Scholar] [CrossRef] [Green Version]
  31. Chalermskulrat, W.; Sood, N.; Neuringer, I.P.; Hecker, T.M.; Chang, L.; Rivera, M.P.; Paradowski, L.J.; Aris, R.M. Non-tuberculous mycobacteria in end stage cystic fibrosis: Implications for lung transplantation. Thorax 2006, 61, 507–513. [Google Scholar] [CrossRef]
  32. Elborn, J.S. Cystic fibrosis. Lancet 2016, 388, 2519–2531. [Google Scholar] [CrossRef]
  33. Bernardi, D.M.; Ribeiro, A.F.; Mazzola, T.N.; Vilela, M.M.; Sgarbieri, V.C. The impact of cystic fibrosis on the immunologic profile of pediatric patients. J. Pediatr. (Rio. J.) 2013, 89, 40–47. [Google Scholar] [CrossRef] [Green Version]
  34. Bessich, J.L.; Nymon, A.B.; Moulton, L.A.; Dorman, D.; Ashare, A. Low levels of insulin-like growth factor-1 contribute to alveolar macrophage dysfunction in cystic fibrosis. J. Immunol. 2013, 191, 378–385. [Google Scholar] [CrossRef] [Green Version]
  35. Mueller, C.; Braag, S.A.; Keeler, A.; Hodges, C.; Drumm, M.; Flotte, T.R. Lack of cystic fibrosis transmembrane conductance regulator in CD3+ lymphocytes leads to aberrant cytokine secretion and hyperinflammatory adaptive immune responses. Am. J. Respir. Cell Mol. Biol. 2011, 44, 922–929. [Google Scholar] [CrossRef] [Green Version]
  36. Saiman, L. Improving outcomes of infections in cystic fibrosis in the era of CFTR modulator therapy. Pediatr. Pulmonol. 2019, 54 (Suppl. S3), S18–S26. [Google Scholar] [CrossRef] [Green Version]
  37. Goetz, D.M.; Savant, A.P. Review of CFTR modulators 2020. Pediatr. Pulmonol. 2021, 56, 3595–3606. [Google Scholar] [CrossRef]
  38. Pyarali, F.F.; Schweitzer, M.; Bagley, V.; Salamo, O.; Guerrero, A.; Sharifi, A.; Campos, M.; Quartin, A.; Mirsaeidi, M. Increasing Non-tuberculous Mycobacteria Infections in Veterans With COPD and Association With Increased Risk of Mortality. Front. Med. (Lausanne) 2018, 5, 311. [Google Scholar] [CrossRef]
  39. Akinbami, L.J.; Liu, X. Chronic obstructive pulmonary disease among adults aged 18 and over in the United States, 1998–2009. NCHS Data Brief. 2011, 63, 1–8. [Google Scholar]
  40. Centers for Disease Control and Prevention. Chronic obstructive pulmonary disease among adults--United States, 2011. MMWR Morb. Mortal. Wkly. Rep. 2012, 61, 938–943. [Google Scholar]
  41. Thompson, W.H.; St-Hilaire, S. Prevalence of chronic obstructive pulmonary disease and tobacco use in veterans at Boise Veterans Affairs Medical Center. Respir. Care 2010, 55, 555–560. [Google Scholar]
  42. Leung, C.; Bourbeau, J.; Sin, D.D.; Aaron, S.D.; FitzGerald, J.M.; Maltais, F.; Marciniuk, D.D.; O′Donnell, D.; Hernandez, P.; Chapman, K.R.; et al. The Prevalence of Chronic Obstructive Pulmonary Disease (COPD) and the Heterogeneity of Risk Factors in the Canadian Population: Results from the Canadian Obstructive Lung Disease (COLD) Study. Int. J. Chron. Obstruct. Pulmon. Dis. 2021, 16, 305–320. [Google Scholar] [CrossRef] [PubMed]
  43. Peloquin, C.A.; Berning, S.E.; Nitta, A.T.; Simone, P.M.; Goble, M.; Huitt, G.A.; Iseman, M.D.; Cook, J.L.; Curran-Everett, D. Aminoglycoside toxicity: Daily versus thrice-weekly dosing for treatment of mycobacterial diseases. Clin. Infect. Dis. 2004, 38, 1538–1544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Huang, C.T.; Tsai, Y.J.; Wu, H.D.; Wang, J.Y.; Yu, C.J.; Lee, L.N.; Yang, P.C. Impact of non-tuberculous mycobacteria on pulmonary function decline in chronic obstructive pulmonary disease. Int. J. Tuberc. Lung Dis. 2012, 16, 539–545. [Google Scholar] [CrossRef] [PubMed]
  45. Griffith, D.E.; Aksamit, T.; Brown-Elliott, B.A.; Catanzaro, A.; Daley, C.; Gordin, F.; Holland, S.M.; Horsburgh, R.; Huitt, G.; Iademarco, M.F.; et al. An official ATS/IDSA statement: Diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am. J. Respir. Crit. Care Med. 2007, 175, 367–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Daley, C.L.; Iaccarino, J.M.; Lange, C.; Cambau, E.; Wallace, R.J., Jr.; Andrejak, C.; Bottger, E.C.; Brozek, J.; Griffith, D.E.; Guglielmetti, L.; et al. Treatment of nontuberculous mycobacterial pulmonary disease: An official ATS/ERS/ESCMID/IDSA clinical practice guideline. Eur. Respir. J. 2020, 56, 2000535. [Google Scholar] [CrossRef]
  47. Hwang, J.A.; Kim, S.; Jo, K.W.; Shim, T.S. Natural history of Mycobacterium avium complex lung disease in untreated patients with stable course. Eur. Respir. J. 2017, 49, 1600537. [Google Scholar] [CrossRef] [Green Version]
  48. Abate, G.; Stapleton, J.T.; Rouphael, N.; Creech, B.; Stout, J.E.; El Sahly, H.M.; Jackson, L.; Leyva, F.J.; Tomashek, K.M.; Tibbals, M.; et al. Variability in the Management of Adults With Pulmonary Nontuberculous Mycobacterial Disease. Clin. Infect. Dis. 2021, 72, 1127–1137. [Google Scholar] [CrossRef]
  49. Plotinsky, R.N.; Talbot, E.A.; von Reyn, C.F. Proposed definitions for epidemiologic and clinical studies of Mycobacterium avium complex pulmonary disease. PLoS ONE 2013, 8, e77385. [Google Scholar] [CrossRef]
  50. Prevots, D.R.; Shaw, P.A.; Strickland, D.; Jackson, L.A.; Raebel, M.A.; Blosky, M.A.; Montes de Oca, R.; Shea, Y.R.; Seitz, A.E.; Holland, S.M.; et al. Nontuberculous mycobacterial lung disease prevalence at four integrated health care delivery systems. Am. J. Respir. Crit. Care Med. 2010, 182, 970–976. [Google Scholar] [CrossRef] [Green Version]
  51. Winthrop, K.L.; McNelley, E.; Kendall, B.; Marshall-Olson, A.; Morris, C.; Cassidy, M.; Saulson, A.; Hedberg, K. Pulmonary nontuberculous mycobacterial disease prevalence and clinical features: An emerging public health disease. Am. J. Respir. Crit. Care Med. 2010, 182, 977–982. [Google Scholar] [CrossRef] [Green Version]
  52. Kobashi, Y.; Yoshida, K.; Miyashita, N.; Niki, Y.; Oka, M. Relationship between clinical efficacy of treatment of pulmonary Mycobacterium avium complex disease and drug-sensitivity testing of Mycobacterium avium complex isolates. J. Infect. Chemother. 2006, 12, 195–202. [Google Scholar] [CrossRef]
  53. Kwon, B.S.; Kim, M.N.; Sung, H.; Koh, Y.; Kim, W.S.; Song, J.W.; Oh, Y.M.; Lee, S.D.; Lee, S.W.; Lee, J.S.; et al. In Vitro MIC Values of Rifampin and Ethambutol and Treatment Outcome in Mycobacterium avium Complex Lung Disease. Antimicrob. Agents Chemother. 2018, 62, e00491-18. [Google Scholar] [CrossRef] [Green Version]
  54. Olivier, K.N.; Griffith, D.E.; Eagle, G.; McGinnis, J.P., 2nd; Micioni, L.; Liu, K.; Daley, C.L.; Winthrop, K.L.; Ruoss, S.; Addrizzo-Harris, D.J.; et al. Randomized Trial of Liposomal Amikacin for Inhalation in Nontuberculous Mycobacterial Lung Disease. Am. J. Respir. Crit. Care Med. 2017, 195, 814–823. [Google Scholar] [CrossRef]
  55. Sison, J.P.; Yao, Y.; Kemper, C.A.; Hamilton, J.R.; Brummer, E.; Stevens, D.A.; Deresinski, S.C. Treatment of Mycobacterium avium complex infection: Do the results of in vitro susceptibility tests predict therapeutic outcome in humans? J. Infect. Dis. 1996, 173, 677–683. [Google Scholar] [CrossRef]
  56. Jeon, K.; Kwon, O.J.; Lee, N.Y.; Kim, B.J.; Kook, Y.H.; Lee, S.H.; Park, Y.K.; Kim, C.K.; Koh, W.J. Antibiotic treatment of Mycobacterium abscessus lung disease: A retrospective analysis of 65 patients. Am. J. Respir. Crit. Care Med. 2009, 180, 896–902. [Google Scholar] [CrossRef]
  57. Xu, H.B.; Jiang, R.H.; Li, L. Treatment outcomes for Mycobacterium avium complex: A systematic review and meta-analysis. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 347–358. [Google Scholar] [CrossRef]
  58. Diel, R.; Nienhaus, A.; Ringshausen, F.C.; Richter, E.; Welte, T.; Rabe, K.F.; Loddenkemper, R. Microbiologic Outcome of Interventions Against Mycobacterium avium Complex Pulmonary Disease: A Systematic Review. Chest 2018, 153, 888–921. [Google Scholar] [CrossRef]
  59. Lyu, J.; Jang, H.J.; Song, J.W.; Choi, C.M.; Oh, Y.M.; Lee, S.D.; Kim, W.S.; Kim, D.S.; Shim, T.S. Outcomes in patients with Mycobacterium abscessus pulmonary disease treated with long-term injectable drugs. Respir. Med. 2011, 105, 781–787. [Google Scholar] [CrossRef] [Green Version]
  60. Koh, W.J.; Jeon, K.; Lee, N.Y.; Kim, B.J.; Kook, Y.H.; Lee, S.H.; Park, Y.K.; Kim, C.K.; Shin, S.J.; Huitt, G.A.; et al. Clinical significance of differentiation of Mycobacterium massiliense from Mycobacterium abscessus. Am. J. Respir. Crit. Care Med. 2011, 183, 405–410. [Google Scholar] [CrossRef]
  61. von Reyn, C.F.; Williams, D.E.; Horsburgh, C.R., Jr.; Jaeger, A.S.; Marsh, B.J.; Haslov, K.; Magnusson, M. Dual skin testing with Mycobacterium avium sensitin and purified protein derivative to discriminate pulmonary disease due to M. avium complex from pulmonary disease due to Mycobacterium tuberculosis. J. Infect. Dis. 1998, 177, 730–736. [Google Scholar] [CrossRef] [Green Version]
  62. Schorey, J.S.; Sweet, L. The mycobacterial glycopeptidolipids: Structure, function, and their role in pathogenesis. Glycobiology 2008, 18, 832–841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Hirose, W.; Uchiyama, T.; Nemoto, A.; Harigai, M.; Itoh, K.; Ishizuka, T.; Matsumoto, M.; Yamaoka, K.; Nanki, T. Diagnostic performance of measuring antibodies to the glycopeptidolipid core antigen specific to Mycobacterium avium complex in patients with rheumatoid arthritis: Results from a cross-sectional observational study. Arthritis. Res. Ther. 2015, 17, 273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kitada, S.; Maekura, R.; Toyoshima, N.; Naka, T.; Fujiwara, N.; Kobayashi, M.; Yano, I.; Ito, M.; Kobayashi, K. Use of glycopeptidolipid core antigen for serodiagnosis of mycobacterium avium complex pulmonary disease in immunocompetent patients. Clin. Diagn. Lab. Immunol. 2005, 12, 44–51. [Google Scholar] [CrossRef] [PubMed]
  65. Griffith, D.E.; Thomson, R.; Flume, P.A.; Aksamit, T.R.; Field, S.K.; Addrizzo-Harris, D.J.; Morimoto, K.; Hoefsloot, W.; Mange, K.C.; Yuen, D.W.; et al. Amikacin Liposome Inhalation Suspension for Refractory Mycobacterium avium Complex Lung Disease: Sustainability and Durability of Culture Conversion and Safety of Long-term Exposure. Chest 2021, 160, 831–842. [Google Scholar] [CrossRef] [PubMed]
  66. Griffith, D.E.; Eagle, G.; Thomson, R.; Aksamit, T.R.; Hasegawa, N.; Morimoto, K.; Addrizzo-Harris, D.J.; O’Donnell, A.E.; Marras, T.K.; Flume, P.A.; et al. Amikacin Liposome Inhalation Suspension for Treatment-Refractory Lung Disease Caused by Mycobacterium avium Complex (CONVERT): A Prospective, Open-Label, Randomized Study. Am. J. Respir. Crit. Care Med. 2018, 198, 1559–1569. [Google Scholar] [CrossRef]
  67. Clinical Trials: NCT04616924: RHB-204 for the Treatment of Pulmonary Mycobacterium Avium Complex Disease (CleaR-MAC). RedHill Biopharma Limited. Available online: https://clinicaltrials.gov/ct2/show/NCT04616924 (accessed on 3 October 2022).
  68. Miwa, S.; Shirai, M.; Toyoshima, M.; Shirai, T.; Yasuda, K.; Yokomura, K.; Yamada, T.; Masuda, M.; Inui, N.; Chida, K.; et al. Efficacy of clarithromycin and ethambutol for Mycobacterium avium complex pulmonary disease. A preliminary study. Ann. Am. Thorac. Soc. 2014, 11, 23–29. [Google Scholar] [CrossRef]
  69. Winthrop, K. Clinical Trials NCT03672630: Comparison of Two-Versus Three-antibiotic Therapy for Pulmonary Mycobacterium Avium Complex Disease. Oregon Health and Science University. Available online: https://clinicaltrials.gov/ct2/show/NCT03672630 (accessed on 1 October 2022).
  70. Lin, S.; Hua, W.; Wang, S.; Zhang, Y.; Chen, X.; Liu, H.; Shao, L.; Chen, J.; Zhang, W. In vitro assessment of 17 antimicrobial agents against clinical Mycobacterium avium complex isolates. BMC Microbiol. 2022, 22, 175. [Google Scholar] [CrossRef]
  71. Quang, N.T.; Jang, J. Current Molecular Therapeutic Agents and Drug Candidates for Mycobacterium abscessus. Front. Pharmacol. 2021, 12, 724725. [Google Scholar] [CrossRef]
  72. Crilly, N.P.; Ayeh, S.K.; Karakousis, P.C. The New Frontier of Host-Directed Therapies for Mycobacterium avium Complex. Front. Immunol. 2020, 11, 623119. [Google Scholar] [CrossRef]
  73. Choi, S.R.; Switzer, B.; Britigan, B.E.; Narayanasamy, P. Gallium Porphyrin and Gallium Nitrate Synergistically Inhibit Mycobacterial Species by Targeting Different Aspects of Iron/Heme Metabolism. ACS Infect. Dis. 2020, 6, 2582–2591. [Google Scholar] [CrossRef]
  74. Gpss, C. Clinical Trials #NCT04294043. IV Gallium Study for Patients with Cystic Fibrosis Who Have NTM (ABATE Study). Available online: https://clinicaltrials.gov/ct2/show/NCT04294043 (accessed on 1 October 2022).
  75. Milanes-Virelles, M.T.; Garcia-Garcia, I.; Santos-Herrera, Y.; Valdes-Quintana, M.; Valenzuela-Silva, C.M.; Jimenez-Madrigal, G.; Ramos-Gomez, T.I.; Bello-Rivero, I.; Fernandez-Olivera, N.; Sanchez-de la Osa, R.B.; et al. Adjuvant interferon gamma in patients with pulmonary atypical Mycobacteriosis: A randomized, double-blind, placebo-controlled study. BMC Infect. Dis. 2008, 8, 17. [Google Scholar] [CrossRef] [Green Version]
  76. Karakousis, P.C.; Moore, R.D.; Chaisson, R.E. Mycobacterium avium complex in patients with HIV infection in the era of highly active antiretroviral therapy. Lancet Infect. Dis. 2004, 4, 557–565. [Google Scholar] [CrossRef]
  77. Havlir, D.V.; Schrier, R.D.; Torriani, F.J.; Chervenak, K.; Hwang, J.Y.; Boom, W.H. Effect of potent antiretroviral therapy on immune responses to Mycobacterium avium in human immunodeficiency virus-infected subjects. J. Infect. Dis. 2000, 182, 1658–1663. [Google Scholar] [CrossRef]
  78. Yaacoby-Bianu, K.; Gur, M.; Toukan, Y.; Nir, V.; Hakim, F.; Geffen, Y.; Bentur, L. Compassionate Nitric Oxide Adjuvant Treatment of Persistent Mycobacterium Infection in Cystic Fibrosis Patients. Pediatr. Infect. Dis. J. 2018, 37, 336–338. [Google Scholar] [CrossRef]
  79. Goldbart, A.; Gatt, D.; Golan Tripto, I. Non-nuberculous mycobacteria infection treated with intermittently inhaled high-dose nitric oxide. BMJ Case. Rep. 2021, 14, e243979. [Google Scholar] [CrossRef]
  80. Clinical Trials NCT03208764: Inhaled Nitric Oxide for Patients with MABSC. Beyond Air Inc. Available online: https://clinicaltrials.gov/ct2/show/NCT03208764 (accessed on 3 October 2022).
  81. Mishra, A.; Singh, V.K.; Actor, J.K.; Hunter, R.L.; Jagannath, C.; Subbian, S.; Khan, A. GM-CSF Dependent Differential Control of Mycobacterium tuberculosis Infection in Human and Mouse Macrophages: Is Macrophage Source of GM-CSF Critical to Tuberculosis Immunity? Front. Immunol. 2020, 11, 1599. [Google Scholar] [CrossRef]
  82. Rachel, M.; Thomson, G.W.; Loebinger, M.R.; Ganslandt, C. Use of inhaled GM-CSF in treatment-refractory NTM infection. An open-label, exploratory clinical trial. Eur. Respir. J. 2021, 58, OA1603. [Google Scholar] [CrossRef]
  83. Scott, J.P.; Ji, Y.; Kannan, M.; Wylam, M.E. Inhaled granulocyte-macrophage colony-stimulating factor for Mycobacterium abscessus in cystic fibrosis. Eur. Respir. J. 2018, 51, 1702127. [Google Scholar] [CrossRef] [Green Version]
  84. Netea, M.G.; van Crevel, R. BCG-induced protection: Effects on innate immune memory. Semin. Immunol. 2014, 26, 512–517. [Google Scholar] [CrossRef]
  85. Jensen, K.J.; Larsen, N.; Biering-Sorensen, S.; Andersen, A.; Eriksen, H.B.; Monteiro, I.; Hougaard, D.; Aaby, P.; Netea, M.G.; Flanagan, K.L.; et al. Heterologous immunological effects of early BCG vaccination in low-birth-weight infants in Guinea-Bissau: A randomized-controlled trial. J. Infect. Dis. 2015, 211, 956–967. [Google Scholar] [CrossRef]
  86. Kleinnijenhuis, J.; Quintin, J.; Preijers, F.; Joosten, L.A.; Ifrim, D.C.; Saeed, S.; Jacobs, C.; van Loenhout, J.; de Jong, D.; Stunnenberg, H.G.; et al. Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc. Natl. Acad. Sci. USA 2012, 109, 17537–17542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Kleinnijenhuis, J.; Quintin, J.; Preijers, F.; Benn, C.S.; Joosten, L.A.; Jacobs, C.; van Loenhout, J.; Xavier, R.J.; Aaby, P.; van der Meer, J.W.; et al. Long-lasting effects of BCG vaccination on both heterologous Th1/Th17 responses and innate trained immunity. J. Innate. Immun. 2014, 6, 152–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. de Gijsel, D.; von Reyn, C.F. A Breath of Fresh Air: BCG Prevents Adult Pulmonary Tuberculosis. Int. J. Infect. Dis. 2019, 80S, S6–S8. [Google Scholar] [CrossRef]
  89. von Reyn, C.F. BCG versus rBCG: What is the way forward? Vaccine 2021, 39, 7319–7320. [Google Scholar] [CrossRef] [PubMed]
  90. Setia, M.S.; Steinmaus, C.; Ho, C.S.; Rutherford, G.W. The role of BCG in prevention of leprosy: A meta-analysis. Lancet Infect. Dis. 2006, 6, 162–170. [Google Scholar] [CrossRef]
  91. Portaels, F.; Aguiar, J.; Debacker, M.; Steunou, C.; Zinsou, C.; Guedenon, A.; Meyers, W.M. Prophylactic effect of mycobacterium bovis BCG vaccination against osteomyelitis in children with Mycobacterium ulcerans disease (Buruli Ulcer). Clin. Diagn. Lab. Immunol. 2002, 9, 1389–1391. [Google Scholar] [CrossRef] [Green Version]
  92. Zimmermann, P.; Finn, A.; Curtis, N. Does BCG Vaccination Protect Against Nontuberculous Mycobacterial Infection? A Systematic Review and Meta-Analysis. J. Infect. Dis. 2018, 218, 679–687. [Google Scholar] [CrossRef] [Green Version]
  93. Abate, G.; Hamzabegovic, F.; Eickhoff, C.S.; Hoft, D.F. BCG Vaccination Induces M. avium and M. abscessus Cross-Protective Immunity. Front. Immunol. 2019, 10, 234. [Google Scholar] [CrossRef] [Green Version]
  94. Scriba, T.J.; Kaufmann, S.H.; Henri Lambert, P.; Sanicas, M.; Martin, C.; Neyrolles, O. Vaccination Against Tuberculosis With Whole-Cell Mycobacterial Vaccines. J. Infect. Dis. 2016, 214, 659–664. [Google Scholar] [CrossRef]
Figure 1. Radiologic abnormalities of patients with pulmonary MAC. (A) Chest CT of a 60-year-old female patient diagnosed of nodular pulmonary MAC. She completed treatment and remained culture negative for several months. (B,C), a chest X-ray and chest CT of a 60-year-old female patient recently diagnosed with fibrocavitary pulmonary MAC. Arrows show specific lesions.
Figure 1. Radiologic abnormalities of patients with pulmonary MAC. (A) Chest CT of a 60-year-old female patient diagnosed of nodular pulmonary MAC. She completed treatment and remained culture negative for several months. (B,C), a chest X-ray and chest CT of a 60-year-old female patient recently diagnosed with fibrocavitary pulmonary MAC. Arrows show specific lesions.
Microorganisms 11 00047 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hendrix, C.; McCrary, M.; Hou, R.; Abate, G. Diagnosis and Management of Pulmonary NTM with a Focus on Mycobacterium avium Complex and Mycobacterium abscessus: Challenges and Prospects. Microorganisms 2023, 11, 47. https://doi.org/10.3390/microorganisms11010047

AMA Style

Hendrix C, McCrary M, Hou R, Abate G. Diagnosis and Management of Pulmonary NTM with a Focus on Mycobacterium avium Complex and Mycobacterium abscessus: Challenges and Prospects. Microorganisms. 2023; 11(1):47. https://doi.org/10.3390/microorganisms11010047

Chicago/Turabian Style

Hendrix, Christian, Myah McCrary, Rong Hou, and Getahun Abate. 2023. "Diagnosis and Management of Pulmonary NTM with a Focus on Mycobacterium avium Complex and Mycobacterium abscessus: Challenges and Prospects" Microorganisms 11, no. 1: 47. https://doi.org/10.3390/microorganisms11010047

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop