Skip to main content
Erschienen in: Respiratory Research 1/2014

Open Access 01.12.2014 | Commentary

Shifting paradigms of nontuberculous mycobacteria in cystic fibrosis

verfasst von: Tavs Qvist, Tania Pressler, Niels Høiby, Terese L Katzenstein

Erschienen in: Respiratory Research | Ausgabe 1/2014

Abstract

Important paradigms of pulmonary disease with nontuberculous mycobacteria (NTM) are currently shifting based on an increasing attention within the field of cystic fibrosis (CF). These shifts are likely to benefit the management of all patients with pulmonary NTM, regardless of underlying pathology. Currently several key areas are being revised: The first outbreak of human NTM transmission has been proven and new evidence of biofilm growth in vivo has been demonstrated. A better understanding of the clinical impact of NTM infection has led to increased diagnostic vigilance and new recommendations for lung transplantation are under way. While recent changes have reinvigorated the interest in NTM disease, the challenge remains, whether such advances can be successfully translated into improved management and care.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Manuscript was drafted by TQ and TLK. Concept was developed in collaboration with TP and NH who assisted in revision of the manuscript. All authors have seen and approved the finished manuscript. The manuscript has not previously been published nor is it being considered for publication elsewhere.
Abkürzungen
CF
Cystic fibrosis
NTM
Nontuberculous mycobacteria
MABSC
Mycobacterium abscessus complex
MAC
Mycobacterium avium complex
ATS
American Thoracic Society
IDSA
Infectious Disease Society of America
TB
Tuberculosis
IV
Intravenous.

Background

Announced as the most important emerging threat to cystic fibrosis (CF) patients at the 2013 European Cystic Fibrosis Conference, the perception of nontuberculous mycobacteria (NTM) disease has once again undergone a notable shift. Previously thought of as a group of rather benign environmental bacteria associated with random colonization and only rarely with genuine infection of the airway [1], perceptions have now changed. Mycobacterium abscessus Complex (MABSC) and Mycobacterium avium Complex (MAC) are now recognized as insidious opportunists that can seriously affect morbidity and mortality in CF. Fundamental paradigms of NTM pulmonary disease concerning clinical impact, patient susceptibility, transmission, bacterial modes of growth and implications for lung transplantation are currently being revised. Such issues are not unique to CF and can be expected to affect NTM management in patients with other underlying pulmonary diseases. While great leaps have been made in understanding the scope and impact of NTM infections, advances in diagnostics and antibiotic management have been less impressive and still await the benefits of the heightened attention NTM disease is now receiving. Since 2007, the American Thoracic Society (ATS)’s and the Infectious Disease Society of America (IDSA)’s comprehensive statement on NTM disease has been the principle guide of management [2]. 2014 appears to be the year where the eagerly expected CF specific NTM guidelines will be published, hopefully building further consensus in a field void of solid empirical evidence and highlighting areas in urgent need of attention.

Biofilm growth

New evidence that rapidly growing NTM can grow in vivo as biofilms on intravascular catheters [3] could be an indication that biofilm growth in pulmonary NTM disease might also be a concern. Emerging evidence that pulmonary Mycobacterium tuberculosis (TB) is a biofilm infection [4] and our group’s recent examination of explanted CF lungs from patients with MABSC [5], suggests that this might indeed be the case. MABSC has been shown to be capable of structured cord formation, a biofilm mode of growth, associated with the morphologically distinct rough growth pattern [6, 7]. This pattern is assumed to be similar to that proven for TB [8], suggesting that biofilm formation, is an inherent part of NTM pathogenesis in pulmonary disease. While phenotype switching of MABSC from smooth to rough has been linked to increased virulence [9], data on the genetic determinants of cord formation remain sparse. Future strategies might include using transposons for insertional mutagenesis [10], experimental lung models and sequential sequencing [11]. Addressing whether in vivo biofilm growth takes place in the CF lung could prove pivotal in explaining the notorious unreliability of in vitro susceptibility testing. The discrepancy between in vitro antibiotic resistance and the clinical benefit of treatment suggests that more than just antibiotic resistance is involved and that antibiotic tolerance, inferred by a protective biofilm coat could be present, in parallel with what is observed in other chronic infections [12, 13]. Demonstrating NTM biofilm growth in the human lung could also open up new treatment opportunities as biofilm disrupting targets could be systematically examined, something that has not previously been attempted for NTM.

Diagnostic challenges and opportunities

Identifying patients with NTM and clinical deterioration is difficult, but important, as these are the once most likely to benefit from antimycobacterial treatment. While conventional culture and acid-fast microscopy remain the backbone of mycobacterial diagnostics, some changes are under way, which could affect both the sensitivity and specificity of NTM diagnostics. New recommendations on NTM diagnostics are likely to include shorter time from sampling to culture, increased focus on routine screening, centralization of testing to larger mycobacteriological reference laboratories and validated pretreatment of samples to avoid Gram negative bacterial or fungal overgrowth. Several of these principles are already recommended, but not universally implemented [2]. MABSC, also called Mycobacterium abscessus sensu lato comprises the closely related species Mycobacterium asbcessus sensu stricto, Mycobacterium massiliense and Mycobacterium bolletii. Differentiation of the MABSC subspecies is difficult [14], and most clinical laboratories report infection with any of the three merely as M. abscessus. Historical variability in NTM taxonomy has only added to the confusion of how to correctly classify members of MABSC and MAC. Advances in molecular diagnostics and whole genome sequencing in particular, promise a brighter future for speciation, which is important in light of new insights into differences in virulence between subspecies. Importantly the subspecies M. massiliense remains susceptible to macrolides even after prolonged exposure, resulting from an inactivating erm (41) deletion [15]. Finally, immunological assays could prove useful as a means of monitoring NTM disease progression in a fashion similar to assays used in Pseudomonas aeruginosa infections in CF [16, 17]. Thus, assays utilizing MABSC antigens-to-patient serum IgG have been shown to correlate with clinical disease [18, 19] and are currently being explored further for clinical use [20].

Prevalence and clinical impact

Reported prevalence rates have varied over time and geographically and are summarized in Table 1.
Table 1
Studies reporting the prevalence of nontuberculous mycobacteria in cystic fibrosis populations
Study
Study year
Location
Design
Included CF patients
NTM cases
NTM revalence
% MABSC
% MAC
Boxerbaum B.
1980
OH, USA
Prospective
430
8
1.8
75*
0
Smith MJ et al. [21]
1984
UK
Prospective
223
4
1.8
25*
0
Hjelte L et al. [22]
1990
Sweden
Prospective
54
5
9.2
0
60
Kilby JM et al. [23]
1992
NC, USA
Prospective
87
17
19.5
29*
76
Aitken ML et al. [24]
1993
WA, USA
Prospective
64
8
12.5
0
88
Hjelt K et al. [25]
1994
Denmark
Prospective
185
7
3.8
71*
29
Sermet-Gaudelus I et al. [26]
1996-99
France
Prospective
296
29
9.8
52
21
Fauroux B et al. [27]
1997
France
Prospective
106
7
6.6
43*
0
Mussaffi H et al. [28]
1997-02
Israel
Retrospective
139
12
8.6
67§
25§
Esther CR Jr et al. [29]
2000-07
NC, USA
Registry
1,216
166
13.7
41
59
Pierre-Audigier C et al. [30]
2000
France
Prospective
385
31
8.1
42
23
Oliver A et al. [19]
2000
Spain
Prospective
37
6
16.2
50*
33
Radhakrishnan et al. [31]
2004
Canada
Prospective
98
6
6.1
33
67
Levy I et al. [32]
2001-03
Israel
Cross-sectional
186
42
22.6
31
14
Olivier KN et al. [33]
2002
USA
Prospective
986
128
13.0
20
72
Roux AL et al. [34]
2004
France
Prospective
1,582
104
6.6
48
22
Valenza et al. [35]
2006
Germany
Prospective
60
8
13.3
50
50
Chalermskulrat W et al. [36]
2006
OH, USA
Retrospective
132
26
19.7
46
50
Binder AM et al. [37]
2011
USA
Registry
5,403
191
3.5
36
64
* = M. chelonae reported, but is here included as MABSC due to historical changes in taxonomy. § = percentage based on 6 chronic NTM cases.
Generally there has been a tendency towards higher prevalence rates being reported over time with a median prevalence rate of 9% (IQR: 3 – 11%) in the 9 studies initiated before 2000 and 13% (IQR: 7 – 17%) for the 10 post millennium prevalence studies. The reasons for this possible rise in NTM prevalence remain unknown, but could include changes in surveillance strategies although many centers are adamant that is not the case. Other reasons could include greater exposure [38], changes in the lung flora due to inhaled antibiotics [39], and reduced host immunity through autophagy inhibition during azithromycin treatment [40] or patient-to-patient transmission [15].
North American studies have consistently reported a higher proportion of MAC compared to MABSC [41], whereas the opposite is true for Western European studies [42], but the reasons for these geographical differences remain elusive. Another trend with some consistency, is that MABSC species, while isolated at all ages, seem to peak in prevalence in children, while MAC reaches the highest prevalence among adults [30, 43, 44], although this paradigm has been challenged [41]. Catherinot et al. have proposed that patient susceptibility might play a bigger role than previously thought, specifically pointing to an association between MABSC and more severe forms of CF [43]. Improved epidemiological surveillance such as the recent inclusion of NTM microbiology in the European CF registry will certainly be helpful in designing studies with the size required to address these questions.
While chronic MAC is not considered a critical threat to patient health, isolation of MABSC is often a real cause for concern. Some patients are diagnosed early on a routine mycobacterial culture, suggesting an initial phase of low to no clinical impact, which predates later loss of lung function [45]. Others develop chronic MABSC infection which can cause severe, sometimes fatal lung disease, even after an indolent period of several years [46]. A 2010 study from the US found that those infected with MABSC had a more rapid decline in lung function than their uninfected counterparts (3% per year vs. 2% per year) [29]. While the association with poor lung function is well established, the question of whether infection is mostly a consequence of, or a predisposing factor for poor lung function, remains unanswered. A 2014 study suggests that the rate of decline in FEV1 in the year leading up to the first positive culture may help distinguish patients who progress to active NTM disease and require treatment [41]. This study also reported that a quarter of patients cleared their NTM spontaneously [41] suggesting three phases of infection: Transient, persistent and active. Why some patients clear NTM suddenly without treatment, while others deteriorate rapidly warrants further investigation, as identifying who and when to treat remains the clinician’s central challenge in NTM management.

Transmission

The first solid evidence of patient-to-patient transmission of NTM in a UK CF center was published in 2013 [15]. The authors used whole-genome sequencing and showed, with a reasonable degree of certainty that the MABSC subsp. massiliense had spread within their center. Out of 31 patients nine shared one genetically near-identical strain and two patients shared another strain. A convincing case for in- and out-patient overlap as well as careful elimination of potential sources of contamination (such as the water supply and bronchoscopy equipment) was performed. The study thus confirms suspicions raised in a report from Seattle, USA, that indicated possible transmission of a single strain of M. massiliense, identified through the use of pulse-field gel electrophoresis and PCR [47] and a similar, but less cited report from Sweden [48]. Mapping the genetic population structure of MABSC is an urgent issue currently being examined by Floto et al. in a multicenter study. This is expected to have significant impact on how infection control is managed in the in- and out-patient clinic.

Lung transplantation

The view that NTM constitute an absolute contraindication for lung transplantation is being modified [49, 50]. Reports on outcome following lung transplantation in CF patients with NTM infection are few and the impact on mortality remains unknown, although increased morbidity is consistently reported [49, 51]. While aggressive treatment is recommended [2], eradication prior to transplantation is often unrealistic. Despite this, NTM disease does not preclude successful recovery after transplantation and new recommendations are expected to reflect this, bringing hope to a small, but growing number of patients, in desperate need of lung transplantation.

Antibiotic treatment

Since the important shift in the 1990s away from antituberculous regimes towards macrolide based multidrug therapy [52], not much new has been accomplished in the field of antibiotic treatment of NTM. A part from a current phase 2 trial of inhaled amikacin in the US, a recent Cochrane Review concluded, that not a single randomized trial comparing antibiotic treatment of NTM lung infection in CF has been completed [53]. NTM are notoriously difficult to treat and require multidrug treatment for 12 months or more. At present, ATS guidelines are widely accepted as the standard of care [2], but there are several ongoing controversies about MAC and MABSC disease that could benefit from an updated consensus. The superiority of one macrolide over another has not been demonstrated for MAC lung disease, nor has the benefit of routinely including an IV drug (amikacin) early on been demonstrated [2] and both issues remain undecided for newly diagnosed MAC. A preliminary trial from a non-CF setting suggests that a two-drug regimen of clarithromycin and ethambutol may be non-inferior to the recommended three-drug regimen, which today includes rifampicin [54].
The duration and intensity of antibiotic treatment for MABSC, especially in patients that do not clear the bacteria, remains a headache for most clinicians. Inhaled therapy is increasingly used in MABSC regimens and a recent retrospective study of inhaled amikacin found that while reductions in smear positivity and some symptomatic improvement were observed, toxicity was not uncommon [55]. Hopefully, preliminary results from the ongoing phase 2 trial of inhaled liposomal amikacin will be released in 2014. New CF specific treatment guidelines under way are not expected to change the fundamental structure of past MABSC recommendations, which consist of an initial induction phase followed by long maintenance therapy, but new regimens will likely include longer induction phases and generally more antibiotics, with azithromycin as the macrolide of choice. NTM specific drug-drug interactions, medication side-effects and nonadherence are also areas receiving fresh attention.

Conclusions

NTM seem to have found a successful niche in the structurally complex and often antibiotic rich environment of the CF lung. The central paradigms of how pulmonary NTM infection is initiated and how the disease progresses and interacts with the host have all shifted within the last years and upcoming consensus documents are eagerly awaited. While recent changes in the understanding of NTM disease have reinvigorated the field, the challenge remains, if such advances can be successfully translated into improved management and care.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Manuscript was drafted by TQ and TLK. Concept was developed in collaboration with TP and NH who assisted in revision of the manuscript. All authors have seen and approved the finished manuscript. The manuscript has not previously been published nor is it being considered for publication elsewhere.
Literatur
1.
2.
Zurück zum Zitat Griffith DE, Aksamit T, Brown-Elliott B a, Catanzaro A, Daley C, Gordin F, Holland SM, Horsburgh R, Huitt G, Iademarco MF, Iseman M, Olivier K, Ruoss S, von Reyn CF, Wallace RJ, Winthrop K, Wallace RJ: An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007, 175: 367-416. 10.1164/rccm.200604-571ST.PubMedCrossRef Griffith DE, Aksamit T, Brown-Elliott B a, Catanzaro A, Daley C, Gordin F, Holland SM, Horsburgh R, Huitt G, Iademarco MF, Iseman M, Olivier K, Ruoss S, von Reyn CF, Wallace RJ, Winthrop K, Wallace RJ: An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007, 175: 367-416. 10.1164/rccm.200604-571ST.PubMedCrossRef
3.
Zurück zum Zitat El HG, Viola GM, Hachem R, Han XY, Raad II: Rapidly growing mycobacterial bloodstream infections. Lancet Infect Dis. 2013, 13 (1474–4457 (Electronic)): 166-174. El HG, Viola GM, Hachem R, Han XY, Raad II: Rapidly growing mycobacterial bloodstream infections. Lancet Infect Dis. 2013, 13 (1474–4457 (Electronic)): 166-174.
4.
Zurück zum Zitat Kulka K, Hatfull G, Ojha AK: Growth of Mycobacterium tuberculosis Biofilms. J Vis Exp. 2012, 60 (60): e3820-doi:10.3791/3820 Kulka K, Hatfull G, Ojha AK: Growth of Mycobacterium tuberculosis Biofilms. J Vis Exp. 2012, 60 (60): e3820-doi:10.3791/3820
5.
Zurück zum Zitat Qvist T, Eickhardt-Sørensen S, Pressler T, Katzenstein TL, Andersen CB, Iversen M, Bjarnsholt T, Hoiby N: First evidence of Mycobacterium abscessus biofilm in the lungs of chronically infected CF patients. J Cyst Fibros. 2013, 12 (Suppl. 1): S2-S2.CrossRef Qvist T, Eickhardt-Sørensen S, Pressler T, Katzenstein TL, Andersen CB, Iversen M, Bjarnsholt T, Hoiby N: First evidence of Mycobacterium abscessus biofilm in the lungs of chronically infected CF patients. J Cyst Fibros. 2013, 12 (Suppl. 1): S2-S2.CrossRef
6.
Zurück zum Zitat Jönsson BE, Bylund J, Johansson BR, Telemo E, Wold AE: Cord-forming mycobacteria induce DNA meshwork formation by human peripheral blood mononuclear cells. Pathog Dis. 2013, 67: 54-66. 10.1111/2049-632X.12007.PubMedCrossRef Jönsson BE, Bylund J, Johansson BR, Telemo E, Wold AE: Cord-forming mycobacteria induce DNA meshwork formation by human peripheral blood mononuclear cells. Pathog Dis. 2013, 67: 54-66. 10.1111/2049-632X.12007.PubMedCrossRef
7.
Zurück zum Zitat Howard ST, Rhoades E, Recht J, Pang X, Alsup A, Kolter R, Lyons CR, Byrd TF: Spontaneous reversion of Mycobacterium abscessus from a smooth to a rough morphotype is associated with reduced expression of glycopeptidolipid and reacquisition of an invasive phenotype. Microbiology. 2006, 152 (Pt 6): 1581-1590.PubMedCrossRef Howard ST, Rhoades E, Recht J, Pang X, Alsup A, Kolter R, Lyons CR, Byrd TF: Spontaneous reversion of Mycobacterium abscessus from a smooth to a rough morphotype is associated with reduced expression of glycopeptidolipid and reacquisition of an invasive phenotype. Microbiology. 2006, 152 (Pt 6): 1581-1590.PubMedCrossRef
8.
Zurück zum Zitat Giovannini D, Cappelli G, Jiang L, Castilletti C, Colone A, Serafino A, Wannenes F, Giaco L, Quintiliani G, Fraziano M, Nepravishta R, Colizzi V, Mariani F: A new Mycobacterium tuberculosis smooth colony reduces growth inside human macrophages and represses PDIM Operon gene expression. Does an heterogeneous population exist in intracellular mycobacteria?. Microb Pathog. 2012, 53 (1096–1208 (Electronic)): 135-146.PubMedCrossRef Giovannini D, Cappelli G, Jiang L, Castilletti C, Colone A, Serafino A, Wannenes F, Giaco L, Quintiliani G, Fraziano M, Nepravishta R, Colizzi V, Mariani F: A new Mycobacterium tuberculosis smooth colony reduces growth inside human macrophages and represses PDIM Operon gene expression. Does an heterogeneous population exist in intracellular mycobacteria?. Microb Pathog. 2012, 53 (1096–1208 (Electronic)): 135-146.PubMedCrossRef
9.
Zurück zum Zitat Catherinot E, Clarissou J, Etienne G, Ripoll F, Emile J-FF, Daffé M, Perronne C, Soudais C, Gaillard J-LL, Rottman M, Daffe M: Hypervirulence of a rough variant of the Mycobacterium abscessus type strain. Infect Immun. 2007, 75: 1055-1058. 10.1128/IAI.00835-06.PubMedPubMedCentralCrossRef Catherinot E, Clarissou J, Etienne G, Ripoll F, Emile J-FF, Daffé M, Perronne C, Soudais C, Gaillard J-LL, Rottman M, Daffe M: Hypervirulence of a rough variant of the Mycobacterium abscessus type strain. Infect Immun. 2007, 75: 1055-1058. 10.1128/IAI.00835-06.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Pang JM, Layre E, Sweet L, Sherrid A, Moody DB, Ojha A, Sherman DR: The polyketide Pks1 contributes to biofilm formation in Mycobacterium tuberculosis. J Bacteriol. 2012, 194: 715-721. 10.1128/JB.06304-11.PubMedPubMedCentralCrossRef Pang JM, Layre E, Sweet L, Sherrid A, Moody DB, Ojha A, Sherman DR: The polyketide Pks1 contributes to biofilm formation in Mycobacterium tuberculosis. J Bacteriol. 2012, 194: 715-721. 10.1128/JB.06304-11.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Kreutzfeldt KM, McAdam PR, Claxton P, Holmes A, Seagar AL, Laurenson IF, Fitzgerald JR: Molecular longitudinal tracking of Mycobacterium abscessus spp. during chronic infection of the human lung. PLoS One. 2013, 8: e63237-10.1371/journal.pone.0063237.PubMedPubMedCentralCrossRef Kreutzfeldt KM, McAdam PR, Claxton P, Holmes A, Seagar AL, Laurenson IF, Fitzgerald JR: Molecular longitudinal tracking of Mycobacterium abscessus spp. during chronic infection of the human lung. PLoS One. 2013, 8: e63237-10.1371/journal.pone.0063237.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Bjarnsholt T, Jensen PO, Fiandaca MJ, Pedersen J, Hansen CR, Andersen CB, Pressler T, Givskov M, Hoiby N: Pseudomonas aeruginosa biofilms in the respiratory tract of cystic fibrosis patients. Pediatr Pulmonol. 2009, 44 (1099–0496 (Electronic)): 547-558.PubMedCrossRef Bjarnsholt T, Jensen PO, Fiandaca MJ, Pedersen J, Hansen CR, Andersen CB, Pressler T, Givskov M, Hoiby N: Pseudomonas aeruginosa biofilms in the respiratory tract of cystic fibrosis patients. Pediatr Pulmonol. 2009, 44 (1099–0496 (Electronic)): 547-558.PubMedCrossRef
13.
Zurück zum Zitat Hoiby N, Ciofu O, Johansen HK, Song ZJ, Moser C, Jensen PO, Molin S, Givskov M, Tolker-Nielsen T, Bjarnsholt T: The clinical impact of bacterial biofilms. Int J Oral Sci. 2011, 3 (1674–2818 (Print)): 55-65.PubMedPubMedCentralCrossRef Hoiby N, Ciofu O, Johansen HK, Song ZJ, Moser C, Jensen PO, Molin S, Givskov M, Tolker-Nielsen T, Bjarnsholt T: The clinical impact of bacterial biofilms. Int J Oral Sci. 2011, 3 (1674–2818 (Print)): 55-65.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Zelazny AM, Root JM, Shea YR, Colombo RE, Shamputa IC, Stock F, Conlan S, McNulty S, Brown-Elliott BA, Wallace RJ, Olivier KN, Holland SM, Sampaio EP: Cohort study of molecular identification and typing of Mycobacterium abscessus, Mycobacterium massiliense, and Mycobacterium bolletii. J Clin Microbiol. 2009, 47 (1098-660X (Electronic)): 1985-1995.PubMedPubMedCentralCrossRef Zelazny AM, Root JM, Shea YR, Colombo RE, Shamputa IC, Stock F, Conlan S, McNulty S, Brown-Elliott BA, Wallace RJ, Olivier KN, Holland SM, Sampaio EP: Cohort study of molecular identification and typing of Mycobacterium abscessus, Mycobacterium massiliense, and Mycobacterium bolletii. J Clin Microbiol. 2009, 47 (1098-660X (Electronic)): 1985-1995.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Bryant JM, Grogono DM, Greaves D, Foweraker J, Roddick I, Inns T, Reacher M, Haworth CS, Curran MD, Harris SR, Peacock SJ, Parkhill J, Floto RA: Whole-genome sequencing to identify transmission of Mycobacterium abscessus between patients with cystic fibrosis: a retrospective cohort study. Lancet. 2013, 381 (1474-547X (Electronic)): 1551-1560.PubMedPubMedCentralCrossRef Bryant JM, Grogono DM, Greaves D, Foweraker J, Roddick I, Inns T, Reacher M, Haworth CS, Curran MD, Harris SR, Peacock SJ, Parkhill J, Floto RA: Whole-genome sequencing to identify transmission of Mycobacterium abscessus between patients with cystic fibrosis: a retrospective cohort study. Lancet. 2013, 381 (1474-547X (Electronic)): 1551-1560.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Pressler T, Karpati F, Granström M, Knudsen PK, Lindblad A, Hjelte L, Olesen HV, Meyer P, Høiby N, Granstrom M, Hoiby N: Diagnostic significance of measurements of specific IgG antibodies to Pseudomonas aeruginosa by three different serological methods. J Cyst Fibros. 2009, 8 (1569–1993 (Print)): 37-42.PubMedCrossRef Pressler T, Karpati F, Granström M, Knudsen PK, Lindblad A, Hjelte L, Olesen HV, Meyer P, Høiby N, Granstrom M, Hoiby N: Diagnostic significance of measurements of specific IgG antibodies to Pseudomonas aeruginosa by three different serological methods. J Cyst Fibros. 2009, 8 (1569–1993 (Print)): 37-42.PubMedCrossRef
17.
Zurück zum Zitat Pressler T, Bohmova C, Conway S, Dumcius S, Hjelte L, Hoiby N, Kollberg H, Tummler B, Vavrova V: Chronic Pseudomonas aeruginosa infection definition: EuroCareCF Working Group report. J Cyst Fibros. 2011, 10 Suppl (10 Suppl): S75-S78. 1873–5010 (Electronic)CrossRef Pressler T, Bohmova C, Conway S, Dumcius S, Hjelte L, Hoiby N, Kollberg H, Tummler B, Vavrova V: Chronic Pseudomonas aeruginosa infection definition: EuroCareCF Working Group report. J Cyst Fibros. 2011, 10 Suppl (10 Suppl): S75-S78. 1873–5010 (Electronic)CrossRef
18.
Zurück zum Zitat Ferroni A, Sermet-Gaudelus I, Le Bourgeois M, Pierre-Audigier C, Offredo C, Rottman M, Guillemot D, Bernède C, Vincent V, Berche P, Gaillard J-LL, Le BM, Bernede C: Measurement of immunoglobulin G against Mycobacterial antigen A60 in patients with cystic fibrosis and lung infection due to Mycobacterium abscessus. Clin Infect Dis. 2005, 40 (1537–6591 (Electronic)): 58-66. 1537–6591 (Electronic)PubMedCrossRef Ferroni A, Sermet-Gaudelus I, Le Bourgeois M, Pierre-Audigier C, Offredo C, Rottman M, Guillemot D, Bernède C, Vincent V, Berche P, Gaillard J-LL, Le BM, Bernede C: Measurement of immunoglobulin G against Mycobacterial antigen A60 in patients with cystic fibrosis and lung infection due to Mycobacterium abscessus. Clin Infect Dis. 2005, 40 (1537–6591 (Electronic)): 58-66. 1537–6591 (Electronic)PubMedCrossRef
19.
Zurück zum Zitat Oliver A, Maiz L, Canton R, Escobar H, Baquero F, Gomez-Mampaso E: Nontuberculous mycobacteria in patients with cystic fibrosis. Clin Infect Dis. 2001, 32 (1058–4838 (Print)): 1298-1303.PubMedCrossRef Oliver A, Maiz L, Canton R, Escobar H, Baquero F, Gomez-Mampaso E: Nontuberculous mycobacteria in patients with cystic fibrosis. Clin Infect Dis. 2001, 32 (1058–4838 (Print)): 1298-1303.PubMedCrossRef
20.
Zurück zum Zitat Qvist T, Pressler T, Katzenstein TL, Hoiby N: Elevated levels of antibodies against Mycobacterium abscessus in cystic fibrosis patients is associated with active lung disease. Pediatr Pulmonol. 2012, 3 (47 Suppl): 335-1099–0496 (Electronic) Qvist T, Pressler T, Katzenstein TL, Hoiby N: Elevated levels of antibodies against Mycobacterium abscessus in cystic fibrosis patients is associated with active lung disease. Pediatr Pulmonol. 2012, 3 (47 Suppl): 335-1099–0496 (Electronic)
21.
Zurück zum Zitat Smith MJ, Efthimiou J, Hodson ME, Batten JC: Mycobacterial isolations in young adults with cystic fibrosis. Thorax. 1984, 39: 369-375. 10.1136/thx.39.5.369.PubMedPubMedCentralCrossRef Smith MJ, Efthimiou J, Hodson ME, Batten JC: Mycobacterial isolations in young adults with cystic fibrosis. Thorax. 1984, 39: 369-375. 10.1136/thx.39.5.369.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Hjelte L, Petrini B, Källenius G, Strandvik B, Kallenius G: Prospective study of mycobacterial infections in patients with cystic fibrosis. Thorax. 1990, 45 (0040–6376 (Print)): 397-400.PubMedPubMedCentralCrossRef Hjelte L, Petrini B, Källenius G, Strandvik B, Kallenius G: Prospective study of mycobacterial infections in patients with cystic fibrosis. Thorax. 1990, 45 (0040–6376 (Print)): 397-400.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Kilby JM, Gilligan PH, Yankaskas JR, Highsmith WE, Edwards LJ, Knowles MR: Nontuberculous mycobacteria in adult patients with cystic fibrosis. Chest. 1992, 102 (0012–3692 (Print)): 70-75.PubMedCrossRef Kilby JM, Gilligan PH, Yankaskas JR, Highsmith WE, Edwards LJ, Knowles MR: Nontuberculous mycobacteria in adult patients with cystic fibrosis. Chest. 1992, 102 (0012–3692 (Print)): 70-75.PubMedCrossRef
24.
Zurück zum Zitat Aitken ML: Nontuberculous mycobacterial disease in adult cystic fibrosis patients. CHEST J. 1993, 103: 1096-10.1378/chest.103.4.1096.CrossRef Aitken ML: Nontuberculous mycobacterial disease in adult cystic fibrosis patients. CHEST J. 1993, 103: 1096-10.1378/chest.103.4.1096.CrossRef
25.
Zurück zum Zitat Hjelt K, Hojlyng N, Howitz P, Illum N, Munk E, Valerius NH, Fursted K, Hansen KN, Heltberg I, Koch C: The role of Mycobacteria Other Than Tuberculosis (MOTT) in patients with cystic fibrosis. Scand J Infect Dis. 1994, 26 (0036–5548 (Print)): 569-576.PubMedCrossRef Hjelt K, Hojlyng N, Howitz P, Illum N, Munk E, Valerius NH, Fursted K, Hansen KN, Heltberg I, Koch C: The role of Mycobacteria Other Than Tuberculosis (MOTT) in patients with cystic fibrosis. Scand J Infect Dis. 1994, 26 (0036–5548 (Print)): 569-576.PubMedCrossRef
26.
Zurück zum Zitat Sermet-Gaudelus I, Le Bourgeois M, Pierre-Audigier C, Offredo C, Guillemot D, Halley S, Akoua-Koffi C, Vincent V, Sivadon-Tardy V, Ferroni A, Berche P, Scheinmann P, Lenoir G, Gaillard J-LL, Le BM: Mycobacterium abscessus and children with cystic fibrosis. Emerg Infect Dis. 2003, 9: 1587-1591. 10.3201/eid0912.020774.PubMedPubMedCentralCrossRef Sermet-Gaudelus I, Le Bourgeois M, Pierre-Audigier C, Offredo C, Guillemot D, Halley S, Akoua-Koffi C, Vincent V, Sivadon-Tardy V, Ferroni A, Berche P, Scheinmann P, Lenoir G, Gaillard J-LL, Le BM: Mycobacterium abscessus and children with cystic fibrosis. Emerg Infect Dis. 2003, 9: 1587-1591. 10.3201/eid0912.020774.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Fauroux B, Delaisi B, Clément A, Saizou C, Moissenet D, Truffot-Pernot C, Tournier G, Vu Thien H: Mycobacterial lung disease in cystic fibrosis: a prospective study. Pediatr Infect Dis J. 1997, 16: 354-358. 10.1097/00006454-199704000-00004.PubMedCrossRef Fauroux B, Delaisi B, Clément A, Saizou C, Moissenet D, Truffot-Pernot C, Tournier G, Vu Thien H: Mycobacterial lung disease in cystic fibrosis: a prospective study. Pediatr Infect Dis J. 1997, 16: 354-358. 10.1097/00006454-199704000-00004.PubMedCrossRef
28.
Zurück zum Zitat Mussaffi H, Rivlin J, Shalit I, Ephros M, Blau H: Nontuberculous mycobacteria in cystic fibrosis associated with allergic bronchopulmonary aspergillosis and steroid therapy. Eur Respir J. 2005, 25 (0903–1936 (Print)): 324-328.PubMedCrossRef Mussaffi H, Rivlin J, Shalit I, Ephros M, Blau H: Nontuberculous mycobacteria in cystic fibrosis associated with allergic bronchopulmonary aspergillosis and steroid therapy. Eur Respir J. 2005, 25 (0903–1936 (Print)): 324-328.PubMedCrossRef
29.
Zurück zum Zitat Esther CR, Esserman DA, Gilligan P, Kerr A, Noone PG, Esther CR: Chronic Mycobacterium abscessus infection and lung function decline in cystic fibrosis. J Cyst Fibros. 2010, 9: 117-123. 10.1016/j.jcf.2009.12.001.PubMedCrossRef Esther CR, Esserman DA, Gilligan P, Kerr A, Noone PG, Esther CR: Chronic Mycobacterium abscessus infection and lung function decline in cystic fibrosis. J Cyst Fibros. 2010, 9: 117-123. 10.1016/j.jcf.2009.12.001.PubMedCrossRef
30.
Zurück zum Zitat Pierre-Audigier C, Ferroni A, Sermet-Gaudelus I, Le BM, Offredo C, Vu-Thien H, Fauroux B, Mariani P, Munck A, Bingen E, Guillemot D, Quesne G, Vincent V, Berche P, Gaillard JL: Age-related prevalence and distribution of nontuberculous mycobacterial species among patients with cystic fibrosis. J Clin Microbiol. 2005, 43 (0095–1137 (Print)): 3467-3470.PubMedPubMedCentralCrossRef Pierre-Audigier C, Ferroni A, Sermet-Gaudelus I, Le BM, Offredo C, Vu-Thien H, Fauroux B, Mariani P, Munck A, Bingen E, Guillemot D, Quesne G, Vincent V, Berche P, Gaillard JL: Age-related prevalence and distribution of nontuberculous mycobacterial species among patients with cystic fibrosis. J Clin Microbiol. 2005, 43 (0095–1137 (Print)): 3467-3470.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Radhakrishnan DK, Yau Y, Corey M, Richardson S, Chedore P, Jamieson F, Dell SD: Non-tuberculous mycobacteria in children with cystic fibrosis: isolation, prevalence, and predictors. Pediatr Pulmonol. 2009, 44: 1100-1106. 10.1002/ppul.21106.PubMedCrossRef Radhakrishnan DK, Yau Y, Corey M, Richardson S, Chedore P, Jamieson F, Dell SD: Non-tuberculous mycobacteria in children with cystic fibrosis: isolation, prevalence, and predictors. Pediatr Pulmonol. 2009, 44: 1100-1106. 10.1002/ppul.21106.PubMedCrossRef
32.
Zurück zum Zitat Levy I, Grisaru-Soen G, Lerner-Geva L, Kerem E, Blau H, Bentur L, Aviram M, Rivlin J, Picard E, Lavy A, Yahav Y, Rahav G: Multicenter cross-sectional study of nontuberculous mycobacterial infections among cystic fibrosis patients, Israel. Emerg Infect Dis. 2008, 14: 378-384. 10.3201/eid1403.061405.PubMedPubMedCentralCrossRef Levy I, Grisaru-Soen G, Lerner-Geva L, Kerem E, Blau H, Bentur L, Aviram M, Rivlin J, Picard E, Lavy A, Yahav Y, Rahav G: Multicenter cross-sectional study of nontuberculous mycobacterial infections among cystic fibrosis patients, Israel. Emerg Infect Dis. 2008, 14: 378-384. 10.3201/eid1403.061405.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Olivier KN, Weber DJ, Wallace RJ, Faiz AR, Lee JH, Zhang Y, Brown-Elliot BA, Handler A, Wilson RW, Schechter MS, Edwards LJ, Chakraborti S, Knowles MR: Nontuberculous mycobacteria. I: multicenter prevalence study in cystic fibrosis. Am J Respir Crit Care Med. 2003, 167 (1073-449X (Print)): 828-834.PubMedCrossRef Olivier KN, Weber DJ, Wallace RJ, Faiz AR, Lee JH, Zhang Y, Brown-Elliot BA, Handler A, Wilson RW, Schechter MS, Edwards LJ, Chakraborti S, Knowles MR: Nontuberculous mycobacteria. I: multicenter prevalence study in cystic fibrosis. Am J Respir Crit Care Med. 2003, 167 (1073-449X (Print)): 828-834.PubMedCrossRef
34.
Zurück zum Zitat Roux AL, Catherinot E, Ripoll F, Soismier N, Macheras E, Ravilly S, Bellis G, Vibet MA, Le RE, Lemonnier L, Gutierrez C, Vincent V, Fauroux B, Rottman M, Guillemot D, Gaillard JL: Multicenter study of prevalence of nontuberculous mycobacteria in patients with cystic fibrosis in france. J Clin Microbiol. 2009, 47 (1098-660X (Electronic)): 4124-4128.PubMedPubMedCentralCrossRef Roux AL, Catherinot E, Ripoll F, Soismier N, Macheras E, Ravilly S, Bellis G, Vibet MA, Le RE, Lemonnier L, Gutierrez C, Vincent V, Fauroux B, Rottman M, Guillemot D, Gaillard JL: Multicenter study of prevalence of nontuberculous mycobacteria in patients with cystic fibrosis in france. J Clin Microbiol. 2009, 47 (1098-660X (Electronic)): 4124-4128.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Valenza G, Tappe D, Turnwald D, Frosch M, König C, Hebestreit H, Abele-Horn M: Prevalence and antimicrobial susceptibility of microorganisms isolated from sputa of patients with cystic fibrosis. J Cyst Fibros. 2008, 7: 123-127. 10.1016/j.jcf.2007.06.006.PubMedCrossRef Valenza G, Tappe D, Turnwald D, Frosch M, König C, Hebestreit H, Abele-Horn M: Prevalence and antimicrobial susceptibility of microorganisms isolated from sputa of patients with cystic fibrosis. J Cyst Fibros. 2008, 7: 123-127. 10.1016/j.jcf.2007.06.006.PubMedCrossRef
36.
Zurück zum Zitat Chalermskulrat W, Sood N, Neuringer IP, Hecker TM, Chang L, Rivera MP, Paradowski LJ, Aris RM: Non-tuberculous mycobacteria in end stage cystic fibrosis: implications for lung transplantation. Thorax. 2006, 61 (0040–6376 (Print)): 507-513.PubMedPubMedCentralCrossRef Chalermskulrat W, Sood N, Neuringer IP, Hecker TM, Chang L, Rivera MP, Paradowski LJ, Aris RM: Non-tuberculous mycobacteria in end stage cystic fibrosis: implications for lung transplantation. Thorax. 2006, 61 (0040–6376 (Print)): 507-513.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Binder AM, Adjemian J, Olivier KN, Prevots DR: Epidemiology of nontuberculous mycobacterial infections and associated chronic macrolide use among persons with cystic fibrosis. Am J Respir Crit Care Med. 2013, 188: 807-812. 10.1164/rccm.201307-1200OC.PubMedPubMedCentralCrossRef Binder AM, Adjemian J, Olivier KN, Prevots DR: Epidemiology of nontuberculous mycobacterial infections and associated chronic macrolide use among persons with cystic fibrosis. Am J Respir Crit Care Med. 2013, 188: 807-812. 10.1164/rccm.201307-1200OC.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Feazel LM, Baumgartner LK, Peterson KL, Frank DN, Harris JK, Pace NR: Opportunistic pathogens enriched in showerhead biofilms. Proc Natl Acad Sci USA. 2009, 106 (1091–6490 (Electronic)): 16393-16399.PubMedPubMedCentralCrossRef Feazel LM, Baumgartner LK, Peterson KL, Frank DN, Harris JK, Pace NR: Opportunistic pathogens enriched in showerhead biofilms. Proc Natl Acad Sci USA. 2009, 106 (1091–6490 (Electronic)): 16393-16399.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Renna M, Schaffner C, Brown K, Shang S, Tamayo MH, Hegyi K, Grimsey NJ, Cusens D, Coulter S, Cooper J, Bowden AR, Newton SM, Kampmann B, Helm J, Jones A, Haworth CS, Basaraba RJ, Degroote MA, Ordway DJ, Rubinsztein DC, Floto RA: Azithromycin blocks autophagy and may predispose cystic fibrosis patients to mycobacterial infection. J Clin Invest. 2011, 121 (1558–8238 (Electronic)): 3554-3563.PubMedPubMedCentralCrossRef Renna M, Schaffner C, Brown K, Shang S, Tamayo MH, Hegyi K, Grimsey NJ, Cusens D, Coulter S, Cooper J, Bowden AR, Newton SM, Kampmann B, Helm J, Jones A, Haworth CS, Basaraba RJ, Degroote MA, Ordway DJ, Rubinsztein DC, Floto RA: Azithromycin blocks autophagy and may predispose cystic fibrosis patients to mycobacterial infection. J Clin Invest. 2011, 121 (1558–8238 (Electronic)): 3554-3563.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Martiniano SL, Sontag MK, Daley CL, Nick JA, Sagel SD: Clinical significance of a first positive nontuberculous mycobacteria culture in cystic fibrosis. Ann Am Thorac Soc. 2014, 11: 36-44. 10.1513/AnnalsATS.201309-310OC.PubMedPubMedCentralCrossRef Martiniano SL, Sontag MK, Daley CL, Nick JA, Sagel SD: Clinical significance of a first positive nontuberculous mycobacteria culture in cystic fibrosis. Ann Am Thorac Soc. 2014, 11: 36-44. 10.1513/AnnalsATS.201309-310OC.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Russell CD, Claxton P, Doig C, Seagar A-L, Rayner A, Laurenson IF: Non-tuberculous mycobacteria: a retrospective review of Scottish isolates from 2000 to 2010. Thorax. 2013, Epub ahead of print Russell CD, Claxton P, Doig C, Seagar A-L, Rayner A, Laurenson IF: Non-tuberculous mycobacteria: a retrospective review of Scottish isolates from 2000 to 2010. Thorax. 2013, Epub ahead of print
43.
Zurück zum Zitat Catherinot E, Roux AL, Vibet MA, Bellis G, Ravilly S, Lemonnier L, Le RE, Bernede-Bauduin C, Le BM, Herrmann JL, Guillemot D, Gaillard JL: Mycobacterium avium and Mycobacterium abscessus complex target distinct cystic fibrosis patient subpopulations. J Cyst Fibros. 2013, 12 (1873–5010 (Electronic)): 74-80.PubMedCrossRef Catherinot E, Roux AL, Vibet MA, Bellis G, Ravilly S, Lemonnier L, Le RE, Bernede-Bauduin C, Le BM, Herrmann JL, Guillemot D, Gaillard JL: Mycobacterium avium and Mycobacterium abscessus complex target distinct cystic fibrosis patient subpopulations. J Cyst Fibros. 2013, 12 (1873–5010 (Electronic)): 74-80.PubMedCrossRef
44.
Zurück zum Zitat Esther CR, Henry MM, Molina PL, Leigh MW: Nontuberculous mycobacterial infection in young children with cystic fibrosis. Pediatr Pulmonol. 2005, 40 (8755–6863 (Print)): 39-44.PubMedCrossRef Esther CR, Henry MM, Molina PL, Leigh MW: Nontuberculous mycobacterial infection in young children with cystic fibrosis. Pediatr Pulmonol. 2005, 40 (8755–6863 (Print)): 39-44.PubMedCrossRef
45.
Zurück zum Zitat Olivier KN, Weber DJ, Lee J-HH, Handler A, Tudor G, Molina PL, Tomashefski J, Knowles MR: Nontuberculous mycobacteria. II: nested-cohort study of impact on cystic fibrosis lung disease. Am J RespirCrit Care Med. 2003, 167 (1073-449X (Print)): 835-840.CrossRef Olivier KN, Weber DJ, Lee J-HH, Handler A, Tudor G, Molina PL, Tomashefski J, Knowles MR: Nontuberculous mycobacteria. II: nested-cohort study of impact on cystic fibrosis lung disease. Am J RespirCrit Care Med. 2003, 167 (1073-449X (Print)): 835-840.CrossRef
46.
Zurück zum Zitat Cullen AR, Cannon CL, Mark EJ, Colin AA: Mycobacterium abscessus infection in cystic fibrosis. Colonization or infection?. Am J RespirCrit Care Med. 2000, 161 (1073-449X (Print)): 641-645.CrossRef Cullen AR, Cannon CL, Mark EJ, Colin AA: Mycobacterium abscessus infection in cystic fibrosis. Colonization or infection?. Am J RespirCrit Care Med. 2000, 161 (1073-449X (Print)): 641-645.CrossRef
47.
Zurück zum Zitat Aitken ML, Limaye A, Pottinger P, Whimbey E, Goss CH, Tonelli MR, Cangelosi GA, Dirac MA, Olivier KN, Brown-Elliott BA, McNulty S, Wallace RJ: Respiratory outbreak of Mycobacterium abscessus subspecies massiliense in a lung transplant and cystic fibrosis center. Am J RespirCrit Care Med. 2012, 185 (1535–4970 (Electronic)): 231-232.CrossRef Aitken ML, Limaye A, Pottinger P, Whimbey E, Goss CH, Tonelli MR, Cangelosi GA, Dirac MA, Olivier KN, Brown-Elliott BA, McNulty S, Wallace RJ: Respiratory outbreak of Mycobacterium abscessus subspecies massiliense in a lung transplant and cystic fibrosis center. Am J RespirCrit Care Med. 2012, 185 (1535–4970 (Electronic)): 231-232.CrossRef
48.
Zurück zum Zitat Jonsson BE, Gilljam M, Lindblad A, Ridell M, Wold AE, Welinder-Olsson C: Molecular epidemiology of Mycobacterium abscessus, with focus on cystic fibrosis. J Clin Microbiol. 2007, 45 (0095–1137 (Print)): 1497-1504.PubMedPubMedCentralCrossRef Jonsson BE, Gilljam M, Lindblad A, Ridell M, Wold AE, Welinder-Olsson C: Molecular epidemiology of Mycobacterium abscessus, with focus on cystic fibrosis. J Clin Microbiol. 2007, 45 (0095–1137 (Print)): 1497-1504.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Qvist T, Pressler T, Thomsen VO, Skov M, Iversen M, Katzenstein TL: Nontuberculous Mycobacterial Disease Is Not a Contraindication to Lung Transplantation in Patients with Cystic Fibrosis: A Retrospective Analysis in a Danish Patient Population. Transplant Proc. 2012, 45 (1873–2623 (Electronic)): 342-345.PubMed Qvist T, Pressler T, Thomsen VO, Skov M, Iversen M, Katzenstein TL: Nontuberculous Mycobacterial Disease Is Not a Contraindication to Lung Transplantation in Patients with Cystic Fibrosis: A Retrospective Analysis in a Danish Patient Population. Transplant Proc. 2012, 45 (1873–2623 (Electronic)): 342-345.PubMed
50.
Zurück zum Zitat Gilljam M, Schersten H, Silverborn M, Jonsson B, Ericsson HA: Lung transplantation in patients with cystic fibrosis and Mycobacterium abscessus infection. J Cyst Fibros. 2010, 9 (1873–5010 (Electronic)): 272-276.PubMedCrossRef Gilljam M, Schersten H, Silverborn M, Jonsson B, Ericsson HA: Lung transplantation in patients with cystic fibrosis and Mycobacterium abscessus infection. J Cyst Fibros. 2010, 9 (1873–5010 (Electronic)): 272-276.PubMedCrossRef
51.
Zurück zum Zitat Chernenko SM, Humar A, Hutcheon M, Chow CW, Chaparro C, Keshavjee S, Singer LG: Mycobacterium abscessus infections in lung transplant recipients: the international experience. J Heart Lung Transplant. 2006, 25 (1557–3117 (Electronic)): 1447-1455.PubMedCrossRef Chernenko SM, Humar A, Hutcheon M, Chow CW, Chaparro C, Keshavjee S, Singer LG: Mycobacterium abscessus infections in lung transplant recipients: the international experience. J Heart Lung Transplant. 2006, 25 (1557–3117 (Electronic)): 1447-1455.PubMedCrossRef
52.
Zurück zum Zitat Griffith DE: Therapy of nontuberculous mycobacterial disease. Curr Opin Infect Dis. 2007, 20 (0951–7375 (Print)): 198-203.PubMedCrossRef Griffith DE: Therapy of nontuberculous mycobacterial disease. Curr Opin Infect Dis. 2007, 20 (0951–7375 (Print)): 198-203.PubMedCrossRef
53.
Zurück zum Zitat Waters V, Ratjen F: Antibiotic treatment for nontuberculous mycobacteria lung infection in people with cystic fibrosis. Cochrane Database Syst Rev. 2012, 12 (1469-493X (Electronic)): CD010004-PubMed Waters V, Ratjen F: Antibiotic treatment for nontuberculous mycobacteria lung infection in people with cystic fibrosis. Cochrane Database Syst Rev. 2012, 12 (1469-493X (Electronic)): CD010004-PubMed
54.
Zurück zum Zitat Miwa S, Shirai M, Toyoshima M, Shirai T, Yasuda K, Yokomura K, Yamada T, Masuda M, Inui N, Chida K, Suda T, Hayakawa H: Efficacy of clarithromycin and ethambutol for Mycobacterium avium complex pulmonary disease. A preliminary study. Ann Am Thorac Soc. 2014, 11: 23-9. 10.1513/AnnalsATS.201308-266OC.PubMedCrossRef Miwa S, Shirai M, Toyoshima M, Shirai T, Yasuda K, Yokomura K, Yamada T, Masuda M, Inui N, Chida K, Suda T, Hayakawa H: Efficacy of clarithromycin and ethambutol for Mycobacterium avium complex pulmonary disease. A preliminary study. Ann Am Thorac Soc. 2014, 11: 23-9. 10.1513/AnnalsATS.201308-266OC.PubMedCrossRef
55.
Zurück zum Zitat Olivier KN, Shaw PA, Glaser TS, Bhattacharyya D, Fleshner M, Brewer CC, Zalewski CK, Folio LR, Siegelman JR, Shallom S, Park IK, Sampaio EP, Zelazny AM, Holland SM, Prevots DR: Inhaled amikacin for treatment of refractory pulmonary nontuberculous mycobacterial disease. Ann Am Thorac Soc. 2014, 11: 30-5. 10.1513/AnnalsATS.201307-231OC.PubMedPubMedCentralCrossRef Olivier KN, Shaw PA, Glaser TS, Bhattacharyya D, Fleshner M, Brewer CC, Zalewski CK, Folio LR, Siegelman JR, Shallom S, Park IK, Sampaio EP, Zelazny AM, Holland SM, Prevots DR: Inhaled amikacin for treatment of refractory pulmonary nontuberculous mycobacterial disease. Ann Am Thorac Soc. 2014, 11: 30-5. 10.1513/AnnalsATS.201307-231OC.PubMedPubMedCentralCrossRef
Metadaten
Titel
Shifting paradigms of nontuberculous mycobacteria in cystic fibrosis
verfasst von
Tavs Qvist
Tania Pressler
Niels Høiby
Terese L Katzenstein
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2014
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/1465-9921-15-41

Weitere Artikel der Ausgabe 1/2014

Respiratory Research 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.