Skip to main content

Meeting the challenges of NTM-PD from the perspective of the organism and the disease process: innovations in drug development and delivery

Abstract

Non-tuberculous mycobacterial pulmonary disease (NTM-PD) poses a substantial patient, healthcare, and economic burden. Managing NTM-PD remains challenging, and factors contributing to this include morphological, species, and patient characteristics as well as the treatment itself. This narrative review focusses on the challenges of NTM-PD from the perspective of the organism and the disease process. Morphological characteristics of non-tuberculous mycobacteria (NTM), antimicrobial resistance mechanisms, and an ability to evade host defences reduce NTM susceptibility to many antibiotics. Resistance to antibiotics, particularly macrolides, is of concern, and is associated with high mortality rates in patients with NTM-PD. New therapies are desperately needed to overcome these hurdles and improve treatment outcomes in NTM-PD. Amikacin liposome inhalation suspension (ALIS) is the first therapy specifically developed to treat refractory NTM-PD caused by Mycobacterium avium complex (MAC) and is approved in the US, EU and Japan. It provides targeted delivery to the lung and effective penetration of macrophages and biofilms and has demonstrated efficacy in treating refractory MAC pulmonary disease (MAC-PD) in the Phase III CONVERT study. Several other therapies are currently being developed including vaccination, bacteriophage therapy, and optimising host defences. Newly developed antibiotics have shown potential activity against NTM-PD and include benzimidazole, delamanid, and pretomanid. Antibiotics commonly used to treat other infections have also been repurposed for NTM-PD, including clofazimine and bedaquiline. Data from larger-scale studies are needed to determine the potential of many of these therapies for treating NTM-PD.

Background

Non-tuberculous mycobacterial pulmonary disease (NTM-PD) is a difficult-to-treat condition that is increasing in prevalence globally and presents a substantial burden to patients [1]. NTM-PD can have a significant impact on patients, causing lung function decline, worsening comorbidities, and reduced health-related quality of life and social functioning compared with the general population [2,3,4,5,6,7,8,9,10]. All-cause mortality in patients with NTM-PD can be up to four times higher than the general population, independent of other factors [8, 11,12,13]. NTM-PD is also associated with substantial economic burden, significantly greater risk of all-cause hospitalisation, and increased healthcare expenditure [13,14,15].

Many factors contribute to the challenges of treating NTM-PD; these include characteristics of the non-tuberculous mycobacteria (NTM) species and its intrinsic resistance capabilities [16, 17] as well as the ability of NTM to evade host defences through sequestration in biofilms and macrophages in the lung, making effective antibiotic penetration and treatment difficult [18]. In addition, symptoms of NTM-PD are non-specific and mirror those of underlying conditions, and diagnosis is often delayed for a number of years for some patients who have moderate-to-severe symptoms at the time of diagnosis [19,20,21]. The decision to treat is challenging and depends on the severity of disease, causative NTM species, and the patient’s goals [22]. Treatment is also lengthy, typically lasting for more than 12 months with multidrug regimens [19, 22].

The objective of this narrative review is to outline many of these factors and their implications for the treatment of NTM-PD, specifically focusing on challenges from the perspective of the NTM organism and disease process, and to discuss new treatment approaches already available or in development that aim to overcome these challenges.

Methods

We conducted a narrative review of literature retrieved from PubMed. The authors selected publications related to NTM based on title and abstract, published between 1990 and 2021. Relevant information was also retrieved from clinicaltrials.gov. Each publication was reviewed subjectively, and publications considered most relevant or robust were included in this narrative review.

Overview of the challenges of NTM infection—species virulence, at-risk patients, and treatment outcomes

The prevalence of NTM-PD is increasing globally [23], with recent reports estimating a prevalence of 2.3–6.5 per 100,000 in Europe [24,25,26]. In Japan, prevalence rates are even higher at an estimated 33–65 cases per 100,000 [27], and incidence rates in the United States of 3.1 per 100,000 in 2008 increasing to 4.7 per 100,000 in 2015 [28]. Predictive modelling studies using machine learning with United Kingdom and German databases have not only revealed an increase in NTM-PD prevalence but also a higher prevalence of potentially undiagnosed patients [29, 30].

Despite the ubiquitous nature of NTM in the environment, exposure does not equate to infection and NTM-PD remains a rare disease. The clinical relevance of mycobacterial species and their ability to cause disease differs, with the most clinically relevant species being Mycobacterium avium complex (MAC) (e.g., M. intracellulare, M. avium and M. chimaera), M. kansasii, and M. abscessus complex (M. abscessus subsp. abscessus, M. abscessus subsp. massiliense and M. abscessus subsp. bolletii) [31,32,33] (Fig. 1).

Fig. 1
figure 1

Clinical relevance of non-tuberculous mycobacteria species [31]

It is the interplay of factors of host susceptibility, NTM species virulence, and environmental exposure that determine the disease trajectory. Host susceptibility factors including underlying lung conditions, immunosuppression, and a selection of morphological patient characteristics are shown in Table 1 [23, 34,35,36,37,38]. Frequent exposure to environmental sources of NTM such as household water, soil, and bathrooms can also increase risk of infection, and reinfection from these sources is common [39, 40].

Table 1 Predisposing risk factors for non-tuberculous mycobacterial pulmonary disease [23, 35, 41,42,43,44]

Treatment goals for NTM-PD are to improve clinical, radiologic, and microbiological aspects of the disease and to achieve sputum culture conversion [7, 22, 45, 46]. Treatment outcomes for NTM-PD are intimately linked with infecting NTM species (Fig. 2) [46], and treatment recommendations for the most clinically relevant species causing NTM-PD—MAC, M. abscessus, M. xenopi, and M. kansasii—are provided in the 2020 guidelines [22]. A major challenge in treating NTM-PD is the high level of treatment failure, which can range from approximately 25% to almost 60% depending on the NTM species [7, 9, 47], and in macrolide-resistant NTM-PD potentially more than 70% [48]. Treatment failure can also increase the risk of further lung damage, reduce quality of life, and increase mortality [1, 48, 49]. Treatment itself is also challenging, with the need for extended treatment duration of 12 months post-culture conversion for some species of NTM [22].

Fig. 2
figure 2

Relationship between the virulence of non-tuberculous mycobacteria species, treatment outcomes, and drug susceptibility [42]

Overview of the challenges of NTM organisms—biology, structure, and antibiotic resistance

The life cycle and morphological characteristics of NTM bacteria create challenges for treatment as they exist as planktonic bacteria, can form biofilms, and invade eukaryotic cells [50, 51]. NTM are characterised by thick, hydrophobic cell walls, an ability to evade host defences through sequestration in and manipulation of macrophages, and an array of antimicrobial resistance mechanisms (Table 2; Fig. 3).

Table 2 Considerations and challenges to overcome in developing drugs to treat non-tuberculous mycobacterial pulmonary disease
Fig. 3
figure 3

Mycobacterial resistance mechanisms to antibiotics used against non-tuberculous mycobacteria [17]

NTM are non-motile, rod-shaped, aerobic Gram-positive bacilli, with specific physiological characteristics such as long-chain mycolic acids in their cell wall that make NTM extremely hydrophobic and impenetrable [16]. Because of these characteristic cell wall features, NTM are intrinsically resistant to many antibiotics, making penetration into the bacteria extremely difficult, and those reaching the bacterial cell may be subject to efflux pumps or metabolising processes that modify either the antibiotic itself or its target [17] (Fig. 3). In addition, some species of NTM may harbour inducible resistance by activating certain genes upon exposure to antibiotics and can also acquire genetic mutations responsible for antibiotic resistance [17].

NTM are ubiquitous in the environment, rendering avoidance impossible [53]. Typically, NTM infection arises from inhalation of contaminated environmental particles such as aerosols and dust or aspiration of contaminated substances [18, 40, 50]. In the environment, biofilms containing NTM can be found in water distribution systems, while examples of intracellular niches include amoeba in water [54, 55].

In infected individuals, NTM can form biofilms on the alveolar wall and invade cells including epithelial cells and alveolar macrophages [54, 55]. Alveolar macrophages are believed to be the main reservoir of NTM in NTM-PD [54, 55]. Once inside alveolar macrophages, NTM augment macrophage functions including cytokine production and release, as well as phagosome–lysosome fusion inhibition. This allows bacteria to survive and replicate intracellularly before macrophages undergo apoptosis, releasing the bacteria to infect neighbouring macrophages and triggering a proinflammatory response [34, 56,57,58,59,60]. Antibiotic penetration of intracellular spaces is variable, with some antibiotics such as macrolides able to penetrate macrophages and biofilms whereas others, such as amikacin, poorly penetrate thereby limiting access to bacteria and effectively reducing their bactericidal potential despite most NTM being susceptible [61,62,63,64].

The life cycle of NTM bacteria contributes to a reduced susceptibility to antibiotics. Under conditions of nutrient starvation M. intracellulare and M. avium demonstrate a biphasic approach: an adaptive phase lasting around one week when bacterial viability plummets by 50% followed by a metabolically dormant phase—the persistence phase [65]. In these two phases, upregulation of genes and accumulation of proteins drive antibiotic susceptibility decline as changes in lipid metabolism gain traction, reducing cell wall permeability beyond that afforded by the cell wall to reduce antibiotic permeability, rendering bacteria ‘tolerant’ to antibiotics [65]. In in vitro biofilms, extracellular DNA has been shown to be integral to the structural integrity of M. avium subsp. hominissuis, increasing tolerance to antibiotics [65]. Additionally, upregulation of expression of efflux pumps contributes to reduced antibiotic susceptibility and a study of efflux pump inhibitors (such as verapamil) has demonstrated increased antibiotic susceptibility [66] suggesting that efflux pump inhibitors could, potentially, provide adjunctive therapeutic support to target intracellular and extracellular antibiotic-tolerant mycobacteria. M. avium contains phosphate-sensing genes, that are comparable with those in M. tuberculosis [65]. In M. tuberculosis, phosphate sensing, which is upregulated during phases of nutrient starvation, is an important mechanism that provides organisms with antibiotic tolerance [67]. Whether gene homology in other mycobacteria confers similar tolerance effects is yet unknown and further studies are required.

Antibiotic resistance is a key concern in the treatment of NTM-PD, as patients with resistant disease have poor culture conversion rates and high 5-year mortality rates [48, 49, 68]. Resistance to macrolides is of particular concern as this forms the backbone therapy for NTM-PD caused by MAC and M. abscessus, and acts as an alternative therapy to isoniazid in M. kansasii-PD and moxifloxacin in M. xenopi-PD [22]. In MAC, macrolide resistance can result from modifications of drug binding sites through mutations in the 23S rRNA gene that prevent macrolides binding to ribosomes [69].

Prophylactic macrolide therapy and macrolide monotherapy in the presence of NTM infection are risk factors for macrolide resistance [69] and recent guidelines for bronchiectasis recommend testing for, and excluding, NTM before long-term macrolide therapy is put in place for exacerbations [70]. In M. abscessus, macrolide resistance can be intrinsic owing to the presence of the ribosomal methyltransferase gene erm(41). Erm(41) can also be induced to provide resistance to macrolides over time, whereas in M. kansasii, resistance to rifampicin can be acquired via mutations in the gene coding for RNA polymerase [69].

NTM guidelines recommend susceptibility testing before initiating regimens with drugs for which there are clear correlations between in vitro activity and treatment outcomes, such as macrolides and amikacin for MAC and M. abscessus, and rifampicin for M. kansasii [22]. However, differences in growth conditions for NTM in vitro and in the lung environment can result in a poor correlation between minimum inhibitory concentration (MIC) and clinical outcomes [52].

To be effective, antimicrobial treatment must overcome all these challenges to reach bacteria and facilitate eradication (Fig. 2). Development of new therapies for NTM-PD need to consider these multiple hurdles provided by NTM organisms (Table 2) [52].

Meeting the challenges of NTM-PD

International 2020 guidelines outline therapeutic options for four of the most common NTM species that cause pulmonary disease: MAC, M. abscessus, M. xenopi and M. kansasii (Table 3) [22]. Although, due to relatively high rates of treatment failure, development of further treatment options for NTM-PD are a priority. Treatment options include new therapeutic delivery approaches or new therapies to treat NTM-PD [71].

Table 3 Overview of guideline-based therapy for pulmonary disease caused by common NTM pathogens

Antibiotic delivery via inhalation

As a pulmonary disease, one approach to treating NTM-PD has been to target the lung directly via inhalation. Inhalation of drugs for lung conditions provides precise, direct delivery that can provide high lung concentrations with the potential for reduced systemic exposure and reduced selection pressure for multidrug resistant (MDR) organisms [18, 72].

Most NTM species, particularly MAC, are susceptible to aminoglycoside antibiotics and amikacin has been shown to be an effective concentration-dependent antibiotic against MAC in vitro [64]. Systemic amikacin in multidrug regimens has been associated with higher rates of culture conversion in MAC and M. abscessus infections than regimens where amikacin is absent [73, 74] and is recommended as part of current guideline-based therapy (GBT) for those with severe, cavitary or macrolide-resistant MAC-PD [22, 75]. However, systemic administration of amikacin is limited for prolonged use by the emergence of ototoxicity, vestibular toxicity, and renal toxicity, and the correlation between clinical outcomes and MIC is not well established [64]. Similarly, systemic penetration of antibiotics to the lung, including amikacin, is limited [76] requiring increased dosing in order to achieve effective lung concentration [77], which can lead to an increased risk of serious adverse events [78]. Many patients cannot safely reach high enough concentrations for optimal efficacy and are at risk of treatment failure [78]. This presents challenges for an effective drug concentration to combat MAC and M. abscessus.

Penetration of some antibiotics, including amikacin, into macrophages and biofilms is low and accumulation in cells such as macrophages is poor [76, 77, 79, 80]. However, for infections like NTM-PD where entry into macrophages and other cells, as well as the formation of biofilms, is common and provides potential reservoirs of infection, penetration of and accumulation in intracellular spaces is essential. Liposomes, as neutral carriers constructed of mammalian membrane-like components, can effectively penetrate both macrophages and biofilms. Liposomes are small, artificial, enclosed spherical vesicles composed of a phospholipid bilayer, which effectively encapsulate hydrophilic molecules or sequester hydrophobic drugs in the lipid bilayer and provide a controlled release system [18]. Liposomes are widely used as drug delivery nanocarriers, with the ability to transport agents to target sites while minimising systemic exposure [18].

Currently, the only treatment specifically developed for the treatment of refractory MAC-PD and approved in the USA, EU and Japan is amikacin liposome inhalation suspension (ALIS) [81]. ALIS is a nebulised liposomal formulation of amikacin which has been specifically designed to meet the three major challenges for MAC-PD: effective antimicrobial activity against MAC; effective and targeted distribution to the point of infection; and effective penetration of intracellular spaces including macrophages and biofilms, where MAC are sequestered [82, 83]. The breakpoint for amikacin resistance for MAC has changed to ≥ 128 μg/mL for liposomal encapsulated formulation due to direct delivery of ALIS to the lung (resistance breakpoint is ≥ 64 μg/mL for IV amikacin) [22, 84, 85] and this should be considered when undertaking amikacin susceptibility testing as recommended by guidelines [22]. ALIS is recommended to be added in adults with MAC-PD who fail to achieve culture conversion after 6 months of oral GBT alone by 2020 international guidelines [22].

ALIS consists of amikacin encapsulated in liposomes composed of dipalmitoylphosphatidylcholine (DPPC) and cholesterol [81, 82]. ALIS is administered using PARI’s Lamira® Nebuliser System, which was optimised for ALIS based on PARI Pharma’s eFlow® nebuliser [82, 83].

Clinical studies demonstrated effective lung penetration of amikacin with ALIS in healthy volunteers and patients with NTM-PD [86, 87]. ALIS also demonstrated effective penetration of macrophages in preclinical studies (in vitro and in vivo animal studies), compared with non-liposomal delivery, along with an ability to penetrate NTM biofilms [88]. In the Phase III randomized controlled clinical study CONVERT, culture conversion was strictly defined as three consecutive monthly negative sputum cultures. ALIS achieved culture conversion in 29% (65 of 224) of patients at month six compared with 9% (10 of 112) treated with oral GBT alone (P < 0.0001), with a serious adverse events rate comparable in both treatment groups (20.2% vs 17.9% at 6 months). Culture conversion was also sustained at 12 months of treatment (18.3% vs 2.7%; P < 0.0001) and durable 3 (16.1% vs 0; P < 0.0001) and 12 months (13.4% vs 0; P < 0.0001) following the end of treatment [89, 90]. ALIS is now being evaluated in newly diagnosed MAC-PD patients in the post-approval studies ARISE and ENCORE (trial registrations: NCT04677543 and NCT04677569).

Using existing antibiotics

There has been a long history of managing NTM-PD with antimycobacterial agents typically used for the treatment of TB and leprosy. Clofazimine has been historically used for the treatment of leprosy, but its use has been increasing in the treatment of NTM-PD, despite limited data to support efficacy. Recently, data from various retrospective observational studies have suggested efficacy, supported by a recent meta-analysis which demonstrated a treatment success rate of 56.8% when clofazimine was part of the treatment regimen [91,92,93,94]. However, regimens containing clofazimine demonstrated lower rates of treatment success compared with non-clofazimine containing regimens [93]. A Phase II trial is currently underway that will evaluate the efficacy of clofazimine for the treatment of MAC-PD (trial registration: NCT02968212). Novel formulations of clofazimine are currently under investigation for the treatment of NTM-PD, including dry powder inhalation [95], and data are awaited for a new fixed-dose formulation (RHB-204, Redhill Biopharma) of clarithromycin, rifabutin, and clofazimine, which is in a Phase III trial (trial registration: NCT04616924).

Bedaquiline is a diarylquinoline antibiotic indicated for MDR TB. Although less active against NTM compared with M. tuberculosis, bedaquiline has demonstrated in vitro bacteriostatic activity against MAC and M. abscessus. However, a real-world case series with a limited number of patients (n = 10) suggested that although it was able to improve symptoms and decrease bacterial load, sustained culture conversion after 6 months of treatment was not observed [96]. A Phase II/III trial to evaluate the efficacy and safety of treatment regimens containing bedaquiline in patients with refractory MAC-PD is currently underway (trial registration: NCT04630145).

Antibiotics more commonly used to treat non-mycobacterial infections have also shown some efficacy in NTM-PD. One example is tedizolid, an oxazolidinone typically used to treat acute bacterial skin and skin structure infections (ABSSSI), which has demonstrated efficacy in a macrophage model and in a case study of an immunocompromised patient with M. abscessus infection. Omadacycline, also more commonly used for ABSSSI, has similarly demonstrated significant in vitro activity against M. abscessus, but clinical data are currently limited to case series [96, 97].

Using previously untried antibiotic combinations in NTM-PD is another approach to repurposing antibiotics; these include vancomycin–clarithromycin for M. abscessus-PD. Dual β-lactam combinations have also demonstrated in vitro efficacy against M. abscessus-PD in macrophages as well as animal models [98].

Novel non-antibiotic therapies and approaches in development

Several novel approaches to the treatment of NTM-PD are being developed without the use of antibiotics. In a prospective pilot study in nine patients with cystic fibrosis who have M. abscessus infection, nitric oxide (NO) demonstrated improvements in both forced expiratory volume in one second and six-minute walking distance, and reductions in bacterial load following treatment with inhaled NO [99]. A Phase II proof-of-concept study of inhaled NO in patients with NTM-PD has also been completed (trial registration: NCT03748992) and an open-label study of the at-home NO generator LungFit® GO is currently taking place (trial registration: NCT04685720). In vitro studies have shown potent antibacterial activity against M. abscessus following perfusion with NO in combination with clofazimine and amikacin [100]. Further studies are needed to assess the efficacy of NO against M. abscessus infection as part of combination therapy, and also its ability to reach bacteria sequestered in biofilms and macrophages.

Another candidate in development is granulocyte–macrophage colony stimulating factor (GM-CSF), which contributes to macrophage activation. Inhalation of GM-CSF may have the potential to enhance the host defence mechanism against M. abscessus [96]. A study to explore its utility in M. abscessus infection (ENCORE) was terminated in 2021 because of COVID-19 limitations and another (OPTIMA) was completed in 2020, with initial results demonstrating that in patients with severe disease, inhaled GM-CSF did not significantly improve culture conversion rates, although a slight reduction in bacterial load was observed (trial registration: NCT03597347; NCT03421743) [101].

One novel approach to treating MAC and M. abscessus infections is that of vaccination. Current data demonstrates that Bacillus Calmette-Guérin (BCG) vaccination-induced immunity exhibits cross-reactivity to MAC and M. abscessus and may be effective as a potential prophylaxis or treatment for NTM-PD [102]. In vitro studies have shown that immunity caused by BCG vaccination or latent tuberculosis (TB) infection induces NTM cross-reactive T cells that can inhibit NTM replication within macrophages. In addition, an immune response is elicited when BCG-expanded T cells are exposed to macrophages infected with M. avium and M. abscessus [102]. Studies in BCG-vaccinated mice and humans have further emphasised these findings that BCG vaccination provides cross-protective immunity against M. avium and M. abscessus [102]. A Phase II open-label study is currently underway that will assess the role of BCG vaccination in the prevention of infections including those caused by NTM (trial registration: NCT04884308).

Bacteriophage therapy provides another potential novel approach to treat NTM-PD [96, 98], which uses viruses that infect and neutralise infecting bacteria. Although clinical data are currently lacking, a case report of a patient with disseminated M. abscessus infection where pulmonary disease predominated demonstrated clearance of infection after receiving treatment with multiple phages [98]. However, a limitation to this therapy includes its poor efficacy against mycobacteria without laboratory manipulation, meaning that practical usage of this method remains far from realised.

Optimising host defences against NTM infection also provides a possible avenue to effective therapy; targeting the inflammatory and immune pathways is currently under exploration. These experimental approaches include enhancing autophagy with mammalian target of rapamycin (mTOR) inhibitors; blocking programmed cell death protein-1 expressed on the surface of macrophages, which may improve host immune defence; and boosting the immune system with interferon-γ (IFN-γ) where in vivo mouse models suggest IFN-γ therapy may enhance the bactericidal capacity of clofazimine [103].

Novel antibiotics in development

Several novel antibiotics are also in development for the treatment of NTM-PD. For example, the novel benzimidazole has demonstrated potent bacteriostatic activity in vitro against MAC and M. kansasii, with MIC50 values ranging from 0.25 to 4 μg/mL for several species of NTM [104, 105]. A Phase IIa study to assess the efficacy and safety of SPR719 for the treatment of M. avium complex pulmonary disease (MAC-PD) was put on hold pending discussions with the US Food and Drug Administration, and is due to restart in the second half of 2022 (trial registration: NCT04553406). Two newly developed anti-TB drugs, delamanid and pretomanid, have also been evaluated for activity against M. abscessus. Although current data are not encouraging, more in vitro and in vivo data are required to determine their potential for treating M. abscessus infections [98].

Future perspectives

NTM-PD is increasing in prevalence and is a growing public health concern. A better understanding of the microbiology, pathogenesis, and epidemiology is needed to optimise patient care. A recent survey by EMBARC of patient perspectives indicated that development of new, effective drugs with improved tolerability was an imperative [106]. Before recent guideline updates [22], treatment outcomes for NTM-PD were seen to be suboptimal [48], and for patients who failed first-line treatment, options were limited [107]. Development of therapies for NTM-PD requires a focus on overcoming structural barriers of NTM for effective bacterial penetration and penetrating intracellular spaces including phagocytic cells (e.g., macrophages, biofilms) where NTM are sequestered to evade host defences and antimicrobial therapy.

A range of approaches are emerging and are in development to treat NTM-PD that focus mainly on novel antimicrobial therapy but with a view to also capitalise on existing technologies [71, 108]. A key advancement in NTM-PD management was achieved with the approval of ALIS, the first tailored approach for the treatment of refractory MAC-PD in combination with oral GBT. While ALIS is an important therapeutic advance for MAC-PD, both M. abscessus and M. kansasii remain as challenging pathogens, and a focus to treat these debilitating infections is urgently needed.

Availability of data and materials

This manuscript does not include data and material that can be shared.

Abbreviations

ABSSSI:

Acute bacterial skin and skin structure infections

ALIS:

Amikacin liposome inhalation suspension

BCG:

Bacillus Calmette-Guérin

DPPC:

Dipalmitoylphosphatidylcholine

GBT:

Guideline-based therapy

GM-CSF:

Granulocyte–macrophage colony stimulating factor

IFN-γ:

Interferon-γ

MAC:

Mycobacterium avium Complex

MAC-PD:

Mycobacterium avium Complex pulmonary disease

MDR:

Multidrug resistant

MIC:

Minimum inhibitory concentration

NO:

Nitric oxide

NTM:

Non-tuberculous mycobacteria

NTM-PD:

Non-tuberculous mycobacterial pulmonary disease

TB:

Tuberculosis

References

  1. van Ingen J, Obradovic M, Hassan M, Lesher B, Hart E, Chatterjee A, et al. Nontuberculous mycobacterial lung disease caused by Mycobacterium avium complex - disease burden, unmet needs, and advances in treatment developments. Expert Rev Respir Med. 2021;15(11):1387–401.

    Google Scholar 

  2. Asakura T, Ishii M, Ishii K, Suzuki S, Namkoong H, Okamori S, et al. Health-related QOL of elderly patients with pulmonary M. avium complex disease in a university hospital. Int J Tuberc Lung Dis. 2018;22(6):695–703.

    CAS  Google Scholar 

  3. Mehta M, Marras TK. Impaired health-related quality of life in pulmonary nontuberculous mycobacterial disease. Respir Med. 2011;105(11):1718–25.

    Google Scholar 

  4. Gochi M, Takayanagi N, Kanauchi T, Ishiguro T, Yanagisawa T, Sugita Y. Retrospective study of the predictors of mortality and radiographic deterioration in 782 patients with nodular/bronchiectatic Mycobacterium avium complex lung disease. BMJ Open. 2015;5(8): e008058.

    Google Scholar 

  5. Park TY, Chong S, Jung JW, Park IW, Choi BW, Lim C, et al. Natural course of the nodular bronchiectatic form of Mycobacterium avium complex lung disease: long-term radiologic change without treatment. PLoS ONE. 2017;12(10): e0185774.

    Google Scholar 

  6. Park HY, Jeong BH, Chon HR, Jeon K, Daley CL, Koh WJ. Lung function decline according to clinical course in nontuberculous mycobacterial lung disease. Chest. 2016;150(6):1222–32.

    Google Scholar 

  7. 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(1):120–42.

    Google Scholar 

  8. Diel R, Lipman M, Hoefsloot W. High mortality in patients with Mycobacterium avium complex lung disease: a systematic review. BMC Infect Dis. 2018;18(1):206.

    Google Scholar 

  9. Diel R, Nienhaus A, Ringshausen FC, Richter E, Welte T, Rabe KF, et al. Microbiologic outcome of interventions against Mycobacterium avium complex pulmonary disease: a systematic review. Chest. 2018;153(4):888–921.

    Google Scholar 

  10. Kwak N, Kim SA, Choi SM, Lee J, Lee CH, Yim JJ. Longitudinal changes in health-related quality of life according to clinical course among patients with non-tuberculous mycobacterial pulmonary disease: a prospective cohort study. BMC Pulm Med. 2020;20(1):126.

    Google Scholar 

  11. Marras TK, Vinnard C, Zhang Q, Hamilton K, Adjemian J, Eagle G, et al. Relative risk of all-cause mortality in patients with nontuberculous mycobacterial lung disease in a US managed care population. Respir Med. 2018;145:80–8.

    Google Scholar 

  12. Fleshner M, Olivier KN, Shaw PA, Adjemian J, Strollo S, Claypool RJ, et al. Mortality among patients with pulmonary non-tuberculous mycobacteria disease. Int J Tuberc Lung Dis. 2016;20(5):582–7.

    CAS  Google Scholar 

  13. Diel R, Jacob J, Lampenius N, Loebinger M, Nienhaus A, Rabe KF, et al. Burden of non-tuberculous mycobacterial pulmonary disease in Germany. Eur Respir J. 2017;49:4.

    Google Scholar 

  14. Goring SM, Wilson JB, Risebrough NR, Gallagher J, Carroll S, Heap KJ, et al. The cost of Mycobacterium avium complex lung disease in Canada, France, Germany, and the United Kingdom: a nationally representative observational study. BMC Health Serv Res. 2018;18(1):700.

    CAS  Google Scholar 

  15. Marras TK, Mirsaeidi M, Chou E, Eagle G, Zhang R, Leuchars M, et al. Health care utilization and expenditures following diagnosis of nontuberculous mycobacterial lung disease in the United States. J Manag Care Spec Pharm. 2018;24(10):964–74.

    Google Scholar 

  16. Johansen MD, Herrmann JL, Kremer L. Non-tuberculous mycobacteria and the rise of Mycobacterium abscessus. Nat Rev Microbiol. 2020;18(7):392–407.

    CAS  Google Scholar 

  17. Nasiri MJ, Haeili M, Ghazi M, Goudarzi H, Pormohammad A, Imani Fooladi AA, et al. New insights in to the intrinsic and acquired drug resistance mechanisms in mycobacteria. Front Microbiol. 2017;8:681.

    Google Scholar 

  18. Chalmers JD, van Ingen J, van der Laan R, Herrmann JL. Liposomal drug delivery to manage nontuberculous mycobacterial pulmonary disease and other chronic lung infections. Eur Respir Rev. 2021;30:161.

    Google Scholar 

  19. Kotilainen H, Valtonen V, Tukiainen P, Poussa T, Eskola J, Jarvinen A. Clinical findings in relation to mortality in non-tuberculous mycobacterial infections: patients with Mycobacterium avium complex have better survival than patients with other mycobacteria. Eur J Clin Microbiol Infect Dis. 2015;34(9):1909–18.

    CAS  Google Scholar 

  20. Wagner D, van Ingen J, Adjemian J, Lange C, Prevots D, Griffith D, et al. Annual prevalence and treatment estimates of nontuberculous mycobacterial pulmonary disease in Europe: a NTM-NET collaborative study. Eur Respir J. 2014;44(Suppl 58):P1067.

    Google Scholar 

  21. van Ingen J, Wagner D, Gallagher J, Morimoto K, Lange C, Haworth CS, et al. Poor adherence to management guidelines in nontuberculous mycobacterial pulmonary diseases. Eur Respir J. 2017;49:2.

    Google Scholar 

  22. Daley CL, Iaccarino JM, Lange C, Cambau E, Wallace RJ Jr, Andrejak C, et al. Treatment of nontuberculous mycobacterial pulmonary disease: an official ATS/ERS/ESCMID/IDSA clinical practice guideline. Eur Respir J. 2020;56(1):20.

    Google Scholar 

  23. Prevots DR, Marras TK. Epidemiology of human pulmonary infection with nontuberculous mycobacteria: a review. Clin Chest Med. 2015;36(1):13–34.

    Google Scholar 

  24. Ringshausen FC, Wagner D, de Roux A, Diel R, Hohmann D, Hickstein L, et al. Prevalence of nontuberculous mycobacterial pulmonary disease, Germany, 2009–2014. Emerg Infect Dis. 2016;22(6):1102–5.

    Google Scholar 

  25. Chalmers J, Aksamit T, Carvalho A, Rendon A, Franco I. Non-tuberculous mycobacterial pulmonary infections. Pulmonol. 2018;24(2):120–31.

    Google Scholar 

  26. Schildkraut JA, Zweijpfenning SMH, Nap M, He K, Dacheva E, Overbeek J, et al. The epidemiology of nontuberculous mycobacterial pulmonary disease in the Netherlands. ERJ Open Res. 2021;7:3.

    Google Scholar 

  27. Morimoto K, Iwai K, Uchimura K, Okumura M, Yoshiyama T, Yoshimori K, et al. A steady increase in nontuberculous mycobacteriosis mortality and estimated prevalence in Japan. Ann Am Thorac Soc. 2014;11(1):1–8.

    Google Scholar 

  28. Winthrop KL, Marras TK, Adjemian J, Zhang H, Wang P, Zhang Q. Incidence and prevalence of nontuberculous mycobacterial lung disease in a large U.S. managed care health plan, 2008–2015. Ann Am Thorac Soc. 2020;17(2):178–85.

    Google Scholar 

  29. Doyle OM, van der Laan R, Obradovic M, McMahon P, Daniels F, Pitcher A, et al. Identification of potentially undiagnosed patients with nontuberculous mycobacterial lung disease using machine learning applied to primary care data in the UK. Eur Respir J. 2020;56:4.

    Google Scholar 

  30. Ringshausen FC, Ewen R, Multmeier J, Monga B, Obradovic M, van der Laan R, et al. Predictive modeling of nontuberculous mycobacterial pulmonary disease epidemiology using German health claims data. Int J Infect Dis. 2021;104:398–406.

    Google Scholar 

  31. Zweijpfenning SMH, Ingen JV, Hoefsloot W. Geographic distribution of nontuberculous mycobacteria isolated from clinical specimens: a systematic review. Semin Respir Crit Care Med. 2018;39(3):336–42.

    Google Scholar 

  32. Vande Weygaerde Y, Cardinaels N, Bomans P, Chin T, Boelens J, Andre E, et al. Clinical relevance of pulmonary non-tuberculous mycobacterial isolates in three reference centres in Belgium: a multicentre retrospective analysis. BMC Infect Dis. 2019;19(1):1061.

    Google Scholar 

  33. Lee MR, Sheng WH, Hung CC, Yu CJ, Lee LN, Hsueh PR. Mycobacterium abscessus complex infections in humans. Emerg Infect Dis. 2015;21(9):1638–46.

    CAS  Google Scholar 

  34. Honda JR, Knight V, Chan ED. Pathogenesis and risk factors for nontuberculous mycobacterial lung disease. Clin Chest Med. 2015;36(1):1–11.

    Google Scholar 

  35. Andrejak C, Nielsen R, Thomsen VO, Duhaut P, Sorensen HT, Thomsen RW. Chronic respiratory disease, inhaled corticosteroids and risk of non-tuberculous mycobacteriosis. Thorax. 2013;68(3):256–62.

    Google Scholar 

  36. Chu H, Zhao L, Xiao H, Zhang Z, Zhang J, Gui T, et al. Prevalence of nontuberculous mycobacteria in patients with bronchiectasis: a meta-analysis. Arch Med Sci. 2014;10(4):661–8.

    Google Scholar 

  37. Reich JM, Johnson RE. Mycobacterium avium complex pulmonary disease presenting as an isolated lingular or middle lobe pattern, The Lady Windermere syndrome. Chest. 1992;101(6):1605–9.

    CAS  Google Scholar 

  38. Dirac MA, Horan KL, Doody DR, Meschke JS, Park DR, Jackson LA, et al. Environment or host?: A case-control study of risk factors for Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2012;186(7):684–91.

    Google Scholar 

  39. Nishiuchi Y, Iwamoto T, Maruyama F. Infection sources of a common non-tuberculous mycobacterial pathogen, Mycobacterium avium complex. Front Med (Lausanne). 2017;4:27.

    Google Scholar 

  40. Falkinham JO 3rd. Environmental sources of nontuberculous mycobacteria. Clin Chest Med. 2015;36(1):35–41.

    Google Scholar 

  41. Olivier KN, Weber DJ, Wallace RJ Jr, Faiz AR, Lee JH, Zhang Y, et al. Nontuberculous mycobacteria. I: multicenter prevalence study in cystic fibrosis. Am J Respir Crit Care Med. 2003;167(6):828–34.

    Google Scholar 

  42. Roux AL, Catherinot E, Ripoll F, Soismier N, Macheras E, Ravilly S, et al. Multicenter study of prevalence of nontuberculous mycobacteria in patients with cystic fibrosis in France. J Clin Microbiol. 2009;47(12):4124–8.

    Google Scholar 

  43. Hojo M, Iikura M, Hirano S, Sugiyama H, Kobayashi N, Kudo K. Increased risk of nontuberculous mycobacterial infection in asthmatic patients using long-term inhaled corticosteroid therapy. Respirology. 2012;17(1):185–90.

    Google Scholar 

  44. Schweitzer MD, Salamo O, Campos M, Schraufnagel DE, Sadikot R, Mirsaeidi M. Body habitus in patients with and without bronchiectasis and non-tuberculous mycobacteria. PLoS ONE. 2017;12(9): e0185095.

    Google Scholar 

  45. Griffith DE, Aksamit T, Brown-Elliott BA, Catanzaro A, Daley C, Gordin F, et al. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175(4):367–416.

    CAS  Google Scholar 

  46. van Ingen J, Boeree MJ, van Soolingen D, Iseman MD, Heifets LB, Daley CL. Are phylogenetic position, virulence, drug susceptibility and in vivo response to treatment in mycobacteria interrelated? Infect Genet Evol. 2012;12(4):832–7.

    Google Scholar 

  47. Wallace RJ Jr, Brown-Elliott BA, McNulty S, Philley JV, Killingley J, Wilson RW, et al. Macrolide/azalide therapy for nodular/bronchiectatic mycobacterium avium complex lung disease. Chest. 2014;146(2):276–82.

    CAS  Google Scholar 

  48. Moon SM, Park HY, Kim SY, Jhun BW, Lee H, Jeon K, et al. Clinical characteristics, treatment outcomes, and resistance mutations associated with macrolide-resistant Mycobacterium avium complex lung disease. Antimicrob Agents Chemother. 2016;60(11):6758–65.

    CAS  Google Scholar 

  49. Morimoto K, Namkoong H, Hasegawa N, Nakagawa T, Morino E, Shiraishi Y, et al. Macrolide-resistant Mycobacterium avium complex lung disease: analysis of 102 consecutive cases. Ann Am Thorac Soc. 2016;13(11):1904–11.

    Google Scholar 

  50. Falkinham JO 3rd. Surrounded by mycobacteria: nontuberculous mycobacteria in the human environment. J Appl Microbiol. 2009;107(2):356–67.

    CAS  Google Scholar 

  51. Honda JR, Virdi R, Chan ED. Global environmental nontuberculous mycobacteria and their contemporaneous man-made and natural niches. Front Microbiol. 2018;9:2029.

    Google Scholar 

  52. Chin KL, Sarmiento ME, Alvarez-Cabrera N, Norazmi MN, Acosta A. Pulmonary non-tuberculous mycobacterial infections: current state and future management. Eur J Clin Microbiol Infect Dis. 2020;39(5):799–826.

    Google Scholar 

  53. DeFlorio-Barker S, Egorov A, Smith GS, Murphy MS, Stout JE, Ghio AJ, et al. Environmental risk factors associated with pulmonary isolation of nontuberculous mycobacteria, a population-based study in the southeastern United States. Sci Total Environ. 2021;763: 144552.

    CAS  Google Scholar 

  54. McGarvey J, Bermudez LE. Pathogenesis of nontuberculous mycobacteria infections. Clin Chest Med. 2002;23(3):569–83.

    Google Scholar 

  55. Qvist T, Eickhardt S, Kragh KN, Andersen CB, Iversen M, Hoiby N, et al. Chronic pulmonary disease with Mycobacterium abscessus complex is a biofilm infection. Eur Respir J. 2015;46(6):1823–6.

    CAS  Google Scholar 

  56. Sousa S, Bandeira M, Carvalho PA, Duarte A, Jordao L. Nontuberculous mycobacteria pathogenesis and biofilm assembly. Int J Mycobacteriol. 2015;4(1):36–43.

    Google Scholar 

  57. Sousa S, Borges V, Joao I, Gomes JP, Jordao L. Nontuberculous mycobacteria persistence in a cell model mimicking alveolar macrophages. Microorganisms. 2019;7:5.

    Google Scholar 

  58. Chiplunkar SS, Silva CA, Bermudez LE, Danelishvili L. Characterization of membrane vesicles released by Mycobacterium avium in response to environment mimicking the macrophage phagosome. Future Microbiol. 2019;14:293–313.

    CAS  Google Scholar 

  59. Lee KI, Whang J, Choi HG, Son YJ, Jeon HS, Back YW, et al. Mycobacterium avium MAV2054 protein induces macrophage apoptosis by targeting mitochondria and reduces intracellular bacterial growth. Sci Rep. 2016;6:37804.

    CAS  Google Scholar 

  60. Appelberg R. Pathogenesis of Mycobacterium avium infection: typical responses to an atypical mycobacterium? Immunol Res. 2006;35(3):179–90.

    CAS  Google Scholar 

  61. van Ingen J, Egelund EF, Levin A, Totten SE, Boeree MJ, Mouton JW, et al. The pharmacokinetics and pharmacodynamics of pulmonary Mycobacterium avium complex disease treatment. Am J Respir Crit Care Med. 2012;186(6):559–65.

    Google Scholar 

  62. Luedtke NW, Carmichael P, Tor Y. Cellular uptake of aminoglycosides, guanidinoglycosides, and poly-arginine. J Am Chem Soc. 2003;125(41):12374–5.

    CAS  Google Scholar 

  63. Yamabe K, Arakawa Y, Shoji M, Onda M, Miyamoto K, Tsuchiya T, et al. Direct anti-biofilm effects of macrolides on Acinetobacter baumannii: comprehensive and comparative demonstration by a simple assay using microtiter plate combined with peg-lid. Biomed Res. 2020;41(6):259–68.

    CAS  Google Scholar 

  64. Brown-Elliott BA, Iakhiaeva E, Griffith DE, Woods GL, Stout JE, Wolfe CR, et al. In vitro activity of amikacin against isolates of Mycobacterium avium complex with proposed MIC breakpoints and finding of a 16S rRNA gene mutation in treated isolates. J Clin Microbiol. 2013;51(10):3389–94.

    CAS  Google Scholar 

  65. Parker H, Lorenc R, Ruelas Castillo J, Karakousis PC. Mechanisms of antibiotic tolerance in Mycobacterium avium complex: lessons from related mycobacteria. Front Microbiol. 2020;11: 573983.

    Google Scholar 

  66. Rodrigues L, Sampaio D, Couto I, Machado D, Kern WV, Amaral L, et al. The role of efflux pumps in macrolide resistance in Mycobacterium avium complex. Int J Antimicrob Agents. 2009;34(6):529–33.

    CAS  Google Scholar 

  67. Namugenyi SB, Aagesen AM, Elliott SR, Tischler AD. Mycobacterium tuberculosis PhoY proteins promote persister formation by mediating Pst/SenX3-RegX3 phosphate sensing. MBio. 2017;8:4.

    Google Scholar 

  68. Griffith DE, Brown-Elliott BA, Langsjoen B, Zhang Y, Pan X, Girard W, et al. Clinical and molecular analysis of macrolide resistance in Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;174(8):928–34.

    CAS  Google Scholar 

  69. Kwon YS, Daley CL, Koh WJ. Managing antibiotic resistance in nontuberculous mycobacterial pulmonary disease: challenges and new approaches. Expert Rev Respir Med. 2019;13(9):851–61.

    CAS  Google Scholar 

  70. Smith D, Du Rand I, Addy CL, Collyns T, Hart SP, Mitchelmore PJ, et al. British Thoracic Society guideline for the use of long-term macrolides in adults with respiratory disease. Thorax. 2020;75(5):370–404.

    Google Scholar 

  71. Wu ML, Aziz DB, Dartois V, Dick T. NTM drug discovery: status, gaps and the way forward. Drug Discov Today. 2018;23(8):1502–19.

    Google Scholar 

  72. Palmer LB. Aerosolized antibiotics in the intensive care unit. Clin Chest Med. 2011;32(3):559–74.

    Google Scholar 

  73. Kobashi Y, Matsushima T, Oka M. A double-blind randomized study of aminoglycoside infusion with combined therapy for pulmonary Mycobacterium avium complex disease. Respir Med. 2007;101(1):130–8.

    Google Scholar 

  74. Namkoong H, Morimoto K, Nishimura T, Tanaka H, Sugiura H, Yamada Y, et al. Clinical efficacy and safety of multidrug therapy including thrice weekly intravenous amikacin administration for Mycobacterium abscessus pulmonary disease in outpatient settings: a case series. BMC Infect Dis. 2016;16:396.

    Google Scholar 

  75. Haworth CS, Banks J, Capstick T, Fisher AJ, Gorsuch T, Laurenson IF, et al. British Thoracic Society guidelines for the management of non-tuberculous mycobacterial pulmonary disease (NTM-PD). Thorax. 2017;72(2):1–64.

    Google Scholar 

  76. Honeybourne D. Antibiotic penetration into lung tissues. Thorax. 1994;49(2):104–6.

    CAS  Google Scholar 

  77. Wenzler E, Fraidenburg DR, Scardina T, Danziger LH. Inhaled antibiotics for gram-negative respiratory infections. Clin Microbiol Rev. 2016;29(3):581–632.

    Google Scholar 

  78. Raaijmakers J, Schildkraut JA, Hoefsloot W, van Ingen J. The role of amikacin in the treatment of nontuberculous mycobacterial disease. Expert Opin Pharmacother. 2021;22(15):1961–74.

    CAS  Google Scholar 

  79. Kesavalu L, Goldstein JA, Debs RJ, Duzgunes N, Gangadharam PR. Differential effects of free and liposome encapsulated amikacin on the survival of Mycobacterium avium complex in mouse peritoneal macrophages. Tubercle. 1990;71(3):215–7.

    CAS  Google Scholar 

  80. Greendyke R, Byrd TF. Differential antibiotic susceptibility of Mycobacterium abscessus variants in biofilms and macrophages compared to that of planktonic bacteria. Antimicrob Agents Chemother. 2008;52(6):2019–26.

    CAS  Google Scholar 

  81. Arikayce liposomal 590 mg nebuliser dispersion. Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/arikayce-liposomal-product-information_en.pdf. Accessed 24 Feb 2022.

  82. Shirley M. Amikacin liposome inhalation suspension: a review in Mycobacterium avium complex lung disease. Drugs. 2019;79(5):555–62.

    CAS  Google Scholar 

  83. Zhang Y, Hill AT. Amikacin liposome inhalation suspension as a treatment for patients with refractory mycobacterium avium complex lung infection. Expert Rev Respir Med. 2021;15(6):737–44.

    CAS  Google Scholar 

  84. M62 performance standards for susceptibility testing of mycobacteria, nocardia spp. and other aerobic actinomycetes. 1st ed. Wayne: Clinical Laboratory Standards Institute; 2018.

  85. Brown-Elliott BA, Woods GL. Antimycobacterial susceptibility testing of nontuberculous mycobacteria. J Clin Microbiol. 2019;57:10.

    Google Scholar 

  86. Olivier K, Maass-Moreno R, Whatley M, Cheng K, Lee J-H, Folio L, et al. Airway deposition and retention of liposomal amikacin for inhalation in patients with pulmonary nontuberculous mycobacterial disease. Am Thorac Soc. 2016; Abstract: A3732.

  87. Weers J, Metzheiser B, Taylor G, Warren S, Meers P, Perkins WR. A gamma scintigraphy study to investigate lung deposition and clearance of inhaled amikacin-loaded liposomes in healthy male volunteers. J Aerosol Med Pulm Drug Deliv. 2009;22(2):131–8.

    CAS  Google Scholar 

  88. Zhang J, Leifer F, Rose S, Chun DY, Thaisz J, Herr T, et al. Amikacin liposome inhalation suspension (ALIS) penetrates non-tuberculous mycobacterial biofilms and enhances amikacin uptake into macrophages. Front Microbiol. 2018;9:915.

    Google Scholar 

  89. Griffith DE, Eagle G, Thomson R, Aksamit TR, Hasegawa N, Morimoto K, 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(12):1559–69.

    CAS  Google Scholar 

  90. Griffith DE, Thomson R, Flume PA, Aksamit TR, Field SK, Addrizzo-Harris DJ, 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(3):831–42.

    CAS  Google Scholar 

  91. Martiniano SL, Wagner BD, Levin A, Nick JA, Sagel SD, Daley CL. Safety and effectiveness of clofazimine for primary and refractory nontuberculous mycobacterial infection. Chest. 2017;152(4):800–9.

    Google Scholar 

  92. Carey G, Tebas P, Vinnard C, Kim D, Hadjiliadis D, Hansen-Flaschen J, et al. Clinical outcomes of clofazimine use for rapidly growing mycobacterial infections. Open Forum Infect Dis. 2019;6(11):ofz456.

    Google Scholar 

  93. Nasiri MJ, Calcagno T, Hosseini SS, Hematian A, Nojookambari NY, Karimi-Yazdi M, et al. Role of clofazimine in treatment of Mycobacterium avium complex. Front Med (Lausanne). 2021;8: 638306.

    Google Scholar 

  94. Pfaeffle HOI, Alameer RM, Marshall MH, Houpt ER, Albon DP, Heysell SK. Clofazimine for treatment of multidrug-resistant non-tuberculous mycobacteria. Pulm Pharmacol Ther. 2021;70: 102058.

    CAS  Google Scholar 

  95. Valinetz E, Stankiewicz Karita H, Pottinger PS, Jain R. Novel administration of clofazimine for the treatment of Mycobacterium avium infection. Open Forum Infect Dis. 2020;7(6):ofaa183.

    Google Scholar 

  96. Laudone TW, Garner L, Kam CW, Esther CR Jr, McKinzie CJ. Novel therapies for treatment of resistant and refractory nontuberculous mycobacterial infections in patients with cystic fibrosis. Pediatr Pulmonol. 2021;56(Suppl 1):S55–68.

    Google Scholar 

  97. Kumar K, Daley CL, Griffith DE, Loebinger MR. Management of Mycobacterium avium complex and Mycobacterium abscessus pulmonary disease: therapeutic advances and emerging treatments. Eur Respir Rev. 2022;31:163.

    Google Scholar 

  98. Meir M, Barkan D. Alternative and experimental therapies of mycobacterium abscessus infections. Int J Mol Sci. 2020;21:18.

    Google Scholar 

  99. Bentur L, Gur M, Ashkenazi M, Livnat-Levanon G, Mizrahi M, Tal A, et al. Pilot study to test inhaled nitric oxide in cystic fibrosis patients with refractory Mycobacterium abscessus lung infection. J Cyst Fibros. 2020;19(2):225–31.

    CAS  Google Scholar 

  100. Chalmers JD, Balavoine C, Castellotti PF, Hugel C, Payet A, Wat D, et al. European Respiratory Society International Congress, Madrid, 2019: nontuberculous mycobacterial pulmonary disease highlights. ERJ Open Res. 2020;6:4.

    Google Scholar 

  101. Thomson R, Waterer G, Loebinger M, Ganslandt C, editors. Use of inhaled GM-CSF in treatment-refractory NTM infection. An open-label, exploratory clinical trial. Presented at European Respiratory Society Congress (virtual); 2021; Abstract 537144

  102. Abate G, Stapleton JT, Rouphael N, Creech B, Stout JE, El Sahly HM, et al. Variability in the management of adults with pulmonary nontuberculous mycobacterial disease. Clin Infect Dis. 2021;72(7):1127–37.

    CAS  Google Scholar 

  103. Crilly NP, Ayeh SK, Karakousis PC. The new frontier of host-directed therapies for Mycobacterium avium complex. Front Immunol. 2020;11: 623119.

    CAS  Google Scholar 

  104. Pennings LJ, Ruth MM, Wertheim HFL, van Ingen J. The benzimidazole SPR719 shows promising concentration-dependent activity and synergy against nontuberculous mycobacteria. Antimicrob Agents Chemother. 2021;65:4.

    Google Scholar 

  105. Brown-Elliott BA, Rubio A, Wallace RJ Jr. In vitro susceptibility testing of a novel benzimidazole, SPR719, against nontuberculous mycobacteria. Antimicrob Agents Chemother. 2018;62:11.

    Google Scholar 

  106. Shteinberg M, Boyd J, Aliberti S, Polverino E, Harris B, Berg T, et al. What is important for people with nontuberculous mycobacterial disease? An EMBARC-ELF patient survey. ERJ Open Res. 2021;7:1.

    Google Scholar 

  107. Griffith DE, Aksamit TR. Therapy of refractory nontuberculous mycobacterial lung disease. Curr Opin Infect Dis. 2012;25(2):218–27.

    CAS  Google Scholar 

  108. Abate G, Hamzabegovic F, Eickhoff CS, Hoft DF. BCG vaccination induces M. avium and M. abscessus cross-protective immunity. Front Immunol. 2019;10:234.

    CAS  Google Scholar 

Download references

Acknowledgements

Medical writing assistance and editorial support was provided by Highfield, Oxford, UK. This assistance was sponsored by Insmed.

Funding

The development of this manuscript has been supported by Insmed.

Author information

Authors and Affiliations

Authors

Contributions

RvdL, AS and MO equally contributed to the conception and outline of the manuscript. All authors critically reviewed each draft and equally contributed to the revisions. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Roald van der Laan.

Ethics declarations

Ethics approval and consent to participate

This manuscript does not include information that requires ethical approval.

Consent for publication

Not applicable.

Competing interests

Marko Obradovic is an employee of Insmed.

Andy Snabilié is an employee of Insmed.

Roald van der Laan is an employee of Insmed.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

van der Laan, R., Snabilié, A. & Obradovic, M. Meeting the challenges of NTM-PD from the perspective of the organism and the disease process: innovations in drug development and delivery. Respir Res 23, 376 (2022). https://doi.org/10.1186/s12931-022-02299-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-022-02299-w

Keywords