Skip to main content

Clinical course of nontuberculous mycobacterial pulmonary disease in patients with rheumatoid arthritis

Abstract

Objectives

The impact of rheumatoid arthritis (RA) on nontuberculous mycobacterial pulmonary disease (NTM-PD) has not been well established. In this study, we investigated the clinical course of NTM-PD in patients with RA and the impact of RA on the prognosis of NTM-PD.

Methods

We analyzed patients who developed NTM-PD after being diagnosed with RA from January 2004 to August 2023 at a tertiary referral hospital in South Korea. The patient’s baseline characteristics, clinical course, and prognosis were evaluated. An optimal matching analysis was performed to measure the impact of RA on the risk of mortality.

Results

During the study period, 18 patients with RA [median age, 68 years; interquartile range (IQR) 59–73; female, 88.9%] developed NTM-PD. The median interval between RA diagnosis and subsequent NTM-PD development was 14.8 years (IQR, 8.6–19.5). At a median of 30 months (IQR, 27–105) after NTM-PD diagnosis, 10 of 18 (55.6%) patients received anti-mycobacterial treatment for NTM-PD and 5 (50.0%) patients achieved microbiological cure. When matched to patients with NTM-PD but without RA, patients with both RA and NTM-PD had a higher risk of mortality (adjusted hazard ratio, 8.14; 95% confidence interval, 2.43–27.2).

Conclusion

NTM-PD occurring after RA is associated with a higher risk of mortality than NTM-PD in the absence of RA.

Background

Nontuberculous mycobacteria (NTM), comprising more than 200 mycobacterial species other than Mycobacterium tuberculosis and M. leprae, are ubiquitous organisms found in soil, dust, and municipal water [1]. NTM can lead to chronic infections in humans, with pulmonary disease (PD) being the most common presentation [1]. Most patients with NTM-PD present with cough, sputum production, fatigue, malaise, dyspnea and weight loss [2]. The epidemiological importance of NTM-PD has been emphasized during the past few decades. In South Korea, the annual prevalence of NTM-PD increased approximately five-fold from 2010 to 2021 [3].

Pre-existing respiratory comorbidities significantly increase the risk of NTM-PD. Conditions such as bronchiectasis, chronic obstructive pulmonary disease, interstitial lung disease (ILD), or previous tuberculosis infection have been associated with the development of NTM-PD [4]. Although structural lung disease increases the susceptibility to NTM infection, the use of immunosuppressive agents further enhances the risk of NTM-PD [5, 6]. Exposure to anti-tumor necrosis factor agents has been linked to an increased risk of both tuberculosis and NTM infection. Moreover, the administration of systemic steroids raises the risk of NTM infection [5].

Rheumatoid arthritis (RA) is a chronic autoimmune disease affecting 0.27–1.85% of the general population in South Korea [7]. Several studies have demonstrated a close association between RA and NTM-PD. The structural lung abnormalities in RA can predispose individuals to NTM acquisition [8]. Restriction of the T-cell repertoire in patients with RA impairs the ability to respond to NTM [8]. Additionally, the immunomodulation induced by biologic and non-biologic disease-modifying anti-rheumatic drugs (DMARDs) contributes to the development of NTM-PD [5]. As a result, the incidence of NTM-PD in patients with RA is higher than that in the general population [9]. According to a population-based study, the risk of developing NTM-PD in patients with RA was 4.86 times higher than in the general population, resulting in an incidence rate of 41.6 cases per 100,000 person-years [10].

Although the association between RA and the development of NTM-PD has been confirmed, the impact of RA on the course of NTM-PD is not well-established. In some studies, the type of DMARDs used after the diagnosis of NTM-PD did not affect clinical or radiographic deterioration [11, 12], and RA did not increase the risk of mortality in patients with NTM-PD [13]. Notably, however, these findings were obtained from retrospective studies. Additional data accumulation is warranted to gain a more comprehensive understanding of how RA affects the clinical course of NTM-PD.

In this study, we investigated the clinical course of NTM-PD in patients with RA and the impact of RA on the prognosis of NTM-PD at a tertiary referral center in South Korea.

Methods

Study design

The study has two arms. First, the clinical course of NTM-PD in patients with RA was analyzed using the detailed data obtained from patients initially diagnosed with RA who were subsequently diagnosed with NTM-PD. Second, the impact of RA on the prognosis of NTM-PD was determined based on the case-control study, with the control being patients with NTM-PD but without RA. The study protocol was approved by the Institutional Review Board at Seoul National University Hospital (No. 2309-032-1463), and the need to obtain informed consent was waived. This study was performed in accordance with the Declaration of Helsinki.

Patient selection

We screened patients with RA who were aged ≥ 18 years and fulfilled the 1987 American College of Rheumatology criteria or the 2010 American College of Rheumatology/European League Against Rheumatism criteria for a diagnosis of RA [14, 15] from 1 January 2004 to 31 August 2023 at Seoul National University Hospital. Among these patients, we retrospectively analyzed those who met the diagnostic criteria for NTM-PD (having all of the following: pulmonary or systemic symptoms, nodular or cavitary lesions on chest radiographs, and positive culture results from at least two sputum samples or at least one bronchial wash or lavage) as suggested by the American Thoracic Society/European Respiratory Society/European Society of Clinical Microbiology and Infectious Diseases and the Infectious Diseases Society of America [1].

Data collection

We collected the patients’ demographic and clinical data, including age, sex, body mass index, acid-fast bacilli (AFB) smear results, mycobacterial species, and the presence of cavities on chest computed tomography scans at the time of NTM-PD diagnosis. Additionally, we recorded the presence of rheumatoid factor or anti-citrullinated peptide antibodies at the time of RA diagnosis, the duration of RA, and the DMARDs used for at least 1 month before and after NTM-PD diagnosis. To determine the incidence rate of NTM-PD after the onset of RA, we measured the duration of follow-up until the occurrence of NTM-PD or until the last visit, which was censored on 31 August, 2023. Once anti-mycobacterial treatments were initiated, we measured the time to the start of anti-mycobacterial treatment, the administered drugs, and the treatment outcomes.

Definitions

The progression of NTM-PD was determined by the initiation of anti-mycobacterial treatment as decided by the attending physician [16]. Microbiological cure of NTM-PD was defined as three or more consecutive negative cultures of respiratory samples after achieving culture conversion until the completion of anti-mycobacterial treatment [17]. Information regarding the date of death was obtained from the Ministry of the Interior and Safety of South Korea, and the time to death was calculated as the duration from the date of NTM-PD diagnosis to the date of death.

Case-control study

To assess the impact of RA on the clinical course of NTM-PD, we matched cases [patients with both RA and NTM-PD, referred to as RA (+) NTM-PD] to controls [patients with NTM-PD but without RA who were diagnosed from 1 January 2011 to 31 August 2023, referred to as RA (−) NTM-PD] from a prospective cohort of patients with NTM-PD in our institution [16, 18,19,20,21]. The cases and controls were matched in a 1:5 ratio. NTM-PD progression and mortality were compared between the RA (+) NTM-PD and RA (−) NTM-PD groups.

Statistical analysis

Data are summarized as median with interquartile range (IQR) for continuous variables and as proportion for categorical variables. Optimal matching using the network flow methodology was performed to match the cases and controls for age, sex, body mass index, AFB smear results, mycobacterial species, and presence of cavities [22]. The Kruskal–Wallis test and Fisher’s exact test were used for continuous and categorical variables, respectively. Survival data were analyzed using Kaplan–Meier analysis with a log-rank test and multivariable Cox proportional hazard regression. Variables with a P-value of < 0.20 in the univariate analysis were entered into the multivariate analysis. All statistical analyses were performed using Stata version 17.0 (StataCorp, College Station, TX, USA).

Results

Patient characteristics

During the study period, 9,908 patients were diagnosed with RA, and 18 of those patients developed NTM-PD. The incidence rate of NTM-PD after RA diagnosis was 21.8 per 100,000 person-years. The patients’ median age was 68 years (IQR, 59–73), and 16 (88.9%) patients were female. The median interval from RA diagnosis to subsequent NTM-PD development was 14.8 years (IQR, 8.6–19.5) (Table 1). Eight (44.4%) patients had RA-associated ILD before the diagnosis of NTM-PD. At the time of NTM-PD diagnosis, glucocorticoids were administered to 14 (77.8%) patients at a median daily prednisolone-equivalent dose of 4.7 mg (IQR, 2.5–7.5). Methotrexate (17 patients, 94.4%) was the most commonly used DMARD, followed by hydroxychloroquine (12 patients, 66.7%) and sulfasalazine (11 patients, 61.1%). Biologic agents were used in four patients (adalimumab, n = 2; infliximab, n = 1; and tocilizumab, n = 1). The time to NTM-PD development was not different between patients treated with biologics (median, 14.5 years; IQR, 10.6–17.3) and those not treated with biologics (median, 14.8 years; IQR, 8.6–20.9) (P = 0.632).

Table 1 Baseline characteristics of the study population

Clinical course of NTM-PD

After the diagnosis of NTM-PD, 16 of 18 (88.9%) patients received DMARDs for RA. Biologic agents were used for two (11.1%) patients. Glucocorticoids were administered to 16 (88.9%) patients with a median monthly prednisolone-equivalent dose of 150 mg (IQR, 75–318). During the median follow-up of 60 months (IQR, 8-102), eight (44.4%) patients were followed up without anti-mycobacterial treatment. This was dictated by the absence of symptom worsening or radiographic deterioration attributable to NTM-PD. However, ten of 18 (55.6%) patients received anti-mycobacterial treatment for NTM-PD at a median interval of 30 months (IQR, 27–105) after NTM-PD diagnosis. This was due to disease progression, with two patients experiencing symptomatic worsening, three experiencing radiographic deterioration, and five experiencing both. All patients were treated with macrolide-based regimens. Three patients received both amikacin and clofazimine. Five of 10 patients achieved microbiological cure for NTM-PD. During a median follow-up period of 40 months (IQR, 19–105), six patients died (four in patients who received anti-mycobacterial treatment and two in patients who did not receive anti-mycobacterial treatment), and all deaths were attributed to respiratory disease (pneumonia, n = 4; RA-ILD exacerbation, n = 1; and progression of NTM-PD, n = 1). Detailed information on the patients’ clinical outcomes following NTM-PD diagnosis is provided in Table 2.

Table 2 Detailed clinical information of each patient after diagnosis of NTM-PD

Prognostic impact of RA on NTM-PD

After optimal matching, every patient with RA (+) NTM-PD was paired with 90 patients with RA (−) NTM-PD. The characteristics of the two groups are presented in Table 3. The time to NTM-PD progression was not different between the two groups (log-rank P = 0.073), and the presence of RA did not affect the progression of NTM-PD (Table 4). The microbiological cure rate also showed no difference between the RA (+) NTM-PD and RA (−) NTM-PD groups (50.0% and 53.1%, respectively; P > 0.999). However, the patients in the RA (+) NTM-PD group had worse survival rates than those in the RA (−) NTM-PD group (log-rank P = 0.001). According to the multivariate Cox proportional hazard analysis, patients with RA and NTM-PD had a higher risk of mortality compared to those with NTM-PD alone (adjusted hazard ratio, 8.14; 95% confidence interval, 2.43–27.2) (Table 5). The Kaplan–Meier plots for disease progression and survival are shown in Fig. 1.

Table 3 Characteristics of study group and matched control group
Table 4 Factors associated with disease progression in patients with nontuberculous mycobacterial pulmonary disease
Table 5 Factors associated with mortality in patients with nontuberculous mycobacterial pulmonary disease
Fig. 1
figure 1

Kaplan–Meier curves for (A) time to disease progression and (B) time to death

Discussion

In this study, we investigated the clinical course of patients initially diagnosed with RA who later developed NTM-PD. Most patients continued to receive DMARDs and glucocorticoids for RA after the diagnosis of NTM-PD. More than half of the patients received anti-mycobacterial treatment for NTM-PD, and among them, 50% of the patients achieved microbiological cure. When RA coexisted with NTM-PD, the risk of mortality was higher than that in the absence of RA. The primary causes of death were predominantly related to respiratory complications.

With the increasing use of various biologics in clinical practice, there is a growing interest in the development of mycobacterial infection in patients with connective tissue diseases [23,24,25]. RA has been actively studied in this context, and several studies have demonstrated an increased risk of NTM-PD in patients with RA [9, 26]. However, few studies have provided insights into the clinical course of NTM-PD after its occurrence in patients with RA. Therefore, in this study, we rigorously focused on the clinical course of NTM-PD in 18 patients who were diagnosed with both RA and NTM-PD.

All 18 patients were diagnosed with NTM-PD approximately 15 years after their initial diagnosis of RA. Considering the incidence rate of 21.8 cases per 100,000 person-years in this study, the occurrence of NTM-PD in patients with RA is relatively uncommon. Although most patients had a history of prolonged exposure to glucocorticoids and DMARDs prior to NTM-PD development, their age at the time of NTM-PD diagnosis, distribution of mycobacterial species, and AFB smear positivity were comparable with those of patients with NTM-PD but without RA in South Korea [27, 28]. These results imply that the clinical phenotypes of NTM-PD are not markedly different between patients with and without RA.

The decision to initiate anti-mycobacterial treatment is individualized based on the clinical contexts. Approximately one-third of patients with non-cavitary nodular bronchiectatic NTM-PD can achieve spontaneous culture conversion without treatment [29]. However, about half of patients eventually require treatment for NTM-PD due to symptomatic worsening or radiographic aggravation [30]. In this study, anti-mycobacterial treatment was administered in 10 of 18 patients. The interval between diagnosis and treatment for NTM-PD was 30 months, which was comparable to that of patients without RA. Moreover, once anti-mycobacterial treatment was initiated, the treatment outcomes were also comparable to those in patients without RA.

While RA itself and the subsequent use of immunosuppressive agent increase the risk of infection [31], our findings suggest that the presence of RA and the use of immunosuppressive agents do not dictate the severity or progression of NTM-PD. This disparity may be explained by the distinct immune mechanism underlying systemic NTM infection and NTM-PD, a localized infection [32]. NTM-PD appears to be more influenced by local immunity than systemic immunity [32]. Indeed, the impact of corticosteroid on the development of NTM-PD has been more extensively studied with inhaled corticosteroid than with systemic corticosteroid [33, 34]. These observations are further supported by another Korean study that showed no discernible difference in comorbidities or the use of immunosuppressive agents between patients with progressive NTM-PD and patients with stable NTM-PD [35].

Although RA did not impact the progression of NTM-PD, RA increased the risk of mortality in patients with NTM-PD. Male sex, older age, and the presence of cavity are established risk factors for mortality [18]. In our study, the impact of RA on mortality exceeded that of these variables. This finding contradicts a Japanese study showing that RA did not increase the risk of mortality in patients with NTM-PD [13]. Importantly, our study showed that only one of six deaths was attributed to the progression of NTM-PD; the other five deaths were directly related to respiratory diseases, including pneumonia and exacerbation of ILD. These results suggest that the co-existence of ILD or the subsequent infection due to immunosuppressive agents, rather than NTM-PD itself, increased the risk of respiratory complications, which may then lead to death in patients with NTM-PD. However, due to the small sample size in this study, the impact of ILD or immunosuppressive agents should be interpreted with caution.

This study has several limitations. First, the sample size was small because of the relative rarity of both NTM and RA. However, we mitigated this limitation through optimal matching. Second, we were unable to comprehensively adjust for RA disease activity, which is an inherent limitation of retrospective studies. Third, because the cause of death in patients with RA (−) NTM-PD was not fully available, the risk of mortality attributed to NTM-PD could not be compared.

In conclusion, NTM-PD occurring after RA is associated with a higher risk of mortality than NTM-PD without RA, and respiratory diseases are a predominant cause of death.

Data availability

The dataset used are available from the corresponding author on reasonable request.

References

  1. Daley CL, Iaccarino JM, Lange C, Cambau E, Wallace RJ Jr, Andrejak C, Böttger EC, Brozek J, Griffith DE, Guglielmetti L. Treatment of nontuberculous mycobacterial pulmonary disease: an official ATS/ERS/ESCMID/IDSA clinical practice guideline. Clin Infect Dis. 2020;71(4):e1–e36.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Griffith DE, Aksamit T, Brown-Elliott BA, Catanzaro A, Daley C, Gordin F, Holland SM, Horsburgh R, Huitt G, Iademarco MF, 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.

    Article  CAS  PubMed  Google Scholar 

  3. Kim J-Y, Kwak N, Yim J-J. The rise in prevalence and related costs of nontuberculous mycobacterial diseases in South Korea, 2010–2021. Open Forum Infect Diss: 2022. Oxford University Press US; 2022. p. ofac649.

  4. Loebinger MR, Quint JK, van der Laan R, Obradovic M, Chawla R, Kishore A, van Ingen J. Risk Factors for Nontuberculous Mycobacterial Pulmonary Disease: A Systematic Literature Review and Meta-Analysis. Chest 2023.

  5. Brode SK, Jamieson FB, Ng R, Campitelli MA, Kwong JC, Paterson JM, Li P, Marchand-Austin A, Bombardier C, Marras TK. Increased risk of mycobacterial infections associated with anti-rheumatic medications. Thorax. 2015;70(7):677–82.

    Article  PubMed  Google Scholar 

  6. Chao W-C, Lin C-H, Liao T-L, Chen Y-M, Hsu C-Y, Chen J-P, Chen D-Y, Chen H-H. The risk of nontuberculous mycobacterial infection in patients with Sjögren’s syndrome: a nationwide, population-based cohort study. BMC Infect Dis. 2017;17(1):1–8.

    Article  Google Scholar 

  7. Kim H, Sung YK. Epidemiology of rheumatoid arthritis in Korea. J Rheum Dis. 2021;28(2):60–7.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Winthrop KL, Iseman M. Bedfellows: mycobacteria and rheumatoid arthritis in the era of biologic therapy. Nat Rev Rheumatol. 2013;9(9):524–31.

    Article  PubMed  Google Scholar 

  9. Yeh J-J, Wang Y-C, Sung F-C, Kao C-H. Rheumatoid arthritis increases the risk of nontuberculosis mycobacterial disease and active pulmonary tuberculosis. PLoS ONE. 2014;9(10):e110922.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  10. Brode SK, Jamieson FB, Ng R, Campitelli MA, Kwong JC, Paterson JM, Li P, Marchand-Austin A, Bombardier C, Marras TK. Risk of mycobacterial infections associated with rheumatoid arthritis in Ontario. Can Chest. 2014;146(3):563–72.

    Article  PubMed  Google Scholar 

  11. Yamakawa H, Takayanagi N, Miyahara Y, Ishiguro T, Kanauchi T, Hoshi T, Yanagisawa T, Sugita Y. Prognostic factors and radiographic outcomes of nontuberculous mycobacterial lung disease in rheumatoid arthritis. J Rheumatol. 2013;40(8):1307–15.

    Article  PubMed  Google Scholar 

  12. Takei H, Nishina N, Namkoong H, Suzuki K, Uwamino Y, Hasegawa N, Takeuchi T. Rheumatoid arthritis with nontuberculous mycobacterial pulmonary disease: a retrospective, single-centre cohort study. Mod Rheumatol. 2022;32(3):534–40.

    Article  PubMed  Google Scholar 

  13. Mori S, Koga Y, Nakamura K, Hirooka S, Matsuoka T, Uramoto H, Sakamoto O, Ueki Y. Mortality in rheumatoid arthritis patients with pulmonary nontuberculous mycobacterial disease: a retrospective cohort study. PLoS ONE. 2020;15(12):e0243110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO III, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League against Rheumatism collaborative initiative. Arthritis Rheum. 2010;62(9):2569–81.

    Article  PubMed  Google Scholar 

  15. Arnett FC, Edworthy SM, Bloch DA, Mcshane DJ, Fries JF, Cooper NS, Healey LA, Kaplan SR, Liang MH, Luthra HS. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31(3):315–24.

    Article  CAS  PubMed  Google Scholar 

  16. Kwak N, Hwang HW, Kim H-J, Lee HW, Yim J-J, Lee C-H. The Association between Bacille Calmette-Guérin Vaccination and Nontuberculous Mycobacterial Pulmonary Disease. J Korean Med Sci. 2022;37(26):e206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Van Ingen J, Aksamit T, Andrejak C, Böttger EC, Cambau E, Daley CL, Griffith DE, Guglielmetti L, Holland SM, Huitt GA. Treatment outcome definitions in nontuberculous mycobacterial pulmonary disease: an NTM-NET consensus statement. Eur Respir J. 2018;51(3):1800170.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Kim H-J, Kwak N, Hong H, Kang N, Im Y, Jhun BW, Yim J-J. BACES score for predicting mortality in nontuberculous mycobacterial pulmonary disease. Am J Respir Crit Care Med. 2021;203(2):230–6.

    Article  CAS  PubMed  Google Scholar 

  19. Kwak N, Kim SA, Choi SM, Lee J, Lee C-H, Yim J-J. 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):1–7.

    Article  Google Scholar 

  20. Kwak N, Whang J, Yang JS, Kim TS, Kim SA, Yim J-J. Minimal inhibitory concentration of clofazimine among clinical isolates of nontuberculous mycobacteria and its impact on treatment outcome. Chest. 2021;159(2):517–23.

    Article  CAS  PubMed  Google Scholar 

  21. Jung HI, Kim SA, Kim H-J, Yim J-J, Kwak N. Anxiety and depression in patients with nontuberculous mycobacterial pulmonary disease: a prospective cohort study in South Korea. Chest. 2022;161(4):918–26.

    Article  PubMed  Google Scholar 

  22. Rosenbaum PR. Optimal matching for observational studies. J Am Stat Assoc. 1989;84(408):1024–32.

    Article  Google Scholar 

  23. Cobo-Ibáñez T, Descalzo MÁ, Loza-Santamaría E, Carmona L, Muñoz-Fernández S. Serious infections in patients with rheumatoid arthritis and other immune-mediated connective tissue diseases exposed to anti-TNF or rituximab: data from the Spanish registry BIOBADASER 2.0. Rheumatol int. 2014;34:953–61.

    Article  PubMed  Google Scholar 

  24. Winthrop KL, Chang E, Yamashita S, Iademarco MF, LoBue PA. Nontuberculous mycobacteria infections and anti–tumor necrosis factor-α therapy. Emerg Infect Dis. 2009;15(10):1556.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Winthrop KL, Yamashita S, Beekmann S, Polgreen P, Network IDSAEI. Mycobacterial and other serious infections in patients receiving anti-tumor necrosis factor and other newly approved biologic therapies: case finding through the Emerging Infections Network. Clin Infect Dis. 2008;46(11):1738–40.

    Article  PubMed  Google Scholar 

  26. Park DW, Kim YJ, Sung Y-K, Chung SJ, Yeo Y, Park TS, Lee H, Moon J-Y, Kim S-H, Kim T-H. TNF inhibitors increase the risk of nontuberculous mycobacteria in patients with seropositive rheumatoid arthritis in a mycobacterium tuberculosis endemic area. Sci Rep. 2022;12(1):4003.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jhun BW, Moon SM, Jeon K, Kwon OJ, Yoo H, Carriere KC, Huh HJ, Lee NY, Shin SJ, Daley CL. Prognostic factors associated with long-term mortality in 1445 patients with nontuberculous mycobacterial pulmonary disease: a 15-year follow-up study. Eur Respir J 2020, 55(1).

  28. Kim J-Y, Kim NY, Jung H-W, Yim J-J, Kwak N. Old age is associated with worse treatment outcome and frequent adverse drug reaction in Mycobacterium avium complex pulmonary disease. BMC Pulm Med. 2022;22(1):1–9.

    Article  Google Scholar 

  29. Moon SM, Jhun BW, Baek S-Y, Kim S, Jeon K, Ko R-E, Shin SH, Lee H, Kwon OJ, Huh HJ. Long-term natural history of non-cavitary nodular bronchiectatic nontuberculous mycobacterial pulmonary disease. Respir Med. 2019;151:1–7.

    Article  PubMed  Google Scholar 

  30. Kim J-Y, Choi Y, Park J, Goo JM, Kim TS, Seong M-W, Kwak N, Yim J-J. Impact of treatment on long-term survival of patients with Mycobacterium avium complex pulmonary disease. Clin Infect Dis 2023:ciad108.

  31. Listing J, Gerhold K, Zink A. The risk of infections associated with rheumatoid arthritis, with its comorbidity and treatment. Rheumatol. 2013;52(1):53–61.

    Article  CAS  Google Scholar 

  32. Shu C-C, Wu M-F, Pan S-W, Wu T-S, Lai H-C, Lin M-C. Host immune response against environmental nontuberculous mycobacteria and the risk populations of nontuberculous mycobacterial lung disease. J Formos Med Assoc. 2020;119:13–S22.

    Article  Google Scholar 

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

    Article  PubMed  Google Scholar 

  34. Brode SK, Campitelli MA, Kwong JC, Lu H, Marchand-Austin A, Gershon AS, Jamieson FB, Marras TK. The risk of mycobacterial infections associated with inhaled corticosteroid use. Eur Respir J 2017, 50(3).

  35. Hwang JA, Kim S, Jo K-W, Shim TS. Natural history of Mycobacterium avium complex lung disease in untreated patients with stable course. Eur Respir J 2017, 49(3).

Download references

Acknowledgements

Not applicable.

Funding

There was no funding for this study.

Author information

Authors and Affiliations

Authors

Contributions

NK, JM and JJY participated in the conceptualization. NK and JM reviewed the medical records of the study patients and contributed to the data curation. NK, JM, JYK and JWP developed the methodology and performed the formal analysis under the supervision of JJY. JWP and JYK critically appraised the results. NK and JM wrote the original draft of the manuscript, and JYK, JWP, and JJY critically reviewed and edited the manuscript. All authors had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. All authors have read and approved the final version of the submitted manuscript for publication.

Corresponding author

Correspondence to Jae-Joon Yim.

Ethics declarations

Ethics approval and consent to participate

The study protocol was approved by the institutional review board (approval number No. 2309-032-1463) at Seoul National University Hospital, which waived the written informed consent requirement due to the retrospective design of the study. The study was performed in accordance with the Declaration of Helsinki.

Consent for publication

Not applicable.

Competing interests

All authors declare no conflicts of interest.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kwak, N., Moon, J., Kim, JY. et al. Clinical course of nontuberculous mycobacterial pulmonary disease in patients with rheumatoid arthritis. Adv Rheumatol 64, 20 (2024). https://doi.org/10.1186/s42358-024-00357-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s42358-024-00357-z

Keywords