Skip directly to site content Skip directly to page options Skip directly to A-Z link Skip directly to A-Z link Skip directly to A-Z link
Volume 22, Number 6—June 2016
Letter

Interferon-γ Autoantibodies as Predisposing Factor for Nontuberculous Mycobacterial Infection

On This Page
Tables
Article Metrics
37
citations of this article
EID Journal Metrics on Scopus

Cite This Article

To the Editor: Recent advances in the understanding of antimycobacterial immune response have led to descriptions of predisposing conditions to dissemination of nontuberculous mycobacteria (NTM) infection. The interferon- γ (IFN-γ)/interleukin-12 (IL-12) axis is a critical pathway for intracellular killing of mycobacteria (Technical Appendix Figure) (1).We report a case of disseminated NTM infection in a woman from Laos who showed IFN-γ autoantibodies. We also conducted a literature review to review similar cases.

A previously healthy 50-year-old woman from Laos was referred to Hospices Civils de Lyon, France, for fever and generalized lymphadenopathies; pathological examination revealed a nonnecrotizing granuloma. Culture yielded Mycobacterium fortuitum. After a 3-month azithromycin/ciprofloxacin regimen, symptoms resolved. A few months later, a computed tomography scan showed persistence of enlarged mediastinal lymph nodes; biopsy disclosed nonspecific sinus histiocytosis. Mycobacterial cultures were negative. One month later, the patient reported intense dorsal spine pain. She was diagnosed with multifocal vertebral osteomyelitis, and tissue specimens tested positive for M. intracellulare. Azithromycin/ciprofloxacin was prescribed again. At 6 months, lesions worsened, and epidural abscess led to spinal cord compression, requiring decompressive laminectomy. Postoperative cultures were positive for M. intracellulare. Treatment was changed to azithromycin, rifabutin, ethambutol, and amikacin for 2 weeks, then rifabutin and moxifloxacin, which led to clinical improvement.

The patient experienced 2 episodes of thoracic herpes zoster concomitantly to each episode of illness. The recurrent NTM infection led us to investigate her immune status. Results of standard immunologic tests (leukocyte, serum level of immunoglobulin isotypes, and T-, B-, and NK0cell counts) and in vitro T-cell proliferation were normal; HIV testing was negative. Given its critical role in Mycobacteria clearance by host cells, we used IFN-γ whole-blood activation to investigate further. IFN-γ plasma level was undetectable, excluding complete forms of IFN-γ R1/R2. IL-12 p40 subunit plasmatic level was decreased, but the IL-12 receptor was present and functional. In addition, both BCG and IL-12 stimulations failed to trigger IFN-γ production. Ultimately, the detection of IFN-γ autoantibodies in high levels explained these abnormalities. Antimycobacterial treatment was continued for 2 years, then changed to azithromycin suppressive therapy; IFN-γ autoantibodies remained positive at the time of this report, 2 years after azithromycin initiation.

We reviewed the literature and found 63 other cases of IFN-γ autoantibody-related NTM infection with sufficient detail to provide clinically relevant material for optimizing disease management (Table; Technical Appendix). These cases overwhelmingly occurred among Asian populations (92.2%) (2). Disease incidence may be underestimated, as suggested by large studies describing Asian patients with disseminated NTM infection without evidence of impaired immunity but for whom the IL-12/IFN-γ axis was not investigated (3). This acquired autoantibodies–mediated immunodeficiency is more frequent among women, in whom the disease typically manifests in the second half of adult life (median 48 [IQR 44.8–60.0] years of age). Reported female:male sex ratios are 23/36 in Asian-born (n = 59) and 4/1 in non-Asian (n = 5) case-patients, respectively. Mechanism of the disease is briefly discussed in the Technical Appendix.

M. avium complex predominated in the literature review, accounting for 40.4% of cases, followed by M. abscessus (31.6%) and M. fortuitum (12.3%). Infections were mostly multifocal, affecting lymph nodes (n = 51, 79.7%), osteoarticular tissue (n = 37, 57.8%), lungs (n = 30, 46.9%), and skin and/or soft tissues (n = 24, 37.5%). Aside from NTM infections, other opportunistic infections were reported in 39 (75.0%) patients, mostly Herpesviridae reactivations (44.2%) and Salmonella infections (25.0%) (36).

Specific treatment for IFN-γ autoantibodies–associated NTM infection is not codified and required prolonged, multiple-drug regimens. The median treatment duration for the studies we reviewed was 31 (IQR 22.8–60.0) months. In some studies, clinicians used IFN-γ administration (5 patients, 1 of whom was cured), but treatment likely was invalidated by the autoimmune-driven inhibitory activity (5,2,7). Other strategies included intravenous immunoglobulin (n = 2), plasmapheresis (n = 2), and cyclophosphamide (n = 1) (79). The use of rituximab, a chimeric anti-CD20 monoclonal antibody targeting B-cells, has been recently associated with clinical response and decrease in IFN-γ autoantibody levels as well as neutralizing ability (6,7).

Final outcome was available for 56 patients who completed the intensive treatment phase; 21 (37.5%) were declared cured. Six (10.7%) patients died, and 29 (51.8%) had persistent or relapsing infections. At the time of this report, additional patients were still being treated and showed improvement of symptoms. Despite this high rate of failure, long-term antimicrobial drug suppressive therapy has rarely been proposed as a causal factor. The origin of the case we report was related to the use of azithromycin suppressive therapy, similarly to disseminated NTM disease prophylaxis in HIV-infected patients before the era of highly active antiretroviral therapies (10), assuming the risk/benefit balance including the possibility of NTM macrolide-resistant strain selection.

IFN-γ autoantibodies are evidence of acquired immunodeficiency that should be considered in cases of unexplained disseminated NTM infections in Asian-born persons. Use of immunomodulation strategies is still debated, and long-term suppressive treatment should be considered for persisting high levels of neutralizing antibodies.

Top

Acknowledgment

We thank Lyon tuberculosis study group members F. Ader, F. Biron, A. Boibieux, A. Bouaziz, E. Braun, G. Catho, N. Charhon, C. Chidiac, W. Chumbi-Flores, S. Couraud, G. Devouassoux, O. Dumitrescu, S. Ernesto, T. Ferry, D. Floret, N. Freymond, S. Gardes, S. Gerbier-Colomban, Y. Gillet, S. Goutelle, J. Grando, R. Grima, L. Hees, J. Karsenty, L. Kiakouama-Maleka, G. Lina, J. M. Maury, P. Miailhes, P. Nesme, T. Perpoint, E. Perrot, A. S. Ronnaux-Baron, S. Roux, J. Saison, A. Senechal, P. J. Souquet, H. Thai Van, F. Tronc, F. Valour, and P. Vanhems for choosing research field priorities and editing one another’s reports article for this manuscript.

Top

Florent ValourComments to Author , Thomas Perpoint, Agathe Sénéchal, Xiao-Fei Kong, Jacinta Bustamante, Tristan Ferry, Christian Chidiac, Florence Ader, on behalf of the Lyon TB study group
Author affiliations: Hospices Civils de Lyon, International Center for Research in Infectiology, Université Claude Bernard Lyon 1, INSERM U1111, Lyon, France (F. Valour, T. Perpoint, A. Sénéchal, T. Ferry, C. Chidiac, F. Ader); Necker-Enfants Malades Hospital, INSERM, Paris Descartes University, Paris, France (J. Bustamante); St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University, New York, New York, USA (X.-E. Kong, J. Bustamante)

Top

References

  1. Wu  U-I, Holland  SM. Host susceptibility to non-tuberculous mycobacterial infections. Lancet Infect Dis. 2015;15:96880. DOIPubMedGoogle Scholar
  2. Kampmann  B, Hemingway  C, Stephens  A, Davidson  R, Goodsall  A, Anderson  S, Acquired predisposition to mycobacterial disease due to autoantibodies to IFN-γ. J Clin Invest. 2005;115:24808. DOIPubMedGoogle Scholar
  3. Browne  SK, Burbelo  PD, Chetchotisakd  P, Suputtamongkol  Y, Kiertiburanakul  S, Shaw  PA, Adult-onset immunodeficiency in Thailand and Taiwan. N Engl J Med. 2012;367:72534. DOIPubMedGoogle Scholar
  4. Chi  C-Y, Chu  C-C, Liu  J-P, Lin  C-H, Ho  M-W, Lo  W-J, Anti-IFN-γ autoantibodies in adults with disseminated nontuberculous mycobacterial infections are associated with HLA-DRB1*16:02 and HLA-DQB1*05:02 and the reactivation of latent varicella-zoster virus infection. Blood. 2013;121:135766. DOIPubMedGoogle Scholar
  5. Döffinger  R, Helbert  MR, Barcenas-Morales  G, Yang  K, Dupuis  S, Ceron-Gutierrez  L, Autoantibodies to interferon-gamma in a patient with selective susceptibility to mycobacterial infection and organ-specific autoimmunity. Clin Infect Dis. 2004;38:e104. DOIPubMedGoogle Scholar
  6. Czaja  CA, Merkel  PA, Chan  ED, Lenz  LL, Wolf  ML, Alam  R, Rituximab as successful adjunct treatment in a patient with disseminated nontuberculous mycobacterial infection due to acquired anti-interferon-γ autoantibody. Clin Infect Dis. 2014;58:e1158. DOIPubMedGoogle Scholar
  7. Browne  SK, Zaman  R, Sampaio  EP, Jutivorakool  K, Rosen  LB, Ding  L, Anti-CD20 (rituximab) therapy for anti-IFN-γ autoantibody-associated nontuberculous mycobacterial infection. Blood. 2012;119:39339. DOIPubMedGoogle Scholar
  8. Koya  T, Tsubata  C, Kagamu  H, Koyama  K, Hayashi  M, Kuwabara  K, Anti-interferon-gamma autoantibody in a patient with disseminated Mycobacterium avium complex. J Infect Chemother. 2009;15:11822. DOIPubMedGoogle Scholar
  9. Baerlecken  N, Jacobs  R, Stoll  M, Schmidt  RE, Witte  T. Recurrent, multifocal Mycobacterium avium-intercellulare infection in a patient with interferon-gamma autoantibody. Clin Infect Dis. 2009;49:e768. DOIPubMedGoogle Scholar
  10. Havlir  DV, Dubé  MP, Sattler  FR, Forthal  DN, Kemper  CA, Dunne  MW, Prophylaxis against disseminated Mycobacterium avium complex with weekly azithromycin, daily rifabutin, or both. California Collaborative Treatment Group. N Engl J Med. 1996;335:3928. DOIPubMedGoogle Scholar

Top

Table

Top

Cite This Article

DOI: 10.3201/eid2206.151860

Related Links

Top

Table of Contents – Volume 22, Number 6—June 2016

EID Search Options
presentation_01 Advanced Article Search – Search articles by author and/or keyword.
presentation_01 Articles by Country Search – Search articles by the topic country.
presentation_01 Article Type Search – Search articles by article type and issue.

Top

Comments

Please use the form below to submit correspondence to the authors or contact them at the following address:

Florent Valour, Department of Infectious Diseases, Hospices Civils de Lyon, 103 Grande-Rue de la Croix-Rousse, 69004 Lyon, France

Send To

10000 character(s) remaining.

Top

Page created: May 17, 2016
Page updated: May 17, 2016
Page reviewed: May 17, 2016
The conclusions, findings, and opinions expressed by authors contributing to this journal do not necessarily reflect the official position of the U.S. Department of Health and Human Services, the Public Health Service, the Centers for Disease Control and Prevention, or the authors' affiliated institutions. Use of trade names is for identification only and does not imply endorsement by any of the groups named above.
file_external