1
|
Menon AR, Prest RJ, Tobin DM, Champion PA. Mycobacterium marinum as a model for understanding principles of mycobacterial pathogenesis. J Bacteriol 2025; 207:e0004725. [PMID: 40304497 PMCID: PMC12096832 DOI: 10.1128/jb.00047-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Mycobacterium marinum is a fish pathogen that has become a powerful and well-established model that has accelerated our understanding of the mechanisms of mycobacterial disease. M. marinum is a versatile surrogate for understanding the closely related human pathogen M. tuberculosis, which causes tuberculosis in humans. M. marinum has defined key mechanisms of pathogenesis, both shared with M. tuberculosis and unique to this species. In this review, we discuss the discovery of M. marinum as an occasional human pathogen, the shared aspects of pathogenesis with M. tuberculosis, and how M. marinum has been exploited as a model to define the molecular mechanisms of mycobacterial pathogenesis across several phases of infection.
Collapse
Affiliation(s)
- Aruna R. Menon
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Rebecca J. Prest
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
2
|
Xu X, Wang L, Sun Y, Yang C, Wang X, Guo P, Mei D. Unveiling the differences: infection disorders associated with tumor necrosis factor α inhibitors in pediatric patients-a pharmacovigilance study (2004-2023). Eur J Pediatr 2025; 184:324. [PMID: 40317305 DOI: 10.1007/s00431-025-06152-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
The increasing use of tumor necrosis factor inhibitors (TNFi) in pediatric patients has raised concerns about their potential impact on the immune system and related adverse events. Infection-related adverse events (AEs) caused by TNFi have already raised widespread concerns in real-world settings. This study aims to comprehensively analyze and summarize the infection-related AEs associated with TNFi in pediatric patients. A retrospective pharmacovigilance study was conducted to identify cases of TNFi-related infections reported to the FDA Adverse Event Reporting System (FAERS) database between Q1 2004 and Q1 2023. TNFi reports were carefully reviewed to exclude confounding factors like other AEs, concomitant medications, and prescription indications. Proportionality analysis was conducted by comparing TNFi reports to the entire FAERS database to identify infection-related AEs significantly associated with TNFi use. Infection-related AEs accounted for 8.36% of all TNFi-related adverse event reports in the FAERS database. A total of 8050 cases of TNFi-associated infections were identified in the pediatric population, with 2.57% of reports resulting in fatalities. Infliximab and golimumab showed a stronger association with infection-related AEs compared to other TNFi. Notably, only adalimumab shows a lower risk of viral infections, while it exhibits an increased risk of bacterial and mycobacterial infections, similar to other TNFi. CONCLUSIONS This study identified a significant association between TNFi use and infection-related AEs in pediatric patients, providing the foothold for further research. However, due to its retrospective nature, further investigations are warranted to confirm these findings and identify potential risk factors in a controlled, prospective study setting. WHAT IS KNOWN • There is sufficient evidence to demonstrate the infection risk associated with TNFi in adult patients. • Pediatric patients, whose immune systems are still developing, are more vulnerable to certain infections. WHAT IS NEW • There is a significant association between TNFi use and infection-related adverse events in pediatric patients, and different TNFi have distinct infection profiles.
Collapse
Affiliation(s)
- Xiaolin Xu
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Luquan Wang
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yixin Sun
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Changqing Yang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoling Wang
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Peng Guo
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
| | - Dong Mei
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
| |
Collapse
|
3
|
Goldman A, Raschi E, Druyan A, Sharif K, Lahat A, Ben‐Zvi I, Ben‐Horin S. Gastrointestinal Perforations Associated With JAK Inhibitors: A Disproportionality Analysis of the FDA Adverse Event Reporting System. United European Gastroenterol J 2025; 13:566-575. [PMID: 39736095 PMCID: PMC12090834 DOI: 10.1002/ueg2.12736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Gastrointestinal perforations have been reported in a small number of rheumatoid arthritis (RA) patients treated with Janus kinase (JAK) inhibitors in clinical trials. However, large-scale postmarketing data repositories are needed to further investigate this potentially rare but serious adverse event. METHODS A retrospective, pharmacovigilance study of the FDA adverse event reporting system (July 2014 to September 2023) assessing the reporting of gastrointestinal perforations following JAK inhibitors compared to biological disease-modifying antirheumatic drugs (bDMARDs) in RA patients. The adjusted reporting odds ratio (adj.ROR) was calculated using a multivariable logistic regression model. RESULTS Of 399,983 RA patients included in the study, 76,446 were treated with JAK inhibitors (tofacitinib, n = 52,365; upadacitinib, n = 21,856; baricitinib, n = 2225) and 323,537 were treated with bDMARDs (TNF inhibitors, rituximab, and abatacept). Overall, 230 cases of gastrointestinal perforation following JAK inhibitors were identified, with a median time of 9 (IQR: 4-22) months from treatment initiation. Compared with bDMARDs, JAK inhibitors were associated with a higher-than-expected reporting of gastrointestinal perforations (adj.ROR = 1.98[1.69-2.31]). Increased reporting of gastrointestinal perforations was observed among recipients of JAK inhibitors or bDMARDs who used steroids or non-steroidal anti-inflammatory drugs concurrently (adj.ROR = 2.82 [2.41-3.31]). Perforations of both the upper and lower gastrointestinal tract were significantly over-reported (n = 51, adj.ROR = 1.55 [1.12-2.14], n = 143, adj.ROR = 1.78 [1.46-2.17], respectively). Furthermore, the safety signal was significant across all JAK inhibitors: tofacitinib (n = 125, adj.ROR = 1.52 [1.25-1.85]), upadacitinib (n = 84, adj.ROR = 2.73 [2.17-3.44]), and baricitinib (n = 21, adj.ROR = 5.38 [3.46-8.37]). CONCLUSION In this global pharmacovigilance study, all JAK inhibitors were associated with increased reporting of gastrointestinal perforations compared with bDMARDs in RA patients. Until more data on IBD patients emerge, careful surveillance and increased clinicians' awareness should also be advocated for this population.
Collapse
Affiliation(s)
- Adam Goldman
- Department of Internal MedicineSheba Medical CenterRamat‐GanIsrael
- Department of Epidemiology and Preventive MedicineSchool of Public HealthSchool of MedicineTel Aviv UniversityTel AvivIsrael
- The Sheba Talpiot Medical Leadership ProgramSheba Medical CenterRamat‐GanIsrael
| | - Emanuel Raschi
- Pharmacology UnitDepartment of Medical and Surgical SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Amit Druyan
- Department of Internal MedicineSheba Medical CenterRamat‐GanIsrael
- School of MedicineFaculty of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| | - Kassem Sharif
- School of MedicineFaculty of MedicineTel‐Aviv UniversityTel‐AvivIsrael
- Department of GastroenterologySheba Medical CenterRamat‐GanIsrael
| | - Adi Lahat
- School of MedicineFaculty of MedicineTel‐Aviv UniversityTel‐AvivIsrael
- Department of GastroenterologySheba Medical CenterRamat‐GanIsrael
| | - Ilan Ben‐Zvi
- Department of Internal MedicineSheba Medical CenterRamat‐GanIsrael
- The Sheba Talpiot Medical Leadership ProgramSheba Medical CenterRamat‐GanIsrael
- School of MedicineFaculty of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| | - Shomron Ben‐Horin
- The Sheba Talpiot Medical Leadership ProgramSheba Medical CenterRamat‐GanIsrael
- School of MedicineFaculty of MedicineTel‐Aviv UniversityTel‐AvivIsrael
- Department of GastroenterologySheba Medical CenterRamat‐GanIsrael
| |
Collapse
|
4
|
Muzanyi G, Bark CM. Severe Tuberculosis. Med Clin North Am 2025; 109:641-650. [PMID: 40185552 DOI: 10.1016/j.mcna.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
The global tuberculosis (TB) pandemic continues despite living in the era of the greatest advances in human medicine. Severe TB in the form of meningitis and miliary TB kills and disables people around the world every day. It disproportionately affects the most vulnerable populations, especially children. Diagnosis and treatment are challenging, and most have poor outcomes, often never receiving treatment at all. Dedicated TB researchers continue to make improvements in the management of severe TB, but the long-term success will require a global commitment to improving health care for poor people around the world.
Collapse
Affiliation(s)
- Grace Muzanyi
- Uganda-Case Western Reserve University Research Collaboration, PO Box 663, Kampala, Uganda
| | - Charles M Bark
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
5
|
Penã Avila J, Simmons J, Figueiredo MC, Turner M, Cordeiro-Santos M, Rolla VC, Kristki AL, Gangula R, Nochowicz C, Ram R, Bailin S, Mallal S, Gaudieri S, Alves E, Barreto-Duarte BB, Queiroz ATL, Nakaya HI, Andrade BB, Sterling TR, Kalams SA. Single-cell immune profiling at time of M. tuberculosis exposure reveals antigen-reactive programs that predict progression to active disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.29.25326433. [PMID: 40343021 PMCID: PMC12060959 DOI: 10.1101/2025.04.29.25326433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Early delineation of host immune responses at the moment of Mycobacterium tuberculosis (Mtb) exposure and infection is critical to identify individuals at risk of progressing to active tuberculosis (TB). We performed single-cell transcriptional profiling of over 500,000 peripheral blood mononuclear cells from 57 HIV-negative close contacts of TB cases in Brazil, including 25 individuals who developed active disease within two years (progressors) and 32 matched controls who remained disease-free (non-progressors). Cells were stimulated separately with the MTB300 peptide pool or irradiated Mtb (gRV), enabling resolution of antigen-reactive states across adaptive (CD4⁺ T-cells expressing abundant cytokines including IFNG, TNF, and IL17F) and trained-innate lineages, such as NK cells (producing GM-CSF, IFNG, CCL3, CCL4) and monocytes (GM-CSF, IL12B, IL36G). Progressors exhibited early hyper-metabolic CD4⁺ T-cell programs and proliferative NK cell signatures, whereas non-progressors preferentially upregulated complement activation and CCL3/4-driven chemokine signaling in monocytes. Notably, among progressors, gene expression profiles within antigen-reactive CD4⁺ T-cells and monocytes predicted the timing of progression to active TB. Together, these findings reveal high frequencies and functional diversity of antigen-reactive cells in Mtb-exposed individuals and nominate tractable immune correlates for the rational design of next-generation TB vaccines.
Collapse
|
6
|
Sismanlar Eyuboglu T, Aslan AT, Medeni V, Can S, Ata N, Ulgu MM, Birinci S. Tuberculosis in children and adolescents using biological agents: a nationwide cohort study from Turkey. BMC Pulm Med 2025; 25:196. [PMID: 40281511 PMCID: PMC12023584 DOI: 10.1186/s12890-025-03616-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The use of biological agents in various diseases in children has been increasing and the risk of tuberculosis (TB) increases with them. We aimed to investigate the role of biological agents in children diagnosed with TB in a moderate level of TB country where TB screening is mandatory before and during biological agent treatment. STUDY DESIGN AND METHODS This was a retrospective cohort study. All patients who were 0-18 years old and diagnosed with TB-related ICD-10 in the national health database system between 2018 and 2023 were included in the study. The number of patients, demographic characteristics, treatments used by the patients, underlying diseases, and organ involvement of TB were recorded. Children using and not using biological agents were compared. RESULTS A total of 4351 children were diagnosed with TB, and 1.9% of them were treated with biological agents. The age of diagnosis was older (p = 0.001), and both pulmonary and extrapulmonary involvement was more frequent in children using biological agents (p = 0.001). Pulmonary involvement was more frequent in rheumatological diseases (p = 0.001), and naproxen usage was higher in children with pulmonary involvement (p = 0.014). Naproxen was found to increase the risk of pulmonary TB in children using biological agents (OR:3.824, p = 0.033). CONCLUSIONS The low frequency of TB may be due to effective TB screening before and during the therapy. The age of diagnosis was older, pulmonary and extrapulmonary TB involvement was more common in children using biological agents, which may be related to the immunosuppressive effects. Children using biological agents who are also using naproxen should be closely followed up in terms of pulmonary TB.
Collapse
Affiliation(s)
| | - Ayse Tana Aslan
- Faculty of Medicine, Department of Pediatric Pulmonology, Gazi University, Ankara, Turkey
| | - Volkan Medeni
- Faculty of Medicine, Department of Public Health, Gazi University, Ankara, Turkey
| | - Sinem Can
- Republic of Turkey Ministry of Health, General Directorate of Health Information Systems, Ankara, Turkey
| | - Naim Ata
- Republic of Turkey Ministry of Health, General Directorate of Health Information Systems, Ankara, Turkey
| | - Mustafa Mahir Ulgu
- Republic of Turkey Ministry of Health, General Directorate of Health Information Systems, Ankara, Turkey
| | | |
Collapse
|
7
|
Day CL, Njuguna IN, Cranmer LM, Whatney WE, Pearson RA, Lindestam Arlehamn CS, Sette A, LaCourse SM, Escudero JN, Sasser LE, Mugo C, Okinyi HM, Maleche-Obimbo E, Wamalwa DC, John-Stewart GC. Patterns and Cofactors of Polyfunctional Mycobacteria-Specific T-Cell Response Restoration Following 6-Month Antiretroviral Treatment in Children With HIV. J Infect Dis 2025; 231:957-966. [PMID: 39693245 PMCID: PMC11998549 DOI: 10.1093/infdis/jiae630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Despite immune restoration after initiation of antiretroviral treatment (ART), the risk of tuberculosis (TB) persists in children with HIV (CHIV). We determined patterns of immune restoration of mycobacteria-specific T cells following ART in CHIV. METHODS CD4 and CD8 T-cell activation and memory phenotype and functional profiles before and 6 months after ART were evaluated in peripheral blood mononuclear cells from CHIV enrolled in the PUSH study (NCT02063880) in Nairobi, Kenya. T-cell expression of cytokines and activation-induced markers were measured following stimulation of peripheral blood mononuclear cells with a pool of 300 peptides from TB (MTB300) or staphylococcal enterotoxin B. RESULTS Among 47 CHIV (median age, 1.5 years), staphylococcal enterotoxin B-induced Th1 cytokine+ and activation-induced marker+ CD4 cell frequencies increased significantly after 6 months of ART. Although MTB300-specific CD4 and CD8 cell frequency did not increase after ART, polyfunctional capacity of MTB300-specific CD4 cells expressing combinations of Th1 cytokines with CD40L increased significantly after ART. Baseline age, immune activation, and effector memory CD4 levels were associated with less restoration of MTB300-specific polyfunctional CD4 cells, whereas CD4 percentage and levels of naive CD4 cells following ART were associated with improved MTB300-specific polyfunctional capacity. CONCLUSIONS Despite increases in Th1 cytokine production, deficits in mycobacteria-specific CD4 cells persisted 6 months after ART, with higher deficits in older CHIV with more immunosuppression, higher immune activation, and lower proportion of naive CD4 cells. These findings may explain persistent TB risk during early ART among CHIV and identify those at highest risk.
Collapse
Affiliation(s)
- Cheryl L Day
- Department of Microbiology and Immunology, Emory University School of Medicine
- Emory Vaccine Center, Emory University, Atlanta, Georgia
| | - Irene N Njuguna
- Department of Medical Research, Kenyatta National Hospital, Nairobi, Kenya
- Department of Global Health, University of Washington, Seattle
| | - Lisa Marie Cranmer
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta
- Children's Healthcare of Atlanta, Georgia
| | - Wendy E Whatney
- Department of Microbiology and Immunology, Emory University School of Medicine
- Emory Vaccine Center, Emory University, Atlanta, Georgia
| | - Rachel A Pearson
- Department of Microbiology and Immunology, Emory University School of Medicine
- Emory Vaccine Center, Emory University, Atlanta, Georgia
| | - Cecilia S Lindestam Arlehamn
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, California
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, California
- Department of Medicine, University of California San Diego
| | - Sylvia M LaCourse
- Department of Global Health, University of Washington, Seattle
- Division of Allergy and Infectious Diseases, Department of Medicine
- Department of Epidemiology, University of Washington, Seattle
| | | | - Loren E Sasser
- Department of Microbiology and Immunology, Emory University School of Medicine
- Emory Vaccine Center, Emory University, Atlanta, Georgia
| | - Cyrus Mugo
- Department of Medical Research, Kenyatta National Hospital, Nairobi
| | | | | | - Dalton C Wamalwa
- Department of Pediatrics and Child Health, University of Nairobi, Kenya
| | - Grace C John-Stewart
- Department of Global Health, University of Washington, Seattle
- Division of Allergy and Infectious Diseases, Department of Medicine
- Department of Epidemiology, University of Washington, Seattle
- Department of Pediatrics, University of Washington, Seattle
| |
Collapse
|
8
|
Warren WG, Osborn M, Yates A, O'Sullivan SE. ART26.12, an FABP5 Inhibitor, Shows Efficacy in Preclinical Psoriasis Models. J Invest Dermatol 2025:S0022-202X(25)00393-8. [PMID: 40210114 DOI: 10.1016/j.jid.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/12/2025]
Abstract
FABP5 is upregulated in psoriasis. This study assessed the efficacy of the potent, selective, and orally active FABP5 inhibitor ART26.12 in preclinical psoriasis models. In vitro, reconstructed human epidermis was stimulated with cytokines (IL-17 + IL-22 + TNF at 3 ng/ml each) and treated with ART26.12 (1, 3, or 10 μM) or Jak1 inhibitor (10 μM) for 24 hours; after which, 64 psoriasis-related genes were measured. ART26.12 (3 and 10 μM) treatment reduced cytokines, chemokines, and markers of keratinocyte proliferation/differentiation and increased certain antimicrobial peptides. In vivo, ART26.12 (25 or 100 mg/kg twice a day) or BMS-986165 (TYK2 inhibitor; 10 mg/kg once a day) was given orally for 10 days in the imiquimod mouse model. Imiquimod increased psoriasis-like symptoms. ART26.12 (25 mg/kg twice a day) and BMS-986165 comparably reduced psoriasis-like symptoms by day 6 of imiquimod treatment. Histopathology showed that ART26.12 reduced symptom severity, for example, hyperkeratosis, parakeratosis, epidermal acanthosis, and inflammatory infiltrates. Proteomic analysis indicated that ART26.12 rescued the expression of FLG-2; promoted epidermal differentiation complex-associated proteins; and modulated peroxisome proliferator-activated receptor, NF-kB, and protein kinase C pathways likely downstream of lipid modulation. Lipidomic analysis showed widespread modulation, including ceramides and linoleic acid derivatives. These data suggest that ART26.12 may be a potential psoriasis treatment.
Collapse
|
9
|
O'Garra A. From Cytokines to Tuberculosis and Back: My Journey to Understanding the Immune Response to Infection. Annu Rev Immunol 2025; 43:1-28. [PMID: 40279305 DOI: 10.1146/annurev-immunol-010824-041601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
I felt honored by the invitation to write this autobiography, although it was an arduous task to describe my journey through science: first bacterial adhesion, then cytokine function, and then immune responses in tuberculosis. Since only seven women had been authors of autobiographies for the Annual Review of Immunology, I felt I couldn't refuse to contribute to Volume 43 of the journal. Moreover, this was a good occasion to record my appreciation to all the lab members and collaborators for their contributions over the last 40 years, to remember the exciting times, and to reflect on the obstacles we faced. I often reflect on this line that is commonly attributed to Winston Churchill: Success is not final; failure is not fatal: It is the courage to continue that counts. What kept me going was a burning desire to know how things work and find enjoyment in the discovery. This passion to understand immune responses to infection remains with me to this day. I thank all those I have interacted with for the support and friendship they provided.
Collapse
Affiliation(s)
- Anne O'Garra
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
10
|
Minagi A, Nawa H, Goda M, Niimura T, Miyata K, Hamano H, Zamami Y, Ishizawa K. Evaluation of Interstitial Lung Disease Complications Caused by Biologic Agents Using a Spontaneous Adverse Drug Reaction Reporting Database. Pharmacol Res Perspect 2025; 13:e70063. [PMID: 39984304 PMCID: PMC11845275 DOI: 10.1002/prp2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 02/23/2025] Open
Abstract
Interstitial lung disease (ILD) is a clinically relevant adverse event associated with biologic agent use. However, the current incidence of ILD remains unclear as large-scale risk assessments of biologic agents have not been conducted. The aim of this study was to clarify the association between biologic agent use and ILD development in clinical practice by detecting adverse event signals using a spontaneous adverse drug reaction database. The VigiBase database is used for spontaneous adverse event reporting. The analysis focused on nine biologics used to treat psoriasis, rheumatoid arthritis, and Crohn's disease. The safety of each biologic agent was evaluated using the information component signal detection method. There were 32,520,983 reports in VigiBase, of which 68,489 (0.21%) were for ILD. Signals were mainly detected for tumor necrosis factor-α inhibitors when the information component for ILD caused by biologic agents was calculated. Comorbidity analysis in patients who developed ILD and analysis of the time from the start of treatment with each drug to ILD onset showed differences for each biologic agent. ILD is a serious adverse effect of biologic agents, and there are several cases in which a causal relationship with ILD development cannot be ruled out. The occurrence of interstitial ILD should be noted when using biologics, particularly TNF-α inhibitors.
Collapse
Affiliation(s)
- Ayu Minagi
- Department of Pharmacy, Faculty of PharmacyShujitsu UniversityOkayamaJapan
| | - Hideki Nawa
- Department of Pharmacy, Faculty of PharmacyShujitsu UniversityOkayamaJapan
- Department of PharmacyOkayama University HospitalOkayamaJapan
- Department of PharmacyOkayama Kyokuto HospitalOkayamaJapan
| | - Mitsuhiro Goda
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Takahiro Niimura
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Koji Miyata
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Hirofumi Hamano
- Department of PharmacyOkayama University HospitalOkayamaJapan
| | - Yoshito Zamami
- Department of PharmacyOkayama University HospitalOkayamaJapan
| | - Keisuke Ishizawa
- Department of PharmacyTokushima University HospitalTokushimaJapan
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| |
Collapse
|
11
|
de Souza VA, Caparroz ALMA, Trevisani VFM, Gomes Tavares ACFM, de Melo AKG, Trajman A, Medeiros-Ribeiro ACD, Pinheiro MDM, Xavier RM, Monticielo OA, Guimarães MFBDR, Sztajnbok F, Bombarda S, Chebli LA, Kakehasi AM, Bierrenbach AL, Gomides Reis APM, Gomes Bica BER, Marques CDL, Flores C, Rodrigues DS, Paiva EDS, Matos ED, Costa Johansen FD, Bacha HA, de Carvalho JS, Provenza JR, Lira Machado KLL, da Mota LMH, Valadares LDDA, Rocha Loures MAAD, Pretti Dalcolmo MM, Bortoletto MCDC, Ferreira Lopes MIB, Abreu Vieira RMRD, Romiti R, Saad-Hossne R, Ciconelli RM, Feijó Azevedo V, Augusto VM, Alves Cruz V, Salviato Pileggi GC. Brazilian recommendations for the management of tuberculosis infection in immune-mediated inflammatory diseases. Adv Rheumatol 2025; 65:18. [PMID: 40114289 DOI: 10.1186/s42358-025-00449-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The risk of tuberculosis infection (TBI) and its progression to tuberculosis disease (TBD) among persons with immune-mediated inflammatory diseases (IMID) results from a complex interplay of patient and disease characteristics, immunosuppression level, and the epidemiological context. Brazilian recommendations are unclear about TBI screening and its preventive treatment (TPT) in persons with IMID. OBJECTIVE To provide a comprehensive and evidence-based guideline for managing TBI in persons with IMID in Brazil. METHODS This task force was constituded by 42 specialists with interest in IMID and TBD. A core leadership team (CLT) drafted fourteen clinical questions on the risk of tuberculosis and indications of TPT among persons with IMID who started, or are about to start immunosuppressive drugs. The CLT supervised the systematic reviews and formulated the recommendations. The experts voted using the Delphi Method. RESULTS Nine recommendations were established. More than 80% of panelists voted "agree" and "strongly agree" with all statements. In brief, all persons with IMID starting or about to start immunosuppressive treatment should undergo tuberculin skin testing (TST) or interferon-gamma release assays (IGRAs), a chest imaging test and investigation of contact with active pulmonary or laryngeal TBD. TPT is mandatory for those with any positive result after excluding TBD. Exceptions include individuals with a history of TBD or a past positive TBI infection test. IGRA is preferred only in persons BCG-vaccinated in the past 2 years. Those with inconclusive IGRA results can have the test repeated once, and TPT should be offered if it remains indeterminate. TST or IGRA should be repeated yearly, for three years, when the previous test was negative, when starting or changing to a different class of immunosuppressive drug. Overall, the included studies had a low quality of evidence and high risk of bias. CONCLUSIONS These guidelines are meant to improve the management of TBI in IMID. Health professionals must consider the epidemiological risk, host features, the social scenario, the characteristics of the disease, the access to health resources, and the development of an individualized plan for every patient.
Collapse
Affiliation(s)
| | | | | | | | | | - Anete Trajman
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | | | - Ricardo Machado Xavier
- Hospital de Clínicas de Porto Alegre - Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Odirlei Andre Monticielo
- Hospital de Clínicas de Porto Alegre - Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Flavio Sztajnbok
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Sidney Bombarda
- Secretaria de Estado da Saúde de São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | - Cristina Flores
- Centro de Tratamento de Doenças Inflamatórias Intestinais e Imunomediadas, Porto Alegre, Brazil
| | | | | | | | - Fernanda Dockhorn Costa Johansen
- Coordenação-Geral de Vigilância da Tuberculose, Micoses Endêmicas e Micobactérias não Tuberculosas, (Ministério da Saúde), Brasília, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | - Ricardo Romiti
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Rogerio Saad-Hossne
- Faculdade de Medicina de Botucatu - Universidade Estadual Paulista - UNESP, São Paulo, Brazil
| | - Rozana Mesquita Ciconelli
- Universidade Federal de São Paulo (UNIFESP/ EPM), São Paulo, Brazil
- Hospital Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
12
|
Chancharoenthana W, Kamolratanakul S, Rotcheewaphan S, Leelahavanichkul A, Schultz MJ. Recent advances in immunopathogenesis and clinical practice: mastering the challenge-managing of non-tuberculous mycobacteria. Front Immunol 2025; 16:1554544. [PMID: 40176807 PMCID: PMC11961655 DOI: 10.3389/fimmu.2025.1554544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/26/2025] [Indexed: 04/04/2025] Open
Abstract
Non-tuberculous mycobacteria (NTM) are widespread environmental pathogens that can lead to significant disease burden, particularly in immunocompromised individuals, but also in those with a normal immune system. The global incidence of NTM is increasing rapidly, with Mycobacterium avium complex (MAC) being one of the most common types. The immunopathogenesis of the MAC involves a complex interaction between the bacteria and the host immune system. MAC survives and replicates within macrophages by preventing the fusion of phagosomes and lysosomes. The mycobacteria can neutralize reactive oxygen and nitrogen species produced by the macrophages through their own enzymes. Additionally, MAC modulates cytokine production, allowing it to suppress or regulate the immune response. Diagnosing MAC infections can be challenging, and the effectiveness of available treatments may be limited due to MAC's unpredictable resistance to various antimycobacterial drugs in different regions. Treating MAC infection requires a collaborative approach involving different healthcare professionals and ensuring patient compliance. This review aims to shed light on the complexities of MAC infection treatment, discussing the challenges of MAC infection diagnosis, pharmacological considerations, such as drug regimens, drug monitoring, drug interactions, and the crucial role of a multidisciplinary healthcare team in achieving the best possible treatment outcomes for patients.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Supitcha Kamolratanakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence on Translational Research in Inflammatory and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Marcus J. Schultz
- Department of Intensive Care & Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A), Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Hoerter A, Petrucciani A, Bonifacio J, Arnett E, Schlesinger LS, Pienaar E. Timing matters in macrophage/CD4+ T cell interactions: an agent-based model comparing Mycobacterium tuberculosis host-pathogen interactions between latently infected and naïve individuals. mSystems 2025; 10:e0129024. [PMID: 39918314 PMCID: PMC11915833 DOI: 10.1128/msystems.01290-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 03/19/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant health challenge. Clinical manifestations of TB exist across a spectrum with a majority of infected individuals remaining asymptomatic, commonly referred to as latent TB infection (LTBI). In vitro models have demonstrated that cells from individuals with LTBI can better control Mtb growth and form granuloma-like structures more quickly, compared to cells from uninfected (Mtb-naïve) individuals. These in vitro results agree with animal and clinical evidence that LTBI protects, to some degree, against reinfection. However, the mechanisms by which LTBI might offer protection against reinfection remain unclear, and quantifying the relative contributions of multiple control mechanisms is challenging using experimental methods alone. To complement in vitro models, we have developed an in silico agent-based model to help elucidate host responses that might contribute to protection against reinfection. Our simulations indicate that earlier contact between macrophages and CD4+ T cells leads to LTBI simulations having more activated CD4+ T cells and, in turn, more activated infected macrophages, all of which contribute to a decreased bacterial load early on. Our simulations also demonstrate that granuloma-like structures support this early macrophage activation in LTBI simulations. We find that differences between LTBI and Mtb-naïve simulations are driven by TNFα and IFNγ-associated mechanisms as well as macrophage phagocytosis and killing mechanisms. Together, our simulations show how important the timing of the first interactions between innate and adaptive immune cells is, how this impacts infection progression, and why cells from LTBI individuals might be faster to respond to reinfection.IMPORTANCETuberculosis (TB) remains a significant global health challenge, with millions of new infections and deaths annually. Despite extensive research, the mechanisms by which latent TB infection (LTBI) confers protection against reinfection remain unclear. In this study, we developed an in silico agent-based model to simulate early immune responses to Mycobacterium tuberculosis infection based on experimental in vitro infection of human donor cells. Our simulations reveal that early interactions between macrophages and CD4+ T cells, driven by TNFα and IFNγ, are critical for bacterial control and granuloma formation in LTBI. These findings offer new insights into the immune processes involved in TB, which could inform the development of targeted vaccines and host-directed therapies. By integrating experimental data with computational predictions, our research provides a robust framework for understanding TB immunity and guiding future interventions to mitigate the global TB burden.
Collapse
Affiliation(s)
- Alexis Hoerter
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Alexa Petrucciani
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | | | - Eusondia Arnett
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | | | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
14
|
Stephens M, Keane K, Roizes S, Defaye M, Altier C, von der Weid PY. Uncovering the therapeutic potential of anti-tuberculoid agent Isoniazid in a model of microbial-driven Crohn's disease. J Crohns Colitis 2025; 19:jjaf032. [PMID: 39987456 PMCID: PMC11920797 DOI: 10.1093/ecco-jcc/jjaf032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Indexed: 02/24/2025]
Abstract
AIMS TNFα has long stood as a hallmark feature of both inflammatory bowel disease and arthritis with its therapeutic potential demonstrated in neutralizing monoclonal antibody treatments such as Infliximab. Due to the high global burden of latent Mycobacterium tuberculosis (TB) infections, prior to receiving anti-TNF therapy, patients testing positive for latent TB are given prophylactic treatment with anti-tuberculoid medications including the first described TB-selective antibiotic, Isoniazid. While this is common clinical practice to prevent the emergence of TB, little is known about whether Isoniazid modifies intestinal inflammation alone. The aim of this study, therefore, was to determine whether Isoniazid presents a novel TB-independent therapeutic option for the treatment of Crohn's disease (CD)-like ileitis and uncover new mechanisms predisposing the host to intestinal inflammation. METHODS The transgenic TNFΔARE mouse model of Crohn's-like terminal ileitis was used. The impact of Isoniazid administration (10 mg/kg/day dose in drinking water) on disease development was monitored between 8 and 12 weeks of age using a variety of behavioral and serological assays. Behavioral and motor functions were assessed using the LABORAS automated monitoring system while systemic and local tissue inflammation were determined at experimental termination using multiplex cytokine analysis. Whole-mount tissue immunofluorescence and fluorescent in situ hybridization were used to qualify changes within the host as well as the microbial compartment of the ileum and associated mesentery. Proposed cellular mechanisms of altered cytokine decay were performed on isolated primary splenocytes in vitro using selective pharmacological agents. RESULTS Compared to age-matched wild-type littermates, TNFΔARE mice display prominent progressive sickness behaviors from 8 through 12 weeks of age indicated by reduced movement, climbing, and rearing. Prophylactic administration of Isoniazid (10 mg/kg/day) is effectively able to protect TNFΔARE mice from this loss of function during the same period. Analysis revealed that Isoniazid was able to significantly reduce both systemic and intestinal inflammation compared to untreated vehicle controls impacting the epithelial colonization of known pathobiont segmented filamentous bacteria (SFB). Reduction in terminal ileal inflammation was also associated to the diminished formation of precursor-tertiary lymphoid organs within the associated ileal mesentery which were found to be associated with endospores derived SFB itself. Finally, we reveal that due to their genetic manipulation, TNFΔARE mice display accelerated posttranscriptional decay of IL-22 mRNA resulting in diminished IL-22 protein production and associated downstream antimicrobial peptide production. CONCLUSIONS Isoniazid protects against the development of intestinal and systemic inflammation in the TNFΔARE model of terminal ileitis by limiting the expansion of mucosal SFB and progression of the associated microbial-driven inflammation. This work highlights a possible mycobacterial-independent function of Isoniazid in limiting CD pathophysiology through limiting the mucosal establishment of pathobionts such as SFB and the association of such microbe-derived endospores linked to the formation of ectopic tertiary lymphoid organs seen commonly in patients.
Collapse
Affiliation(s)
- Matthew Stephens
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| | - Keith Keane
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| | - Simon Roizes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Pierre-Yves von der Weid
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Inflammation Research Network Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N4N1, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Drive NW, Calgary, Alberta T2N4N1, Canada
| |
Collapse
|
15
|
Isa M, Ramos MRR, Kamal S. Infection Risk in Biological Disease-Modifying Anti-rheumatic Drugs. Cureus 2025; 17:e80634. [PMID: 40236366 PMCID: PMC11998624 DOI: 10.7759/cureus.80634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Rheumatology patients on biological disease-modifying anti-rheumatic drugs (bDMARDs) have been proposed to be at a higher risk of infections. Our review summarizes the current evidence behind this theory as well as explores which factors predispose patients to various infections, which agents are more likely to cause infections, and which infections are common in these patients. We also aim to explore updated guidelines on infection prevention in patients on bDMARDs.
Collapse
Affiliation(s)
- Mourushi Isa
- General Medicine, Northern Health, Melbourne, AUS
| | | | - Shahed Kamal
- General Medicine, Northern Health, Melbourne, AUS
| |
Collapse
|
16
|
Barry B, Stewart D, Brownback KR. Acute Lung Injury in Immunocompromised Patients. Clin Chest Med 2025; 46:105-114. [PMID: 39890282 DOI: 10.1016/j.ccm.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
Acute lung injury is a devastating complication when occurring in immunocompromised patients. The incidence appears to be increasing as more patients survive for longer in this susceptible state. Being aware of potential causes of acute lung injury may lead to earlier recognition and diagnosis. Infection is a common cause of acute lung injury and needs to be considered in the diagnostic algorithm. Management involves use of supportive ventilatory strategies and potentially pharmacologic therapies.
Collapse
Affiliation(s)
- Brogan Barry
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, MailStop 3007, Kansas City, KS 66160, USA
| | - Dane Stewart
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, MailStop 3007, Kansas City, KS 66160, USA
| | - Kyle R Brownback
- Division of Pulmonary and Critical Care Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, MailStop 3007, Kansas City, KS 66160, USA.
| |
Collapse
|
17
|
Picchianti-Diamanti A, Aiello A, De Lorenzo C, Migliori GB, Goletti D. Management of tuberculosis risk, screening and preventive therapy in patients with chronic autoimmune arthritis undergoing biotechnological and targeted immunosuppressive agents. Front Immunol 2025; 16:1494283. [PMID: 39963138 PMCID: PMC11830708 DOI: 10.3389/fimmu.2025.1494283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Tuberculosis (TB) is the leading cause of death in the world from an infectious disease. Its etiologic agent, the Mycobacterium tuberculosis (Mtb), is a slow-growing bacterium that has coexisted in humans for thousands of years. According to the World Health Organization, 10.6 million new cases of TB and over 1 million deaths were reported in 2022. It is widely recognized that patients affected by chronic autoimmune arthritis such as rheumatoid arthritis (RA), psoriatic arthritis (PsA), and ankylosing spondylitis (AS) have an increased incidence rate of TB disease compared to the general population. As conceivable, the risk is associated with age ≥65 years and is higher in endemic regions, but immunosuppressive therapy plays a pivotal role. Several systematic reviews have analysed the impact of anti-TNF-α agents on the risk of TB in patients with chronic autoimmune arthritis, as well as for other biologic disease-modifying immunosuppressive anti-rheumatic drugs (bDMARDs) such as rituximab, abatacept, tocilizumab, ustekinumab, and secukinumab. However, the data are less robust compared to those available with TNF-α inhibitors. Conversely, data on anti-IL23 agents and JAK inhibitors (JAK-i), which have been more recently introduced for the treatment of RA and PsA/AS, are limited. TB screening and preventive therapy are recommended in Mtb-infected patients undergoing bDMARDs and targeted synthetic (ts)DMARDs. In this review, we evaluate the current evidence from randomized clinical trials, long-term extension studies, and real-life studies regarding the risk of TB in patients with RA, PsA, and AS treated with bDMARDs and tsDMARDs. According to the current evidence, TNF-α inhibitors carry the greatest risk of TB progression among bDMARDs and tsDMARDs, such as JAK inhibitors and anti-IL-6R agents. The management of TB screening and the updated preventive therapy are reported.
Collapse
Affiliation(s)
- Andrea Picchianti-Diamanti
- Department of Clinical and Molecular Medicine, “Sapienza” University, S. Andrea University Hospital, Rome, Italy
| | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Chiara De Lorenzo
- Department of Clinical and Molecular Medicine, “Sapienza” University, S. Andrea University Hospital, Rome, Italy
| | - Giovanni Battista Migliori
- Istituti Clinici Scientifici Maugeri, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Tradate, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
18
|
Baker H, Fine R, Suter F, Allore H, Hsiao B, Chowdhary V, Lavelle E, Chen P, Hintz R, Suter LG, Danve A. Implementation of a Best Practice Advisory to Improve Infection Screening Prior to New Prescriptions of Biologics and Targeted Synthetic Drugs. Arthritis Care Res (Hoboken) 2025; 77:273-281. [PMID: 37382043 DOI: 10.1002/acr.25181] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/07/2023] [Accepted: 06/27/2023] [Indexed: 06/30/2023]
Abstract
OBJECTIVE Use of biologic and targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs) in patients with preexisting tuberculosis (TB), hepatitis B virus (HBV), or hepatitis C virus (HCV) infection can have serious consequences. Although various society guidelines recommend routine screening for these infections before initiating certain b/tsDMARDs, adherence to these recommendations varies widely. This quality improvement initiative evaluated local compliance with screening and assessed whether an automated computerized decision support system in the form of a best practice advisory (BPA) in the electronic health record could improve patient screening. METHODS Established patients with autoimmune rheumatic disease (ARD) aged 18 years or older with at least one visit to our rheumatology practice between October 1, 2017, and March 3, 2022, were included. When prescribing a new b/tsDMARD, clinicians were alerted via a BPA that showed the most recent results for TB, HBV, and HCV. Screening proportions for TB, HBV, and HCV before BPA initiation were compared with those of eligible patients after the BPA implementation. RESULTS A total of 711 patients pre-BPA and 257 patients post-BPA implementation were included in the study. The BPA implementation was associated with statistically significant improvement in screening for TB from 66% to 82% (P ≤ 0.001), HCV from 60% to 79% (P ≤ 0.001), hepatitis B core antibody 32% to 51% (P ≤ 0.001), and hepatitis B surface antigen from 51% to 70% (P ≤ 0.001). CONCLUSION Implementation of a BPA can improve infectious disease screening for patients with ARD who are started on b/tsDMARDs and has potential to improve patient safety.
Collapse
Affiliation(s)
- Hailey Baker
- Yale New Haven Health System, New Haven, Connecticut
| | - Rebecca Fine
- Yale New Haven Health System, New Haven, Connecticut
| | | | | | | | | | | | - Ping Chen
- Yale New Haven Hospital, New Haven, Connecticut
| | | | - Lisa G Suter
- Yale University and West Haven Veterans Affairs Medical Center, New Haven, Connecticut
| | | |
Collapse
|
19
|
Telefon AH, Cil MK, Sonmez G, Turgut EA, Gulmez TK, Taskin DG, Garip S, Ekinci RMK, Celik U. Balancing Effective Treatments With Potential Threats: The Impact of Biologic Agent Use on Tuberculosis Development in Children With Chronic Inflammatory Disorders. Int J Rheum Dis 2025; 28:e70145. [PMID: 39989306 DOI: 10.1111/1756-185x.70145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/29/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
AIM We aimed to investigate the frequency and clinical characteristics of tuberculosis in pediatric patients receiving both anti-TNF-alpha and other biological agents. MATERIALS AND METHODS The data of 270 patients who used biological agents due to rheumatologic diseases and inflammatory bowel disease (IBD) and were followed between January 2021 and October 2023 were retrospectively collected from their files. RESULTS Of the patients, 138(51.1%) were female, 132(48.9%) were male. The mean age at diagnosis was 107 months (min 8-max 215 months). Patients were most commonly followed for juvenile idiopathic arthritis (JIA) (191 patients, 70.7%), followed by other autoinflammatory diseases (49 patients) and IBD (26 patients, 9.6%). Treatments included adalimumab (97 patients, 35.9%), etanercept (85 patients, 31.5%), canakinumab (33 patients, 12.2%), tocilizumab (31 patients, 11.5%), infliximab (16 patients, 5.9%), anakinra (5 patients, 1.9%), tofacitinib (2 patients, 0.7%), and rituximab (1 patient, 0.4%). During follow-up, latent tuberculosis infection (LTBI) developed in 5 (1.9%) patients. Three of these patients had JIA (using adalimumab, etanercept and tocilizumab), one had Familial Mediterranean Fever (using canakinumab), and one had IBD (using adalimumab). The mean duration of biological agent treatment until LTBI development was 14.6 months (min 6-max 29 months). Tuberculosis disease was diagnosed in two patients. Both were patients followed up due to IBD. One of the patients was diagnosed with miliary tuberculosis after infliximab treatment, while the other was followed for tuberculosis lymphadenitis after adalimumab treatment. Quadruple anti-tuberculosis therapy was initiated for the patients. CONCLUSION The use of biological agents has revolutionized the course of chronic inflammatory diseases in childhood. However, in these patients, monitoring for tuberculosis risk is crucial.
Collapse
Affiliation(s)
- Ayse Hitay Telefon
- Department of Pediatric Infectious Diseases, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| | - Merve Kilic Cil
- Department of Pediatric Infectious Diseases, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| | - Gulsum Sonmez
- Department of Pediatric Infectious Diseases, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| | - Elif Afat Turgut
- Department of Pediatric Infectious Diseases, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| | - Tugba Kandemir Gulmez
- Department of Pediatric Infectious Diseases, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| | - Didem Gulcu Taskin
- Department of Pediatric Gastroenterology, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| | - Sevinc Garip
- Department of Pediatric Gastroenterology, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| | - Rabia Miray Kisla Ekinci
- Department of Pediatric Rheumatology, Cukurova University Faculty of Medicine: Cukurova Universitesi Tip Fakultesi, Adana, Turkey
| | - Umit Celik
- Department of Pediatric Infectious Diseases, Ministry of Health Adana City Training & Research Hospital: TC Saglik Bakanligi Adana Sehir Egitim ve Arastirma Hastanesi, Adana, Turkey
| |
Collapse
|
20
|
Capoferri G, Ghielmetti G, Glatz B, Mutke MR, Tzankov A, Stephan R, Keller PM, Labhardt ND. Disseminated, fatal reactivation of bovine tuberculosis in a patient treated with adalimumab: a case report and review of the literature. Infection 2025; 53:481-487. [PMID: 39143434 PMCID: PMC11825525 DOI: 10.1007/s15010-024-02364-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE Tumor necrosis factor inhibitors (TNFi) are known to increase the risk of tuberculosis (TB) reactivation, though cases involving Mycobacterium bovis are rarely reported. CASE PRESENTATION/RESULTS We describe a case of disseminated TB with M. bovis in a 78-year-old woman with a negative Interferon-Gamma-Release Assay (IGRA), taking adalimumab due to rheumatoid polyarthritis, which resulted in a fatal outcome. The atypical clinical and histopathological features were initially interpreted as sarcoidosis. The case occurred in Switzerland, an officially bovine tuberculosis-free country. The whole genome sequence of the patient's cultured M. bovis isolate was identified as belonging to the animal lineage La1.2, the main genotype in continental Europe, but showed significant genetic distance from previously sequenced Swiss cattle strains. In a literature review, four cases of bovine tuberculosis reactivation under TNFi treatment were identified, with pulmonal, oral and intestinal manifestations. Similar to our patient, two cases presented a negative IGRA before TNFi initiation, which later converted to positive upon symptomatic presentation of M. bovis infection. CONCLUSION This case highlights the diagnostic challenges of TB in immunosuppressed patients, the limited sensitivity of IGRA, and the importance of considering TB reactivation even in regions declared free of bovine tuberculosis. Detailed patient histories, including potential exposure to unpasteurized dairy products, are essential for guiding preventive TB treatment before TNFi initiation.
Collapse
Affiliation(s)
- Gioele Capoferri
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland.
| | - Giovanni Ghielmetti
- Section of Veterinary Bacteriology, Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Bettina Glatz
- Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Markus R Mutke
- Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Roger Stephan
- Section of Veterinary Bacteriology, Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Peter M Keller
- Division of Clinical Bacteriology and Mycology, University Hospital of Basel, Basel, Switzerland
| | - Niklaus D Labhardt
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
21
|
Vallejos OP, Bueno SM, Kalergis AM. Probiotics in inflammatory bowel disease: microbial modulation and therapeutic prospects. Trends Mol Med 2025:S1471-4914(24)00338-1. [PMID: 39814640 DOI: 10.1016/j.molmed.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder that represents a significant public health challenge worldwide. This multifactorial condition results from complex interactions among genetic, environmental, immune, and microbial factors. Some beneficial microbes, known as probiotics, have been identified as promising therapeutic agents for inflammatory conditions, such as IBD. In this review, we explore the potential of probiotics as a therapeutic strategy for managing IBD. Probiotics have shown promise due to their ability to modulate the gut microbiota, regulate histamine levels, and enhance vitamin D metabolism, thereby promoting a tolerant immune profile and reducing inflammation. While the exact mechanisms underlying these benefits remain incompletely understood, probiotics represent a novel and emerging approach for alleviating the exacerbated inflammation characteristic of this disorder.
Collapse
Affiliation(s)
- Omar P Vallejos
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
22
|
Cifaldi C, Sgrulletti M, Cesare SD, Rivalta B, Emanuele A, Colucci L, Moscato GMF, Matraxia M, Perrone C, Di Matteo G, Cancrini C, Moschese V. Partial Loss of NEMO Function in a Female Carrier with No Incontinentia Pigmenti. J Clin Med 2025; 14:363. [PMID: 39860371 PMCID: PMC11765721 DOI: 10.3390/jcm14020363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/20/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The nuclear factor (NF)-kB essential modulator (NEMO) has a crucial role in the NFκB pathway. Hypomorphic IKBKG pathogenic variants cause ectodermal dysplasia with immunodeficiency (EDA-ID) in affected males. However, heterozygous amorphic IKBKG variants could be responsible for Incontinentia Pigmenti (IP) in female carriers. Typically, IP patients do not exhibit immunodeficiency, although hypomorphic variants might lead to immunodeficiency in female IP patients. Here, we report the case of an IKBKG female carrier, with no IP but an unexpected picture of immunodeficiency. She had a positive family history for the same genetic condition. Methods: We performed immunological, molecular, and functional analysis to evaluate NEMO contribution. Results: The patient was healthy until the age of 25 when severe asthma and Hashimoto thyroiditis occurred. She had HLAB27-positive ankylosing spondylitis, non-tubercular mycobacteriosis, and pulmonary aspergillosis infections. We found CD19+ B cell lymphopenia and T cell subset alterations. Sanger sequencing revealed a heterozygous IKBKG variant at position +1 of the 5' UTR of the gene which disrupted the normal pre-mRNA splicing. We observed a decreased NEMO protein expression, a reduced level of mRNA, and a defective NF-κB pathway. Conclusions: These findings suggest a possible correlation between the partial loss of NEMO function and the immunodeficiency observed in this patient. This case could expand our understanding of NEMO deficiency in female carriers.
Collapse
Affiliation(s)
- Cristina Cifaldi
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (G.M.F.M.); (G.D.M.); (C.C.)
| | - Mayla Sgrulletti
- Pediatric Immunopathology and Allergology Unit, Policlinico Tor Vergata, University of Rome Tor Vergata, 00133 Rome, Italy;
- PhD Program in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Silvia Di Cesare
- Unit of Clinical Immunology and Vaccinology, IRCCS Bambino Gesù Children Hospital, 00165 Rome, Italy;
| | - Beatrice Rivalta
- Research Unit of Primary Immunodeficiency, IRCCS Bambino Gesù Children Hospital, 00165 Rome, Italy; (B.R.); (L.C.)
| | - Agolini Emanuele
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital IRCCS, 00165 Rome, Italy; (A.E.); (M.M.); (C.P.)
| | - Lucia Colucci
- Research Unit of Primary Immunodeficiency, IRCCS Bambino Gesù Children Hospital, 00165 Rome, Italy; (B.R.); (L.C.)
| | - Giusella Maria Francesca Moscato
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (G.M.F.M.); (G.D.M.); (C.C.)
- Pediatric Immunopathology and Allergology Unit, Policlinico Tor Vergata, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Marta Matraxia
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital IRCCS, 00165 Rome, Italy; (A.E.); (M.M.); (C.P.)
| | - Chiara Perrone
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital IRCCS, 00165 Rome, Italy; (A.E.); (M.M.); (C.P.)
| | - Gigliola Di Matteo
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (G.M.F.M.); (G.D.M.); (C.C.)
- Research Unit of Primary Immunodeficiency, IRCCS Bambino Gesù Children Hospital, 00165 Rome, Italy; (B.R.); (L.C.)
| | - Caterina Cancrini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (G.M.F.M.); (G.D.M.); (C.C.)
- Research Unit of Primary Immunodeficiency, IRCCS Bambino Gesù Children Hospital, 00165 Rome, Italy; (B.R.); (L.C.)
| | - Viviana Moschese
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (C.C.); (G.M.F.M.); (G.D.M.); (C.C.)
- Pediatric Immunopathology and Allergology Unit, Policlinico Tor Vergata, University of Rome Tor Vergata, 00133 Rome, Italy;
| |
Collapse
|
23
|
Njagi LN, Nduba V, Murithi WB, Mwongera Z, Cook K, Mecha J, Chacha R, Fennelly KP, Horne DJ, Hawn TR. Association of Mycobacterium tuberculosis aerosolization and HIV coinfection in the index case with T cell responses in household contacts. Sci Rep 2025; 15:224. [PMID: 39748008 PMCID: PMC11695726 DOI: 10.1038/s41598-024-83965-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Exposure to pulmonary tuberculosis (PTB) culminates in heterogeneous outcomes, including variation in Mtb antigen-specific interferon-gamma (IFN-γ) T-cell responses. IFN-γ-independent cytokines, including tumor necrosis factor (TNF) and interleukin (IL-2), offer potential diagnostic improvements and insights into pathogenesis. We hypothesized that ESAT6/CFP10 TNF and IL-2 responses improve Mtb infection detection among exposed household contacts (HHCs) and are associated with index case Mtb aerosolization (i.e., cough aerosol culture positive for Mtb growth, CAC+) and HIV co-infection. We enrolled individuals with PTB and their HHCs in a longitudinal study in Nairobi, Kenya. We measured TNF and IL-2 in HHCs from QuantiFERON-TB Plus TB1 tube supernatants. An additional 9.2% (25) HHCs beyond the 58.6% (129) with an IFN-γ response demonstrated an antigen-specific increase in IL-2 and TNF. HHCs of CAC + participants were more likely to have positive IL-2 (84.6% vs. 53.8%, p = 0.02) and IFN-γ (88.0% vs. 54.9%, p = 0.01), but not TNF responses, compared to CAC-negative individuals. While HIV co-infection in the index was negatively associated with IFN-γ responses in HHCs (35.7% vs. 62.3%, p = 0.03), IL-2 and TNF responses did not differ. Antigen-specific ESAT6/CFP10 IL-2 and TNF may increase rates of Mtb infection detection and provide insights into Mtb transmission and pathogenesis.
Collapse
Affiliation(s)
- Lilian N Njagi
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya.
| | - Videlis Nduba
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Wilfred Bundi Murithi
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Zipporah Mwongera
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kennadi Cook
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jerphason Mecha
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Robi Chacha
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kevin P Fennelly
- Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD, USA
| | - David J Horne
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Thomas R Hawn
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
24
|
Nicolò S, Faggiani I, Errico C, D'Amico F, Parigi TL, Danese S, Ungaro F. Translational characterization of immune pathways in inflammatory bowel disease: insights for targeted treatments. Expert Rev Clin Immunol 2025; 21:55-72. [PMID: 39313992 DOI: 10.1080/1744666x.2024.2400300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION The pathogenesis of inflammatory bowel disease (IBD) involves the dysregulation of multiple inflammatory pathways. The understanding of these mechanisms allows their selective targeting for therapeutic purposes. The discovery of Tumor Necrosis Factor-alpha's (TNF-α) role in mucosal inflammation ushered an exciting new era of drug development which now comprises agents targeting multiple pro-inflammatory signaling pathways, integrins, and leukocyte trafficking regulators. AREA COVERED This review provides an overview of the main molecular players of IBD, their translation into therapeutic targets and the successful development of the advanced agents modulating them. We combine basic science with clinical trials data to present a critical review of both the successful and failed drug development programs. A PubMed literature search was conducted to delve into the available literature and clinical trials. EXPERT OPINION The treatment landscape for IBD has rapidly expanded, particularly with the development of biologics targeting TNF-α, integrins, and S1P modulators, as well as newer agents such as IL-12/IL-23 inhibitors and JAK inhibitors, offering robust efficacy and safety profiles. However, challenges persist in understanding and effectively treating difficult-to-treat IBD, highlighting the need for continued research to uncover novel therapeutic targets and optimize patient outcomes.
Collapse
Affiliation(s)
- Sabrina Nicolò
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ilaria Faggiani
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Carmela Errico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ferdinando D'Amico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Lorenzo Parigi
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Danese
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Federica Ungaro
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
25
|
Sakai Y, Asa M, Hirose M, Kusuhara W, Fujiwara N, Tamashima H, Ikazaki T, Oka S, Kuraba K, Tanaka K, Yoshiyama T, Nagae M, Hoshino Y, Motooka D, Van Rhijn I, Lu X, Ishikawa E, Moody DB, Kato T, Inuki S, Hirai G, Yamasaki S. A conserved human CD4+ T cell subset recognizing the mycobacterial adjuvant trehalose monomycolate. J Clin Invest 2024; 135:e185443. [PMID: 39718834 PMCID: PMC11910211 DOI: 10.1172/jci185443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Mycobacterium tuberculosis causes human tuberculosis (TB). As mycobacteria are protected by a thick lipid cell wall, humans have developed immune responses against diverse mycobacterial lipids. Most of these immunostimulatory lipids are known as adjuvants acting through innate immune receptors, such as C-type lectin receptors. Although a few mycobacterial lipid antigens activate unconventional T cells, the antigenicity of most adjuvantic lipids is unknown. Here, we identified that trehalose monomycolate (TMM), an abundant mycobacterial adjuvant, activated human T cells bearing a unique αβ T cell receptor (αβTCR). This recognition was restricted by CD1b, a monomorphic antigen-presenting molecule conserved in primates but not mice. Single-cell TCR-RNA-Seq using newly established CD1b-TMM tetramers revealed that TMM-specific T cells were present as CD4+ effector memory T cells in the periphery of uninfected donors but expressed IFN-γ, TNF, and anti-mycobacterial effectors upon TMM stimulation. TMM-specific T cells were detected in cord blood and PBMCs of donors without bacillus Calmette-Guérin vaccination but were expanded in patients with active TB. A cryo-electron microscopy study of CD1b-TMM-TCR complexes revealed unique antigen recognition by conserved features of TCRs, positively charged CDR3α, and long CDR3β regions. These results indicate that humans have a commonly shared and preformed CD4+ T cell subset recognizing a typical mycobacterial adjuvant as an antigen. Furthermore, the dual role of TMM justifies reconsideration of the mechanism of action of adjuvants.
Collapse
MESH Headings
- Humans
- Mycobacterium tuberculosis/immunology
- CD4-Positive T-Lymphocytes/immunology
- Adjuvants, Immunologic/pharmacology
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Antigens, CD1/immunology
- Antigens, CD1/genetics
- Animals
- Mice
- Tuberculosis/immunology
- T-Lymphocyte Subsets/immunology
- Antigens, Bacterial/immunology
- Cord Factors
- Trehalose/immunology
- Trehalose/analogs & derivatives
Collapse
Affiliation(s)
- Yuki Sakai
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Minori Asa
- Department of Molecular Immunology, Research Institute for Microbial Diseases
| | - Mika Hirose
- Laboratory for CryoEM Structural Biology, Institute for Protein Research, Osaka University, Suita, Japan
| | - Wakana Kusuhara
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Nagatoshi Fujiwara
- Department of Food and Nutrition, Faculty of Contemporary Human Life Science, Tezukayama University, Nara, Japan
| | - Hiroto Tamashima
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Ikazaki
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shiori Oka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kota Kuraba
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kentaro Tanaka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Takashi Yoshiyama
- Respiratory Disease Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Yoshihiko Hoshino
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Ildiko Van Rhijn
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, University Utrecht, Utrecht, Netherlands
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Xiuyuan Lu
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
| | - D. Branch Moody
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takayuki Kato
- Laboratory for CryoEM Structural Biology, Institute for Protein Research, Osaka University, Suita, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Go Hirai
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), and
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
- Center for Advanced Modalities and Drug Delivery Systems (CAMaD), Osaka University, Suita, Japan
| |
Collapse
|
26
|
Scriba TJ, Maseeme M, Young C, Taylor L, Leslie AJ. Immunopathology in human tuberculosis. Sci Immunol 2024; 9:eado5951. [PMID: 39671470 DOI: 10.1126/sciimmunol.ado5951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/15/2024] [Indexed: 12/15/2024]
Abstract
Mycobacterium tuberculosis (M.tb) is a bacterial pathogen that has evolved in humans, and its interactions with the host are complex and best studied in humans. Myriad immune pathways are involved in infection control, granuloma formation, and progression to tuberculosis (TB) disease. Inflammatory cells, such as macrophages, neutrophils, conventional and unconventional T cells, B cells, NK cells, and innate lymphoid cells, interact via cytokines, cell-cell communication, and eicosanoid signaling to contain or eliminate infection but can alternatively mediate pathological changes required for pathogen transmission. Clinical manifestations include pulmonary and extrapulmonary TB, as well as post-TB lung disease. Risk factors for TB progression, in turn, largely relate to immune status and, apart from traditional chemotherapy, interventions primarily target immune mechanisms, highlighting the critical role of immunopathology in TB. Maintaining a balance between effector mechanisms to achieve protective immunity and avoid detrimental inflammation is central to the immunopathogenesis of TB. Many research gaps remain and deserve prioritization to improve our understanding of human TB immunopathogenesis.
Collapse
Affiliation(s)
- Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Mahlatse Maseeme
- Africa Health Research Institute, Durban, South Africa
- College of Heath Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Carly Young
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Laura Taylor
- Forensic Pathology Services, Western Cape Government/University of Cape Town, Cape Town, South Africa
| | - Alasdair J Leslie
- Africa Health Research Institute, Durban, South Africa
- University College London, London, UK
| |
Collapse
|
27
|
Lyu J, Narum DE, Baldwin SL, Larsen SE, Bai X, Griffith DE, Dartois V, Naidoo T, Steyn AJC, Coler RN, Chan ED. Understanding the development of tuberculous granulomas: insights into host protection and pathogenesis, a review in humans and animals. Front Immunol 2024; 15:1427559. [PMID: 39717773 PMCID: PMC11663721 DOI: 10.3389/fimmu.2024.1427559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Granulomas, organized aggregates of immune cells which form in response to Mycobacterium tuberculosis (Mtb), are characteristic but not exclusive of tuberculosis (TB). Despite existing investigations on TB granulomas, the determinants that differentiate host-protective granulomas from granulomas that contribute to TB pathogenesis are often disputed. Thus, the goal of this narrative review is to help clarify the existing literature on such determinants. We adopt the a priori view that TB granulomas are host-protective organelles and discuss the molecular and cellular determinants that induce protective granulomas and those that promote their failure. While reports about protective TB granulomas and their failure may initially seem contradictory, it is increasingly recognized that either deficiencies or excesses of the molecular and cellular components in TB granuloma formation may be detrimental to the host. More specifically, insufficient or excessive expression/representation of the following components have been reported to skew granulomas toward the less protective phenotype: (i) epithelioid macrophages; (ii) type 1 adaptive immune response; (iii) type 2 adaptive immune response; (iv) tumor necrosis factor; (v) interleukin-12; (vi) interleukin-17; (vii) matrix metalloproteinases; (viii) hypoxia in the TB granulomas; (ix) hypoxia inducible factor-1 alpha; (x) aerobic glycolysis; (xi) indoleamine 2,3-dioxygenase activity; (xii) heme oxygenase-1 activity; (xiii) immune checkpoint; (xiv) leukotriene A4 hydrolase activity; (xv) nuclear-factor-kappa B; and (xvi) transforming growth factor-beta. Rather, more precise and timely coordinated immune responses appear essential for eradication or containment of Mtb infection. Since there are several animal models of infection with Mtb, other species within the Mtb complex, and the surrogate Mycobacterium marinum - whether natural (cattle, elephants) or experimental (zebrafish, mouse, guinea pig, rabbit, mini pig, goat, non-human primate) infections - we also compared the TB granulomatous response and other pathologic lung lesions in various animals infected with one of these mycobacteria with that of human pulmonary TB. Identifying components that dictate the formation of host-protective granulomas and the circumstances that result in their failure can enhance our understanding of the macrocosm of human TB and facilitate the development of novel remedies - whether they be direct therapeutics or indirect interventions - to efficiently eliminate Mtb infection and prevent its pathologic sequelae.
Collapse
Affiliation(s)
- Jiwon Lyu
- Division of Pulmonary and Critical Medicine, Soon Chun Hyang University Cheonan Hospital, Seoul, Republic of Korea
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Drew E. Narum
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Susan L. Baldwin
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Sasha E. Larsen
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Xiyuan Bai
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - David E. Griffith
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Threnesan Naidoo
- Departments of Forensic & Legal Medicine and Laboratory Medicine & Pathology, Faculty of Medicine & Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology and Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rhea N. Coler
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Edward D. Chan
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
28
|
Khelghati F, Rahmanian M, Eghbal E, Seghatoleslami ZS, Goudarzi M, Keramatinia A, Ong CWM, Goletti D, D'Ambrosio L, Centis R, Nasiri MJ, Migliori GB. Risk of tuberculosis disease in patients receiving TNF-α antagonist therapy: A meta-analysis of randomized controlled trials. New Microbes New Infect 2024; 62:101533. [PMID: 39639969 PMCID: PMC11617757 DOI: 10.1016/j.nmni.2024.101533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Tuberculosis (TB) risk associated with tumor necrosis factor-alpha (TNF-α) antagonist therapy in patients with autoimmune diseases is a significant concern. This study aims to evaluate the risk of TB disease associated with TNF-α antagonist therapy. Methods An extensive search of PubMed/MEDLINE, EMBASE, and the Cochrane CENTRAL databases was conducted to identify randomized controlled trials (RCTs) assessing TB disease risk in patients receiving TNF-α antagonist therapy available until November 1, 2024. The pooled statistic used was the weighted odds ratio (OR) and a corresponding 95 % confidence interval (CI). Statistical analysis was performed using Comprehensive Meta-Analysis software, version 3.0 (Biostat Inc., Englewood, NJ, USA). Results Fifty-six RCTs, totaling 22,212 adult patients, met the specified eligibility criteria. Pooled analysis revealed an increased risk of TB disease associated with TNF-α antagonist therapy (OR 1.52, 95 % CI 1.03-2.26, p = 0.03). Subgroup analyses indicated a higher risk in patients with rheumatoid arthritis (RA) (OR 2.25, 95 % CI 1.13-4.45, p = 0.02), while no significant associations were found in patients with ankylosing spondylitis (AS) or psoriasis (Ps). Analyses by specific TNF-α antagonist drugs did not yield significant associations with risk of TB disease. Conclusion Our study highlights an increased risk of TB disease associated with TNF-α antagonist therapy, particularly in patients with RA. However, the absence of significant associations in AS or Ps patients suggests disease-specific variations in risk of TB disease. Further research is needed to elucidate the long-term safety profile of TNF-α antagonist drugs and their associations with risk of TB disease in different patient populations.
Collapse
Affiliation(s)
- Fatemeh Khelghati
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmanian
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elaheh Eghbal
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aliasghar Keramatinia
- Department of Community Medicine, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Catherine WM. Ong
- Division of Infectious Diseases, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research National Institute for Infectious Diseases L. Spallanzani-IRCCS, Roma, Italy
| | | | - Rosella Centis
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Giovanni Battista Migliori
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| |
Collapse
|
29
|
Njagi LN, Nduba V, Murithi WB, Mwongera Z, Cook K, Mecha J, Chacha R, Fennelly KP, Horne DJ, Hawn TR. Household contact antigen-speci fic TNF and IL-2 T-cell responses and impact of index case Mycobacterium tuberculosis aerosolization and HIV Co-infection. RESEARCH SQUARE 2024:rs.3.rs-4815117. [PMID: 39606489 PMCID: PMC11601854 DOI: 10.21203/rs.3.rs-4815117/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Exposure to pulmonary tuberculosis (PTB) culminates in heterogeneous outcomes, including variation in Mtb antigen-specific interferon-gamma (IFN-γ) T-cell responses. IFN-γ-independent cytokines, including tumor necrosis factor (TNF) and interleukin (IL-2), offer potential diagnostic improvements and insights into pathogenesis. We hypothesized that ESAT6/CFP10 TNF and IL-2 responses improve Mtb infection detection among exposed household contacts (HHCs) and are associated with index case Mtb aerosolization (i.e., cough aerosol culture positive for Mtb growth, CAC+]) and HIV co-infection. We enrolled individuals with PTB and their HHCs in a longitudinal study in Nairobi, Kenya. We measured TNF and IL-2 in HHCs from QuantiFERON-TB Plus TB1 tube supernatants. An additional 9.2% (25) HHCs beyond the 58.6% (129) with an IFN-γ response demonstrated an antigen-specific increase in IL-2 and TNF. HHCs of CAC + participants were more likely to have positive IL-2 (84.6% vs. 53.8%, p = 0.02) and IFN-γ (88.0% vs. 54.9%, p = 0.01), but not TNF responses, compared to CAC-negative individuals. While HIV co-infection in the index was negatively associated with IFN-γ responses in HHCs (35.7% vs. 62.3%, p = 0.03), IL-2 and TNF responses did not differ. Antigen-specific ESAT6/CFP10 IL-2 and TNF may increase rates of Mtb infection detection and provide insights into Mtb transmission and pathogenesis.
Collapse
|
30
|
Pal R, Maurya V, Borah S, Mukhopadhyay S. The SH3-binding domain of chorismate mutase protein of Mycobacterium tuberculosis contributes to mycobacterial virulence. iScience 2024; 27:111044. [PMID: 39507252 PMCID: PMC11539714 DOI: 10.1016/j.isci.2024.111044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 05/27/2024] [Accepted: 09/23/2024] [Indexed: 11/08/2024] Open
Abstract
Crystal structure of the secretory chorismate mutase protein of Mycobacterium tuberculosis (MtbCM) reveals presence of a proline rich region on its surface that serve as a recognition site for protein-protein interaction. This study shows that MtbCM upregulates IL-10 which favors M. tuberculosis by affecting PKCε-MKP-1-p38 MAPK signaling. MtbCM translocates to the Golgi-network where it interacts with AKAP9 via its SH3-binding domain to inhibit AKAP9-PKCε interaction and reducing PKCε phosphorylation. In the absence of phosphorylated PKCε, IRAK3 fails to stabilize MKP-1 resulting in higher p38 MAPK activation and IL-10 production. M. smegmatis expressing MtbCM survived better in infected mice. Mutation in SH3-binding domain ablated MtbCM-AKAP9 interaction resulting in IL-10 production and decreased bacterial survival. This study highlights the importance of SH3-binding domain in host-pathogen interaction and a role of MtbCM in modulation of cytokine response and mycobacterial virulence in addition to its role in shikimate pathway.
Collapse
Affiliation(s)
- Ravi Pal
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, Telangana 500039, India
- Graduate Studies, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Vandana Maurya
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, Telangana 500039, India
- Graduate Studies, Regional Center for Biotechnology, Faridabad, Haryana (NCR Delhi) 121001, India
| | - Supriya Borah
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, Telangana 500039, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, Telangana 500039, India
| |
Collapse
|
31
|
Qui M, Salazar E. Beyond Suppression: Peripheral T Cell Responses to Vaccination in Inflammatory Bowel Disease Patients Undergoing Anti-Tumor-Necrosis-Factor Therapy. Vaccines (Basel) 2024; 12:1280. [PMID: 39591183 PMCID: PMC11599089 DOI: 10.3390/vaccines12111280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/29/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Alimentary tract inflammation in inflammatory bowel disease (IBD) is treated by systemically administered drugs that alter fundamental host immune responses. Biologics that target tumor necrosis factor (TNF) are first-line biologics in IBD, used widely for their effectiveness, steroid-sparing quality, and lower cost. While they enable a significant proportion of patients to achieve clinical remission, they carry an increased risk of infection and poor serological responses to vaccination. Conversely, our understanding of adaptive T cell responses in anti-TNF-treated IBD patients remains limited. The introduction of COVID-19 vaccines has prompted research that both challenges and refines our view on immunomodulatory therapy and its potential implications for immunity and protection. Here, we review these emergent findings, evaluate how they shape our understanding of vaccine-induced T cell responses in the context of anti-TNF therapy in IBD, and provide a perspective highlighting the need for a holistic evaluation of both cellular and humoral immunity in this population.
Collapse
Affiliation(s)
- Martin Qui
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Ennaliza Salazar
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore 169608, Singapore
| |
Collapse
|
32
|
McAuliffe E, Renton B. TB or Not TB, That is the Question? Br J Hosp Med (Lond) 2024; 85:1-7. [PMID: 39475044 DOI: 10.12968/hmed.2024.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
A 26-year-old female presented with a 3-month history of dry cough, unintentional weight loss, night sweats and fatigue. Her background history was significant for ulcerative colitis, managed with Adalimumab for almost 2 years. Clinical examination was unremarkable, apart from some mild pallor. Abnormal chest x-ray findings prompted a computerised tomography (CT) thorax which demonstrated multifocal peri-bronchial consolidation. The differential diagnosis was multifocal organising pneumonia and tuberculosis (TB). Extensive investigations, including invasive bronchial imaging and biopsy, ultimately ruled out TB. This paper reports a case of Adalimumab-induced organising pneumonia and discusses its clinical implications.
Collapse
Affiliation(s)
- Ellen McAuliffe
- Acute Medical Unit, Galway University Hospital, Galway, Ireland
| | - Bryan Renton
- Acute Medical Unit, Galway University Hospital, Galway, Ireland
| |
Collapse
|
33
|
Santos AP, Mello FCDQ. Latent tuberculosis infection and biologic agents other than TNF-α inhibitors: "over-screening and over-treatment?". J Bras Pneumol 2024; 50:e20240277. [PMID: 39356917 PMCID: PMC11449617 DOI: 10.36416/1806-3756/e20240277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Affiliation(s)
- Ana Paula Santos
- . Instituto de Doenças do Tórax - IDT - Universidade Federal do Rio de Janeiro - UFRJ - Rio de Janeiro (RJ) Brasil
- . Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro - UERJ - Rio de Janeiro (RJ) Brasil
| | | |
Collapse
|
34
|
Feakins RM. Inflammatory disorders of the large intestine. MORSON AND DAWSON'S GASTROINTESTINAL PATHOLOGY 2024:709-857. [DOI: 10.1002/9781119423195.ch35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Herling A, Perluk TM, Freund O, Maharshak N, Cohen NA. Pulmonary Manifestations of IBD: Case Report and Review of the Literature. J Clin Med 2024; 13:5401. [PMID: 39336887 PMCID: PMC11432544 DOI: 10.3390/jcm13185401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
This article explores the pulmonary complications associated with inflammatory bowel disease (IBD). It presents a detailed case study of a 22-year-old male with Crohn's disease exhibiting pulmonary symptoms. The review delves into the spectrum of pulmonary involvement in IBD, covering clinical presentations, diagnostic challenges, underlying pathophysiology, and management strategies. It highlights the significance of these extraintestinal manifestations on patient outcomes and quality of life. The article underscores the need for heightened clinical awareness and a systematic approach to diagnosis and management, integrating the expertise of multiple specialists. The review identifies gaps in current research, suggesting avenues for future investigation to enhance the understanding and treatment of these complex manifestations.
Collapse
Affiliation(s)
- Amit Herling
- Faculty of Medicine, Ben-Gurion University of the Negev, Be'er Sheva 8410501, Israel
| | - Tal Moshe Perluk
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6139001, Israel
- The Pulmonary Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Ophir Freund
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6139001, Israel
- The Pulmonary Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Nitsan Maharshak
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6139001, Israel
- IBD Unit, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Nathaniel Aviv Cohen
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6139001, Israel
- IBD Unit, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| |
Collapse
|
36
|
Hansen SDH, Rudolf F, Gregersen NS, Norman A. Disseminated MDR-TB missed in a patient treated with TNF inhibitor. BMJ Case Rep 2024; 17:e260058. [PMID: 39231557 DOI: 10.1136/bcr-2024-260058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
A previously healthy man developed pulmonary symptoms 2 weeks after starting treatment with a tumour necrosis factor (TNF) inhibitor. A negative interferon-gamma release assay (IGRA) test was obtained prior to TNF inhibitor exposure, without consideration of the fact that the patient was already immunosuppressed and had a previous positive IGRA test 17 months earlier. The patient was treated for pneumonia twice but did not achieve remission. His physical health progressively deteriorated over the following months. Malignancy was suspected but not found. Eight months after the onset of symptoms, Mycobacterium tuberculosis was found in samples from mediastinal lymph nodes, and the patient was diagnosed with multidrug-resistant tuberculosis (MDR-TB).This case illustrates the diagnostic challenge of TB, the need to raise awareness of the increased risk of TB in patients treated with TNF inhibitors and the need to increase knowledge regarding the effect of immunosuppressive agents on IGRA tests.
Collapse
Affiliation(s)
| | - Frauke Rudolf
- Department of Infectious Diseases, Aarhus Universitetshospital, Aarhus, Denmark
| | | | | |
Collapse
|
37
|
Arias AA, Neehus AL, Ogishi M, Meynier V, Krebs A, Lazarov T, Lee AM, Arango-Franco CA, Yang R, Orrego J, Corcini Berndt M, Rojas J, Li H, Rinchai D, Erazo-Borrás L, Han JE, Pillay B, Ponsin K, Chaldebas M, Philippot Q, Bohlen J, Rosain J, Le Voyer T, Janotte T, Amarajeeva K, Soudée C, Brollo M, Wiegmann K, Marquant Q, Seeleuthner Y, Lee D, Lainé C, Kloos D, Bailey R, Bastard P, Keating N, Rapaport F, Khan T, Moncada-Vélez M, Carmona MC, Obando C, Alvarez J, Cataño JC, Martínez-Rosado LL, Sanchez JP, Tejada-Giraldo M, L'Honneur AS, Agudelo ML, Perez-Zapata LJ, Arboleda DM, Alzate JF, Cabarcas F, Zuluaga A, Pelham SJ, Ensser A, Schmidt M, Velásquez-Lopera MM, Jouanguy E, Puel A, Krönke M, Ghirardello S, Borghesi A, Pahari S, Boisson B, Pittaluga S, Ma CS, Emile JF, Notarangelo LD, Tangye SG, Marr N, Lachmann N, Salvator H, Schlesinger LS, Zhang P, Glickman MS, Nathan CF, Geissmann F, Abel L, Franco JL, Bustamante J, Casanova JL, Boisson-Dupuis S. Tuberculosis in otherwise healthy adults with inherited TNF deficiency. Nature 2024; 633:417-425. [PMID: 39198650 PMCID: PMC11390478 DOI: 10.1038/s41586-024-07866-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024]
Abstract
Severe defects in human IFNγ immunity predispose individuals to both Bacillus Calmette-Guérin disease and tuberculosis, whereas milder defects predispose only to tuberculosis1. Here we report two adults with recurrent pulmonary tuberculosis who are homozygous for a private loss-of-function TNF variant. Neither has any other clinical phenotype and both mount normal clinical and biological inflammatory responses. Their leukocytes, including monocytes and monocyte-derived macrophages (MDMs) do not produce TNF, even after stimulation with IFNγ. Blood leukocyte subset development is normal in these patients. However, an impairment in the respiratory burst was observed in granulocyte-macrophage colony-stimulating factor (GM-CSF)-matured MDMs and alveolar macrophage-like (AML) cells2 from both patients with TNF deficiency, TNF- or TNFR1-deficient induced pluripotent stem (iPS)-cell-derived GM-CSF-matured macrophages, and healthy control MDMs and AML cells differentiated with TNF blockers in vitro, and in lung macrophages treated with TNF blockers ex vivo. The stimulation of TNF-deficient iPS-cell-derived macrophages with TNF rescued the respiratory burst. These findings contrast with those for patients with inherited complete deficiency of the respiratory burst across all phagocytes, who are prone to multiple infections, including both Bacillus Calmette-Guérin disease and tuberculosis3. Human TNF is required for respiratory-burst-dependent immunity to Mycobacterium tuberculosis in macrophages but is surprisingly redundant otherwise, including for inflammation and immunity to weakly virulent mycobacteria and many other infectious agents.
Collapse
MESH Headings
- Adult
- Female
- Humans
- Male
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Homozygote
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/immunology
- Induced Pluripotent Stem Cells/cytology
- Inflammation/immunology
- Interferon-gamma/immunology
- Loss of Function Mutation
- Lung/cytology
- Lung/drug effects
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Macrophages, Alveolar/cytology
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/microbiology
- Macrophages, Alveolar/pathology
- Mycobacterium tuberculosis/immunology
- Phenotype
- Reactive Oxygen Species/metabolism
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Respiratory Burst
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/microbiology
- Tuberculosis, Pulmonary/genetics
- Tumor Necrosis Factor Inhibitors/pharmacology
- Tumor Necrosis Factors/deficiency
- Tumor Necrosis Factors/genetics
- Adolescent
- Young Adult
Collapse
Affiliation(s)
- Andrés A Arias
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- School of Microbiology, University of Antioquia UdeA, Medellín, Colombia
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.
- Paris Cité University, Imagine Institute, Paris, France.
| | - Masato Ogishi
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Vincent Meynier
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Adam Krebs
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Angela M Lee
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Carlos A Arango-Franco
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Rui Yang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Julio Orrego
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Melissa Corcini Berndt
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Julian Rojas
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Hailun Li
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Darawan Rinchai
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Lucia Erazo-Borrás
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Ji Eun Han
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Bethany Pillay
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, New South Wales, Australia
| | - Khoren Ponsin
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Matthieu Chaldebas
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Paris Cité University, Imagine Institute, Paris, France
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Clinical Immunology Department, AP-HP, Saint-Louis Hospital, Paris, France
| | - Till Janotte
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Krishnajina Amarajeeva
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Marion Brollo
- Lab VIM Suresnes, UMR 0892, Paris Saclay University, INRAe UVSQ, Suresnes, France
| | - Katja Wiegmann
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Quentin Marquant
- Lab VIM Suresnes, UMR 0892, Paris Saclay University, INRAe UVSQ, Suresnes, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Danyel Lee
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Candice Lainé
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Doreen Kloos
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover, Germany
| | - Rasheed Bailey
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Paul Bastard
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Narelle Keating
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Franck Rapaport
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | | | - Marcela Moncada-Vélez
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - María Camila Carmona
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Catalina Obando
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Jesús Alvarez
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Juan Carlos Cataño
- Infectious Diseases Section, Department of Internal Medicine, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Larry Luber Martínez-Rosado
- Latin American Research Team in Infectiology and Public Health (ELISAP), La Maria Hospital, Medellín, Colombia
| | - Juan P Sanchez
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Manuela Tejada-Giraldo
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Anne-Sophie L'Honneur
- Department of Virology, Paris Cité University and Cochin Hospital, AP-HP, Paris, France
| | - María L Agudelo
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Lizet J Perez-Zapata
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Diana M Arboleda
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Juan Fernando Alzate
- National Center for Genome Sequencing (CNSG), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Felipe Cabarcas
- National Center for Genome Sequencing (CNSG), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- SISTEMIC Group, Department of Electronic Engineering, Faculty of Engineering, University of Antioquia UdeA, Medellín, Colombia
| | | | - Simon J Pelham
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Armin Ensser
- University Hospital Erlangen, Institute of Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Monika Schmidt
- University Hospital Erlangen, Institute of Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Margarita M Velásquez-Lopera
- Dermatology Section, Department of Internal Medicine, School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Dermatological Research Center (CIDERM), Medellín, Colombia
| | - Emmanuelle Jouanguy
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Anne Puel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Martin Krönke
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | - Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia, Italy
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Susanta Pahari
- Host Pathogen Interactions program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Bertrand Boisson
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Stefania Pittaluga
- Center for Cancer Research, Laboratory of Pathology, NCI, NIH, Bethesda, MD, USA
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, New South Wales, Australia
| | - Jean-François Emile
- Department of Pathology, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, New South Wales, Australia
| | - Nico Marr
- Department of Human Immunology, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Nico Lachmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational Regenerative Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Hélène Salvator
- Clinical Immunology Department, AP-HP, Saint-Louis Hospital, Paris, France
- Respiratory Diseases Department, FOCH Hospital, Suresnes, France
- Simone Veil Department of Health Sciences, Versailles Saint Quentin University, Montigny le Bretonneux, France
| | - Larry S Schlesinger
- Host Pathogen Interactions program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Peng Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Michael S Glickman
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Carl F Nathan
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Frédéric Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Laurent Abel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - José Luis Franco
- Inborn Errors of Immunity Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia UdeA, Medellín, Colombia.
| | - Jacinta Bustamante
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France.
- Paris Cité University, Imagine Institute, Paris, France.
- Howard Hughes Medical Institute, New York, NY, USA.
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France.
| | - Stéphanie Boisson-Dupuis
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
38
|
Pyle CJ, Tobin DM. People who lack the immune protein TNF can still fight infection. Nature 2024; 633:293-294. [PMID: 39198607 DOI: 10.1038/d41586-024-02657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
|
39
|
Maeda T, Connolly M, Thevenet-Morrison K, Levy P, Utell M, Munsiff S, Croft D. Tuberculosis screening for patients on biologic Medications: A Single-Center experience and Society guideline Review, Monroe County, New York, 2018-2021. J Clin Tuberc Other Mycobact Dis 2024; 36:100460. [PMID: 39021381 PMCID: PMC11254483 DOI: 10.1016/j.jctube.2024.100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Abstract
Rationale Biologic medications for immune-mediated inflammatory diseases may increase the risk of tuberculosis (TB) reactivation, but data on screening for TB in low TB prevalence areas are limited. Objective To assess the real-world practice patterns of TB screening among prescribers of biologic medications. Methods We conducted a retrospective observational study at a single, university-based healthcare facility in a low TB prevalence area. We enrolled adult patients prescribed a biologic medication between October 2018 and December 2021, and collected data on demographics, biologic medications and TB test results. For patients with positive TB tests, further data including prescriber specialty and response to positive tests were obtained. We reviewed pertinent major society guidelines/ consensus statements regarding TB screening among patients treated with biologic medications. Results 4,085 patients were included. 3024 (74.0%) had at least one screening TB test and 42 were positive. Among patients treated with tumor necrosis factor-alpha (TNFα) inhibitors, 1779 of 2129 patients (83.6%) underwent TB testing and 25 (1.4%) were positive. Most with positive TB test results were prescribed biologic medication by gastroenterology (11 patients, 26%), dermatology (12, 29%), or rheumatology (15, 36%) providers. 32 (76%) patients had imaging and roughly half were treated for latent TB infection. Biologic medications were temporarily held for 27 patients (67%). Nine out of 13 society guidelines recommend TB screening for TNFα inhibitors but have differing recommendations for other biologic medications. Conclusions Significant practice pattern differences in TB screening for patients receiving biologic medications exist. Multiple society guidelines continue to recommend TB screening even for drugs with no known increased risk of TB reactivation.
Collapse
Affiliation(s)
- Tetsuro Maeda
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, United States
| | - Margaret Connolly
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, United States
| | - Kelly Thevenet-Morrison
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, United States
| | - Paul Levy
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, United States
| | - Mark Utell
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, United States
| | - Sonal Munsiff
- Division of Infectious Diseases, University of Rochester Medical Center, United States
| | - Daniel Croft
- Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, United States
| |
Collapse
|
40
|
Tumba MC, Silva RL, Arevalo AB, Sattui SE. Current perspective on infections and mitigation strategies in primary systemic vasculitis. Curr Rheumatol Rep 2024; 26:279-289. [PMID: 38668813 DOI: 10.1007/s11926-024-01149-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2024] [Indexed: 07/05/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize and evaluate most recent evidence on the epidemiology of infections and associated risk factors in patients with primary systemic vasculitides (PSV), as well as discuss mitigation strategies including the risk of antibiotic prophylaxis. RECENT FINDINGS Infections remain one of the leading causes of mortality in patients with PSV, with rates of severe infection ranging from 16 to 40% in different cohorts. Older age, frailty, renal and pulmonary involvement, and higher burden of comorbidities have been recognized as important patient-associated risk factors. Treatments including higher cumulative doses of glucocorticoids are associated with an increased risk of infections, and recent studies show the potential benefit of interventions such as reduced-dose glucocorticoid regimens. Existing mitigation strategies include screening, vaccination, and infection prophylaxis. The latter remains particularly important for Pneumocystis jirovecii pneumonia; however, the benefit-risk ratio seems to be less clear outside of induction phase (i.e., high dose of glucocorticoids) and optimal treatment duration remains less clear. Patients with PSV are at increased risk of infections, due to disease itself, comorbidities, and treatment side effects. Awareness of the timing and types of infection, as well as mitigation strategies are imperative to ensure treatment success and survival for patients.
Collapse
Affiliation(s)
- Manuel Carpio Tumba
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Raisa Lomanto Silva
- Division of General Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana B Arevalo
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Massachusetts, Boston, MA, USA
| | - Sebastian E Sattui
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
41
|
Tsai CY, Oo M, Peh JH, Yeo BCM, Aptekmann A, Lee B, Liu JJJ, Tsao WS, Dick T, Fink K, Gengenbacher M. Splenic marginal zone B cells restrict Mycobacterium tuberculosis infection by shaping the cytokine pattern and cell-mediated immunity. Cell Rep 2024; 43:114426. [PMID: 38959109 PMCID: PMC11307145 DOI: 10.1016/j.celrep.2024.114426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/29/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
Understanding the role of B cells in tuberculosis (TB) is crucial for developing new TB vaccines. However, the changes in B cell immune landscapes during TB and their functional implications remain incompletely explored. Using high-dimensional flow cytometry to map the immune landscape in response to Mycobacterium tuberculosis (Mtb) infection, our results show an accumulation of marginal zone B (MZB) cells and other unconventional B cell subsets in the lungs and spleen, shaping an unconventional B cell landscape. These MZB cells exhibit activated and memory-like phenotypes, distinguishing their functional profiles from those of conventional B cells. Notably, functional studies show that MZB cells produce multiple cytokines and contribute to systemic protection against TB by shaping cytokine patterns and cell-mediated immunity. These changes in the immune landscape are reversible upon successful TB chemotherapy. Our study suggests that, beyond antibody production, targeting the regulatory function of B cells may be a valuable strategy for TB vaccine development.
Collapse
Affiliation(s)
- Chen-Yu Tsai
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Myo Oo
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Jih Hou Peh
- Biosafety Level 3 Core, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Level 15, Centre for Translational Medicine (MD6), NUS, 14 Medical Drive, Singapore 117599, Singapore
| | - Benjamin C M Yeo
- Infectious Diseases Translational Research Programme and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Level 2, Blk MD4, 5 Science Drive 2, Singapore 117545, Singapore
| | - Ariel Aptekmann
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research, Biopolis, 8A Biomedical Grove, Level 3 & 4, Immunos Building, Singapore 138648, Singapore; Centre for Biomedical Informatics, Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; A(∗)STAR Infectious Diseases Labs, Agency for Science, Technology and Research, 8A Biomedical Grove #05-13, Immunos, Singapore 138648, Singapore
| | - Joe J J Liu
- Biosafety Level 3 Core, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Level 15, Centre for Translational Medicine (MD6), NUS, 14 Medical Drive, Singapore 117599, Singapore
| | - Wen-Shan Tsao
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Thomas Dick
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA; Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Katja Fink
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research, Biopolis, 8A Biomedical Grove, Level 3 & 4, Immunos Building, Singapore 138648, Singapore
| | - Martin Gengenbacher
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA; Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA.
| |
Collapse
|
42
|
Reid VA, Ramos EI, Veerapandian R, Carmona A, Gadad SS, Dhandayuthapani S. Differential Expression of lncRNAs in HIV Patients with TB and HIV-TB with Anti-Retroviral Treatment. Noncoding RNA 2024; 10:40. [PMID: 39051374 PMCID: PMC11270221 DOI: 10.3390/ncrna10040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Tuberculosis (TB) is the leading cause of death among people with HIV-1 infection. To improve the diagnosis and treatment of HIV-TB patients, it is important to understand the mechanisms underlying these conditions. Here, we used an integrated genomics approach to analyze and determine the lncRNAs that are dysregulated in HIV-TB patients and HIV-TB patients undergoing anti-retroviral therapy (ART) using a dataset available in the public domain. The analyses focused on the portion of the genome transcribed into non-coding transcripts, which historically have been poorly studied and received less focus. This revealed that Mtb infection in HIV prominently up-regulates the expression of long non-coding RNA (lncRNA) genes DAAM2-AS1, COL4A2-AS1, LINC00599, AC008592.1, and CLRN1-AS1 and down-regulates the expression of lncRNAs AC111000.4, AC100803.3, AC016168.2, AC245100.7, and LINC02073. It also revealed that ART down-regulates the expression of some lncRNA genes (COL4A2-AS1, AC079210.1, MFA-AS1, and LINC01993) that are highly up-regulated in HIV-TB patients. Furthermore, the interrogation of the genomic regions that are associated with regulated lncRNAs showed enrichment for biological processes linked to immune pathways in TB-infected conditions. However, intriguingly, TB patients treated with ART showed completely opposite and non-overlapping pathways. Our findings suggest that lncRNAs could be used to identify critical diagnostic, prognostic, and treatment targets for HIV-TB patients.
Collapse
Affiliation(s)
- Victoria A. Reid
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
| | - Enrique I. Ramos
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Raja Veerapandian
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
| | - Areanna Carmona
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
| | - Shrikanth S. Gadad
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Frederick L. Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Subramanian Dhandayuthapani
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Frederick L. Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
43
|
Amoura A, Frapard T, Treton X, Surgers L, Beaugerie L, Lafaurie M, Gornet JM, Lepeule R, Amiot A, Canouï E, Abitbol V, Froissart A, Vidon M, Nguyen Y, Lefort A, Zarrouk V. Tuberculosis and Immune Reconstitution Inflammatory Syndrome in Patients With Inflammatory Bowel Disease and Anti-TNFα Treatment: Insights From a French Multicenter Study and Systematic Literature Review With Emphasis on Paradoxical Anti-TNFα Resumption. Open Forum Infect Dis 2024; 11:ofae327. [PMID: 38957691 PMCID: PMC11218776 DOI: 10.1093/ofid/ofae327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
Background The advent of anti-tumor necrosis factor α (anti-TNFα) has revolutionized the treatment of inflammatory bowel disease (IBD). However, susceptibility to active tuberculosis (TB) is associated with this therapy and requires its discontinuation. The risk of immune reconstitution inflammatory syndrome (IRIS) in this population is poorly understood, as is the safety of resuming anti-TNFα. Methods This French retrospective study (2010-2022) included all TB cases in patients with IBD who were treated with anti-TNFα in 6 participating centers. A systematic literature review was performed on TB-IRIS and anti-TNFα exposure. Results Thirty-six patients were included (median age, 35 years; IQR, 27-48). TB was disseminated in 86% and miliary in 53%. IRIS occurred in 47% after a median 45 days (IQR, 18-80). Most patients with TB-IRIS (93%) had disseminated TB. Miliary TB was associated with IRIS risk in univariate analysis (odds ratio, 7.33; 95% CI, 1.60-42.82; P = .015). Anti-TB treatment was longer in this population (median [IQR], 9 [9-12] vs 6 [6-9] months; P = .049). Anti-TNFα was resumed in 66% after a median 4 months (IQR, 3-10) for IBD activity (76%) or IRIS treatment (24%), with only 1 case of TB relapse. Fifty-two cases of TB-IRIS in patients treated with anti-TNFα were reported in the literature, complicating disseminating TB (85%) after a median 42 days (IQR, 21-90), with 70% requiring anti-inflammatory treatment. Forty cases of TB-IRIS or paradoxical reaction treated with anti-TNFα were also reported. IRIS was neurologic in 64%. Outcome was mostly favorable (93% recovery). Conclusions TB with anti-TNFα treatment is often complicated by IRIS of varying severity. Restarting anti-TNFα is a safe and effective strategy.
Collapse
Affiliation(s)
- Ariane Amoura
- Service de Médecine Interne, Hôpital Beaujon, Assistance Publique des Hôpitaux de Paris, Clichy, France
- Sorbonne Université, Paris, France
| | - Thomas Frapard
- Service de Médecine Intensive et Réanimation, Hôpital Henri Mondor, DHU ATVB, Assistance Publique des Hôpitaux de Paris, Créteil, France
- Faculté de Médecine de Créteil, Université Paris Est Créteil, Institut Mondor de Recherche Biomédicale-Groupe de Recherche Clinique CARMAS, Créteil, France
| | - Xavier Treton
- Institut des MICI, Groupe hospitalier privé Ambroise-Paré-Hartmann, Neuilly, France
| | - Laure Surgers
- Service des Maladies Infectieuses et Tropicales, Hôpital Saint-Antoine, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
- Sorbonne Université, INSERM, Institut Pierre Louis d’Épidémiologie et de Santé Publique, Paris, France
| | - Laurent Beaugerie
- Service de Gastroentérologie, Hôpital Saint Antoine, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Matthieu Lafaurie
- Service de Maladies infectieuses et Tropicales, Hôpital Saint-Louis-Hôpital Lariboisière, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Jean Marc Gornet
- Service de Gastroentérologie, Hôpital Saint-Louis-Hôpital, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Raphaël Lepeule
- Unité Transversale de Traitement des Infections, Assistance Publique des Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Aurélien Amiot
- Service de Gastroentérologie, Hôpitaux Universitaires Henri Mondor, Assistance Publique des Hôpitaux de Paris, Créteil, France
| | - Etienne Canouï
- Équipe Mobile d’Infectiologie, Assistance Publique des Hôpitaux de Paris, APHP-CUP, Hôpital Cochin, Paris, France
| | - Vered Abitbol
- Service de gastroentérologie, Hôpital Cochin, Assistance Publique des Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Antoine Froissart
- Service de Médecine interne, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Mathias Vidon
- Service de Gastroentérologie, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Yann Nguyen
- Service de Médecine Interne, Hôpital Beaujon, Assistance Publique des Hôpitaux de Paris, Clichy, France
- Centre de recherche en immunologie des maladies, INSERM U1184, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Agnès Lefort
- Service de Médecine Interne, Hôpital Beaujon, Assistance Publique des Hôpitaux de Paris, Clichy, France
- Groupe de recherche Infection Antimicrobials Modelling Evolution, Inserm U1137, Université Paris Cité, Paris, France
| | - Virginie Zarrouk
- Service de Médecine Interne, Hôpital Beaujon, Assistance Publique des Hôpitaux de Paris, Clichy, France
| |
Collapse
|
44
|
Roberts ET, Schmajuk G, Li J, Murrill M, Yazdany J. Latent Tuberculosis Screening Among New Users of a Biologic or Targeted Synthetic Disease-Modifying Antirheumatic Drug: Gaps in Screening Overall and Among Janus Kinase Inhibitors. Arthritis Care Res (Hoboken) 2024; 76:1037-1044. [PMID: 38412872 PMCID: PMC11209809 DOI: 10.1002/acr.25318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/29/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVE We combined claims and electronic health record (EHR) data to provide contemporary and accurate estimates of latent tuberculosis (TB) screening among new users of a biologic or targeted synthetic disease-modifying antirheumatic drug (b/tsDMARD) and assess potential gaps in testing by drug type, patient characteristics, and practice. METHODS Our denominator population was patients in the Rheumatology Informatics System for Effectiveness (RISE) registry and Medicare using a b/tsDMARD in 2018 without a claim or prescription in the year prior. TB screening was assessed in both Medicare and RISE 1 and 3 years before the medication start date. We calculated the proportion screened overall, by medication class, and by practice. We tested for demographic differences in screening using logistic regression. RESULTS In the year before drug starts, 65.6% of patients had any TB screening; in a 3-year window, 72.9% had any TB screening. Rates of screening within 1 year by drug type were greater or equal to the overall screening rate for most drugs except for JAK inhibitors (JAKis) (46%) and interleukin-17 inhibitors (IL-17is) (11.5%). A lower proportion of Hispanic and Asian patients were screened compared with White patients. Practice screening rates ranged from 20.0% to 92.9% of patients within 1 year. CONCLUSION We report higher screening rates than have previously been published because of combining claims and EHR data. However, important safety gaps remain, namely, reduced screening among new users of a JAKi or IL-17i and among Asian and Hispanic patients, as well as low-performing practices. Educational initiatives, team-based care delivery, task shifting, and technological interventions to address observed gaps in patient safety procedures are needed.
Collapse
Affiliation(s)
- Eric T Roberts
- University of California San Francisco, San Francisco, CA
| | - Gabriela Schmajuk
- University of California San Francisco, San Francisco, CA
- San Francisco Veterans Affairs Medical Center, San Francisco, CA
| | - Jing Li
- University of California San Francisco, San Francisco, CA
| | | | - Jinoos Yazdany
- University of California San Francisco, San Francisco, CA
- Zuckerberg San Francisco General Hospital, San Francisco, CA
| |
Collapse
|
45
|
Hosseinian K, Gerami A, Bral M, Venketaraman V. Mycobacterium tuberculosis-Human Immunodeficiency Virus Infection and the Role of T Cells in Protection. Vaccines (Basel) 2024; 12:730. [PMID: 39066368 PMCID: PMC11281535 DOI: 10.3390/vaccines12070730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Tuberculosis (TB), primarily caused by Mycobacterium tuberculosis (M. tb), remains a widespread fatal health issue that becomes significantly detrimental when coupled with HIV. This study explores the host's innate and adaptive immune system response to TB in HIV immunocompromised patients, highlighting the significant role of CD8+ T cells. While the crucial role of macrophages and cytokines, like TNF-α and IFN-γ, in managing the host's immune response to M. tb is examined, the emphasis is on the changes that occur as a result of HIV coinfection. With the progression of HIV infection, the primary source of IFN-γ changes from CD4+ to CD8+ T cells, especially when latent TB advances to an active state. This study sheds light on the necessity of developing new preventative measures such as vaccines and new treatment approaches to TB, especially for immunocompromised patients, who are at a higher risk of life-threatening complications due to TB-HIV coinfection.
Collapse
Affiliation(s)
| | | | | | - Vishwanath Venketaraman
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
46
|
Liu YC, Zhou ML, Cheng KJ, Zhou SH, Wen X. Treatment of primary nasal tuberculosis with anti-tumor necrosis factor immunotherapy: A case report. World J Clin Cases 2024; 12:3271-3276. [PMID: 38898839 PMCID: PMC11185384 DOI: 10.12998/wjcc.v12.i17.3271] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/24/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Primary nasal tuberculosis (TB) is a rare form of extrapulmonary TB, particularly in patients receiving anti-tumor necrosis factor (TNF) immunotherapy. As a result, its diagnosis remains challenging. CASE SUMMARY A 58-year-old male patient presented to the ear, nose, and throat department with right-sided nasal obstruction and bloody discharge for 1 month. He was diagnosed with psoriatic arthritis and received anti-TNF immunotherapy for 3 years prior to presentation. Biopsy findings revealed chronic granulomatous inflammation and a few acid-fast bacilli, suggestive of primary nasal TB. He was referred to our TB management department for treatment with oral anti-TB agents. After 9 months, the nasal lesions had disappeared. No recurrence was noted during follow-up. CONCLUSION The diagnosis of primary nasal TB should be considered in patients receiving TNF antagonists who exhibit thickening and crusting of the nasal septum mucosa or inferior turbinate, particularly when pathological findings suggest granulomatous inflammation.
Collapse
Affiliation(s)
- Yong-Cai Liu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, HangZhou 310003, Zhejiang Province, China
| | - Min-Li Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Ke-Jia Cheng
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Shui-Hong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Xue Wen
- Department of Pathology, The First Affiliated Hospital, Medical College, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
47
|
McKay CE, Batish I, Arami S. Infliximab-Induced Improvement in Dercum's Disease. Cureus 2024; 16:e61499. [PMID: 38952592 PMCID: PMC11216115 DOI: 10.7759/cureus.61499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2024] [Indexed: 07/03/2024] Open
Abstract
Dercum's disease (DD) is a rare and poorly understood disease characterized by obesity and painful lipomas throughout the body. Although the entity is well described in the literature, its etiology, prevalence, and treatment remain unclear. Currently, treatment is focused on pain management. We describe a case of a patient with DD who showed improvement with infliximab and methotrexate.
Collapse
Affiliation(s)
- Cailean E McKay
- Internal Medicine-Pediatrics, University of Illinois Chicago, Chicago, USA
| | - Ishaan Batish
- Internal Medicine, Sri Guru Ramdas Institute of Medical Sciences and Research, Amritsar, IND
| | - Shiva Arami
- Rheumatology, University of Illinois Chicago, Chicago, USA
| |
Collapse
|
48
|
Boqaeid A, Layqah L, Alonazy A, Althobaiti M, Almahlawi AZ, Al-Roqy A, Baharoon O, Alsaeedi A, Shamou J, Baharoon S. The risk of tuberculosis infection in Saudi patients receiving adalimumab, etanercept, and tocilizumab therapy. J Infect Public Health 2024; 17:1134-1141. [PMID: 38728834 DOI: 10.1016/j.jiph.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND The risk of infection including tuberculosis (TB) infection or reactivation during biological therapy with the current various clinical application is a major concern. This risk may be higher in countries endemic to TB. Our aim of this study is to determine the risk of TB infection in patients receiving 3 biological treatments, Adalimumab, Etanercept and Tocilizumab. METHODS A retrospective cohort study extending over 2 years follow-up for all patients receiving Adalimumab, Etanercept and Tocilizumab for various clinical indications in a tertiary care center in Saudi Arabia. RESULT Over the period of 2015-2019, A total of 410 patients received Adalimumab, 271 received Etanercept and 58 patients received Tocilizumab. Rheumatoid arthritis was the most common indication for therapy in all groups and for Adalimumab the most common indication was inflammatory bowel disease, for Etanercept was psoriatic arthritis and for Tocilizumab was juvenile idiopathic arthritis. After a mean follow up period of 36 ± 8.9 months for patients receiving Adalimumab, 21.5 ± 8.4 months for patients receiving Etanercept and 21 ± 2.5 months for patients receiving Tocilizumab there were no reported cases of TB infection in all groups. Only one patient was diagnosed with latent TB 7 months later after starting Adalimumab and tow patients after starting Etanercept. The overall Interferon Gamma Release Assays (IGRA) positivity rate was 9.7%. There was significant association between IGRA positivity rate and patient age. The cutoff age in which IGRA positivity has significantly increased was 53.20 years. CONCLUSION In our study, patients receiving Etanercept, Adalimumab and Tocilizumab had no increased risk of TB infection. Only 0.3% of patients treated with Adalimumab and 0.9% of patients treated with Etanercept converted to a positive IGRA during therapy.
Collapse
Affiliation(s)
- Abdulaziz Boqaeid
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia.
| | - Laila Layqah
- King Saud bin Abdul-Aziz University for Health Sciences, Riyadh, Saudi Arabia; Research office, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia.
| | - Amgad Alonazy
- Department of Medicine, King Faisal Specialized Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Mutaz Althobaiti
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia.
| | - Al-Zahraa Almahlawi
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia.
| | - Abdullah Al-Roqy
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia.
| | - Omar Baharoon
- College of Medicine, Dar Al-Uloom University, Riyadh, Saudi Arabia.
| | | | - Jinan Shamou
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia.
| | - Salim Baharoon
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdul-Aziz University for Health Sciences, Riyadh, Saudi Arabia; Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia.
| |
Collapse
|
49
|
Megna M, Lauletta G, Tommasino N, Salsano A, Battista T, Ruggiero A, Martora F, Potestio L. Management of Psoriasis Patients with Serious Infectious Diseases. Adv Ther 2024; 41:2099-2111. [PMID: 38709397 PMCID: PMC11133026 DOI: 10.1007/s12325-024-02873-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024]
Abstract
The management of patients affected by moderate-to-severe psoriasis may be challenging, in particular in patients with serious infectious diseases [tuberculosis (TB), hepatitis B and C, HIV, COVID-19]. Indeed, these infections should be ruled out before starting and during systemic treatment for psoriasis. Currently, four conventional systemic drugs (methotrexate, dimethyl fumarate, acitretin, cyclosporine), four classes of biologics (anti-tumour necrosis factor alpha, anti-interleukin (IL)12/23, anti-IL-17s, and anti-IL-23], and two oral small molecules (apremilast, deucravacitinib) have been licensed for the treatment of moderate-to-severe psoriasis. Each of these drugs is characterized by a unique safety profile which should be considered before starting therapy. Indeed, some comorbidities or risk factors may limit their use. In this context, the aim of this manuscript was to evaluate the management of patients affected by moderate-to-severe psoriasis with serious infectious diseases.
Collapse
Affiliation(s)
- Matteo Megna
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Giuseppe Lauletta
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Nello Tommasino
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Antonia Salsano
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Teresa Battista
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Angelo Ruggiero
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Fabrizio Martora
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Luca Potestio
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
50
|
Choreño-Parra JA, Ramon-Luing LA, Castillejos M, Ortega-Martínez E, Tapia-García AR, Matías-Martínez MB, Cruz-Lagunas A, Ramírez-Martínez G, Gómez-García IA, Ramírez-Noyola JA, Garcia-Padrón B, López-Salinas KG, Jiménez-Juárez F, Guadarrama-Ortiz P, Salinas-Lara C, Bozena-Piekarska K, Muñóz-Torrico M, Chávez-Galán L, Zúñiga J. The rs11684747 and rs55790676 SNPs of ADAM17 influence tuberculosis susceptibility and plasma levels of TNF, TNFR1, and TNFR2. Front Microbiol 2024; 15:1392782. [PMID: 38881671 PMCID: PMC11177089 DOI: 10.3389/fmicb.2024.1392782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction The proteolytic activity of A Disintegrin and Metalloproteinase 17 (ADAM17) regulates the release of tumor necrosis factor (TNF) and TNF receptors (TNFRs) from cell surfaces. These molecules play important roles in tuberculosis (TB) shaping innate immune reactions and granuloma formation. Methods Here, we investigated whether single nucleotide polymorphisms (SNPs) of ADAM17 influence TNF and TNFRs levels in 224 patients with active TB (ATB) and 118 healthy close contacts. Also, we looked for significant associations between SNPs of ADAM17 and ATB status. TNF, TNFR1, and TNFR2 levels were measured in plasma samples by ELISA. Four SNPs of ADAM17 (rs12692386, rs1524668, rs11684747, and rs55790676) were analyzed in DNA isolated from peripheral blood leucocytes. The association between ATB status, genotype, and cytokines was analyzed by multiple regression models. Results Our results showed a higher frequency of rs11684747 and rs55790676 in close contacts than ATB patients. Coincidentally, heterozygous to these SNPs of ADAM17 showed higher plasma levels of TNF compared to homozygous to their respective ancestral alleles. Strikingly, the levels of TNF and TNFRs distinguished participant groups, with ATB patients displaying lower TNF and higher TNFR1/TNFR2 levels compared to their close contacts. Conclusion These findings suggest a role for SNPs of ADAM17 in genetic susceptibility to ATB.
Collapse
Affiliation(s)
- José Alberto Choreño-Parra
- Dirección de Enseñanza, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Lucero A Ramon-Luing
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Manuel Castillejos
- Departamento de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Emmanuel Ortega-Martínez
- Posgrado en Ciencias Quimicobiológicas, SEPI, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Department of Pathology, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Alan Rodrigo Tapia-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Melvin Barish Matías-Martínez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Gustavo Ramírez-Martínez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Jazmín Ariadna Ramírez-Noyola
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Beatriz Garcia-Padrón
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Karen Gabriel López-Salinas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | | | - Citlaltepetl Salinas-Lara
- Department of Pathology, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Karolina Bozena-Piekarska
- Dirección de Enseñanza, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcela Muñóz-Torrico
- Clínica de Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| |
Collapse
|