1
|
Lemson A, van Laarhoven A, Kurver L, Stemkens R, Aarnoutse R, Boeree M, van Ingen J, Hoefsloot W. Treatment of nontuberculous mycobacterial pulmonary disease requires a stepwise and multidisciplinary approach. Expert Rev Respir Med 2025; 19:287-299. [PMID: 40114560 DOI: 10.1080/17476348.2025.2479615] [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: 01/02/2025] [Revised: 03/01/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Nontuberculous mycobacterial pulmonary disease (NTM-PD) occurs in people with inborn or acquired susceptibility factors. Current treatment guideline recommendations include a watchful waiting strategy, antimycobacterial and surgical treatment, with a comprehensive assessment of clinical, microbiological, and radiological factors determining which approach is most suitable. AREAS COVERED Treatment outcomes in NTM-PD are unsatisfactory with culture conversion rates varying from 30 to 80% and recurrence rates up to 50%. Possible explanations include our insufficient knowledge and management of host susceptibility factors, poor guideline adherence by physicians, frequent adverse drug reactions demanding premature discontinuation, inadequate drug exposures due to both drug-drug interactions and inter-patient variability in pharmacokinetics, and a lack of pharmacokinetics/pharmacodynamics targets. EXPERT OPINION We describe a stepwise approach to NTM-PD treatment, complementing existing guidelines, including recommendations for a multidisciplinary assessment, classification of disease severity, personalized supportive care, antimycobacterial treatment, adjuvant surgery, and host-directed therapies. The recommendations are informed by PubMed literature and the authors' clinical expertise. Recognizing that our experience is shaped within a specialized reference clinic, we acknowledge that some of these recommendations may not be applicable in all settings.
Collapse
Affiliation(s)
- Arthur Lemson
- Department of Pulmonary Diseases, Research Institute for Medical Innovation, Radboud University Medical Center, Radboudumc Community for Infectious Diseases, Nijmegen, Netherlands
| | - Arjan van Laarhoven
- Department of Internal Medicine and Radboud Community for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lisa Kurver
- Department of Internal Medicine and Radboud Community for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ralf Stemkens
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rob Aarnoutse
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Martin Boeree
- Department of Pulmonary Diseases, Research Institute for Medical Innovation, Radboud University Medical Center, Radboudumc Community for Infectious Diseases, Nijmegen, Netherlands
| | - Jakko van Ingen
- Department of Medical Microbiology, Radboudumc Community for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Wouter Hoefsloot
- Department of Pulmonary Diseases, Research Institute for Medical Innovation, Radboud University Medical Center, Radboudumc Community for Infectious Diseases, Nijmegen, Netherlands
| |
Collapse
|
2
|
Liu C, Wu W, Wang L, Li J. Case report: co-infection of Scedosporium and Mycobacterium in lungs. AME Case Rep 2024; 9:3. [PMID: 39866262 PMCID: PMC11760516 DOI: 10.21037/acr-24-9] [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: 01/10/2024] [Accepted: 08/22/2024] [Indexed: 01/28/2025]
Abstract
Background There are hundreds of pathogens that cause lung infections. Compared to infections caused by a single pathogen, mixed infections account for a larger proportion of pulmonary infections and have a more severe clinical presentation, while treatment options differ between the two. We aimed to explore the advantages of metagenomic next-generation sequencing (mNGS) in the diagnosis and treatment of mixed infections. Case Description To investigate the specific pathogens in a 79-year-old male pneumonia patient who had recurrent cough with poor empirical treatment, we collected bronchoalveolar lavage fluid (BALF) from the patient and performed mNGS technology, along with Sanger sequencing and polymerase chain reaction (PCR) was carried out to confirm the authenticity of the pathogens detected by mNGS. The findings showed that rare pathogen Scedosporium boydii (S. boydii, reads: 18) and Mycobacterium avium complex (MAC, reads: 19) were detected, and the patient was subsequently transferred to another hospital for the same mNGS with the same results as the first detection. Therefore, combined treatment with voriconazole, ethambutol, azithromycin, and levofloxacin were given to the S. boydii and MAC for 1 week, and then patient's condition improved and discharged. Conclusions mNGS, a non-targeted sequencing technology, could improve the efficiency of clinical diagnosis for mixed infection of rare or atypical pathogens, bring new ideas for clinical pathogen diagnosis, and improve patient prognosis.
Collapse
Affiliation(s)
- Chengying Liu
- Division of Respiratory and Critical Care Medicine, Jiangyin People’s Hospital Affiliated to Nantong, Jiangyin, China
| | | | - Lan Wang
- Division of Respiratory and Critical Care Medicine, Jiangyin People’s Hospital Affiliated to Nantong, Jiangyin, China
| | - Jie Li
- Division of Respiratory and Critical Care Medicine, Jiangyin People’s Hospital Affiliated to Nantong, Jiangyin, China
| |
Collapse
|
3
|
Van Braeckel E, Bosteels C. Growing from common ground: nontuberculous mycobacteria and bronchiectasis. Eur Respir Rev 2024; 33:240058. [PMID: 38960614 PMCID: PMC11220627 DOI: 10.1183/16000617.0058-2024] [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: 03/19/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchiectasis and nontuberculous mycobacteria (NTM) are intricately intertwined, with NTM capable of being both a cause and consequence of bronchiectatic disease. This narrative review focuses on the common ground of bronchiectasis and NTM pulmonary disease (NTM-PD) in terms of diagnostic approach, underlying risk factors and treatment strategies. NTM-PD diagnosis relies on a combination of clinical, radiological and microbiological criteria. Although their epidemiology is complicated by detection and reporting biases, the prevalence and pathogenicity of NTM species vary geographically, with Mycobacterium avium complex and Mycobacterium abscessus subspecies most frequently isolated in bronchiectasis-associated NTM-PD. Diagnosis of nodular bronchiectatic NTM-PD should prompt investigation of host factors, including disorders of mucociliary clearance, connective tissue diseases and immunodeficiencies, either genetic or acquired. Treatment of NTM-PD in bronchiectasis involves a multidisciplinary approach and considers the (sub)species involved, disease severity and comorbidities. Current guideline-based antimicrobial treatment of NTM-PD is considered long, cumbersome and unsatisfying in terms of outcomes. Novel treatment regimens and strategies are being explored, including rifampicin-free regimens and inclusion of clofazimine and inhaled antibiotics. Host-directed therapies, such as immunomodulators and cytokine-based therapies, might enhance antimycobacterial immune responses. Optimising supportive care, as well as pathogen- and host-directed strategies, is crucial, highlighting the need for personalised approaches tailored to individual patient needs. Further research is warranted to elucidate the complex interplay between host and mycobacterial factors, informing more effective management strategies.
Collapse
Affiliation(s)
- Eva Van Braeckel
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Respiratory Infection and Defense Lab (RIDL), Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- European Reference Network on rare respiratory diseases (ERN-LUNG)
| | - Cédric Bosteels
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Respiratory Infection and Defense Lab (RIDL), Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- European Reference Network on rare respiratory diseases (ERN-LUNG)
| |
Collapse
|
4
|
Zhang H, Chen Y, Jiang X, Gu Q, Yao J, Wang X, Wu J. Unveiling the landscape of cytokine research in glioma immunotherapy: a scientometrics analysis. Front Pharmacol 2024; 14:1333124. [PMID: 38259287 PMCID: PMC10800575 DOI: 10.3389/fphar.2023.1333124] [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: 11/04/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Background: Cytokines modulate the glioma tumor microenvironment, influencing occurrence, progression, and treatment response. Strategic cytokine application may improve glioma immunotherapy outcomes. Gliomas remain refractory to standard therapeutic modalities, but immunotherapy shows promise given the integral immunomodulatory roles of cytokines. However, systematic evaluation of cytokine glioma immunotherapy research is absent. Bibliometric mapping of the research landscape, recognition of impactful contributions, and elucidation of evolutive trajectories and hot topics has yet to occur, potentially guiding future efforts. Here, we analyzed the structure, evolution, trends, and hotspots of the cytokine glioma immunotherapy research field, subsequently focusing on avenues for future investigation. Methods: This investigation conducted comprehensive bibliometric analyses on a corpus of 1529 English-language publications, from 1 January 2000, to 4 October 2023, extracted from the Web of Science database. The study employed tools including Microsoft Excel, Origin, VOSviewer, CiteSpace, and the Bibliometrix R package, to systematically assess trends in publication, contributions from various countries, institutions, authors, and journals, as well as to examine literature co-citation and keyword distributions within the domain of cytokines for glioma immunotherapy. The application of these methodologies facilitated a detailed exploration of the hotspots, the underlying knowledge structure, and the developments in the field of cytokines for glioma immunotherapy. Results: This bibliometric analysis revealed an exponential growth in annual publications, with the United States, China, and Germany as top contributors. Reviews constituted 17% and research articles 83% of total publications. Analysis of keywords like "interleukin-13," "TGF-beta," and "dendritic cells" indicated progression from foundational cytokine therapies to sophisticated understanding of the tumor microenvironment and immune dynamics. Key research avenues encompassed the tumor microenvironment, epidermal growth factor receptor, clinical trials, and interleukin pathways. This comprehensive quantitative mapping of the glioma immunotherapy cytokine literature provides valuable insights to advance future research and therapeutic development. Conclusion: This study has identified remaining knowledge gaps regarding the role of cytokines in glioma immunotherapy. Future research will likely focus on the tumor microenvironment, cancer vaccines, epidermal growth factor receptor, and interleukin-13 receptor alpha 2. Glioma immunotherapy development will continue through investigations into resistance mechanisms, microglia and macrophage biology, and interactions within the complex tumor microenvironment.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Chen
- Gamma Knife Center, Department of Oncology, Department of Neurological Surgery, Tianjin Huanhu Hospital, Tianjin Medical University, Tianjin, China
| | - Xinzhan Jiang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Qiang Gu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiahao Yao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuefeng Wang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianghua Wu
- School of Nursing, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong, China
| |
Collapse
|
5
|
Anidi IU, Olivier KN. Host-Directed Therapy in Nontuberculous Mycobacterial Pulmonary Disease: Preclinical and Clinical Data Review. Clin Chest Med 2023; 44:839-845. [PMID: 37890920 PMCID: PMC10614072 DOI: 10.1016/j.ccm.2023.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Standard treatment of nontuberculous mycobacterial pulmonary disease (NTM-PD) infection involves a multi-drug antimicrobial regimen for at least 12 months. The length, complexity, and side effect profile of antibiotic therapy for NTM-PD pose significant difficulties for maintaining patient adherence. Furthermore, physician adherence to NTM guidelines suffers for similar reasons to the extent that a study evaluating treatment approaches across multiple specialties found that only 13% of antibiotic regimens met ATS/IDSA guidelines. For this reason, a great need exists for therapy that augments the current armamentarium of antimicrobial chemotherapeutics or provides an alternative approach for decreasing host mycobacterial burden. As our knowledge of the mechanisms driving protective responses to NTM-PD infections by mammalian hosts expand, these processes provide novel therapeutic targets. These agents, which are commonly referred to as host-directed therapies (HDTs) have the potential of providing the much-needed boost to the nontuberculous mycobacterial therapeutic pipeline. In this review, we will focus on translational research and clinical trial data that detail the creation of therapeutic modalities developed to improve host mechanical protection and immunologic responses to PNTM infection.
Collapse
Affiliation(s)
- Ifeanyichukwu U Anidi
- Pulmonary Division, National Heart, Lung and Blood Institute, National Institutes of Health, 33 North Drive, Room 1W10A, Bethesda, MD 20892, USA.
| | - Kenneth N Olivier
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina School of Medicine, 125 Mason Farm Road, CB#7248, 7214 Marsico Hall, Chapel Hill, NC 27599-7248, USA
| |
Collapse
|
6
|
Kajiwara C, Shiozawa A, Urabe N, Yamaguchi T, Kimura S, Akasaka Y, Ishii Y, Tateda K. Apoptosis Inhibitor of Macrophages Contributes to the Chronicity of Mycobacterium avium Infection by Promoting Foamy Macrophage Formation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:431-441. [PMID: 36602769 DOI: 10.4049/jimmunol.2200306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 12/07/2022] [Indexed: 01/06/2023]
Abstract
In Mycobacterium avium infections, macrophages play a critical role in the host defense response. Apoptosis inhibitor of macrophage (AIM), also known as CD5L, may represent a novel supportive therapy against various diseases, including metabolic syndrome and infectious diseases. The mechanisms of AIM include modulating lipid metabolism in macrophages and other host cells. We investigated the role of AIM in M. avium infections in vitro and in vivo. In a mouse model of M. avium pneumonia, foamy macrophages were induced 6 wk after infection. The bacteria localized in these macrophages. Flow cytometric analysis also confirmed that the percentage of CD11chighMHCclassIIhigh interstitial and alveolar macrophages, a cell surface marker defined as foamy macrophages, increased significantly after infection. AIM in alveolar lavage fluid and serum gradually increased after infection. Administration of recombinant AIM significantly increased the number of bacteria in the lungs of mice, accompanied by the induction of inflammatory cytokine and iNOS expression. In mouse bone marrow-derived macrophages, the mRNA expression of AIM after M. avium infection and the amount of AIM in the supernatant increased prior to the increase in intracellular bacteria. Infected cells treated with anti-AIM Abs had fewer bacteria and a higher percentage of apoptosis-positive cells than infected cells treated with isotype control Abs. Finally, AIM in the sera of patients with M. avium-pulmonary disease was measured and was significantly higher than in healthy volunteers. This suggests that AIM production is enhanced in M. avium-infected macrophages, increasing macrophage resistance to apoptosis and providing a possible site for bacterial growth.
Collapse
Affiliation(s)
- Chiaki Kajiwara
- Department of Microbiology and Infectious Diseases, Faculty of Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Ayako Shiozawa
- Department of Microbiology and Infectious Diseases, Faculty of Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Naohisa Urabe
- Department of Respiratory Medicine, Toho University Omori Medical Center, Tokyo, Japan
| | - Tetsuo Yamaguchi
- Department of Microbiology and Infectious Diseases, Faculty of Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Soichiro Kimura
- Faculty of Pharmaceutical Sciences, Department of Clinical Pharmacy, Shonan University of Medical Sciences, Kanagawa, Japan; and
| | - Yoshikiyo Akasaka
- Department of Diagnostic Pathology, Saiseikai Yokohamashi Tobu Hospital, Kanagawa, Japan
| | - Yoshikazu Ishii
- Department of Microbiology and Infectious Diseases, Faculty of Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Kazuhiro Tateda
- Department of Microbiology and Infectious Diseases, Faculty of Medicine, Toho University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Hendrix C, McCrary M, Hou R, Abate G. Diagnosis and Management of Pulmonary NTM with a Focus on Mycobacterium avium Complex and Mycobacterium abscessus: Challenges and Prospects. Microorganisms 2022; 11:microorganisms11010047. [PMID: 36677340 PMCID: PMC9861392 DOI: 10.3390/microorganisms11010047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Nontuberculous mycobacteria (NTM) are ubiquitous. NTM can affect different organs and may cause disseminated diseases, but the pulmonary form is the most common form. Pulmonary NTM is commonly seen in patients with underlying diseases. Pulmonary Mycobacterium avium complex (MAC) is the most common NTM disease and M. abscessus (MAB) is the most challenging to treat. This review is prepared with the following objectives: (a) to evaluate new methods available for the diagnosis of pulmonary MAC or MAB, (b) to assess advances in developing new therapeutics and their impact on treatment of pulmonary MAC or MAB, and (c) to evaluate the prospects of preventive strategies including vaccines against pulmonary MAC or MAB. METHODS A literature search was conducted using PubMed/MEDLINE and multiple search terms. The search was restricted to the English language and human studies. The database query resulted in a total of 197 publications. After the title and abstract review, 64 articles were included in this analysis. RESULTS The guidelines by the American Thoracic Society (ATS), European Respiratory Society (ERS), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), and Infectious Diseases Society of America (IDSA) are widely applicable. The guidelines are based on expert opinion and there may be a need to broaden criteria to include those with underlying lung diseases who may not fulfill some of the criteria as 'probable cases' for better follow up and management. Some cases with only one culture-positive sputum sample or suggestive histology without a positive culture may benefit from new methods of confirming NTM infection. Amikacin liposomal inhalation suspension (ALIS), gallium containing compounds and immunotherapies will have potential in the management of pulmonary MAC and MAB. CONCLUSIONS the prevalence of pulmonary NTM is increasing. The efforts to optimize diagnosis and treatment of pulmonary NTM are encouraging. There is still a need to develop new diagnostics and therapeutics.
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW This review provides readers with examples of refractory infections due to inborn errors of immunity, highlighting how they may be successfully treated by deducing and targeting the underlying immunodeficiency. RECENT FINDINGS The use of host-directed immunotherapy to treat infectious disease in inborn errors of immunity is currently limited but growing. Different strategies include depleting the cellular reservoir for pathogens with restricted cell-tropism; augmenting the diminished effector response; and restoring molecular equipoise. The immunotherapies illustrated are existing drugs that have been re-purposed and rationally used, depending on the molecular or cellular impact of the mutation. As more biologic response modifiers and molecular targeted therapies are developed for other indications, they open the avenues for their use in inborn errors of immunity. Conversely, as more molecular pathways underlying defective immune responses and refractory infections are elucidated, they lend themselves to tractability with these emerging therapies. SUMMARY Infections that fail appropriate antimicrobial therapy are a harbinger of underlying inborn errors of immunity. Dissecting the mechanism by which the immune system fails provides opportunities to target the host response and make it succeed.
Collapse
|
9
|
Gleeson LE, Waterer G. Beyond antibiotics: recent developments in the diagnosis and management of nontuberculous mycobacterial infection. Breathe (Sheff) 2022; 18:210171. [PMID: 36337137 PMCID: PMC9584569 DOI: 10.1183/20734735.0171-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
Nontuberculous mycobacteria (NTM) pulmonary disease represents a significant clinical challenge with suboptimal therapy and increasing prevalence globally. Although clinical practice guidelines seek to standardise the approach to diagnosis and treatment of NTM disease, a lack of robust evidence limits their utility and significant variability exists in clinical practice. Here we walk through some novel approaches in diagnosis and therapy that are under development to tackle a disease where traditional strategies are failing. Prevalence of NTM disease is rising globally, yet current diagnostic and therapeutic strategies are lacking. This review describes some burgeoning diagnostic and therapeutic approaches, but it is clear that real progress will need more focused attention.https://bit.ly/3O0K2SP
Collapse
|
10
|
Hayes D, Shukla RK, Cheng Y, Gecili E, Merling MR, Szczesniak RD, Ziady AG, Woods JC, Hall-Stoodley L, Liyanage NP, Robinson RT. Tissue-localized immune responses in people with cystic fibrosis and respiratory nontuberculous mycobacteria infection. JCI Insight 2022; 7:157865. [PMID: 35536650 DOI: 10.1172/jci.insight.157865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/29/2022] [Indexed: 11/17/2022] Open
Abstract
Nontuberculous mycobacteria (NTM) are an increasingly common cause of respiratory infection in people with cystic fibrosis (PwCF). Relative to those with no history of NTM infection (CF-NTMNEG), PwCF and a history of NTM infection (CF-NTMPOS) are more likely to develop severe lung disease and experience complications over the course of treatment. In other mycobacterial infections (e.g. tuberculosis), an overexuberant immune response causes pathology and compromises organ function; however, since the immune profiles of CF-NTMPOS and CF-NTMNEG airways are largely unexplored, it is unknown which if any immune responses distinguish these cohorts or concentrate in damaged tissues. Here we evaluated lung lobe-specific immune profiles of three cohorts (CF-NTMPOS, CF-NTMNEG, and non-CF adults) and found that CF-NTMPOS airways are distinguished by a hyper-inflammatory cytokine profile. Importantly, the CF-NTMPOS airway immune profile was dominated by B cells, classical macrophages and the cytokines which support their accumulation. These and other immunological differences between cohorts, including the near absence of NK cells and complement pathway members, were enriched in the most damaged lung lobes. The implications of these findings for our understanding of lung disease in PwCF are discussed, as are how they may inform the development of host-directed therapies to improve NTM disease treatment.
Collapse
Affiliation(s)
- Don Hayes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Rajni Kant Shukla
- Department of Microbial Infection and Immunity, The Ohio State University, columbus, United States of America
| | - Yizi Cheng
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Emrah Gecili
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Marlena R Merling
- Department of Microbial Infection and Immunity, The Ohio State University, columbus, United States of America
| | - Rhonda D Szczesniak
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Assem G Ziady
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Jason C Woods
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Luanne Hall-Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, columbus, United States of America
| | - Namal Pm Liyanage
- Department of Microbial Infection and Immunity, The Ohio State University, columbus, United States of America
| | - Richard T Robinson
- Department of Microbial Infection and Immunity, The Ohio State University, columbus, United States of America
| |
Collapse
|
11
|
Kumar K, Daley CL, Griffith DE, Loebinger MR. Management of Mycobacterium avium complex and Mycobacterium abscessus pulmonary disease: therapeutic advances and emerging treatments. Eur Respir Rev 2022; 31:210212. [PMID: 35140106 PMCID: PMC9488909 DOI: 10.1183/16000617.0212-2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022] Open
Abstract
Nontuberculous mycobacterial pulmonary disease (NTM-PD) remains a challenging condition to diagnose and treat effectively. Treatment of NTM-PD is prolonged, frequently associated with adverse effects and has variable success. In this review, we consider the factors influencing clinicians when treating NTM-PD and discuss outcomes from key studies on the pharmacological management of Mycobacterium avium complex pulmonary disease and M. abscessus pulmonary disease. We highlight issues relating to treatment-related toxicity and provide an overview of repurposed and emerging therapies for NTM-PD.
Collapse
Affiliation(s)
- Kartik Kumar
- National Heart and Lung Institute, Imperial College London, London, UK
- Host Defence Unit, Dept of Respiratory Medicine, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Charles L Daley
- Division of Mycobacterial and Respiratory Infections, Dept of Medicine, National Jewish Health, Denver, CO, USA
- School of Medicine, University of Colorado, Aurora, CO, USA
| | - David E Griffith
- Division of Mycobacterial and Respiratory Infections, Dept of Medicine, National Jewish Health, Denver, CO, USA
| | - Michael R Loebinger
- National Heart and Lung Institute, Imperial College London, London, UK
- Host Defence Unit, Dept of Respiratory Medicine, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
12
|
Autologous Cytokine-Induced Killer Cell Immunotherapy Enhances Chemotherapy Efficacy against Multidrug-Resistant Tuberculosis. J Immunol Res 2022; 2022:2943113. [PMID: 35340584 PMCID: PMC8947923 DOI: 10.1155/2022/2943113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Multidrug-resistant tuberculosis (MDR-TB) causes persistent infection and challenges tuberculosis control worldwide. T cell-mediated immunity plays a critical role in controlling Mycobacterium tuberculosis (Mtb) infection, and therefore, enhancing Mtb-specific T cell immune responses represents a promising therapeutic strategy against TB. Cytokine-induced killer (CIK) immunotherapy is based on autologous infusion of in vitro expanded bulk T cells, which include both pathogen-specific and nonspecific T cells from patient peripheral blood mononuclear cells (PBMC) into TB patients. Preclinical mouse studies have shown that the adoptive T cell therapy inhibited Mtb infection. However, the efficacy of CIK immunotherapy in the treatment of MDR-TB infection has not been evaluated in clinical trials. Methods We performed a retrospective study of MDR-TB patients who received CIK immunotherapy in combination with anti-TB chemotherapy and those who had standard chemotherapy. Results Our results showed that CIK immunotherapy in combination with anti-TB chemotherapy treatment increased the conversion rate of sputum smear and Mtb culture, alleviated symptoms, improved lesion absorption, and increased recovery. The kinetics of serology and immunology index monitoring data showed good safety profiles for the CIK treatment. Conclusion Our study has provided strong evidence that CIK immunotherapy in combination with anti-TB chemotherapy is beneficial for MDR-TB patients. A multicenter clinical trial is warranted to evaluate CIK as a new immune therapy for MDR-TB.
Collapse
|
13
|
Pennington KM, Vu A, Challener D, Rivera CG, Shweta FNU, Zeuli JD, Temesgen Z. Approach to the diagnosis and treatment of non-tuberculous mycobacterial disease. J Clin Tuberc Other Mycobact Dis 2021; 24:100244. [PMID: 34036184 PMCID: PMC8135042 DOI: 10.1016/j.jctube.2021.100244] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Non-tuberculous mycobacteria (NTM) is a collective name given to a group of more than 190 species of Mycobacterium. The clinical presentation for most NTM infections is non-specific, often resulting in delayed diagnosis. Further complicating matters is that NTM organisms can be difficult to isolate. Medications used to treat NTM infection can be difficult for patients to tolerate, and prolonged courses of anti-mycobacterial therapy are often required for adequate suppression or eradication. Herein, we review different NTM syndromes, appropriate diagnostic tests, and treatment regimens.
Collapse
Key Words
- ADR, adverse drug reactions
- AFB, acid fast bacilli
- AST, antimicrobial-susceptibility testing
- ATS, American Thoracic Society
- BCG, Bacille Calmette-Guerin
- CLSI, Clinical and Laboratory Standards Institute
- COPD, chronic obstructive pulmonary disease
- ECG, electrocardiogram
- EMB, ethambutol
- Erm, erythromycin ribosomal methylase
- FDA, Food and Drug Administration
- HIV, human immunodeficiency virus
- HRCT, high resolution computed tomography
- IDSA, Infectious Disease Society of America
- INF-γ, interferon- γ
- INH, isoniazid
- MAC, Mycobacterium avium complex
- MALDI-TOF, matrix-assisted laser desorption ionization time-of-flight mass spectrometry
- MGIT, mycobacteria growth indicator tube
- MIC, minimum inhibitory concentrations
- Mycobacterium abscessus
- Mycobacterium avium
- NTM, non-tuberculous mycobacteria
- Non-tuberculous mycobacteria
- PCR, polymerase chain reaction
- PFT, pulmonary function test
- TB, tuberculosis
- TDM, therapeutic drug monitoring
Collapse
Affiliation(s)
- Kelly M Pennington
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic Rochester, MN, USA
| | - Ann Vu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic Rochester, MN, USA
| | - Douglas Challener
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic Rochester, MN, USA
| | | | - F N U Shweta
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic Rochester, MN, USA
| | - John D Zeuli
- Department of Pharmacy, Mayo Clinic Rochester, MN, USA
| | - Zelalem Temesgen
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
14
|
Kilinç G, Saris A, Ottenhoff THM, Haks MC. Host-directed therapy to combat mycobacterial infections. Immunol Rev 2021; 301:62-83. [PMID: 33565103 PMCID: PMC8248113 DOI: 10.1111/imr.12951] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/27/2020] [Indexed: 12/27/2022]
Abstract
Upon infection, mycobacteria, such as Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria (NTM), are recognized by host innate immune cells, triggering a series of intracellular processes that promote mycobacterial killing. Mycobacteria, however, have developed multiple counter‐strategies to persist and survive inside host cells. By manipulating host effector mechanisms, including phagosome maturation, vacuolar escape, autophagy, antigen presentation, and metabolic pathways, pathogenic mycobacteria are able to establish long‐lasting infection. Counteracting these mycobacteria‐induced host modifying mechanisms can be accomplished by host‐directed therapeutic (HDT) strategies. HDTs offer several major advantages compared to conventional antibiotics: (a) HDTs can be effective against both drug‐resistant and drug‐susceptible bacteria, as well as potentially dormant mycobacteria; (b) HDTs are less likely to induce bacterial drug resistance; and (c) HDTs could synergize with, or shorten antibiotic treatment by targeting different pathways. In this review, we will explore host‐pathogen interactions that have been identified for Mtb for which potential HDTs impacting both innate and adaptive immunity are available, and outline those worthy of future research. We will also discuss possibilities to target NTM infection by HDT, although current knowledge regarding host‐pathogen interactions for NTM is limited compared to Mtb. Finally, we speculate that combinatorial HDT strategies can potentially synergize to achieve optimal mycobacterial host immune control.
Collapse
Affiliation(s)
- Gül Kilinç
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anno Saris
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
15
|
Mendelian Susceptibility to Mycobacterial Disease: The First Case of a Diagnosed Adult Patient in the Czech Republic. Case Reports Immunol 2020; 2020:8836685. [PMID: 33414972 PMCID: PMC7769627 DOI: 10.1155/2020/8836685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/09/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
We present a case of a 42-year-old woman with Mendelian susceptibility to mycobacterial disease. The disease was diagnosed at an adult age with relatively typical clinical manifestations; the skeleton, joints, and soft tissues were affected by nontuberculous mycobacteria: Mycobacterium lentiflavum, M. kansasii, and M. avium. A previously published loss-of-function and functionally validated variant NM_000416.2:c.819_822delTAAT in IFNGR1 in a heterozygous state was detected using whole-exome sequencing. After interferon-γ therapy was started at a dose of 200 µg/m2 three times a week, there was significant clinical improvement, with the need to continue the macrolide-based combination regimen. In the last 4 months, she has been in this therapy without the need for antibiotic treatment.
Collapse
|
16
|
Bruno M, Zweijpfenning SMH, Verhoeven J, Boeree MJ, Netea MG, van de Veerdonk FL, van Ingen J, Hoefsloot W. Subtle immunodeficiencies in nodular-bronchiectatic Mycobacterium avium complex lung disease. ERJ Open Res 2020; 6:00548-2020. [PMID: 33123562 PMCID: PMC7569207 DOI: 10.1183/23120541.00548-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 11/05/2022] Open
Abstract
Patients with nodular-bronchiectatic MAC lung disease have dysregulated adaptive immunity with defective IL-17 and IFN-γ production, and IL-10 overproduction. This suggests a role for adjunctive immunomodulatory treatments. https://bit.ly/33AALwx.
Collapse
Affiliation(s)
- Mariolina Bruno
- Dept of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne M H Zweijpfenning
- Radboudumc Center for Infectious Diseases, Dept of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeske Verhoeven
- Radboudumc Center for Infectious Diseases, Dept of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin J Boeree
- Radboudumc Center for Infectious Diseases, Dept of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Dept of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.,Dept for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Frank L van de Veerdonk
- Dept of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jakko van Ingen
- Radboudumc Center for Infectious Diseases, Dept of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wouter Hoefsloot
- Radboudumc Center for Infectious Diseases, Dept of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Abreu R, Giri P, Quinn F. Host-Pathogen Interaction as a Novel Target for Host-Directed Therapies in Tuberculosis. Front Immunol 2020; 11:1553. [PMID: 32849525 PMCID: PMC7396704 DOI: 10.3389/fimmu.2020.01553] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/12/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) has been a transmittable human disease for many thousands of years, and M. tuberculosis is again the number one cause of death worldwide due to a single infectious agent. The intense 6- to 10-month process of multi-drug treatment, combined with the adverse side effects that can run the spectrum from gastrointestinal disturbances to liver toxicity or peripheral neuropathy are major obstacles to patient compliance and therapy completion. The consequent increase in multidrug resistant TB (MDR-TB) and extensively drug resistant TB (XDR-TB) cases requires that we increase our arsenal of effective drugs, particularly novel therapeutic approaches. Over the millennia, host and pathogen have evolved mechanisms and relationships that greatly influence the outcome of infection. Understanding these evolutionary interactions and their impact on bacterial clearance or host pathology will lead the way toward rational development of new therapeutics that favor enhancing a host protective response. These host-directed therapies have recently demonstrated promising results against M. tuberculosis, adding to the effectiveness of currently available anti-mycobacterial drugs that directly kill the organism or slow mycobacterial replication. Here we review the host-pathogen interactions during M. tuberculosis infection, describe how M. tuberculosis bacilli modulate and evade the host immune system, and discuss the currently available host-directed therapies that target these bacterial factors. Rather than provide an exhaustive description of M. tuberculosis virulence factors, which falls outside the scope of this review, we will instead focus on the host-pathogen interactions that lead to increased bacterial growth or host immune evasion, and that can be modulated by existing host-directed therapies.
Collapse
Affiliation(s)
| | | | - Fred Quinn
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
18
|
Chin KL, Sarmiento ME, Alvarez-Cabrera N, Norazmi MN, Acosta A. Pulmonary non-tuberculous mycobacterial infections: current state and future management. Eur J Clin Microbiol Infect Dis 2020; 39:799-826. [PMID: 31853742 PMCID: PMC7222044 DOI: 10.1007/s10096-019-03771-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022]
Abstract
Currently, there is a trend of increasing incidence in pulmonary non-tuberculous mycobacterial infections (PNTM) together with a decrease in tuberculosis (TB) incidence, particularly in developed countries. The prevalence of PNTM in underdeveloped and developing countries remains unclear as there is still a lack of detection methods that could clearly diagnose PNTM applicable in these low-resource settings. Since non-tuberculous mycobacteria (NTM) are environmental pathogens, the vicinity favouring host-pathogen interactions is known as important predisposing factor for PNTM. The ongoing changes in world population, as well as socio-political and economic factors, are linked to the rise in the incidence of PNTM. Development is an important factor for the improvement of population well-being, but it has also been linked, in general, to detrimental environmental consequences, including the rise of emergent (usually neglected) infectious diseases, such as PNTM. The rise of neglected PNTM infections requires the expansion of the current efforts on the development of diagnostics, therapies and vaccines for mycobacterial diseases, which at present, are mainly focused on TB. This review discuss the current situation of PNTM and its predisposing factors, as well as the efforts and challenges for their control.
Collapse
Affiliation(s)
- Kai Ling Chin
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah (UMS), Kota Kinabalu, Sabah, Malaysia.
| | - Maria E Sarmiento
- School of Health Sciences, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan, Malaysia
| | - Nadine Alvarez-Cabrera
- Center for Discovery and Innovation (CDI), Hackensack Meridian School of Medicine at Seton Hall University, Nutley, NJ, USA
| | - Mohd Nor Norazmi
- School of Health Sciences, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan, Malaysia
| | - Armando Acosta
- School of Health Sciences, Universiti Sains Malaysia (USM), Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
19
|
Ratnatunga CN, Lutzky VP, Kupz A, Doolan DL, Reid DW, Field M, Bell SC, Thomson RM, Miles JJ. The Rise of Non-Tuberculosis Mycobacterial Lung Disease. Front Immunol 2020; 11:303. [PMID: 32194556 PMCID: PMC7062685 DOI: 10.3389/fimmu.2020.00303] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/06/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence and number of deaths from non-tuberculous mycobacterial (NTM) disease have been steadily increasing globally. These lesser known “cousins” of Mycobacterium tuberculosis (TB) were once thought to be harmless environmental saprophytics and only dangerous to individuals with defective lung structure or the immunosuppressed. However, NTM are now commonly infecting seemingly immune competent children and adults at increasing rates through pulmonary infection. This is of concern as the pathology of NTM is difficult to treat. Indeed, NTM have become extremely antibiotic resistant, and now have been found to be internationally dispersed through person-to-person contact. The reasons behind this NTM increase are only beginning to be elucidated. Solutions to the problem are needed given NTM disease is more common in the tropics. Importantly, 40% of the world's population live in the tropics and due to climate change, the Tropics are expanding which will increase NTM infection regions. This review catalogs the global and economic disease burden, at risk populations, treatment options, host-bacterial interaction, immune dynamics, recent developments and research priorities for NTM disease.
Collapse
Affiliation(s)
- Champa N. Ratnatunga
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- *Correspondence: Champa N. Ratnatunga
| | - Viviana P. Lutzky
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Kupz
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Denise L. Doolan
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - David W. Reid
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Matthew Field
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| | - Scott C. Bell
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rachel M. Thomson
- Immunology Department, Gallipoli Medical Research Institute, Brisbane, QLD, Australia
| | - John J. Miles
- The Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
- John J. Miles
| |
Collapse
|
20
|
Nontuberculous Mycobacterium. Respir Med 2020. [DOI: 10.1007/978-3-030-42382-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
21
|
Li X, Yuan X, Wang C. The clinical value of IL-3, IL-4, IL-12p70, IL17A, IFN-γ, MIP-1β, NLR, P-selectin, and TNF-α in differentiating bloodstream infections caused by gram-negative, gram-positive bacteria and fungi in hospitalized patients: An Observational Study. Medicine (Baltimore) 2019; 98:e17315. [PMID: 31568018 PMCID: PMC6756613 DOI: 10.1097/md.0000000000017315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Early differential diagnosis of bloodstream infections (BSIs) caused by different sources and species of bacteria in hospitalized patients is crucial for the timely targeted interventions including appropriate use of antibiotics. The aim of this study was to identify 9 biomarkers for the early differentiation of gram-negative-bloodstream infection (GN-BSI), gram-positive (GP)-BSI, and fungal-BSI.A prospective study was conducted for a total of 390 inpatients who underwent blood culture in the Chinese PLA General Hospital from September 2015 to March 2018. Patients with positive culture of a single pathogen were divided into GN-BSI, GP-BSI, and Fungal-BSI groups, and a culture-negative disease control group was also established. The serum levels of macrophage inflammatory protein 1β (MIP-1β), tumor necrosis factor α (TNF-α), interleukin (IL)-3, interferon (IFN)-γ, IL-17A, IL-4, IL-12p70, and P-selectin were detected and the NLR was calculated from routine blood test. Receiver-operating characteristic analysis was used to determine the efficacy of various indicators in the differential diagnosis of BSIs. Prediction and validation experiments on clinical patient samples (263 cases) were also performed.The level of IL-3 in the GP-BSI group was significantly higher than those in the other 3 groups. The level of IFN-γ in the fungal-BSI group was significantly higher than those in the other 3 groups. NLR, MIP-1β, TNF-α, IL-17A, and IL3 exhibited some efficacy when distinguishing between GN-BSI and GP-BSI and NLR had the largest area under curve (AUC) (0.728), followed by MIP-1β with an AUC of 0.679. IFN-γ and IL-3 exhibited some value in differential diagnosis between GN-BSI and Fungal-BSI. IL-3, MIP-1β, TNF-α, IFN-γ, NLR, IL-17A, and IL-4 exhibited some value in distinguishing fungal-BSI and GP-BSI, with IL-3 had the largest AUC (0.722), followed by MIP-1β with an AUC of 0.703.NLR and MIP-1β may be valuable in differentiating GN-BSI from GP-BSI in hospitalized patients. IFN-γ and IL-3 may be helpful in differential diagnosis GN-BSI and fungal-BSI. IL-3 and MIP-1β exhibited some diagnostic efficacy in distinguishing fungal-BSI and GP-BSI. Additionally, IL-3 with high serum level may be a marker for GP-BSI and IFN-γ with high serum level may be a valuable marker for the prediction of Fungal-BSI. The utility of these biomarkers to predict BSIs owing to different pathogens in hospitalized patients needs to be assessed in further studies.
Collapse
|
22
|
Cowman S, van Ingen J, Griffith DE, Loebinger MR. Non-tuberculous mycobacterial pulmonary disease. Eur Respir J 2019; 54:13993003.00250-2019. [PMID: 31221809 DOI: 10.1183/13993003.00250-2019] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/31/2019] [Indexed: 02/03/2023]
Abstract
Nontuberculous mycobacterial pulmonary disease (NTM-PD) is a challenging infection which is becoming increasingly prevalent, particularly in the elderly, for reasons which are unknown. While underlying lung disease is a well-established risk factor for NTM-PD, it may also occur in apparently healthy individuals. No single common genetic or immunological defect has been identified in this group, and it is likely that multiple pathways contribute towards host susceptibility to NTM-PD which further interact with environmental and microbiological factors leading to the development of disease.The diagnosis of NTM-PD relies on the integration of clinical, radiological and microbiological results. The clinical course of NTM-PD is heterogeneous, with some patients remaining stable without the need for treatment and others developing refractory disease associated with considerable mortality and morbidity. Treatment regimens are based on the identity of the isolated species, drug sensitivity testing (for some agents) and the severity of disease. Multiple antibiotics are typically required for prolonged periods of time and treatment is frequently poorly tolerated. Surgery may be beneficial in selected cases. In some circumstances cure may not be attainable and there is a pressing need for better regimens to treat refractory and drug-resistant NTM-PD.This review summarises current knowledge on the epidemiology, aetiology and diagnosis of NTM-PD and discusses the treatment of two of the most clinically significant species, the M. avium and M. abscessus complexes, with a focus on refractory disease and novel therapies.
Collapse
Affiliation(s)
- Steven Cowman
- Host Defence Unit, Royal Brompton Hospital, London, UK.,Imperial College, London, UK
| | - Jakko van Ingen
- Dept of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David E Griffith
- Dept of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Michael R Loebinger
- Host Defence Unit, Royal Brompton Hospital, London, UK .,Imperial College, London, UK
| |
Collapse
|
23
|
Cowman SA, Jacob J, Hansell DM, Kelleher P, Wilson R, Cookson WOC, Moffatt MF, Loebinger MR. Whole-Blood Gene Expression in Pulmonary Nontuberculous Mycobacterial Infection. Am J Respir Cell Mol Biol 2019; 58:510-518. [PMID: 29206475 DOI: 10.1165/rcmb.2017-0230oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The factors predisposing toward the development of pulmonary nontuberculous mycobacterial (pNTM) disease and influencing disease progression remain unclear. Impaired immune responses have been reported in individuals with pNTM disease, but data are limited and inconsistent. In this study, we sought to use gene expression profiling to examine the host response to pNTM disease. Microarray analysis of whole-blood gene expression was performed on 25 subjects with pNTM disease and 27 uninfected control subjects with respiratory disease. Gene expression results were compared with phenotypic variables and survival data. Compared with uninfected control subjects, pNTM disease was associated with downregulation of 213 transcripts enriched for terms related to T cell signaling, including IFNG. Reduced IFNG expression was associated with more severe computed tomography changes and impaired lung function. Mortality was associated with the expression of transcripts related to the innate immune response and inflammation, whereas transcripts related to T and B cell function were associated with improved survival. These findings suggest that pNTM disease is associated with an aberrant immune response, which may reflect an underlying propensity to infection or result from NTM infection itself. There were important differences in the immune response associated with survival and mortality in pNTM disease.
Collapse
Affiliation(s)
- Steven A Cowman
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - Joseph Jacob
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,3 Department of Radiology, Royal Brompton Hospital, London, United Kingdom
| | - David M Hansell
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,3 Department of Radiology, Royal Brompton Hospital, London, United Kingdom
| | - Peter Kelleher
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - Robert Wilson
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - William O C Cookson
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Miriam F Moffatt
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Michael R Loebinger
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| |
Collapse
|
24
|
Longworth SA, Daly JS. Management of infections due to nontuberculous mycobacteria in solid organ transplant recipients-Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019; 33:e13588. [PMID: 31077618 DOI: 10.1111/ctr.13588] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/06/2019] [Indexed: 01/06/2023]
Abstract
These updated guidelines from the American Society of Transplantation Infectious Diseases Community of Practice review the epidemiology, diagnosis, prevention, and management of nontuberculous mycobacterial infections in the pre- and post-transplant period. NTM commonly cause one of five different clinical syndromes: pleuropulmonary disease, skin and soft tissue infection, osteoarticular infection, disseminated disease, including that caused by catheter-associated infection, and lymphadenitis. Diagnosis of these infections can be challenging, particularly when they are isolated from nonsterile spaces, owing to their ubiquity in nature. Consequently, diagnosis of pulmonary infections with these pathogens requires fulfillment of microbiologic, radiographic, and clinical criteria to address this concern. A combination of culture and molecular diagnostic techniques is often required to make a species-level identification. Treatment varies depending on the species isolated and is complex, owing to drug toxicities, need for long-term multidrug regimens, and consideration of complex drug-drug interactions between antimicrobials and immunosuppressive agents. Given these treatment challenges, efforts should be made in both the hospital and community settings to limit exposure to these pathogens to the extent feasible.
Collapse
Affiliation(s)
- Sarah A Longworth
- Division of Infectious Disease, Hospital of University of Pennsylvania, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer S Daly
- UMass Memorial Health Care, University of Massachusetts Medical School, Worcester, Massachusetts
| | | |
Collapse
|
25
|
Asakura T, Suzuki S, Fukano H, Okamori S, Kusumoto T, Uwamino Y, Ogawa T, So M, Uno S, Namkoong H, Yoshida M, Kamata H, Ishii M, Nishimura T, Hoshino Y, Hasegawa N. Sitafloxacin-Containing Regimen for the Treatment of Refractory Mycobacterium avium Complex Lung Disease. Open Forum Infect Dis 2019; 6:ofz108. [PMID: 31111076 PMCID: PMC6519390 DOI: 10.1093/ofid/ofz108] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/01/2019] [Indexed: 02/05/2023] Open
Abstract
Background Sitafloxacin (STFX) exhibits potent activity against Mycobacterium avium complex (MAC) in both in vitro and in vivo experiments. However, limited data are available for the clinical efficacy and adverse effects of STFX and the susceptibility of refractory MAC lung disease (MAC-LD) to the drug. Therefore, this study was aimed at evaluating the clinical efficacy and safety of an STFX-containing regimen for the treatment of refractory MAC-LD. Methods We retrospectively evaluated treatment outcomes of 31 patients with refractory MAC-LD, who received an STFX-containing regimen for ≥4 weeks between January 2010 and July 2017. Refractory MAC-LD was defined as persistent positive sputum cultures for >6 months of macrolide-based standard therapy. Results Clarithromycin resistance (minimum inhibitory concentration [MIC] ≥32 μg/mL) was identified in 15 patients (48%). Twelve months after receiving the STFX-containing regimen, 26% and 19% of patients showed symptomatic and radiological responses, respectively. Although STFX-associated adverse effects were noted in 9 patients, their severity was grade 1 (National Cancer Institute Common Terminology Criteria); only 1 patient discontinued STFX because of suspected gastrointestinal disturbance. Negative sputum culture conversion was achieved in 7 patients (23%). Both univariate and multivariate logistic regression analyses revealed that surgery, low STFX MIC (≤1 μg/mL), and macrolide resistance were significant predictors of negative sputum culture conversion. Conclusions Our results demonstrate that STFX may be effective in one-fourth of patients with refractory MAC-LD. Prospective larger studies that include the analyses of MAC are needed to determine the clinical efficacy of STFX against refractory MAC-LD.
Collapse
Affiliation(s)
- Takanori Asakura
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases.,Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine.,Japan Society for the Promotion of Science, Tokyo
| | - Shoji Suzuki
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine.,Japan Society for the Promotion of Science, Tokyo
| | - Hanako Fukano
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases
| | - Satoshi Okamori
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine.,Japan Society for the Promotion of Science, Tokyo
| | - Tatsuya Kusumoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine
| | - Yoshifumi Uwamino
- Department of Laboratory Medicine, Keio University School of Medicine
| | - Takunori Ogawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine
| | - Matsuo So
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine
| | - Shunsuke Uno
- Center for Infectious Diseases and Infection Control, Keio University School of Medicine
| | - Ho Namkoong
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine
| | - Mitsunori Yoshida
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine
| | | | - Yoshihiko Hoshino
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases
| | - Naoki Hasegawa
- Center for Infectious Diseases and Infection Control, Keio University School of Medicine
| |
Collapse
|
26
|
Addy C, Doran G, Jones AL, Wright G, Caskey S, Downey DG. Microscopic polyangiitis secondary to Mycobacterium abscessus in a patient with bronchiectasis: a case report. BMC Pulm Med 2018; 18:170. [PMID: 30453935 PMCID: PMC6245610 DOI: 10.1186/s12890-018-0732-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/29/2018] [Indexed: 11/30/2022] Open
Abstract
Background Non-Tuberculous Mycobacterial–pulmonary disease (NTM-PD) is increasing in incidence and prevalence. Mycobacterium abscessus (M.abscessus) is a rapid growing multi-resistant NTM associated with severe NTM-PD requiring prolonged antibiotic therapy. Complications of therapy are common but reports on direct complications of active NTM-PD are rare. Vasculitis has been described as a rare complication of NTM-PD, most often in individuals with inherited immune defects. This case is the first to describe an ANCA positive vasculitide (Microscopic Polyangiitis) secondary to M.abscessus pulmonary disease. Case presentation A 70 year old female with bronchiectasis underwent a clinical decline associated with the growth of M.abscessus and was diagnosed with NTM-PD. Before treatment could be initiated she developed small joint arthralgia and a glove and stocking axonal loss sensorimotor neuropathy. Positive Perinuclear Anti-Neutrophil Cytoplasmic Antibodies (P-ANCA) and Myeloperoxidase-ANCA (MPO-ANCA) titres led to a diagnosis of microscopic polyangiitis. Further investigation revealed reduced interferon-gamma production but no other significant immune dysfunction. Dual treatment with immunosuppressive therapy (Corticosteroids/Cyclophosphamide) for vasculitis and antimicrobial therapy for M.abscessus NTM-PD was initiated. Clinical stability was difficult to achieve with reductions in immunosuppression triggering vasculitic flares. One flare led to retinal vein occlusion with impending visual loss requiring escalation in immunosuppression to Rituximab infusions. An increase in immunosuppression led to a deterioration in NTM-PD necessitating alterations to antibiotic regimes. Adverse effects including alopecia and Achilles tendonitis have further limited antibiotic choices resulting in a strategy of pulsed intra-venous therapy to stabilise NTM-PD. Conclusions This is the first reported case of an ANCA positive vasculitis secondary to M.abscessus pulmonary disease. This rare but important complication had a significant impact on the patient adding to the complexity of an already significant disease and treatment burden. The potential role of reduced interferon-gamma production in this case highlights the importance of investigating immune function in those with mycobacterial infection and the intricate relationship between mycobacterial infection and immune dysfunction. Immune dysfunction caused by genetic defects or immunosuppressive therapy is a known risk factor for NTM-PD. Balancing immunosuppressive therapy with prolonged antimicrobial treatment is challenging and likely to become more common as the number of individuals being treated with biologics and immunosuppressive agents increases.
Collapse
Affiliation(s)
- C Addy
- Centre for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland. .,Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland.
| | - G Doran
- Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland
| | - A L Jones
- Department of Respiratory Medicine, Royal Brompton Hospitals, Sydney Street, London, SW3 6NP, England
| | - G Wright
- Department of Rheumatology, Musgrave Park Hospital, Stockmans Ln, Belfast, BT9 7JB, Northern Ireland
| | - S Caskey
- Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland
| | - D G Downey
- Centre for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, Northern Ireland.,Regional Respiratory Centre, Belfast City Hospital, 51 Lisburn Road, Belfast, BT9 7AB, Northern Ireland
| |
Collapse
|
27
|
Kak G, Raza M, Tiwari BK. Interferon-gamma (IFN-γ): Exploring its implications in infectious diseases. Biomol Concepts 2018; 9:64-79. [PMID: 29856726 DOI: 10.1515/bmc-2018-0007] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
A key player in driving cellular immunity, IFN-γ is capable of orchestrating numerous protective functions to heighten immune responses in infections and cancers. It can exhibit its immunomodulatory effects by enhancing antigen processing and presentation, increasing leukocyte trafficking, inducing an anti-viral state, boosting the anti-microbial functions and affecting cellular proliferation and apoptosis. A complex interplay between immune cell activity and IFN-γ through coordinated integration of signals from other pathways involving cytokines and Pattern Recognition Receptors (PRRs) such as Interleukin (IL)-4, TNF-α, Lipopolysaccharide (LPS), Type-I Interferons (IFNS) etc. leads to initiation of a cascade of pro-inflammatory responses. Microarray data has unraveled numerous genes whose transcriptional regulation is influenced by IFN-γ. Consequently, IFN-γ stimulated cells display altered expression of many such target genes which mediate its downstream effector functions. The importance of IFN-γ is further reinforced by the fact that mice possessing disruptions in the IFN-γ gene or its receptor develop extreme susceptibility to infectious diseases and rapidly succumb to them. In this review, we attempt to elucidate the biological functions and physiological importance of this versatile cytokine. The functional implications of its biological activity in several infectious diseases and autoimmune pathologies are also discussed. As a counter strategy, many virulent pathogenic species have devised ways to thwart IFN-γ endowed immune-protection. Thus, IFN-γ mediated host-pathogen interactions are critical for our understanding of disease mechanisms and these aspects also manifest enormous therapeutic importance for the annulment of various infections and autoimmune conditions.
Collapse
Affiliation(s)
- Gunjan Kak
- From the Infectious Disease Immunology Lab, Dr. B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Mohsin Raza
- Department of Biochemistry, University of Delhi, South Campus, New Delhi, 110021, India
| | - Brijendra K Tiwari
- From the Infectious Disease Immunology Lab, Dr. B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| |
Collapse
|
28
|
Diel R, Nienhaus A, Ringshausen FC, Richter E, Welte T, Rabe KF, Loddenkemper R. Microbiologic Outcome of Interventions Against Mycobacterium avium Complex Pulmonary Disease: A Systematic Review. Chest 2018; 153:888-921. [PMID: 29410162 DOI: 10.1016/j.chest.2018.01.024] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/20/2017] [Accepted: 01/12/2018] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Pulmonary disease (PD) caused by Mycobacterium avium complex (MAC) is increasing worldwide. We conducted a systematic review of studies that include microbiologic outcomes to evaluate current macrolide-based treatment regimens. METHODS We searched literature published before April 2017 by using the MEDLINE, Cochrane, and Embase databases. Risk of bias in randomized trials was assessed using the Cochrane tool. RESULTS We retrieved 333 citations and evaluated 42 studies including 2,748 patients: 18 studies were retrospective chart reviews, 18 were prospective, and six were randomized. The weighted average proportion of sputum culture conversions in macrolide-containing regimens after subtracting posttreatment microbiologic recurrences was 52.3% (95% CI, 44.7%-59.9%). Using the triple-drug regimens recommended by the American Thoracic Society (ATS) achieved treatment success in 61.4% (95% CI, 49.7%-72.5%), which further increased to 65.7% (95% CI, 53.3%-77.4%) when drugs were taken for at least 1 year by patients who were macrolide susceptible and had previously untreated MAC. The overall risk of bias was low in five of the six randomized trials. However, selective outcome reporting because of a posteriori exclusion of initially included patients (14.0%), uncompleted treatment (17.6%), and inconsistent use of outcome parameters (17 definitions of treatment success) hampered the comparison of nonrandomized trials. CONCLUSIONS To date, randomized studies on treatment outcome in patients with MAC PD are scarce. Long-term treatments with ATS-recommended regimens for patients who are macrolide susceptible are superior to other macrolide-based therapies. A standardized definition of treatment success and genotypic distinction between reinfection and relapse by means of pretreatment and posttreatment identification of MAC species in cases of microbiologic recurrences may help to optimize evaluation of treatment regimens in the future.
Collapse
Affiliation(s)
- Roland Diel
- Institute for Epidemiology, University Medical Hospital Schleswig-Holstein, Kiel, Germany; Lung Clinic Grosshansdorf, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany.
| | - Albert Nienhaus
- Institute for Health Service Research in Dermatology and Nursing, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institution for Statutory Accident Insurance and Prevention in the Health and Welfare Services, Hamburg, Germany
| | - Felix C Ringshausen
- Department of Respiratory Medicine, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | - Elvira Richter
- MVZ Labor Dr. Limbach, TB Laboratory, Heidelberg, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | - Klaus F Rabe
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| | | |
Collapse
|
29
|
Trofimov V, Costa-Gouveia J, Hoffmann E, Brodin P. Host-pathogen systems for early drug discovery against tuberculosis. Curr Opin Microbiol 2017; 39:143-151. [PMID: 29179041 DOI: 10.1016/j.mib.2017.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/17/2017] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB) is a global disease causing 1.8 million deaths each year. The appearance of drug-resistant strains raised the demand for new anti-mycobacterial drugs and therapies, because previously discovered antibiotics are shown to be inefficient. Moreover, the number of newly discovered drugs is not increasing in proportion to the emergence of drug resistance, which suggests that more optimized methodology and screening procedures are required including the incorporation of in vivo properties of TB infection. A way to improve efficacy of screening approaches is by introducing the use of different host-pathogen systems into primary screenings. These include whole cell-based screenings, zebrafish larvae-based screenings and the impact of artificial granuloma research on the drug discovery process. This review highlights current screening attempts and the identified molecular targets and summarizes findings of alternative, not fully explored host-pathogen systems for the characterization of anti-mycobacterial compounds.
Collapse
Affiliation(s)
- Valentin Trofimov
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Joana Costa-Gouveia
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Eik Hoffmann
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France
| | - Priscille Brodin
- CNRS, Inserm, CHU Lille, U1019 - UMR8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Institut Pasteur de Lille, University Lille, Lille, France.
| |
Collapse
|
30
|
Haworth CS, Banks J, Capstick T, Fisher AJ, Gorsuch T, Laurenson IF, Leitch A, Loebinger MR, Milburn HJ, Nightingale M, Ormerod P, Shingadia D, Smith D, Whitehead N, Wilson R, Floto RA. British Thoracic Society guidelines for the management of non-tuberculous mycobacterial pulmonary disease (NTM-PD). Thorax 2017; 72:ii1-ii64. [DOI: 10.1136/thoraxjnl-2017-210927] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2017] [Indexed: 01/18/2023]
|
31
|
Host-directed therapies offer novel opportunities for the fight against tuberculosis. Drug Discov Today 2017; 22:1250-1257. [PMID: 28533187 DOI: 10.1016/j.drudis.2017.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/20/2017] [Accepted: 05/12/2017] [Indexed: 12/24/2022]
Abstract
Tuberculosis (TB) remains a leading global health problem that is exacerbated by the emergence of multidrug and extensively drug-resistant Mycobacterium tuberculosis strains. Control of the disease requires novel therapeutic strategies. Modulating host homeostasis appears to be a promising approach, and recent studies have identified novel potential host targets and compounds that could be investigated for host-directed therapies (HDTs). Moreover, the recent development of intracellular high-throughput phenotypic assays makes it possible to screen large libraries of compounds to identify more rapidly new effectors for mycobacterial elimination. Technological advances combined with the novel HDT concept opens an interesting and promising research area that could ultimately deliver personalized TB treatment.
Collapse
|
32
|
Stout JE, Dicks KV. A Breath of Fresh Air for Patients with Pulmonary Nontuberculous Mycobacterial Infection. Am J Respir Crit Care Med 2017; 195:716-717. [DOI: 10.1164/rccm.201610-2073ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jason E. Stout
- Department of MedicineDuke University Medical CenterDurham, North Carolina
| | - Kristen V. Dicks
- Department of MedicineDuke University Medical CenterDurham, North Carolina
| |
Collapse
|
33
|
Floto RA, Olivier KN, Saiman L, Daley CL, Herrmann JL, Nick JA, Noone PG, Bilton D, Corris P, Gibson RL, Hempstead SE, Koetz K, Sabadosa KA, Sermet-Gaudelus I, Smyth AR, van Ingen J, Wallace RJ, Winthrop KL, Marshall BC, Haworth CS. US Cystic Fibrosis Foundation and European Cystic Fibrosis Society consensus recommendations for the management of non-tuberculous mycobacteria in individuals with cystic fibrosis. Thorax 2016; 71 Suppl 1:i1-22. [PMID: 26666259 PMCID: PMC4717371 DOI: 10.1136/thoraxjnl-2015-207360] [Citation(s) in RCA: 307] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Non-tuberculous mycobacteria (NTM) are ubiquitous environmental organisms that can cause chronic pulmonary infection, particularly in individuals with pre-existing inflammatory lung disease such as cystic fibrosis (CF). Pulmonary disease caused by NTM has emerged as a major threat to the health of individuals with CF but remains difficult to diagnose and problematic to treat. In response to this challenge, the US Cystic Fibrosis Foundation (CFF) and the European Cystic Fibrosis Society (ECFS) convened an expert panel of specialists to develop consensus recommendations for the screening, investigation, diagnosis and management of NTM pulmonary disease in individuals with CF. Nineteen experts were invited to participate in the recommendation development process. Population, Intervention, Comparison, Outcome (PICO) methodology and systematic literature reviews were employed to inform draft recommendations. An anonymous voting process was used by the committee to reach consensus. All committee members were asked to rate each statement on a scale of: 0, completely disagree, to 9, completely agree; with 80% or more of scores between 7 and 9 being considered ‘good’ agreement. Additionally, the committee solicited feedback from the CF communities in the USA and Europe and considered the feedback in the development of the final recommendation statements. Three rounds of voting were conducted to achieve 80% consensus for each recommendation statement. Through this process, we have generated a series of pragmatic, evidence-based recommendations for the screening, investigation, diagnosis and treatment of NTM infection in individuals with CF as an initial step in optimising management for this challenging condition.
Collapse
Affiliation(s)
- R Andres Floto
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK Cambridge Centre for Lung Infection, Papworth Hospital, Cambridge, UK
| | - Kenneth N Olivier
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Lisa Saiman
- Department of Pediatrics, Columbia University Medical Center, Pediatric Infectious Diseases, New York, New York, USA
| | - Charles L Daley
- Division of Mycobacterial and Respiratory Infections, National Jewish Health, Denver, Colorado, USA
| | - Jean-Louis Herrmann
- INSERM U1173, UFR Simone Veil, Versailles-Saint-Quentin University, Saint-Quentin en Yvelines, France AP-HP, Service de Microbiologie, Hôpital Raymond Poincaré, Garches, France
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Peadar G Noone
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Diana Bilton
- Department of Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Paul Corris
- Department of Respiratory Medicine, Freeman Hospital, High Heaton, Newcastle, UK
| | - Ronald L Gibson
- Department of Pediatrics University of Washington School of Medicine, Seattle, Washington, USA
| | - Sarah E Hempstead
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Karsten Koetz
- Department of Pediatrics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kathryn A Sabadosa
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Isabelle Sermet-Gaudelus
- Service de Pneumo-Pédiatrie, Université René Descartes, Hôpital Necker-Enfants Malades, Paris, France
| | - Alan R Smyth
- Division of Child Health, Obstetrics & Gynaecology, University of Nottingham, Nottingham, UK
| | - Jakko van Ingen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Richard J Wallace
- Department of Microbiology, University of Texas Health Science Center, Tyler, Texas, USA
| | | | | | - Charles S Haworth
- Cambridge Centre for Lung Infection, Papworth Hospital, Cambridge, UK
| | | |
Collapse
|
34
|
Lake MA, Ambrose LR, Lipman MCI, Lowe DM. '"Why me, why now?" Using clinical immunology and epidemiology to explain who gets nontuberculous mycobacterial infection. BMC Med 2016; 14:54. [PMID: 27007918 PMCID: PMC4806462 DOI: 10.1186/s12916-016-0606-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 03/18/2016] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The prevalence of nontuberculous mycobacterial (NTM) disease is rising. An understanding of known risk factors for disease sheds light on the immunological and physical barriers to infection, and how and why they may be overcome. This review focuses on human NTM infection, supported by experimental and in vitro data of relevance to the practising clinician who seeks to understand why their patient has NTM infection and how to further investigate. DISCUSSION First, the underlying immune response to NTM disease is examined. Important insights regarding NTM disease susceptibility come from nature's own knockouts, the primary immune deficiency disorders. We summarise the current knowledge surrounding interferon-gamma (IFNγ)-interleukin-12 (IL-12) axis abnormalities, followed by a review of phagocytic defects, T cell lymphopenia and rarer genetic conditions known to predispose to NTM disease. We discuss how these define key immune pathways involved in the host response to NTM. Iatrogenic immunosuppression is also important, and we evaluate the impact of novel biological therapies, as well as bone marrow transplant and chemotherapy for solid organ malignancy, on the epidemiology and presentation of NTM disease, and discuss the host defence dynamics thus revealed. NTM infection and disease in the context of other chronic illnesses including HIV and malnutrition is reviewed. The role of physical barriers to infection is explored. We describe how their compromise through different mechanisms including cystic fibrosis, bronchiectasis and smoking-related lung disease can result in pulmonary NTM colonisation or infection. We also summarise further associations with host factors including body habitus and age. We use the presented data to develop an over-arching model that describes human host defences against NTM infection, where they may fail, and how this framework can be applied to investigation in routine clinical practice.
Collapse
Affiliation(s)
- M Alexandra Lake
- Royal Free London NHS Foundation Trust, London, UK.,Division of Infection and Immunity, University College London, London, UK
| | - Lyn R Ambrose
- Institute of Immunity and Transplantation, University College London, Royal Free Campus, Pond Street, London, NW3 2QG, UK
| | - Marc C I Lipman
- Royal Free London NHS Foundation Trust, London, UK.,UCL Respiratory, Division of Medicine, Faculty of Medical Sciences, University College London, Royal Free Campus, London, UK
| | - David M Lowe
- Royal Free London NHS Foundation Trust, London, UK. .,Institute of Immunity and Transplantation, University College London, Royal Free Campus, Pond Street, London, NW3 2QG, UK.
| |
Collapse
|
35
|
Zhan X, Fang Y, Hu S, Wu Y, Yang K, Liao C, Zhang Y, Huang X, Wu M. IFN-γ differentially regulates subsets of Gr-1+CD11b+ myeloid cells in chronic inflammation. Mol Immunol 2015; 66:451-62. [DOI: 10.1016/j.molimm.2015.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/06/2015] [Accepted: 05/12/2015] [Indexed: 01/02/2023]
|
36
|
van Lieshout MHP, Florquin S, Vanʼt Veer C, de Vos AF, van der Poll T. TIR-Domain-Containing Adaptor-Inducing Interferon-β (TRIF) Mediates Antibacterial Defense during Gram-Negative Pneumonia by Inducing Interferon-x03B3. J Innate Immun 2015; 7:637-46. [PMID: 26065469 DOI: 10.1159/000430913] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/25/2015] [Indexed: 12/13/2022] Open
Abstract
Klebsiella pneumoniae is an important cause of Gram-negative pneumonia and sepsis. Mice deficient for TIR-domain-containing adaptor-inducing interferon-β (TRIF) demonstrate enhanced bacterial growth and dissemination during Klebsiella pneumonia. We show here that the impaired antibacterial defense of TRIF mutant mice is associated with absent interferon (IFN)-x03B3; production in the lungs. IFN-x03B3; production by splenocytes in response to K. pneumoniae in vitro was critically dependent on Toll-like receptor 4 (TLR4), the common TLR adaptor myeloid differentiation primary response gene (MyD88) and TRIF. Reconstitution of TRIF mutant mice with recombinant IFN-x03B3; via the airways reduced bacterial loads in lungs and distant body sites to levels measured in wild-type mice, and partially restored pulmonary cytokine levels. The IFN-x03B3;-induced, improved, enhanced antibacterial response in TRIF mutant mice occurred at the expense of increased hepatocellular injury. These data indicate that TRIF mediates antibacterial defense during Gram-negative pneumonia, at least in part, by inducing IFN-x03B3; at the primary site of infection.
Collapse
Affiliation(s)
- Miriam H P van Lieshout
- Center of Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
37
|
Boe DM, Curtis BJ, Chen MM, Ippolito JA, Kovacs EJ. Extracellular traps and macrophages: new roles for the versatile phagocyte. J Leukoc Biol 2015; 97:1023-35. [PMID: 25877927 DOI: 10.1189/jlb.4ri1014-521r] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/20/2015] [Indexed: 12/13/2022] Open
Abstract
MΦ are multipurpose phagocytes with a large repertoire of well-characterized abilities and functions, including regulation of inflammation, wound healing, maintenance of tissue homeostasis, as well as serving as an integral component of the innate-immune defense against microbial pathogens. Working along with neutrophils and dendritic cells, the other myeloid-derived professional phagocytes, MΦ are one of the key effector cells initiating and directing the host reaction to pathogenic organisms and resolving subsequent responses once the threat has been cleared. ETs are a relatively novel strategy of host defense involving expulsion of nuclear material and embedded proteins from immune cells to immobilize and kill bacteria, fungi, and viruses. As research on ETs expands, it has begun to encompass many immune cell types in unexpected ways, including various types of MΦ, which are not only capable of generating METs in response to various stimuli, but recent preclinical data suggest that they are an important agent in clearing ETs and limiting ET-mediated inflammation and tissue damage. This review aims to summarize historical and recent findings of biologic research regarding ET formation and function and discuss the role of MΦ in ET physiology and associated pathologies.
Collapse
Affiliation(s)
- Devin M Boe
- *Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, Illinois, USA
| | - Brenda J Curtis
- *Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, Illinois, USA
| | - Michael M Chen
- *Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, Illinois, USA
| | - Jill A Ippolito
- *Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, Illinois, USA
| | - Elizabeth J Kovacs
- *Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, Illinois, USA
| |
Collapse
|
38
|
Abstract
A major approach for immunologic intervention in tuberculosis involves redirecting the outcome of the host immune response from the induction of disease to pathogen control. Cytokines and lipid mediators known as eicosanoids play key roles in regulating this balance and as such represent important targets for immunologic intervention. While the evidence for cytokine/eicosanoid function derives largely from the investigation of murine and zebrafish experimental infection models, clinical studies have confirmed the existence of many of the same pathways in tuberculosis patients. Here, we summarize new data that reveal important intersections between the cytokine and eicosanoid networks in the host response to mycobacteria and discuss how targeting this crosstalk can promote resistance to lethal Mycobacterium tuberculosis infection. This approach could lead to new host-directed therapies to be used either as an adjunct for improving the efficacy of standard antibiotic treatment or for the management of drug-resistant infections.
Collapse
Affiliation(s)
- Katrin D Mayer-Barber
- Immunobiology Section, Laboratory of Parasitic Disease, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
39
|
Bag R, Griffith DE. Therapy forMycobacterium aviumComplex Lung Disease. It Ain’t Perfect, but It’s Progress. Am J Respir Crit Care Med 2015; 191:14-6. [DOI: 10.1164/rccm.201411-2078ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
40
|
Abstract
Treatment of tuberculosis (TB) remains challenging, with lengthy treatment durations and complex drug regimens that are toxic and difficult to administer. Similar to the vast majority of antibiotics, drugs for Mycobacterium tuberculosis are directed against microbial targets. Although more effective drugs that target the bacterium may lead to faster cure of patients, it is possible that a biological limit will be reached that can be overcome only by adopting a fundamentally new treatment approach. TB regimens might be improved by including agents that target host pathways. Recent work on host-pathogen interactions, host immunity, and host-directed interventions suggests that supplementing anti-TB therapy with host modulators may lead to shorter treatment times, a reduction in lung damage caused by the disease, and a lower risk of relapse or reinfection. We undertook this review to identify molecular pathways of the host that may be amenable to modulation by small molecules for the treatment of TB. Although several approaches to augmenting standard TB treatment have been proposed, only a few have been explored in detail or advanced to preclinical and clinical studies. Our review focuses on molecular targets and inhibitory small molecules that function within the macrophage or other myeloid cells, on host inflammatory pathways, or at the level of TB-induced lung pathology.
Collapse
|
41
|
Abstract
Evidence has increasingly shown that the lungs are a major site of immune regulation. A robust and highly regulated immune response in the lung protects the host from pathogen infection, whereas an inefficient or deleterious response can lead to various pulmonary diseases. Many cell types, such as epithelial cells, dendritic cells, macrophages, neutrophils, eosinophils, and B and T lymphocytes, contribute to lung immunity. This review focuses on the recent advances in understanding how T lymphocytes mediate pulmonary host defenses against bacterial, viral, and fungal pathogens.
Collapse
Affiliation(s)
- Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15201, USA
| | | |
Collapse
|
42
|
Reljic R, Paul MJ, Arias MA. Cytokine therapy of tuberculosis at the crossroads. Expert Rev Respir Med 2012; 3:53-66. [PMID: 20477282 DOI: 10.1586/17476348.3.1.53] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Drug treatment is the key strategy in TB control. However, the treatment course lasts 6-9 months because the current anti-TB drugs are poorly effective against nondividing (i.e., persistent) bacilli. As a result, completion rates are unsatisfactory, leading to emergence and spread of multidrug-resistant infection. It would, therefore, be very desirable to design a form of complementary treatment that could speed up the recovery process for people afflicted with TB and reduce the relapse rates. With the advancement of our understanding of the immunopathogenesis of TB, it has become increasingly possible to develop novel adjunctive immunotherapies for both drug-susceptible and drug-resistant TB. Notably, cytokines probably offer the most promising prospect of such a therapy being introduced in routine clinical practice. However, in many ways, the cytokine therapy of TB has reached a crossroad, since, although the initial promise failed to live up to expectations, sufficient encouraging evidence exists to warrant further exploration. There are clear arguments in favor as well as against such treatments. This review aims to provide a rationale for cytokine treatment of TB, to describe the current status of several cytokines that have been considered for that purpose and, ultimately, to make a case for the need for further clinical trials.
Collapse
Affiliation(s)
- Rajko Reljic
- Department of Cellular and Molecular Medicine, St George's, University of London, 43 Cranmer Terrace, London SW17 0RE, UK.
| | | | | |
Collapse
|
43
|
McGrath EE, Anderson PB. The therapeutic approach to non-tuberculous mycobacterial infection of the lung. Pulm Pharmacol Ther 2010; 23:389-96. [DOI: 10.1016/j.pupt.2010.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 05/28/2010] [Accepted: 06/03/2010] [Indexed: 11/17/2022]
|
44
|
Abstract
Interferon gamma (IFN-gamma), a pleotropic cytokine, has been shown to be important to the function of virtually all immune cells and both innate and adaptive immune responses. In 1986, early clinical trials of this cytokine began to evaluate its therapeutic potential. The initial studies focused on the tolerability and pharmacology of IFN-gamma and systematically determined its antitumor and anti-infection activities. In the 20-plus years since those first trials, IFN-gamma has been used in a wide variety of clinical indications, which are reviewed in this article.
Collapse
Affiliation(s)
- Catriona H.T. Miller
- Center for Cancer Research, Cancer and Inflammation Program, Laboratory of Experimental Immunology, National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Stephen G. Maher
- Department of Surgery, Trinity Centre for Health Sciences, Trinity College Dublin, St. James’s Hospital, Dublin 8, Ireland
| | - Howard A. Young
- Center for Cancer Research, Cancer and Inflammation Program, Laboratory of Experimental Immunology, National Cancer Institute-Frederick, Frederick, Maryland, USA
| |
Collapse
|
45
|
Zaitzeva S, Matveeva S, Gerasimova T, Pashkov Y, Butov D, Pylypchuk V, Frolov V, Kutsyna G. Treatment of cavitary and infiltrating pulmonary tuberculosis with and without the immunomodulator Dzherelo. Clin Microbiol Infect 2009; 15:1154-62. [DOI: 10.1111/j.1469-0691.2009.02760.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Gupta A, Clauss H. Prosthetic joint infection withMycobacterium aviumcomplex in a solid organ transplant recipient. Transpl Infect Dis 2009; 11:537-40. [DOI: 10.1111/j.1399-3062.2009.00433.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
47
|
Gamma interferon signaling in macrophage lineage cells regulates central nervous system inflammation and chemokine production. J Virol 2009; 83:8604-15. [PMID: 19515766 DOI: 10.1128/jvi.02477-08] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Intracranial (i.c.) infection of mice with lymphocytic choriomeningitis virus (LCMV) results in anorexic weight loss, mediated by T cells and gamma interferon (IFN-gamma). Here, we assessed the role of CD4(+) T cells and IFN-gamma on immune cell recruitment and proinflammatory cytokine/chemokine production in the central nervous system (CNS) after i.c. LCMV infection. We found that T-cell-depleted mice had decreased recruitment of hematopoietic cells to the CNS and diminished levels of IFN-gamma, CCL2 (MCP-1), CCL3 (MIP-1alpha), and CCL5 (RANTES) in the cerebrospinal fluid (CSF). Mice deficient in IFN-gamma had decreased CSF levels of CCL3, CCL5, and CXCL10 (IP-10), and decreased activation of both resident CNS and infiltrating antigen-presenting cells (APCs). The effects of IFN-gamma signaling on macrophage lineage cells was assessed using transgenic mice, called "macrophages insensitive to interferon gamma" (MIIG) mice, that express a dominant-negative IFN-gamma receptor under the control of the CD68 promoter. MIIG mice had decreased levels of CCL2, CCL3, CCL5, and CXCL10 compared to controls despite having normal numbers of LCMV-specific CD4(+) T cells in the CNS. MIIG mice also had decreased recruitment of infiltrating macrophages and decreased activation of both resident CNS and infiltrating APCs. Finally, MIIG mice were significantly protected from LCMV-induced anorexia and weight loss. Thus, these data suggest that CD4(+) T-cell production of IFN-gamma promotes signaling in macrophage lineage cells, which control (i) the production of proinflammatory cytokines and chemokines, (ii) the recruitment of macrophages to the CNS, (iii) the activation of resident CNS and infiltrating APC populations, and (iv) anorexic weight loss.
Collapse
|
48
|
Cello JP, Day LW. Idiopathic AIDS enteropathy and treatment of gastrointestinal opportunistic pathogens. Gastroenterology 2009; 136:1952-65. [PMID: 19457421 PMCID: PMC7094677 DOI: 10.1053/j.gastro.2008.12.073] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 12/17/2008] [Accepted: 12/22/2008] [Indexed: 12/17/2022]
Abstract
Diarrhea in patients with acquired immune deficiency syndrome (AIDS) has proven to be both a diagnostic and treatment challenge since the discovery of the human immunodeficiency virus (HIV) virus more than 30 years ago. Among the main etiologies of diarrhea in this group of patients are infectious agents that span the array of viruses, bacteria, protozoa, parasites, and fungal organisms. In many instances, highly active antiretroviral therapy remains the cornerstone of therapy for both AIDS and AIDS-related diarrhea, but other targeted therapies have been developed as new pathogens are identified; however, some infections remain treatment challenges. Once identifiable infections as well as other causes of diarrhea are investigated and excluded, a unique entity known as AIDS enteropathy can be diagnosed. Known as an idiopathic, pathogen-negative diarrhea, this disease has been investigated extensively. Atypical viral pathogens, including HIV itself, as well as inflammatory and immunologic responses are potential leading causes of it. Although AIDS enteropathy can pose a diagnostic challenge so too does the treatment of it. Highly active antiretroviral therapy, nutritional supplementation, electrolyte replacements, targeted therapy for infection if indicated, and medications for symptom control all are key elements in the treatment regimen. Importantly, a multidisciplinary approach among the gastroenterologist, infectious disease physician, HIV specialists, oncology, and surgery is necessary for many patients.
Collapse
Affiliation(s)
- John P Cello
- Department of Medicine, Gastroenterology Division, University of California, San Francisco, San Francisco General Hospital, San Francisco, California 94110, USA.
| | | |
Collapse
|
49
|
Parrish SC, Myers J, Lazarus A. Nontuberculous mycobacterial pulmonary infections in Non-HIV patients. Postgrad Med 2008; 120:78-86. [PMID: 19020369 DOI: 10.3810/pgm.2008.11.1942] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nontuberculous mycobacteria (NTM) are ubiquitous organisms with nearly 100 different species found in soil and water. The fatty-acid and wax-rich impermeable cell wall of the mycobacteria allow for adherence to solid substrates such as pipes and leaves, allowing the organism to persist despite treatment with common disinfectants. Mycobacteria can cause infection in both humans and animals. It is difficult to assess the incidence or prevalence of NTM disease due to multiple factors. Nontuberculous mycobacteria infection may be difficult to differentiate from colonization, and when NTM infection is diagnosed, it is not a reportable disease. Furthermore, some species such as Mycobacterium gordonae may be a contaminant. Nontuberculous mycobacteria infection is not a communicable disease, although health care-associated outbreaks have been reported, associated with a single facility or procedure. While the nontuberculous infection may affect other organs, the most common site is the lung, and the most common species is Mycobacterium avium complex, commonly referred to as MAC infection. An increasing occurrence of MAC has been reported, especially in certain populations such as middle-aged or elderly thin women, patients with chronic lung disease, human immunodeficiency virus infection, and cystic fibrosis. An association of NTM infection with gastroesophageal reflux disease has also been noted. The clinical presentation often includes chronic productive cough. Other less common symptoms include dyspnea and hemoptysis. With increased use of computed tomography and high-resolution computed tomography, patterns of MAC pulmonary infection have been described. Recently, the American Thoracic Society has outlined guidelines for the diagnosis and management of NTM infection. Treatment of NTM infection requires at least 3 effective drugs for a minimum of 12 months after sputum conversion to negative cultures. Surgical therapy may be considered for localized disease which has failed medical management. In this article, the clinical presentation, radiographic features, diagnostic evaluation, and management are discussed.
Collapse
Affiliation(s)
- Scott C Parrish
- Pulmonary and Critical Care Medicine, National Naval Medical Center, Bethesda, MD 20889, USA
| | | | | |
Collapse
|
50
|
Cytokine profiles of HIV patients with pulmonary tuberculosis resulting from adjunct immunotherapy with herbal phytoconcentrates Dzherelo and Anemin. Cytokine 2008; 44:392-6. [DOI: 10.1016/j.cyto.2008.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2008] [Revised: 10/04/2008] [Accepted: 10/10/2008] [Indexed: 11/22/2022]
|