1
|
Amanati A, Jahromi MG, Jafarian H, Abdipour Mehrian SR, Sajedianfard S, Farokhmanesh S, Maddahi A, Safari F, Nabavizadeh SA, Alinazari MMK. Aspergillus-related immune reconstitution inflammatory syndrome in pediatric cancer patients, clinical characteristics, imaging findings, and survival. BMC Infect Dis 2024; 24:1423. [PMID: 39696082 DOI: 10.1186/s12879-024-10298-z] [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: 07/12/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Aspergillus-related immune reconstitution inflammatory syndrome (IRIS) is a challenge to diagnose in immunocompromised pediatric cancer patients with Invasive Pulmonary Aspergillosis (IPA). If not recognized, it can mimic disease progression and lead to overtreatment. Studies on Aspergillus-related IRIS in the pediatric population are scarce. We prospectively identified four pediatric cancer patients diagnosed with IPA who developed paradoxical worsening of pulmonary symptoms following neutrophil recovery. The clinical course, imaging findings, and response to corticosteroids were also reviewed. All patients had refractory respiratory symptoms, including cough, breathing difficulties (dyspnea), and chest pain, despite antifungal therapy. Serial imaging revealed new or enlarging pulmonary infiltrates. Symptoms improved dramatically with corticosteroids without antifungal escalation, confirming Aspergillus-related IRIS diagnosis. Aspergillus-related IRIS can occur in immunocompromised children with cancer and IPA, mimicking disease progression. Recognition is important for avoiding overtreatment. This is the first report highlighting the features of Aspergillus-related IRIS in pediatric cancer patients.
Collapse
Affiliation(s)
- Ali Amanati
- Shiraz University of Medical Sciences, Shiraz, Iran
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Ghaderian Jahromi
- Department of Radiology, Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadis Jafarian
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Shahdad Farokhmanesh
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arman Maddahi
- Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farima Safari
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, 71-348-45794, Iran.
| | - Seyed Ali Nabavizadeh
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Otolaryngology Research Center, Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran.
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, 71-348-45794, Iran.
| | | |
Collapse
|
2
|
Casutt A, Lamoth F, Lortholary O, Prior JO, Tonglet A, Manuel O, Bergeron A, Beigelman-Aubry C. Atypical imaging patterns during lung invasive mould diseases: lessons for clinicians. Eur Respir Rev 2023; 32:230086. [PMID: 37758271 PMCID: PMC10523149 DOI: 10.1183/16000617.0086-2023] [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: 04/29/2023] [Accepted: 07/13/2023] [Indexed: 09/30/2023] Open
Abstract
Imaging of pulmonary invasive mould diseases (IMDs), which represents a cornerstone in their work-up, is mainly based on computed tomography (CT). The purpose of this review is to discuss their CT features, mainly those related to aspergillosis and mucormycosis. We will especially focus on atypical radiological presentations that are increasingly observed among non-neutropenic emerging populations of patients at risk, such as those receiving novel anticancer therapies or those in the intensive care unit. We will also discuss the interest of other available imaging techniques, mainly positron emission tomography/CT, that may play a role in the diagnosis as well as evaluation of disease extent and follow-up. We will show that any new airway-centred abnormality or caveated lesion should evoke IMDs in mildly immunocompromised hosts. Limitations in their recognition may be due to potential underlying abnormalities that increase the complexity of interpretation of lung imaging, as well as the non-specificity of imaging features. In this way, the differentials of all morphological/metabolic aspects must be kept in mind for the optimal management of patients, as well as the benefit of evaluation of the vascular status.
Collapse
Affiliation(s)
- Alessio Casutt
- Division of Pulmonology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Division of Pulmonology, Ospedale Regionale di Lugano, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | - Frédéric Lamoth
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Institute of Microbiology, Department of Laboratories, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Olivier Lortholary
- University Paris Cité, Necker Enfants Malades University Hospital, AP-HP, IHU Imagine, Paris, France
- Institut Pasteur, National Reference Center for Invasive Mycoses and Antifungals, Paris, France
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Andrea Tonglet
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Oriol Manuel
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Transplantation Center, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Anne Bergeron
- Department of Pulmonology, Geneva University Hospital, University of Geneva, Geneva, Switzerland
- A. Bergeron and C. Beigelman-Aubry contributed equally to this work
| | - Catherine Beigelman-Aubry
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- A. Bergeron and C. Beigelman-Aubry contributed equally to this work
| |
Collapse
|
3
|
Sprute R, Nacov JA, Neofytos D, Oliverio M, Prattes J, Reinhold I, Cornely OA, Stemler J. Antifungal prophylaxis and pre-emptive therapy: When and how? Mol Aspects Med 2023; 92:101190. [PMID: 37207579 DOI: 10.1016/j.mam.2023.101190] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/22/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
The growing pool of critically ill or immunocompromised patients leads to a constant increase of life-threatening invasive infections by fungi such as Aspergillus spp., Candida spp. and Pneumocystis jirovecii. In response to this, prophylactic and pre-emptive antifungal treatment strategies have been developed and implemented for high-risk patient populations. The benefit by risk reduction needs to be carefully weighed against potential harm caused by prolonged exposure against antifungal agents. This includes adverse effects and development of resistance as well as costs for the healthcare system. In this review, we summarise evidence and discuss advantages and downsides of antifungal prophylaxis and pre-emptive treatment in the setting of malignancies such as acute leukaemia, haematopoietic stem cell transplantation, CAR-T cell therapy, and solid organ transplant. We also address preventive strategies in patients after abdominal surgery and with viral pneumonia as well as individuals with inherited immunodeficiencies. Notable progress has been made in haematology research, where strong recommendations regarding antifungal prophylaxis and pre-emptive treatment are backed by data from randomized controlled trials, whereas other critical areas still lack high-quality evidence. In these areas, paucity of definitive data translates into centre-specific strategies that are based on interpretation of available data, local expertise, and epidemiology. The development of novel immunomodulating anticancer drugs, high-end intensive care treatment and the development of new antifungals with new modes of action, adverse effects and routes of administration will have implications on future prophylactic and pre-emptive approaches.
Collapse
Affiliation(s)
- Rosanne Sprute
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Julia A Nacov
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Dionysios Neofytos
- Division of Infectious Diseases, Transplant Infectious Disease Service, University Hospital of Geneva, Geneva, Switzerland
| | - Matteo Oliverio
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Juergen Prattes
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; Medical University of Graz, Department of Internal Medicine, Division of Infectious Disease, Excellence Center for Medical Mycology (ECMM), Graz, Austria
| | - Ilana Reinhold
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, Zurich, Switzerland
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), Cologne, Germany
| | - Jannik Stemler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), Cologne, Germany; German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
| |
Collapse
|
4
|
Justiz-Vaillant AA, Gopaul D, Akpaka PE, Soodeen S, Arozarena Fundora R. Severe Combined Immunodeficiency-Classification, Microbiology Association and Treatment. Microorganisms 2023; 11:1589. [PMID: 37375091 DOI: 10.3390/microorganisms11061589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Severe combined immunodeficiency (SCID) is a primary inherited immunodeficiency disease that presents before the age of three months and can be fatal. It is usually due to opportunistic infections caused by bacteria, viruses, fungi, and protozoa resulting in a decrease in number and impairment in the function of T and B cells. Autosomal, X-linked, and sporadic forms exist. Evidence of recurrent opportunistic infections and lymphopenia very early in life should prompt immunological investigation and suspicion of this rare disorder. Adequate stem cell transplantation is the treatment of choice. This review aimed to provide a comprehensive approach to the microorganisms associated with severe combined immunodeficiency (SCID) and its management. We describe SCID as a syndrome and summarize the different microorganisms that affect children and how they can be investigated and treated.
Collapse
Affiliation(s)
- Angel A Justiz-Vaillant
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Darren Gopaul
- Department of Internal Medicine, Port of Spain General Hospital, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Patrick Eberechi Akpaka
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, Trinidad and Tobago
| | - Sachin Soodeen
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Rodolfo Arozarena Fundora
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, Trinidad and Tobago
- Department of Clinical and Surgical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
5
|
Approach to the diagnosis of invasive fungal infections of the respiratory tract in the immunocompromised host. Curr Opin Pulm Med 2023; 29:149-159. [PMID: 36917216 DOI: 10.1097/mcp.0000000000000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
PURPOSE OF REVIEW The burden of invasive fungal infection is increasing worldwide, largely due to a growing population at-risk. Most serious human fungal pathogens enter the host via the respiratory tract. Early identification and treatment of invasive fungal respiratory infections (IFRIs) in the immunocompromised host saves lives. However, their accurate diagnosis is a difficult challenge for clinicians and mortality remains high. RECENT FINDINGS This article reviews IFRIs, focussing on host susceptibility factors, clinical presentation, and mycological diagnosis. Several new diagnostic tools are coming of age including molecular diagnostics and point-of-care antigen tests. As diagnosis of IFRI relies heavily on invasive procedures like bronchoalveolar lavage and lung biopsy, several novel noninvasive diagnostic techniques are in development, such as metagenomics, 'volatilomics' and advanced imaging technologies. SUMMARY Where IFRI cannot be proven, clinicians must employ a 'weights-of-evidence' approach to evaluate host factors, clinical and mycological data. Implementation studies are needed to understand how new diagnostic tools can be best applied within clinical pathways. Differentiating invasive infection from colonization and identifying antifungal resistance remain key challenges. As our diagnostic arsenal expands, centralized clinical mycology laboratories and efforts to ensure access to new diagnostics in low-resource settings will become increasingly important.
Collapse
|
6
|
Balkrishna A, Sengupta S, Kumari P, Dev R, Haldar S, Varshney A. Anu Taila, an herbal nasal-drop, delays spore germination in Cunninghamella bertholletiae by reducing cAMP-PKA dependent ROS in mucorale pathogen and extrinsic ROS in human host cells. Lett Appl Microbiol 2023; 76:7008501. [PMID: 36708174 DOI: 10.1093/lambio/ovad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/13/2022] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
The rare, fastest-germinating, frequently invasive mucorale, Cunninghamella bertholletiae, is intractable due to its imprecise etiology. Cunninghamella bertholletiae spores can infect both immunocompromised and immunocompetent individuals to cause mucormycosis. Sub-optimal drug-susceptibility further limits its treatment options. The classical nasal drop, Anu Taila, is reported to be effective against the rather prevalent mucorales, Mucor spp., making its anti-mucormycotic effect against C. bertholletiae worth testing. The inhibitory effect of Anu Taila against C. bertholletiae was manifested as microstructural alterations of the spores and their delayed germination. Anu Taila reduced the germination-promoting reactive oxygen species (ROS) levels in both the pathogen, C. bertholletiae, and the human host lung epithelial A549 cells. Expressions of structural (chitin synthase, trehalose synthase) and functional (cAMP-PKA) markers of spore germination were regulated by Anu Taila. cAMP-PKA expression and ROS generation are well-correlated, implicating the role of Anu Taila in delaying C. bertholletiae spore germination by targeting cAMP-PKA-mediated ROS generation. In conclusion, this study demonstrates that Anu Taila can create an opportunity for the host immune system to tackle the onset of C. bertholletiae infection by delaying its pathogenesis. This can be further leveraged to reinforce the host immune system through combinatorial treatment to prevent the establishment of the mucormycosis infection.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, India.,Department of Allied and Applied Sciences, University of Patanjali, Haridwar 249405, India.,Vedic Acharya Samaj Foundation, Inc., NFP, FL 32811, United States.,Patanjali Yog Peeth (UK) Trust, Glasgow G41 1AU, United States
| | - Sohan Sengupta
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, India
| | - Priya Kumari
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, India
| | - Rishabh Dev
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, India
| | - Swati Haldar
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar 249405, India.,Department of Allied and Applied Sciences, University of Patanjali, Haridwar 249405, India.,Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
7
|
Intracellular cytokine expression in invasive fungal sinusitis and its impact on patient outcome. The Journal of Laryngology & Otology 2022; 136:861-865. [DOI: 10.1017/s0022215122000561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractBackgroundCell-mediated immunity plays an important role in host defence against fungal pathogens, regulated by differentiation of lymphocytes towards T-helper 1 or 2 cells. This study reports intracellular cytokine variation in terms of invasive fungal sinusitis type and outcome.MethodsThe mononuclear leukocytes of 15 patients with invasive fungal sinusitis (mucormycosis in 8, aspergillus in 7) were stained with antibodies against intracellular cytokines, after fungal antigen stimulation and culture, and immunophenotyped. Patients were followed up for six months, with clinical course categorised as improvement, worsening or death.ResultsThe mean percentages of mononuclear cells producing interleukins 4, 5, 10 and 12, and interferon-γ, in the mucormycosis group were 0.575, 0.284, 8.661, 4.460 and 1.134, respectively, while percentages in the aspergillosis group were 0.233, 0.492, 4.196, 4.466 and 1.533. Cells producing interleukin 4 and 10 were higher in the mucormycosis group, while those producing interleukin-12 and interferon-γ were lower. Cells producing interleukins 4 and 12 were higher in patients with a poor outcome (p-values of 0.0662 and 0.0373, respectively), while those producing interferon-γ were lower (p = 0.0864).ConclusionAdaptive cell-mediated immunity is expressed differently in two categories of invasive fungal sinusitis, and the cytokine expression pattern is related to prognosis.
Collapse
|
8
|
Hirata A, Miyashita K, Tanaka T, Hirata K, Narazaki T, Utsunomiya H, Ohno H, Nakashima E, Tachikawa Y, Choi I, Taguchi K, Suehiro Y. Serum immunoglobulin G as a discriminator of infection in follicular lymphoma patients undergoing chemotherapy with bendamustine in combination with rituximab. Hematology 2022; 27:384-395. [PMID: 35344477 DOI: 10.1080/16078454.2022.2051863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Chemotherapy, including bendamustine, usually causes lymphocytopaenia and hypogammaglobulinaemia as side effects in patients with haematological malignancies. Therefore, the possibility has been considered that these immunological adverse events induced by bendamustine may lead to infectious diseases. However, lymphocytopaenia and/or hypogammaglobulinaemia have not yet been shown to have a statistically significant association with infection in cancer patients who receive bendamustine. METHODS We retrospectively studied 27 patients with relapsed or refractory indolent follicular lymphoma who were treated with bendamustine and rituximab (BR). In order to elucidate relationships between immune-related laboratory parameters (i.e. peripheral blood leukocyte, neutrophil, lymphocyte and immunoglobulin G [IgG]) and infectious events, receiver operating characteristic (ROC) curve and multivariate logistic regression analyses were performed. RESULTS Infectious diseases occurred in 11 patients (11/27, 41%), including 3 (3/27, 11%) with severe diseases. The area under the ROC curve (AUC) showed that the lowest IgG level during and after BR discriminated infectious events (cut-off value, 603 mg/dL) with 81.8% sensitivity and 68.8% specificity (AUC, 0.76; 95% CI, 0.52-0.90). Furthermore, a multivariate regression analysis revealed that the minimal serum IgG value during and after BR therapy was the only variable that was significantly associated with infection (odds ratio, 8.29; 95% CI, 1.19-57.62; p value, 0.03). CONCLUSION Serum IgG ≤603 mg/dL during and after BR therapy was independently associated with an increased risk of infection. The monitoring of serum IgG during chemotherapy may help to predict the development of infection in blood cancer patients undergoing chemotherapy with bendamustine in combination with rituximab.
Collapse
Affiliation(s)
- Akie Hirata
- Department of Clinical Laboratory Medicine, NHO Kyushu Cancer Center, Fukuoka, Japan.,Medical Information Center, Kyushu University Hospital, Fukuoka, Japan
| | - Kaname Miyashita
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Takafumi Tanaka
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Kiyoko Hirata
- Department of Cell Therapy, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Taisuke Narazaki
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Hayato Utsunomiya
- Department of Cell Therapy, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Hirofumi Ohno
- Department of Cell Therapy, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Eriko Nakashima
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | | | - Ilseung Choi
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Kenichi Taguchi
- Department of Clinical Laboratory Medicine, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Youko Suehiro
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan.,Department of Cell Therapy, NHO Kyushu Cancer Center, Fukuoka, Japan
| |
Collapse
|
9
|
Al Dhaheri F, Lee RA, Sharma TS, Nakamura MM, Marty FM. Epidemiology and outcomes of invasive aspergillosis among pediatric immunocompromised patients: a 12-year single-center experience. Med Mycol 2022; 60:6524907. [PMID: 35138378 DOI: 10.1093/mmy/myac014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 12/11/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Invasive aspergillosis (IA) remains a common cause of mortality in pediatric immunocompromised populations. Much of our knowledge of IA stems from adult literature. We conducted a retrospective evaluation of cases of proven or probable IA, defined according to the 2019 EORTC/MSG criteria, in patients with underlying immunocompromising conditions at Boston Children's Hospital from January 1, 2007 to January 1, 2019. We estimated survival curves over 12 weeks using the Kaplan-Meier method for all-cause mortality, and we used univariate Cox proportional hazards modeling to evaluate for mortality risk factors. We identified 59 cases, 29% with proven and 71% with probable IA. Pulmonary IA was the most common presentation (78%). The median age at diagnosis was 11 years (range, 0.5-28). Hematopoietic cell transplantation (HCT) was the most frequent predisposing underlying condition (41%). Among affected patients, 44.8% were neutropenic and 59.3% were lymphopenic at diagnosis. The 12-week all-cause mortality rate was 25.4%; HCT recipients comprised the majority of deaths (9/15) with a hazard ratio of 2.47 [95% CI, 0.87-6.95]. No patients with congenital immunodeficiencies (n = 8) died within 12 weeks of IA diagnosis. Other risk factors that were significantly associated with mortality included mechanical ventilation at diagnosis, intensive care unit stay, and lymphopenia; treatment with an Aspergillus-active azole was associated with decreased mortality. In conclusion, our study found that in pediatric immunocompromised hosts, IA is associated with a high 12-week all-cause mortality rate, with a particular impact on the HCT population.
Collapse
Affiliation(s)
- Fatima Al Dhaheri
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rose A Lee
- Division of Infectious Disease, Department of Pediatrics, Boston Children's Hospital, Boston, United States.,Harvard Medical School, Boston, United States
| | - Tanvi S Sharma
- Division of Infectious Disease, Department of Pediatrics, Boston Children's Hospital, Boston, United States.,Harvard Medical School, Boston, United States
| | - Mari M Nakamura
- Division of Infectious Disease, Department of Pediatrics, Boston Children's Hospital, Boston, United States.,Harvard Medical School, Boston, United States.,Antimicrobial Stewardship Program, Boston Children's Hospital, Boston, United States
| | - Francisco M Marty
- Harvard Medical School, Boston, United States.,Division of Infectious Diseases, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, United States
| |
Collapse
|
10
|
Central Nervous System Fungal Infections in Paediatric Patients. CURRENT FUNGAL INFECTION REPORTS 2022. [DOI: 10.1007/s12281-021-00427-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
11
|
Radionuclide Imaging of Invasive Fungal Disease in Immunocompromised Hosts. Diagnostics (Basel) 2021; 11:diagnostics11112057. [PMID: 34829403 PMCID: PMC8620393 DOI: 10.3390/diagnostics11112057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Invasive fungal disease (IFD) leads to increased mortality, morbidity, and costs of treatment in patients with immunosuppressive conditions. The definitive diagnosis of IFD relies on the isolation of the causative fungal agents through microscopy, culture, or nucleic acid testing in tissue samples obtained from the sites of the disease. Biopsy is not always feasible or safe to be undertaken in immunocompromised hosts at risk of IFD. Noninvasive diagnostic techniques are, therefore, needed for the diagnosis and treatment response assessment of IFD. The available techniques that identify fungal-specific antigens in biological samples for diagnosing IFD have variable sensitivity and specificity. They also have limited utility in response assessment. Imaging has, therefore, been applied for the noninvasive detection of IFD. Morphologic imaging with computed tomography (CT) and magnetic resonance imaging (MRI) is the most applied technique. These techniques are neither sufficiently sensitive nor specific for the early diagnosis of IFD. Morphologic changes evaluated by CT and MRI occur later in the disease course and during recovery after successful treatment. These modalities may, therefore, not be ideal for early diagnosis and early response to therapy determination. Radionuclide imaging allows for targeting the host response to pathogenic fungi or specific structures of the pathogen itself. This makes radionuclide imaging techniques suitable for the early diagnosis and treatment response assessment of IFD. In this review, we aimed to discuss the interplay of host immunity, immunosuppression, and the occurrence of IFD. We also discuss the currently available radionuclide probes that have been evaluated in preclinical and clinical studies for their ability to detect IFD.
Collapse
|
12
|
Romani L, Williamson PR, Di Cesare S, Di Matteo G, De Luca M, Carsetti R, Figà-Talamanca L, Cancrini C, Rossi P, Finocchi A. Cryptococcal Meningitis and Post-Infectious Inflammatory Response Syndrome in a Patient With X-Linked Hyper IgM Syndrome: A Case Report and Review of the Literature. Front Immunol 2021; 12:708837. [PMID: 34335625 PMCID: PMC8320724 DOI: 10.3389/fimmu.2021.708837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/25/2021] [Indexed: 12/27/2022] Open
Abstract
The hyper IgM syndromes are a rare group of primary immunodeficiency. The X-linked Hyper IgM syndrome (HIGM), due to a gene defect in CD40L, is the commonest variant; it is characterized by an increased susceptibility to a narrow spectrum of opportunistic infection. A few cases of HIGM patients with Cryptococcal meningoencephalitis (CM) have been described in the literature. Herein we report the case of a young male diagnosed in infancy with HIGM who developed CM complicated by a post-infectious inflammatory response syndrome (PIIRS), despite regular immunoglobulin replacement therapy and appropriate antimicrobial prophylaxis. The patient was admitted because of a headache and CM was diagnosed through detection of Cryptococcus neoformans in the cerebrospinal fluid. Despite the antifungal therapy resulting to negative CSF culture, the patient exhibited persistent headaches and developed diplopia. An analysis of inflammatory cytokines on CSF, as well as the brain MRI, suggested a diagnosis of PIIRS. Therefore, a prolonged corticosteroids therapy was started obtaining a complete resolution of symptoms without any relapse.
Collapse
Affiliation(s)
- Lorenza Romani
- Unit of Immune and Infectious Diseases, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | - Peter Richard Williamson
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Health, Bethesda, MD, United States
| | - Silvia Di Cesare
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Gigliola Di Matteo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Maia De Luca
- Unit of Immune and Infectious Diseases, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | - Rita Carsetti
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | - Lorenzo Figà-Talamanca
- Neuroradiology Unit, Imaging Department, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | - Caterina Cancrini
- Unit of Immune and Infectious Diseases, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Rossi
- Unit of Immune and Infectious Diseases, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Finocchi
- Unit of Immune and Infectious Diseases, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
13
|
Abraitytė S, Kotsi E, Devlin LA, Edgar JDM. Unexpected combination: DiGeorge syndrome and myeloperoxidase deficiency. BMJ Case Rep 2020; 13:e232741. [PMID: 32107256 PMCID: PMC7046411 DOI: 10.1136/bcr-2019-232741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
We report a case of a 3-year-old boy who presented with recurrent bacterial and fungal infections and a known diagnosis of partial DiGeorge (22q11.2 deletion) syndrome. The nature and severity of his infections were more than normally expected in partial DiGeorge syndrome with normal T-cell counts and T-cell proliferative response to phytohaemagglutinin. This prompted further investigation of the immune system. An abnormal neutrophil respiratory oxidative burst, but normal protein expression of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase system, led to the identification of myeloperoxidase deficiency. DiGeorge syndrome has a heterogeneous clinical phenotype and may not be an isolated diagnosis. It raises awareness of the possibility of two rare diseases occurring in a single patient and emphasises that even when a rare diagnosis is confirmed, if the clinical features remain atypical or unresponsive, then further investigation for additional cofactors is warranted.
Collapse
Affiliation(s)
| | | | - Lisa Anne Devlin
- Regional Immunology Service, Royal Victoria Hospital, Belfast, UK
| | | |
Collapse
|
14
|
T Cell Antifungal Immunity and the Role of C-Type Lectin Receptors. Trends Immunol 2019; 41:61-76. [PMID: 31813764 PMCID: PMC7427322 DOI: 10.1016/j.it.2019.11.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 10/28/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023]
Abstract
Fungi can cause disease in humans, from mucocutaneous to life-threatening systemic infections. Initiation of antifungal immunity involves fungal recognition by pattern recognition receptors such as C-type lectin receptors (CLRs). These germline-encoded receptors trigger a multitude of innate responses including phagocytosis, fungal killing, and antigen presentation which can also shape the development of adaptive immunity. Recently, studies have shed light on how CLRs directly or indirectly modulate lymphocyte function. Moreover, CLR-mediated recognition of commensal fungi maintains homeostasis and prevents invasion from opportunistic commensals. We present an overview of current knowledge of antifungal T cell immune responses, with emphasis on the role of C-type lectins, and discuss how these receptors modulate these responses at different levels. CLRs are essential pattern recognition receptors involved in fungal recognition and initiation of protective antifungal immunity. CLRs promote the differentiation of mammalian T helper cell subsets essential for the control of systemic (Th1) and mucosal (Th17) fungal infections. CLRs are involved in antigen presentation, the expression of co-stimulatory molecules, and cytokine secretion; therefore, they can regulate lymphocyte function and adaptive immune responses at different levels. Fungal morphological changes, such as the transition from yeast to hyphae in Candida albicans during tissue invasion, affects recognition by CLRs and impacts on adaptive immune responses. CLRs recognize the fungal component of the microbiome that can influence T cell responses during infection at intestinal and peripheral sites.
Collapse
|
15
|
Mark C, McGinn C. From Culture to Fungal Biomarkers: the Diagnostic Route of Fungal Infections in Children with Primary Immunodeficiencies. CURRENT FUNGAL INFECTION REPORTS 2019. [DOI: 10.1007/s12281-019-00356-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
16
|
Suzuki SML, Morelli F, Negri M, Bonfim-Mendonça P, Kioshima ÉS, Salci T, Voidaleski MF, Vicente VA, Svidzinski T. FATAL cryptococcal meningitis in a child with hyper-immunoglobulin M syndrome, with an emphasis on the agent. J Mycol Med 2019; 29:273-277. [PMID: 31409527 DOI: 10.1016/j.mycmed.2019.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/22/2019] [Accepted: 07/03/2019] [Indexed: 10/26/2022]
Abstract
Following a fatal case of Cryptococcus neoformans meningitis in a child with X-linked hyper-immunoglobulin M syndrome (XHIGM), we evaluated the fungal isolate in an experimental infection in a mouse model with respect to microbiology, epidemiology, virulence and response to therapy. The minimum inhibitory concentrations for antifungals in the susceptibility test were 0.5mg/L for amphotericin B, 4.0mg/L for fluconazole and 0.12mg/L for voriconazole. Evaluation of pathogenicity by means of an experimental infection in BALB/c mice showed that fungus isolated from the blood and cerebrospinal fluid of the child was able to disseminate, reaching the spleen, lungs and brain, where it caused significant macroscopic alterations in the size and texture of each organ. Treatment of infected mice with amphotericin B reduced the fungal load in the spleen and lungs, but not in the brain.
Collapse
Affiliation(s)
- S M L Suzuki
- Section of Medical Mycology, Universidade Estadual de Maringá, Maringá, Brazil
| | - F Morelli
- Section of Medical Mycology, Universidade Estadual de Maringá, Maringá, Brazil
| | - M Negri
- Section of Medical Mycology, Universidade Estadual de Maringá, Maringá, Brazil
| | - P Bonfim-Mendonça
- Section of Medical Mycology, Universidade Estadual de Maringá, Maringá, Brazil
| | - É S Kioshima
- Section of Medical Mycology, Universidade Estadual de Maringá, Maringá, Brazil
| | - T Salci
- Section of Medical Mycology, Universidade Estadual de Maringá, Maringá, Brazil; Department of Pharmacy, Faculdade Integrado, Campo Mourão, Paraná, Brazil
| | - M F Voidaleski
- Microbiology, Parasitology and Pathology Post-Graduation Program, Department of Pathology, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - V A Vicente
- Microbiology, Parasitology and Pathology Post-Graduation Program, Department of Pathology, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - T Svidzinski
- Section of Medical Mycology, Universidade Estadual de Maringá, Maringá, Brazil.
| |
Collapse
|
17
|
Clinical features in proven and probable invasive fungal disease in children and adolescents at a pediatric referral center: a 5-year experience. World J Pediatr 2019; 15:270-275. [PMID: 31011987 DOI: 10.1007/s12519-019-00259-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/12/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND There is limited information concerning the overall epidemiology of invasive fungal disease (IFD) in children. The aim of this study was to clarify the clinical features of IFD in a tertiary pediatric care hospital. METHODS Patients diagnosed with proven or probable IFD at our hospital between 2011 and 2015 were retrospectively reviewed. Proven and probable IFD were defined according to the European Organization for Research and Treatment of Cancer/Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group consensus. Patients with possible IFD were excluded. RESULTS The incidence of proven or probable IFD was 26 of 20,079 hospitalized patients (0.13%). The predominant underlying disease was malignancy (54%) and congenital anomaly (27%). The most common diagnosis was candidemia among the patients with proven IFD (8 of 13, 62%). All the isolated pathogens in the candidemia patients were non-albicans Candida spp. The most common site of infection was the lungs in patients with probable IFD (11 of 13 patients, 85%). In probable IFD episodes, positive β-D-glucan and galactomannan were found in 12 of 13 (92%) and 5 of 13 (38%) patients, respectively. All but one patient (96%) received empirical antifungal therapy. No patients underwent surgical resection of residual lesions. The overall mortality was 23% and the attributable mortality of IFD was 12%. CONCLUSION Our results suggest the emergence of non-albicans Candida species as important pathogens in childhood IFD.
Collapse
|
18
|
Warris A, Lehrnbecher T, Roilides E, Castagnola E, Brüggemann RJM, Groll AH. ESCMID-ECMM guideline: diagnosis and management of invasive aspergillosis in neonates and children. Clin Microbiol Infect 2019; 25:1096-1113. [PMID: 31158517 DOI: 10.1016/j.cmi.2019.05.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/19/2019] [Accepted: 05/23/2019] [Indexed: 01/01/2023]
Abstract
SCOPE Presenting symptoms, distributions and patterns of diseases and vulnerability to invasive aspergillosis (IA) are similar between children and adults. However, differences exist in the epidemiology and underlying conditions, the usefulness of newer diagnostic tools, the pharmacology of antifungal agents and in the evidence from interventional phase 3 clinical trials. Therefore, the European Society for Clinical Microbiology and Infectious Diseases (ESCMID) and the European Confederation of Medical Mycology (ECMM) have developed a paediatric-specific guideline for the diagnosis and management of IA in neonates and children. METHODS Review and discussion of the scientific literature and grading of the available quality of evidence was performed by the paediatric subgroup of the ESCMID-ECMM-European Respiratory Society (ERS) Aspergillus disease guideline working group, which was assigned the mandate for the development of neonatal- and paediatric-specific recommendations. QUESTIONS Questions addressed by the guideline included the epidemiology of IA in neonates and children; which paediatric patients may benefit from antifungal prophylaxis; how to diagnose IA in neonates and children; which antifungal agents are available for use in neonates and children; which antifungal agents are suitable for prophylaxis and treatment of IA in neonates and children; what is the role of therapeutic drug monitoring of azole antifungals; and which management strategies are suitable to be used in paediatric patients. This guideline provides recommendations for the diagnosis, prevention and treatment of IA in the paediatric population, including neonates. The aim of this guideline is to facilitate optimal management of neonates and children at risk for or diagnosed with IA.
Collapse
Affiliation(s)
- A Warris
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom; European Society of Clinical Microbiology and Infectious Diseases Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology, the Netherlands.
| | - T Lehrnbecher
- Division of Paediatric Haematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe-University, Frankfurt, Germany; European Society of Clinical Microbiology and Infectious Diseases Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology, the Netherlands
| | - E Roilides
- Infectious Diseases Unit, 3rd Department of Paediatrics, Faculty of Medicine, Aristotle University 96 School of Health Sciences, Thessaloniki, Greece; European Society of Clinical Microbiology and Infectious Diseases Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology, the Netherlands
| | - E Castagnola
- Infectious Diseases Unit, IRCCS Istituto Giannina Gaslini Children's Hospital, Genoa, Italy; European Society of Clinical Microbiology and Infectious Diseases Fungal Infection Study Group (EFISG)
| | - R J M Brüggemann
- Radboud Center for Infectious Diseases, Radboud University Medical Centre, Center of Expertise in Mycology Radboudumc/CWZ, European Confederation of Medical Mycology (ECMM) Excellence Center of Medical Mycology, Nijmegen, the Netherlands; European Society of Clinical Microbiology and Infectious Diseases Fungal Infection Study Group (EFISG)
| | - A H Groll
- Infectious Disease Research Program, Center for Bone Marrow Transplantation and Department of Paediatric Hematology/Oncology, University Children's Hospital Münster, Münster, Germany; European Society of Clinical Microbiology and Infectious Diseases Fungal Infection Study Group (EFISG); European Confederation of Medical Mycology, the Netherlands
| |
Collapse
|
19
|
Efficacy of Olorofim (F901318) against Aspergillus fumigatus, A. nidulans, and A. tanneri in Murine Models of Profound Neutropenia and Chronic Granulomatous Disease. Antimicrob Agents Chemother 2019; 63:AAC.00129-19. [PMID: 30885903 DOI: 10.1128/aac.00129-19] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/14/2019] [Indexed: 12/19/2022] Open
Abstract
The emergence of azole resistance in Aspergillus fumigatus as well as an increasing frequency of multiresistant cryptic Aspergillus spp. necessitates exploration of new classes of antifungals. Olorofim (formerly F901318) is a new fungicidal agent that prevents the growth of ascomycetous mold species via inhibition of de novo pyrimidine biosynthesis, a mechanism of action distinct from that of currently available antifungal drugs. We studied the in vivo efficacy of olorofim intraperitoneal therapy (15 mg/kg of body weight every 8 h for 9 days) against infection with A. fumigatus, A. nidulans, and A. tanneri in both neutropenic CD-1 mice and mice with chronic granulomatous disease (CGD) (gp91 -/- phox mice). In the neutropenic mouse model, 80% to 88% of treated mice survived for 10 days, and in the CGD group, 63% to 88% of treated mice survived for 10 days, depending on the infecting species, while less than 10% of the mice in the control groups survived for 10 days. In the olorofim-treated groups, galactomannan levels were significantly suppressed, with lower organ fungal DNA burdens being seen for all three Aspergillus spp. Histopathological slides revealed a limited number of inflammatory foci with or without detectable fungal elements in the kidneys of neutropenic CD-1 mice and in the lungs of CGD mice. Furthermore, the efficacy of olorofim was unrelated to the triazole MICs of the infecting Aspergillus spp. These results show olorofim to be a promising therapeutic agent for invasive aspergillosis.
Collapse
|
20
|
Estimated Burden of Serious Fungal Infections in Ghana. J Fungi (Basel) 2019; 5:jof5020038. [PMID: 31083531 PMCID: PMC6616901 DOI: 10.3390/jof5020038] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/14/2022] Open
Abstract
Fungal infections are increasingly becoming common and yet often neglected in developing countries. Information on the burden of these infections is important for improved patient outcomes. The burden of serious fungal infections in Ghana is unknown. We aimed to estimate this burden. Using local, regional, or global data and estimates of population and at-risk groups, deterministic modelling was employed to estimate national incidence or prevalence. Our study revealed that about 4% of Ghanaians suffer from serious fungal infections yearly, with over 35,000 affected by life-threatening invasive fungal infections. Incidence of cryptococcal meningitis, Pneumocystis jirovecii pneumonia, and disseminated histoplasmosis cases in AIDS was estimated at 6275, 12,610 and 724, respectively. Oral and esophageal candidiasis collectively affect 27,100 Ghanaians and 42,653 adult asthmatics are estimated to have fungal asthma. We estimate a prevalence of 12,620 cases of chronic pulmonary aspergillosis (CPA and an incidence of 1254 cases of invasive aspergillosis (IA). Estimated cases of candidemia and candida peritonitis cases were 1446 and 217, respectively. The estimated prevalence of recurrent vulvovaginal candidiasis (RVVC) and tinea capitis was 442,621 and 598,840, respectively. Mucormycosis and fungal keratitis each may affect 58 and 810 Ghanaians. These data highlight the urgent need for intensified awareness to improve diagnosis and management.
Collapse
|
21
|
Iosifidis E, Papachristou S, Roilides E. Advances in the Treatment of Mycoses in Pediatric Patients. J Fungi (Basel) 2018; 4:E115. [PMID: 30314389 PMCID: PMC6308938 DOI: 10.3390/jof4040115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/05/2018] [Accepted: 10/07/2018] [Indexed: 12/16/2022] Open
Abstract
The main indications for antifungal drug administration in pediatrics are reviewed as well as an update of the data of antifungal agents and antifungal policies performed. Specifically, antifungal therapy in three main areas is updated as follows: a) Prophylaxis of premature neonates against invasive candidiasis; b) management of candidemia and meningoencephalitis in neonates; and c) prophylaxis, empiric therapy, and targeted antifungal therapy in children with primary or secondary immunodeficiencies. Fluconazole remains the most frequent antifungal prophylactic agent given to high-risk neonates and children. However, the emergence of fluconazole resistance, particularly in non-albicans Candida species, should be considered during preventive or empiric therapy. In very-low birth-weight neonates, although fluconazole is used as antifungal prophylaxis in neonatal intensive care units (NICU's) with relatively high incidence of invasive candidiasis (IC), its role is under continuous debate. Amphotericin B, primarily in its liposomal formulation, remains the mainstay of therapy for treating neonatal and pediatric yeast and mold infections. Voriconazole is indicated for mold infections except for mucormycosis in children >2 years. Newer triazoles-such as posaconazole and isavuconazole-as well as echinocandins, are either licensed or under study for first-line or salvage therapy, whereas combination therapy is kept for refractory cases.
Collapse
Affiliation(s)
- Elias Iosifidis
- Infectious Diseases Unit, 3rd Department of Pediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Konstantinoupoleos 49, 54642, Thessaloniki, Greece.
| | - Savvas Papachristou
- Infectious Diseases Unit, 3rd Department of Pediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Konstantinoupoleos 49, 54642, Thessaloniki, Greece.
| | - Emmanuel Roilides
- Infectious Diseases Unit, 3rd Department of Pediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Konstantinoupoleos 49, 54642, Thessaloniki, Greece.
| |
Collapse
|
22
|
STAT3-Deficient hyperimmunoglobulin E syndrome: report of a case with orofacial granulomatosis-like disease. Oral Surg Oral Med Oral Pathol Oral Radiol 2018; 126:e252-e257. [PMID: 30126807 DOI: 10.1016/j.oooo.2018.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 07/05/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022]
Abstract
Hyperimmunoglobulin E syndrome (HIES) is a rare heterogeneous primary immunodeficiency disorder characterized by infections of the lung and skin, elevated serum immunoglobulin E, and involvement of soft and bony tissues. Autosomal dominant HIES and related disorders are caused by defects in the Janus activated kinase-signal transducer and activator of transcription signaling pathway, leading to reduced numbers of T helper cell type 17 and impaired production of interleukin (IL)-17 A, IL-17 F, and IL-22. In addition, neutrophils have chemotactic defects, resulting in impaired responses at skin and lung sites. We report here a case of orofacial granulomatosis-like disease in a teenage boy ultimately found to have autosomal dominant HIES caused by a heterozygous mutation in the STAT3 gene.
Collapse
|
23
|
Kumar NR, Swaroop DK, Punna N, Sirisha K, Ganapathi T, Kumar CG, Narsaiah B. Synthesis of Novel Pyrido[2′, 3′:3,4]Pyrazolo[1, 5‐
a
]Quinazoline Derivatives, Their Biological Evaluation and Molecular Modelling Studies. ChemistrySelect 2018. [DOI: 10.1002/slct.201801186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Nagiri Ravi Kumar
- Fluoroorganic divisionCSIR-Indian Institute of Chemical Technology, Tarnaka Hyderabad-500607 India
| | | | - Nagender Punna
- Fluoroorganic divisionCSIR-Indian Institute of Chemical Technology, Tarnaka Hyderabad-500607 India
| | - Kanugala Sirisha
- Medicinal Chemistry and Pharmacology DivisionCSIR-Indian Institute of Chemical Technology
| | - Thipparapu Ganapathi
- Stem Cell Research DivisionDepartment of BiochemistryNational Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR) Hyderabad-500007 Telangana India
| | - Chityal Ganesh Kumar
- Medicinal Chemistry and Pharmacology DivisionCSIR-Indian Institute of Chemical Technology
| | - Banda Narsaiah
- Fluoroorganic divisionCSIR-Indian Institute of Chemical Technology, Tarnaka Hyderabad-500607 India
| |
Collapse
|
24
|
Gresnigt MS, Jaeger M, Subbarao Malireddi RK, Rasid O, Jouvion G, Fitting C, Melchers WJG, Kanneganti TD, Carvalho A, Ibrahim-Granet O, van de Veerdonk FL. The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression. Front Immunol 2017; 8:1777. [PMID: 29326692 PMCID: PMC5733348 DOI: 10.3389/fimmu.2017.01777] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/28/2017] [Indexed: 01/01/2023] Open
Abstract
One of the major life-threatening infections for which severely immunocompromised patients are at risk is invasive aspergillosis (IA). Despite the current treatment options, the increasing antifungal resistance and poor outcome highlight the need for novel therapeutic strategies to improve outcome of patients with IA. In the current study, we investigated whether and how the intracellular pattern recognition receptor NOD1 is involved in host defense against Aspergillus fumigatus. When exploring the role of NOD1 in an experimental mouse model, we found that Nod1−/− mice were protected against IA and demonstrated reduced fungal outgrowth in the lungs. We found that macrophages derived from bone marrow of Nod1−/− mice were more efficiently inducing reactive oxygen species and cytokines in response to Aspergillus. Most strikingly, these cells were highly potent in killing A. fumigatus compared with wild-type cells. In line, human macrophages in which NOD1 was silenced demonstrated augmented Aspergillus killing and NOD1 stimulation decreased fungal killing. The differentially altered killing capacity of NOD1 silencing versus NOD1 activation was associated with alterations in dectin-1 expression, with activation of NOD1 reducing dectin-1 expression. Furthermore, we were able to demonstrate that Nod1−/− mice have elevated dectin-1 expression in the lung and bone marrow, and silencing of NOD1 gene expression in human macrophages increases dectin-1 expression. The enhanced dectin-1 expression may be the mechanism of enhanced fungal killing of Nod1−/− cells and human cells in which NOD1 was silenced, since blockade of dectin-1 reversed the augmented killing in these cells. Collectively, our data demonstrate that NOD1 receptor plays an inhibitory role in the host defense against Aspergillus. This provides a rationale to develop novel immunotherapeutic strategies for treatment of aspergillosis that target the NOD1 receptor, to enhance the efficiency of host immune cells to clear the infection by increasing fungal killing and cytokine responses.
Collapse
Affiliation(s)
- Mark S Gresnigt
- Unité de recherche Cytokines and Inflammation, Institut Pasteur, Paris, France.,Laboratory for Experimental Internal Medicine, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Martin Jaeger
- Laboratory for Experimental Internal Medicine, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Orhan Rasid
- Unité de recherche Cytokines and Inflammation, Institut Pasteur, Paris, France
| | - Grégory Jouvion
- Unité Histopathologie Humaine et Modèles Animaux, Département Infection et Epidémiologie, Institut Pasteur, Paris, France
| | - Catherine Fitting
- Unité de recherche Cytokines and Inflammation, Institut Pasteur, Paris, France
| | - Willem J G Melchers
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | | | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Frank L van de Veerdonk
- Laboratory for Experimental Internal Medicine, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
25
|
Abstract
Considerable progress has been made in the prevention, diagnosis, and management of pediatric patients with invasive fungal disease (IFD). The reported decreasing trend in the incidence of invasive candidiasis (IC) over the past 15 years in both neonates and children has been encouraging. Nevertheless, due to the growing number of immunocompromised children at risk for IFD, this disease continues to be associated with significant morbidity and death and with increased financial burden to the health care system. Therefore, it is important to understand the contemporary epidemiology of IFD. Incidence rates of IFD in children are affected by geographical, population, and time variability. There is an ongoing effort to constantly document and update the incidence of IFD and species distribution among different pediatric populations as a means to direct preventative, diagnostic, and therapeutic resources to the most appropriate subset of patients. Children with a hematologic malignancy or a primary or secondary immunodeficiency, those undergoing solid organ or hematopoietic stem cell transplantation, and premature neonates are the major subsets of pediatric patients at risk of developing IFD. In this review, we focus on fungal disease epidemiology with a specific emphasis on the 2 most common pediatric IFDs, IC and invasive aspergillosis (IA).
Collapse
Affiliation(s)
- Zoi Dorothea Pana
- Hospital Epidemiology and Infection Control Department (HEIC), Division of Infectious Diseases, Johns Hopkins Hospital, Baltimore, Maryland
- Third Department of Paediatrics, Infectious Diseases Unit, Aristotle University School of Medicine, Hippokration General Hospital, Thessaloniki, Greece
| | - Emmanuel Roilides
- Third Department of Paediatrics, Infectious Diseases Unit, Aristotle University School of Medicine, Hippokration General Hospital, Thessaloniki, Greece
| | - Adilia Warris
- Aberdeen Fungal Group, MRC Centre for Medical Mycology, Institute of Medical Sciences and the Royal Aberdeen Children’s Hospital, University of Aberdeen, United Kingdom
| | - Andreas H Groll
- Center for Bone Marrow Transplantation and Department of Paediatric Hematology and Oncology, Infectious Disease Research Program, University Children’s Hospital, Muenster, Germany
| | - Theoklis Zaoutis
- Division of Infectious Diseases and Center for Pediatric Clinical Effectiveness Research, Children’s Hospital of Philadelphia, Pennsylvania; and
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia
| |
Collapse
|
26
|
Ni T, Li R, Xie F, Zhao J, Huang X, An M, Zang C, Cai Z, Zhang D, Jiang Y. Synthesis and Biological Evaluation of Novel 2-Aminonicotinamide Derivatives as Antifungal Agents. ChemMedChem 2017; 12:319-326. [PMID: 28071858 DOI: 10.1002/cmdc.201600545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/20/2016] [Indexed: 01/21/2023]
Abstract
Based on the structures of the reported compounds G884 [N-(3-(pentan-2-yloxy)phenyl)nicotinamide], E1210 [3-(3-(4-((pyridin-2-yloxy)methyl)benzyl)isoxazol-5-yl)pyridin-2-amine], and 10 b [2-amino-N-((5-(3-fluorophenoxy)thiophen-2-yl)methyl)nicotinamide], which inhibit the biosynthesis of glycosylphosphatidylinositol (GPI)-anchored proteins in fungi, a series of novel 2-aminonicotinamide derivatives were designed, synthesized, and evaluated for in vitro antifungal activity. Most of these compounds were found to exhibit potent in vitro antifungal activity against Candida albicans, with MIC80 values ranging from 0.0313 to 4.0 μg mL-1 . In particular, compounds 11 g [2-amino-N-((5-(((2-fluorophenyl)amino)methyl)thiophen-2-yl)methyl)nicotinamide] and 11 h [2-amino-N-((5-(((3-fluorophenyl)amino)methyl)thiophen-2-yl)methyl)nicotinamide] displayed excellent activity against C. albicans, with MIC80 values of 0.0313 μg mL-1 , and exhibited broad-spectrum antifungal activity against fluconazole-resistant C. albicans, C. parapsilosis, C. glabrata, and Cryptococcus neoformans, with a MIC80 range of 0.0313-2.0 μg mL-1 . Further studies by electron microscopy and laser confocal microscopy indicated that compound 11 g targets the cell wall and decreases GPI anchor content on the cell surface of C. albicans.
Collapse
Affiliation(s)
- Tingjunhong Ni
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Ran Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Fei Xie
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jing Zhao
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.,School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, 350112, China
| | - Xin Huang
- Department of Pharmacology, Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200092, China
| | - Maomao An
- Department of Pharmacology, Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200092, China
| | - Chengxu Zang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Zhan Cai
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Dazhi Zhang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.,School of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, 350112, China
| | - Yuanying Jiang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| |
Collapse
|
27
|
Myeloperoxidase Attenuates Pathogen Clearance during Plasmodium yoelii Nonlethal Infection. Infect Immun 2016; 85:IAI.00475-16. [PMID: 27795354 DOI: 10.1128/iai.00475-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/06/2016] [Indexed: 01/23/2023] Open
Abstract
Myeloperoxidase (MPO), a leukocyte-derived enzyme mainly secreted by activated neutrophils, is known to be involved in the immune response during bacterial and fungal infection and inflammatory diseases. Nevertheless, the role of MPO in a parasitic disease like malaria is unknown. We hypothesized that MPO contributes to parasite clearance. To address this hypothesis, we used Plasmodium yoelii nonlethal infection in wild-type and MPO-deficient mice as a murine malaria model. We detected high MPO plasma levels in wild-type mice with Plasmodium yoelii infection. Unexpectedly, infected MPO-deficient mice did not show increased parasite loads but were able to clear the infection more rapidly than wild-type mice. Additionally, the presence of neutrophils at the onset of infection seemed not to be essential for the control of the parasitemia. The effect of decreased parasite levels in MPO-deficient mice was absent from animals lacking mature T and B cells, indicating that this effect is most likely dependent on adaptive immune response mechanisms. Indeed, we observed increased gamma interferon and tumor necrosis factor alpha production by T cells in infected MPO-deficient mice. Together, these results suggest that MPO modulates the adaptive immune response during malaria infection, leading to an attenuated parasite clearance.
Collapse
|
28
|
Becker KL, Rösler B, Wang X, Lachmandas E, Kamsteeg M, Jacobs CW, Joosten LA, Netea MG, van de Veerdonk FL. Th2 and Th9 responses in patients with chronic mucocutaneous candidiasis and hyper-IgE syndrome. Clin Exp Allergy 2016; 46:1564-1574. [DOI: 10.1111/cea.12787] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 05/20/2016] [Accepted: 07/16/2016] [Indexed: 01/06/2023]
Affiliation(s)
- K. L. Becker
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
| | - B. Rösler
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
| | - X. Wang
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
- Department of Dermatology; Peking University First Hospital; Beijing China
| | - E. Lachmandas
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
| | - M. Kamsteeg
- Department of Dermatology; Radboud University Medical Center; Nijmegen The Netherlands
| | - C. W. Jacobs
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
| | - L. A. Joosten
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
| | - M. G. Netea
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
| | - F. L. van de Veerdonk
- Department of Internal Medicine; Radboud University Medical Center; Radboud Center for Infectious Diseases (RCI); Nijmegen The Netherlands
| |
Collapse
|
29
|
Wattier RL, Ramirez-Avila L. Pediatric Invasive Aspergillosis. J Fungi (Basel) 2016; 2:jof2020019. [PMID: 29376936 PMCID: PMC5753081 DOI: 10.3390/jof2020019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 02/06/2023] Open
Abstract
Invasive aspergillosis (IA) is a disease of increasing importance in pediatrics due to growth of the immunocompromised populations at risk and improvements in long-term survival for many of these groups. While general principles of diagnosis and therapy apply similarly across the age spectrum, there are unique considerations for clinicians who care for children and adolescents with IA. This review will highlight important differences in the epidemiology, clinical manifestations, diagnosis, and therapy of pediatric IA.
Collapse
Affiliation(s)
- Rachel L Wattier
- Department of Pediatrics, Division of Infectious Diseases and Global Health, University of California-San Francisco, 550 16th St, 4th Floor, Box 0434, San Francisco, CA 94143, USA.
| | - Lynn Ramirez-Avila
- Department of Pediatrics, Division of Infectious Diseases and Global Health, University of California-San Francisco, 550 16th St, 4th Floor, Box 0434, San Francisco, CA 94143, USA.
| |
Collapse
|
30
|
Contou D, Dorison M, Rosman J, Schlemmer F, Gibelin A, Foulet F, Botterel F, Carteaux G, Razazi K, Brun-Buisson C, Mekontso Dessap A, de Prost N. Aspergillus-positive lower respiratory tract samples in patients with the acute respiratory distress syndrome: a 10-year retrospective study. Ann Intensive Care 2016; 6:52. [PMID: 27294891 PMCID: PMC4906097 DOI: 10.1186/s13613-016-0156-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/05/2016] [Indexed: 12/24/2022] Open
Abstract
Background The detection of Aspergillus spp. in endotracheal aspirate cultures of mechanically ventilated patients may reflect either colonization or infection. However, little is known about the prevalence and the impact on outcome of respiratory tract sample positive for Aspergillus during the acute respiratory distress syndrome (ARDS). Methods We conducted a monocentric, retrospective study over a 10-year period (January 2006–December 2015) in the ICU of a university hospital. All consecutive adult patients with ARDS were included, and the diagnosis of invasive pulmonary aspergillosis was assessed using a previously validated algorithm. Results In total, 423 ARDS patients were included with 35 patients [8.3 %, 95 % CI (5.4–10.6)] having at least one respiratory tract sample positive for Aspergillus (Aspergillus+ patients) after a median delay of 3 days (1–11) following ICU admission. Comorbidities did not differ between Aspergillus+ and Aspergillus− patients except for more frequent immunosuppression in Aspergillus+ patients (40 vs. 22 %; p = 0.02). There was no difference between Aspergillus− and Aspergillus+ patients regarding in-ICU mortality, ventilator-free days at day 28, and incidence of ventilator-associated pneumonia, but need for renal replacement therapy was higher in Aspergillus+ patients than in others (49 vs. 27 %; p = 0.01). Seventeen [4.0 %, 95 % CI (2.1–5.9)] patients had putative/proven aspergillosis. After adjusting on covariates associated with ICU mortality, putative/proven aspergillosis was associated with in-ICU mortality [aOR = 9.58 (1.97–46.52); p = 0.005], while Aspergillus colonization was not [aOR = 0.64 (0.21–1.99); p = 0.44]. Conclusions Eight percent of ARDS patients had Aspergillus spp.-positive respiratory tract cultures. These had a higher risk of mortality only when categorized as having putative or proven invasive pulmonary aspergillosis.
Collapse
Affiliation(s)
- Damien Contou
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France.,Groupe de Recherche CARMAS, Faculté de Médecine de Créteil, Université Paris Est Créteil, 94010, Créteil, France
| | - Matthieu Dorison
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France
| | - Jérémy Rosman
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France
| | - Frédéric Schlemmer
- Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Antenne de Pneumologie, Assistance Publique-Hôpitaux de Paris, 94010, Créteil, France
| | - Aude Gibelin
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France
| | - Françoise Foulet
- Unité de Mycologie, Département de Biologie-Pathologie, Centre Hospitalier Universitaire Henri Mondor, DHU VIC, Assistance Publique-Hôpitaux de Paris, 94010, Créteil, France
| | - Françoise Botterel
- Unité de Mycologie, Département de Biologie-Pathologie, Centre Hospitalier Universitaire Henri Mondor, DHU VIC, Assistance Publique-Hôpitaux de Paris, 94010, Créteil, France
| | - Guillaume Carteaux
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France.,Groupe de Recherche CARMAS, Faculté de Médecine de Créteil, Université Paris Est Créteil, 94010, Créteil, France
| | - Keyvan Razazi
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France.,Groupe de Recherche CARMAS, Faculté de Médecine de Créteil, Université Paris Est Créteil, 94010, Créteil, France
| | - Christian Brun-Buisson
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France.,Groupe de Recherche CARMAS, Faculté de Médecine de Créteil, Université Paris Est Créteil, 94010, Créteil, France
| | - Armand Mekontso Dessap
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France.,Groupe de Recherche CARMAS, Faculté de Médecine de Créteil, Université Paris Est Créteil, 94010, Créteil, France
| | - Nicolas de Prost
- Groupe Henri Mondor-Albert Chenevier, Centre Hospitalier Universitaire Henri Mondor, DHU A-TVB, Service de Réanimation Médicale, Assistance Publique-Hôpitaux de Paris, 51, Avenue du Maréchal de Lattre de Tassigny, 94010, Créteil Cedex, France. .,Groupe de Recherche CARMAS, Faculté de Médecine de Créteil, Université Paris Est Créteil, 94010, Créteil, France.
| |
Collapse
|
31
|
Ayatollahi Mousavi SA, Asadikaram G, Nakhaee N, Izadi A. Plasma Levels of IFN-γ, IL-4, IL-6 and IL-17 in HIV-Positive Patients With Oral Candidiasis. Jundishapur J Microbiol 2016; 9:e32021. [PMID: 27127595 PMCID: PMC4842255 DOI: 10.5812/jjm.32021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/10/2015] [Accepted: 12/07/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cell-mediated immunity (CMI) by CD4 + Th (T helper)-type cells is the predominant host defense mechanism against Oral Candidiasis (OC) in HIV-infected individuals. Weakened CMI and depletion of CD4 + T cells are the main factor contributing to the output of OC in HIV-positive individuals. The cytokines produced by Th1, Th2 and Th17 cells play a role in mediating an increased susceptibility to OC during HIV infection. OBJECTIVES The present study investigated plasma concentration of IFN-γ, IL-4, IL-6 and IL-17 in HIV-1 patients suffering from OC. PATIENTS AND METHODS In total, 98 samples in four groups (HIV-positive and HIV-negative persons with and without OC) were obtained from the oral cavities and cultured on Sabouraud's dextrose agar and CHROMagar. Also blood samples were obtained to assess plasma level of IFN-γ, IL-4, IL-6 and IL-17 using ELISA technique. RESULTS There was a statistically significant difference in the plasma concentration of IFN-γ, IL-6 and IL-17 but not about IL-4. Our findings suggest a significant interaction between fungal infection and HIV on expression of assessed cytokines. CONCLUSIONS Fungal infection and HIV alone and together could seriously alter immune system function as assessed by measuring the levels of the plasma cytokines. Therefore, these results provide important new information relative to the putative immune-based factors associated with resistance and/or susceptibility to OC in HIV-positive persons.
Collapse
Affiliation(s)
- Seyyed Amin Ayatollahi Mousavi
- Department of Medical Mycology and Parasitology, School of Medicine, Kerman University of Medical Sciences, Kerman, IR Iran
- Tropical and Infectious Disease Research Center, Kerman University of Medical Sciences, Kerman, IR Iran
| | - Gholamreza Asadikaram
- Institutes of Neuropharmacology, Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, IR Iran
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, IR Iran
| | - Nouzar Nakhaee
- Institutes of Neuropharmacology, Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, IR Iran
| | - Alireza Izadi
- Department of Medical Mycology and Parasitology, School of Medicine, Kerman University of Medical Sciences, Kerman, IR Iran
- Corresponding author: Alireza Izadi, Department of Medical Mycology and Parasitology, School of Medicine, Kerman University of Medical Sciences, Kerman, IR Iran. Tel: +98-3433221661, Fax: +98-3432480680, E-mail:
| |
Collapse
|
32
|
Muhammed M, Arvanitis M, Mylonakis E. Whole animal HTS of small molecules for antifungal compounds. Expert Opin Drug Discov 2015; 11:177-84. [PMID: 26593386 DOI: 10.1517/17460441.2016.1122591] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The high morbidity and mortality among patients with invasive fungal infections and the growing problem of fungal resistance have resulted in an urgent need for new antifungal agents. AREAS COVERED This review covers the importance of antifungal drug discovery with an emphasis on whole-animal high-throughput techniques. More specifically, the authors focus on Caenorhabditis elegans, as a substitute model host and discuss C. elegans as an alternative model host for the study of microbial pathogenesis and the identification of novel antifungal compounds. EXPERT OPINION There are significant advantages from using the substitute model host C. elegans in high-throughput drug discovery. The C. elegans-microbe model provides a whole animal system where host-pathogen interactions can be studied along with the evaluation of antimicrobial efficacy of compounds. This approach allows the study of compound characteristics, such as toxicity and solubility, during the initial screen and compounds discovered using C. elegans are affective in mammalian models.
Collapse
Affiliation(s)
- Maged Muhammed
- a Division of Infectious Diseases , Rhode Island Hospital , Providence , RI , USA.,b Warren Alpert Medical School of Brown University , Providence , RI , USA
| | - Marios Arvanitis
- c Department of Medicine , Boston Medical Center, Boston University , Boston , MA , USA
| | - Eleftherios Mylonakis
- a Division of Infectious Diseases , Rhode Island Hospital , Providence , RI , USA.,b Warren Alpert Medical School of Brown University , Providence , RI , USA
| |
Collapse
|
33
|
Marciano BE, Spalding C, Fitzgerald A, Mann D, Brown T, Osgood S, Yockey L, Darnell DN, Barnhart L, Daub J, Boris L, Rump AP, Anderson VL, Haney C, Kuhns DB, Rosenzweig SD, Kelly C, Zelazny A, Mason T, DeRavin SS, Kang E, Gallin JI, Malech HL, Olivier KN, Uzel G, Freeman AF, Heller T, Zerbe CS, Holland SM. Common severe infections in chronic granulomatous disease. Clin Infect Dis 2015; 60:1176-83. [PMID: 25537876 PMCID: PMC4400412 DOI: 10.1093/cid/ciu1154] [Citation(s) in RCA: 266] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/06/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Chronic granulomatous disease (CGD) is due to defective nicotinamide adenine dinucleotide phosphate oxidase activity and characterized by recurrent infections with a limited spectrum of bacteria and fungi as well as inflammatory complications. To understand the impact of common severe infections in CGD, we examined the records of 268 patients followed at a single center over 4 decades. METHODS All patients had confirmed diagnoses of CGD, and genotype was determined where possible. Medical records were excerpted into a standard format. Microbiologic analyses were restricted to Staphylococcus, Burkholderia, Serratia, Nocardia, and Aspergillus. RESULTS Aspergillus incidence was estimated at 2.6 cases per 100 patient-years; Burkholderia, 1.06 per 100 patient-years; Nocardia, 0.81 per 100 patient-years; Serratia, 0.98 per 100 patient-years, and severe Staphylococcus infection, 1.44 per 100 patient-years. Lung infection occurred in 87% of patients, whereas liver abscess occurred in 32%. Aspergillus incidence was 55% in the lower superoxide-producing quartiles (quartiles 1 and 2) but only 41% in the higher quartiles (rate ratio, <0.0001). Aspergillus and Serratia were somewhat more common in lower superoxide producing gp91phox deficiency. The median age at death has increased from 15.53 years before 1990 to 28.12 years in the last decade. Fungal infection carried a higher risk of mortality than bacterial infection and was the most common cause of death (55%). Gastrointestinal complications were not associated with either infection or mortality. CONCLUSIONS Fungal infections remain a major determinant of survival in CGD. X-linked patients generally had more severe disease, and this was generally in those with lower residual superoxide production. Survival in CGD has increased over the years, but infections are still major causes of morbidity and mortality.
Collapse
Affiliation(s)
- Beatriz E. Marciano
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Christine Spalding
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Alan Fitzgerald
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Daphne Mann
- Clinical Research Directorate/Clinical Monitoring Research Program
| | - Thomas Brown
- Clinical Research Directorate/Clinical Monitoring Research Program
| | - Sharon Osgood
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Lynne Yockey
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Dirk N. Darnell
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Lisa Barnhart
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Janine Daub
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Lisa Boris
- Clinical Research Directorate/Clinical Monitoring Research Program
| | - Amy P. Rump
- Clinical Research Directorate/Clinical Monitoring Research Program
| | - Victoria L. Anderson
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Carissa Haney
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Douglas B. Kuhns
- Clinical Services Program, Applied Developmental Research Directorate, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research
| | | | - Corin Kelly
- Laboratory of Host Defenses, National Institute for Allergy and Infectious Diseases, NIH, Rockville
| | - Adrian Zelazny
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda
| | - Tamika Mason
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Suk See DeRavin
- Laboratory of Host Defenses, National Institute for Allergy and Infectious Diseases, NIH, Rockville
| | - Elizabeth Kang
- Laboratory of Host Defenses, National Institute for Allergy and Infectious Diseases, NIH, Rockville
| | - John I. Gallin
- Laboratory of Host Defenses, National Institute for Allergy and Infectious Diseases, NIH, Rockville
| | - Harry L. Malech
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda
| | - Kenneth N. Olivier
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Gulbu Uzel
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Alexandra F. Freeman
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Theo Heller
- Liver Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland
| | - Christa S. Zerbe
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| | - Steven M. Holland
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda
| |
Collapse
|
34
|
Pana ZD, Kougia V, Roilides E. Therapeutic strategies for invasive fungal infections in neonatal and pediatric patients: an update. Expert Opin Pharmacother 2015; 16:693-710. [PMID: 25676454 DOI: 10.1517/14656566.2015.1013936] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Invasive fungal infections (IFIs) in neonatal and pediatric patients are still associated with high morbidity and mortality, increased length of hospital stay and high healthcare cost. Two key components are prerequisite to combat pediatric IFIs; first, definition of the 'at-risk' populations that could benefit the most from prophylactic treatment and second, prompt initiation of effective antifungal therapy. AREAS COVERED In this article, updated prevention and targeted therapeutic approaches for IFIs in neonates and immunocompromised children are reviewed. Furthermore, European and American guidelines concerning IFI treatment in neonates and children are compared. EXPERT OPINION IFIs in neonates and children present substantial differences from adults in respect to their epidemiology, pharmacokinetics of antifungal agents and dosing as well as absence of interventional Phase III and IV clinical trials for guidance of evidence-based decisions. In the therapeutic armamentarium of these age groups, although amphotericin B formulations remain widely indicated, azoles with broader spectrum activity as well as echinocandins have been added in the updated antifungal treatment algorithm. Recent European guidelines (ESCMID and ECIL) contain specific recommendations for pediatric patients with IFIs. In both age groups, definitive updated guidance for prophylaxis and more importantly targeted treatment need to be further evaluated by large, multicenter, randomized controlled trials.
Collapse
Affiliation(s)
- Zoi-Dorothea Pana
- Aristotle University School of Health Sciences, 3rd Department of Pediatrics, Infectious Diseases Unit, Faculty of Medicine , Thessaloniki , Greece
| | | | | |
Collapse
|
35
|
Wozniak KL, Olszewski MA, Wormley FL. Molecules at the interface of Cryptococcus and the host that determine disease susceptibility. Fungal Genet Biol 2014; 78:87-92. [PMID: 25445308 DOI: 10.1016/j.fgb.2014.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/22/2014] [Accepted: 10/25/2014] [Indexed: 10/24/2022]
Abstract
Cryptococcus neoformans and Cryptococcus gattii, the predominant etiological agents of cryptococcosis, are fungal pathogens that cause disease ranging from a mild pneumonia to life-threatening infections of the central nervous system (CNS). Resolution or exacerbation of Cryptococcus infection is determined following complex interactions of several host and pathogen derived factors. Alternatively, interactions between the host and pathogen may end in an impasse resulting in the establishment of a sub-clinical Cryptococcus infection. The current review addresses the delicate interaction between the host and Cryptococcus-derived molecules that determine resistance or susceptibility to infection. An emphasis will be placed on data highlighted at the recent 9th International Conference on Cryptococcus and Cryptococcosis (ICCC).
Collapse
Affiliation(s)
- Karen L Wozniak
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States; South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Michal A Olszewski
- Veterans Affairs Ann Arbor Health System, Ann Arbor, MI, United States; University of Michigan Medical School, Ann Arbor, MI, United States
| | - Floyd L Wormley
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States; South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States.
| |
Collapse
|
36
|
IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proc Natl Acad Sci U S A 2014; 111:3526-31. [PMID: 24550444 DOI: 10.1073/pnas.1322831111] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patients with chronic granulomatous disease (CGD) have a mutated NADPH complex resulting in defective production of reactive oxygen species; these patients can develop severe colitis and are highly susceptible to invasive fungal infection. In NADPH oxidase-deficient mice, autophagy is defective but inflammasome activation is present despite lack of reactive oxygen species production. However, whether these processes are mutually regulated in CGD and whether defective autophagy is clinically relevant in patients with CGD is unknown. Here, we demonstrate that macrophages from CGD mice and blood monocytes from CGD patients display minimal recruitment of microtubule-associated protein 1 light chain 3 (LC3) to phagosomes. This defect in autophagy results in increased IL-1β release. Blocking IL-1 with the receptor antagonist (anakinra) decreases neutrophil recruitment and T helper 17 responses and protects CGD mice from colitis and also from invasive aspergillosis. In addition to decreased inflammasome activation, anakinra restored autophagy in CGD mice in vivo, with increased Aspergillus-induced LC3 recruitment and increased expression of autophagy genes. Anakinra also increased Aspergillus-induced LC3 recruitment from 23% to 51% (P < 0.01) in vitro in monocytes from CGD patients. The clinical relevance of these findings was assessed by treating CGD patients who had severe colitis with IL-1 receptor blockade using anakinra. Anakinra treatment resulted in a rapid and sustained improvement in colitis. Thus, inflammation in CGD is due to IL-1-dependent mechanisms, such as decreased autophagy and increased inflammasome activation, which are linked pathological conditions in CGD that can be restored by IL-1 receptor blockade.
Collapse
|
37
|
Hirbod-Mobarakeh A, Aghamohammadi A, Rezaei N. Immunoglobulin class switch recombination deficiency type 1 or CD40 ligand deficiency: from bedside to bench and back again. Expert Rev Clin Immunol 2013; 10:91-105. [PMID: 24308834 DOI: 10.1586/1744666x.2014.864554] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The immunoglobulin class switch recombination deficiency or hyper-IgM syndrome is characterized by normal or elevated serum IgM and low serum levels of other immunoglobulins. Since the first reported patient with hyper-IgM, more than 200 patients with this phenotype resulted from CD40 ligand deficiency have been reported. However, in addition to this common finding, they presented with different manifestations like opportunistic infections, autoimmunity and malignancies each of them are worth a detailed look. In this review, we will focus on different underlying mechanisms of these presentations to review what we have learned from our patients. In the end, we will discuss different treatment options available for these patients using this knowledge.
Collapse
Affiliation(s)
- Armin Hirbod-Mobarakeh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
38
|
Rizzetto L, De Filippo C, Rivero D, Riccadonna S, Beltrame L, Cavalieri D. Systems biology of host-mycobiota interactions: dissecting Dectin-1 and Dectin-2 signalling in immune cells with DC-ATLAS. Immunobiology 2013; 218:1428-37. [PMID: 23932568 DOI: 10.1016/j.imbio.2013.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 07/02/2013] [Accepted: 07/06/2013] [Indexed: 01/04/2023]
Abstract
Modelling the networks sustaining the fruitful coexistence between fungi and their mammalian hosts is becoming increasingly important to control emerging fungal pathogens. The C-type lectins Dectin-1 and Dectin-2 are involved in host defense mechanisms against fungal infection driving inflammatory and adaptive immune responses and complement in containing fungal burdens. Recognizing carbohydrate structures in pathogens, their engagement induces maturation of dendritic cells (DCs) into potent immuno-stimulatory cells endowed with the capacity to efficiently prime T cells. Owing to these properties, Dectin-1 and Dectin-2 agonists are currently under investigation as promising adjuvants in vaccination procedures for the treatment of fungal infection. Thus, a detailed understanding of events' cascade specifically triggered in DCs upon engagement is of great interest in translational research. Here, we summarize the current knowledge on Dectin-1 and Dectin-2 signalling in DCs highlighting similarities and differences. Detailed maps are annotated, using the Biological Connection Markup Language (BCML) data model, and stored in DC-ATLAS, a versatile resource for the interpretation of high-throughput data generated perturbing the signalling network of DCs.
Collapse
Affiliation(s)
- Lisa Rizzetto
- Fondazione Edmund Mach, Research and Innovation Centre, San Michele all'Adige (TN), Italy
| | | | | | | | | | | |
Collapse
|
39
|
Targeting iron acquisition blocks infection with the fungal pathogens Aspergillus fumigatus and Fusarium oxysporum. PLoS Pathog 2013; 9:e1003436. [PMID: 23853581 PMCID: PMC3708856 DOI: 10.1371/journal.ppat.1003436] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 05/02/2013] [Indexed: 12/21/2022] Open
Abstract
Filamentous fungi are an important cause of pulmonary and systemic morbidity and mortality, and also cause corneal blindness and visual impairment worldwide. Utilizing in vitro neutrophil killing assays and a model of fungal infection of the cornea, we demonstrated that Dectin-1 dependent IL-6 production regulates expression of iron chelators, heme and siderophore binding proteins and hepcidin in infected mice. In addition, we show that human neutrophils synthesize lipocalin-1, which sequesters fungal siderophores, and that topical lipocalin-1 or lactoferrin restricts fungal growth in vivo. Conversely, we show that exogenous iron or the xenosiderophore deferroxamine enhances fungal growth in infected mice. By examining mutant Aspergillus and Fusarium strains, we found that fungal transcriptional responses to low iron levels and extracellular siderophores are essential for fungal growth during infection. Further, we showed that targeting fungal iron acquisition or siderophore biosynthesis by topical application of iron chelators or statins reduces fungal growth in the cornea by 60% and that dual therapy with the iron chelator deferiprone and statins further restricts fungal growth by 75%. Together, these studies identify specific host iron-chelating and fungal iron-acquisition mediators that regulate fungal growth, and demonstrate that therapeutic inhibition of fungal iron acquisition can be utilized to treat topical fungal infections.
Collapse
|
40
|
Meriden Z, Marr KA, Lederman HM, Illei PB, Villa K, Riedel S, Carroll KC, Zhang SX. Ochroconis gallopavainfection in a patient with chronic granulomatous disease: case report and review of the literature. Med Mycol 2012; 50:883-9. [DOI: 10.3109/13693786.2012.681075] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
41
|
Steele C, Wormley FL. Immunology of fungal infections: lessons learned from animal models. Curr Opin Microbiol 2012; 15:413-9. [PMID: 22763286 DOI: 10.1016/j.mib.2012.05.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/04/2012] [Accepted: 05/08/2012] [Indexed: 01/09/2023]
Abstract
The continuing AIDS epidemic coupled with increased usage of immunosuppressive drugs to prevent organ rejection or treat autoimmune diseases has resulted in an increase in individuals at risk for acquiring fungal diseases. These concerns highlight the need to elucidate mechanisms of inducing protective immune responses against fungal pathogens. Consequently, several experimental models of human mycoses have been developed to study these diseases. The availability of transgenic animal models allows for in-depth analysis of specific components, receptors, and signaling pathways that elicit protection against fungal diseases. This review focuses on recent advances in our understanding of immune responses to fungal infections gained using animal models.
Collapse
Affiliation(s)
- Chad Steele
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | | |
Collapse
|
42
|
Leal SM, Vareechon C, Cowden S, Cobb BA, Latgé JP, Momany M, Pearlman E. Fungal antioxidant pathways promote survival against neutrophils during infection. J Clin Invest 2012; 122:2482-98. [PMID: 22706306 DOI: 10.1172/jci63239] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/09/2012] [Indexed: 01/15/2023] Open
Abstract
Filamentous fungi are a common cause of blindness and visual impairment worldwide. Using both murine model systems and in vitro human neutrophils, we found that NADPH oxidase produced by neutrophils was essential to control the growth of Aspergillus and Fusarium fungi in the cornea. We demonstrated that neutrophil oxidant production and antifungal activity are dependent on CD18, but not on the β-glucan receptor dectin-1. We used mutant A. fumigatus strains to show that the reactive oxygen species-sensing transcription factor Yap1, superoxide dismutases, and the Yap1-regulated thioredoxin antioxidant pathway are each required for protection against neutrophil-mediated oxidation of hyphae as well as optimal survival of fungal hyphae in vivo. We also demonstrated that thioredoxin inhibition using the anticancer drug PX-12 increased the sensitivity of fungal hyphae to both H2O2- and neutrophil-mediated killing in vitro. Additionally, topical application of PX-12 significantly enhanced neutrophil-mediated fungal killing in infected mouse corneas. Cumulatively, our data reveal critical host oxidative and fungal anti-oxidative mediators that regulate hyphal survival during infection. Further, these findings also indicate that targeting fungal anti-oxidative defenses via PX-12 may represent an efficacious strategy for treating fungal infections.
Collapse
Affiliation(s)
- Sixto M Leal
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Unsinger J, Burnham CAD, McDonough J, Morre M, Prakash PS, Caldwell CC, Dunne WM, Hotchkiss RS. Interleukin-7 ameliorates immune dysfunction and improves survival in a 2-hit model of fungal sepsis. J Infect Dis 2012; 206:606-16. [PMID: 22693226 DOI: 10.1093/infdis/jis383] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Secondary hospital-acquired fungal infections are common in critically-ill patients and mortality remains high despite antimicrobial therapy. Interleukin-7 (IL-7) is a potent immunotherapeutic agent that improves host immunity and has shown efficacy in bacterial and viral models of infection. This study examined the ability of IL-7, which is currently in multiple clinical trials (including hepatitis and human immunodeficiency virus), to improve survival in a clinically relevant 2-hit model of fungal sepsis. METHODS Mice underwent cecal ligation and puncture to induce peritonitis. Four days later, surviving mice had intravenous injection with Candida albicans. Following Candida infection, mice were treated with IL-7 or saline control. The effect of IL-7 on host immunity and survival was recorded. RESULTS IL-7 ameliorated the loss of immune effector cells and increased lymphocyte functions, including activation, proliferation, expression of adhesion molecules, and interferon-γ production. These beneficial effects of IL-7 were associated with an increase in global immunity as reflected by an enhanced delayed type hypersensitivity response and a 1.7-fold improvement in survival. CONCLUSIONS The present findings showing that IL-7 improves survival in fungal sepsis, together with its previously reported efficacy in bacterial and viral infectious models, further supports its use as a novel immunotherapeutic in sepsis.
Collapse
Affiliation(s)
- Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Antachopoulos C, Katragkou A, Roilides E. Immunotherapy against invasive mold infections. Immunotherapy 2012; 4:107-20. [PMID: 22150004 DOI: 10.2217/imt.11.159] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Invasive infections due to filamentous fungi, such as Aspergillus spp., Zygomycetes, Scedosporium and Fusarium spp., cause significant morbidity and mortality in immunocompromised patients with hematological malignancies, recipients of hematopoietic stem cell transplants and those with chronic granulomatous disease. Despite antifungal therapy, the outcome is often unfavorable in these patients; immune restoration is considered as the cornerstone of successful treatment. Important aspects of human immune response against fungi include effective innate immune response expressed as effective phagocytic functions and a balance between proinflammatory and regulatory adaptive immune responses. A number of immunomodulatory approaches, including the administration of enhancing cytokines, adoptive transfer of pathogen-specific T lymphocytes and granulocyte transfusions have been investigated as adjunctive treatments against serious mold infections. Despite encouraging in vitro and in vivo data, current clinical evidence is not sufficient to allow firm recommendations on the use of these immunomodulatory modalities in serious mold infections.
Collapse
Affiliation(s)
- Charalampos Antachopoulos
- Infectious Diseases Unit, 3rd Department of Pediatrics, Aristotle University School of Medicine, Hippokration Hospital, Thessaloniki, Greece
| | | | | |
Collapse
|
45
|
C-type lectin receptors and cytokines in fungal immunity. Cytokine 2012; 58:89-99. [DOI: 10.1016/j.cyto.2011.08.031] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 08/20/2011] [Indexed: 12/29/2022]
|
46
|
Bader O, Weig MS, Gross U, Schon MP, Mempel M, Buhl T. A 32-Year-Old Man With Ulcerative Mucositis, Skin Lesions, and Nail Dystrophy. Clin Infect Dis 2012. [DOI: 10.1093/cid/cir943] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
47
|
Smeekens SP, Henriet SSV, Gresnigt MS, Joosten LAB, Hermans PWM, Netea MG, Warris A, van de Veerdonk FL. Low interleukin-17A production in response to fungal pathogens in patients with chronic granulomatous disease. J Interferon Cytokine Res 2011; 32:159-68. [PMID: 22191467 DOI: 10.1089/jir.2011.0046] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Patients with chronic granulomatous disease (CGD) cannot produce reactive oxygen species (ROS) due to a genetic defect in the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase system. Dysregulation of the L-tryptophan metabolism in mice with defects in NADPH oxidase, resulting in overproduction of interleukin (IL)-17, has been proposed to link ROS defects with hyperinflammation and susceptibility to pulmonary aspergillosis. In this study, we assessed the L-tryptophan metabolism and cytokine profiles in response to fungal pathogens in CGD patients. Peripheral blood mononuclear cells (PBMCs) from CGD patients showed increased production of IL-6, tumor necrosis factor-α, and interferon-γ upon stimulation with Aspergillus or Candida species, while IL-17A production was strikingly low compared with healthy controls. Indoleamine 2,3-dioxygenase expression was similar in PBMCs and neutrophils from CGD patients compared with healthy controls. Conversion of L-tryptophan to L-kynurenine, as measured by high-performance liquid chromatography, did not differ between CGD patients and healthy controls. Moreover, adding L-kynurenine to the cell cultures did not suppress fungal-induced IL-17A production. Although PBMCs of CGD patients produced more proinflammatory cytokines after stimulation, IL-17A production was strikingly low in response to fungal pathogens when compared with healthy controls. In addition, cells from CGD patients did not display a defective L-tryptophan metabolism.
Collapse
Affiliation(s)
- Sanne P Smeekens
- Department of Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Wozniak KL, Hardison S, Olszewski M, Wormley FL. Induction of protective immunity against cryptococcosis. Mycopathologia 2011; 173:387-94. [PMID: 22143898 DOI: 10.1007/s11046-011-9505-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 10/22/2011] [Indexed: 02/08/2023]
Abstract
Cryptococcus neoformans, the predominant etiological agent of cryptococcosis, is an encapsulated fungal pathogen that can cause life-threatening infections of the central nervous system in immune compromised individuals resulting in high morbidity and mortality. Consequently, several studies have endeavored to understand those mechanisms that mediate resistance and susceptibility to Cryptococcus infection. In this review, we will examine the contributions of various components of the innate and adaptive immune response toward protection against cryptococcosis. We will focus our discussion on studies presented at the 8th International Conference on Cryptococcus and Cryptococcosis (ICCC). Remarkable progress has been made toward our understanding of host immunity and susceptibility to cryptococcal infection and the potential for vaccine development.
Collapse
Affiliation(s)
- Karen L Wozniak
- Department of Biology, The University of Texas, San Antonio, TX 78249-0062, USA
| | | | | | | |
Collapse
|
49
|
Infections fongiques et déficits immunitaires héréditaires. Arch Pediatr 2011; 18 Suppl 1:S8-14. [DOI: 10.1016/s0929-693x(11)70935-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
Chronic pediatric pulmonary disease and primary humoral antibody based immune disease. Respir Med 2011; 105:511-4. [DOI: 10.1016/j.rmed.2010.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 11/08/2010] [Accepted: 11/12/2010] [Indexed: 11/22/2022]
|