1
|
FitzPatrick AM, Chin AT, Nirenberg S, Cunningham-Rundles C, Sacco K, Perlmutter J, Dasso JF, Tsalatsanis A, Maru J, Creech J, Walter JE, Hartog N, Izadi N, Palmucci M, Butte MJ, Loewy K, Relan A, Rider NL. Piloting an automated query and scoring system to facilitate APDS patient identification from health systems. Front Immunol 2025; 15:1508780. [PMID: 39906746 PMCID: PMC11790479 DOI: 10.3389/fimmu.2024.1508780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/20/2024] [Indexed: 02/06/2025] Open
Abstract
Introduction Patients with activated PI3Kδ syndrome (APDS) may elude diagnoses for nearly a decade. Methods to hasten the identification of these patients, and other patients with inborn errors of immunity (IEIs), are needed. We sought to demonstrate that querying electronic health record (EHR) systems by aggregating disparate signs into a risk score can identify these patients. Methods We developed a structured query language (SQL) script using literature-validated APDS-associated clinical concepts mapped to ICD-10-CM codes. We ran the query across EHRs from 7 large, US-based medical centers encompassing approximately 17 million patients. The query calculated an "APDS Score," which stratified risk for APDS for all individuals in these systems. Scores for all known patients with APDS (n=46) were compared. Results The query identified all but one known patient with APDS (98%; 45/46) as well as patients with other complex disease. Median score for all patients with APDS was 9 (IQR = 5.75; range 1-25). Sensitivity analysis suggested an optimal cutoff score of 7 (sensitivity = 0.70). Conclusion Disease-specific queries are a relatively simple method to foster patient identification across the rare-disease spectrum. Such methods are even more important for disorders such as APDS where an approved, pathway-specific treatment is available in the US.
Collapse
Affiliation(s)
| | - Aaron T. Chin
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sharon Nirenberg
- Division of Informatics and Data Architecture, Icahn School of Medicine, Departments of Scientific Computing and Data, Mount Sinai School of Medicine, New York, NY, United States
| | - Charlotte Cunningham-Rundles
- Division of Clinical Immunology, Icahn School of Medicine, Departments of Medicine and Pediatrics, Mount Sinai School of Medicine, New York, NY, United States
| | - Keith Sacco
- Department of Child Health, University of Arizona College of Medicine and Division of Pulmonology, Section of Allergy-Immunology, Phoenix Children’s Hospital, Phoenix, AZ, United States
| | | | - Joseph F. Dasso
- Department of Pediatric Allergy and Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, United States
| | - Athanasios Tsalatsanis
- Research Methodology and Biostatistics Core, Morsani College of Medicine, University of South Florida Health, St. Petersburg, FL, United States
| | - Jay Maru
- Management Analyst, Research Methodology and Biostatistics Core, Morsani College of Medicine, University of South Florida Health, St. Petersburg, FL, United States
| | - Jessica Creech
- Department of Pediatrics, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, United States
| | - Jolan E. Walter
- Department of Pediatric Allergy and Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, United States
| | - Nicholas Hartog
- Division of Allergy and Immunology, Helen DeVos Children’s Hospital and Corewell Health, Grand Rapids, Michigan State University College of Human Medicine, East Lansing, MI, United States
| | - Neema Izadi
- Division of Clinical Immunology and Allergy, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - Mandy Palmucci
- Division of Information Services, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - Manish J. Butte
- Department of Pediatrics and Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Klaus Loewy
- Department of Information Services, Texas Children’s Hospital, Houston, TX, United States
| | - Anurag Relan
- Pharming Healthcare, Inc., Warren, NJ, United States
| | - Nicholas L. Rider
- Department of Health Systems & Implementation Science, Virginia Tech Carilion School of Medicine, Division of Allergy-Immunology Carilion Clinic, Roanoke, VA, United States
| |
Collapse
|
2
|
Fernandes-Pineda, M, Zea-Vera AF. Lymphoproliferation and hyper-IgM as the first manifestation of activated phosphoinositide 3-kinase δ syndrome: A case report. BIOMEDICA : REVISTA DEL INSTITUTO NACIONAL DE SALUD 2024; 44:10-15. [PMID: 39836850 PMCID: PMC11949422 DOI: 10.7705/biomedica.7436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/06/2024] [Indexed: 01/23/2025]
Abstract
Activated phosphoinositide 3-kinase δ syndrome is an inborn error of immunity due to mutations within the genes responsible for encoding PI3Kδ subunits. This syndrome results in an excessive activation of the phosphoinositide 3-kinase signaling pathway. Gainof-function mutations in the gene PIK3R1 (encoding p85α, p55α, and p50α) lead to the development of the activated PI3K δ syndrome. Notably, the clinical presentations of this syndrome often closely resemble those of other primary immunodeficiencies. We present a case involving a 15-year-old male who displayed an immunological phenotype that bore a striking resemblance to hyper-IgM syndrome. Whole exome sequencing was undertaken to pinpoint the underlying genetic mutation. Our investigation successfully identified a heterozygous splice site mutation previously reported within the well-established hotspot of the PIK3R1 gene (GRCh37, c.1425+1 G>T). The diverse spectrum of inborn errors of immunity underscores the pivotal role of identifying gene mutations, particularly in patients presenting clinical manifestations spanning autoimmune disorders, lymphoproliferative conditions, and antibody deficiencies. Such precise genetic diagnoses hold significant potential for improving patient care and management.
Collapse
Affiliation(s)
- Mónica Fernandes-Pineda,
- Departamento de Medicina Interna, Universidad del Valle, Cali, ColombiaUniversidad del ValleUniversidad del ValleCaliColombia
| | - Andrés F. Zea-Vera
- Departamento de Microbiología, Facultad de Salud, Universidad del Valle, Cali, ColombiaUniversidad del ValleUniversidad del ValleCaliColombia
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disorders, National Institutes of Health, Bethesda, MD, USANational Institutes of HealthNational Institutes of HealthBethesdaUSA
| |
Collapse
|
3
|
IJspeert H, Dalm VASH, van Zelm MC, Edwards ESJ. Hyperactivation of the PI3K pathway in inborn errors of immunity: current understanding and therapeutic perspectives. IMMUNOTHERAPY ADVANCES 2024; 4:ltae009. [PMID: 39679264 PMCID: PMC11638974 DOI: 10.1093/immadv/ltae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/06/2024] [Indexed: 12/17/2024] Open
Abstract
The phosphoinositide-3-kinase (PI3K) pathway function is crucial to the normal development, differentiation, and function of immune cells including B, T, and NK cells. Following the description of two cohorts of patients with an inboirn error of immunity (also known as primary immunodeficiency) with gain-of-function variants in the PIK3CD gene a decade ago, the disease entity activated PI3K delta syndrome (APDS) was named. Since then, many more patients with PIK3CD variants have been described, and loss-of-function variants in PIK3R1 and PTEN have also been linked to APDS. Importantly, the availability of small molecules that inhibit the PI3K pathway has enabled targeted treatment of APDS patients. In this review, we define (i) the PI3K pathway and its role in inborn errors of immunity; (ii) the clinical and immunological presentation of APDS1 (PIK3CD GOF), APDS2 (PIK3R1 LOF), and related disorders; (iii) Diagnostic approaches to identify and functionally validate the genetic causes of disease; (iv) therapeutic interventions to target PI3K hyperactivation; and finally (v) current challenges and future perspectives that require attention for the optimal treatment of patients with APDS and APDS-L diseases.
Collapse
Affiliation(s)
- Hanna IJspeert
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Academic Center for Rare Immunological Diseases (RIDC), Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Virgil A S H Dalm
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Academic Center for Rare Immunological Diseases (RIDC), Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Division of Allergy & Clinical Immunology, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Menno C van Zelm
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, VIC, Australia
| | - Emily S J Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
4
|
Golec DP, Gazzinelli-Guimaraes P, Chauss D, Nagashima H, Yu K, Hill T, Nivelo L, Cannons JL, Perry J, Joshi I, Pereira N, Oliveira FMS, Cruz AC, Druey KM, Lack JB, Nutman TB, Villarino AV, O'Shea JJ, Afzali B, Schwartzberg PL. A PI3Kδ-Foxo1-FasL signaling amplification loop rewires CD4 + T helper cell signaling, differentiation and epigenetic remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620691. [PMID: 39803425 PMCID: PMC11722529 DOI: 10.1101/2024.10.28.620691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
While inputs regulating CD4+ T helper cell (Th) differentiation are well-defined, the integration of downstream signaling with transcriptional and epigenetic programs that define Th-lineage identity remain unresolved. PI3K signaling is a critical regulator of T cell function; activating mutations affecting PI3Kδ result in an immunodeficiency with multiple T cell defects. Using mice expressing activated-PI3Kδ, we found aberrant expression of proinflammatory Th1-signature genes under Th2-inducing conditions, both in vivo and in vitro. This dysregulation was driven by a robust PI3Kδ-IL-2-Foxo1 signaling loop, fueling Foxo1-inactivation, loss of Th2-lineage restriction, altered chromatin accessibility and global impairment of CTCF-DNA interactions. Surprisingly, ablation of Fasl, a Foxo1-repressed gene, restored normal Th2 differentiation, TCR signaling and CTCF expression. BioID revealed Fas interactions with TCR-signaling components, which were supported by Fas-mediated potentiation of TCR signaling. Our results highlight Fas-FasL signaling as a critical intermediate in phenotypes driven by activated-PI3Kδ, thereby linking two key pathways of immune dysregulation.
Collapse
Affiliation(s)
- Dominic P Golec
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Pedro Gazzinelli-Guimaraes
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Science, George Washington University, Washington, DC, USA
| | - Daniel Chauss
- Immunoregulation Section, NIDDK, NIH, Bethesda, MD, USA
| | | | - Kang Yu
- Lymphocyte Cell Biology Section, NIAMS, NIH, Bethesda, MD, USA
| | - Tom Hill
- NIAID Collaborative Bioinformatics Resource (NCBR), NIAID, NIH, Bethesda, MD, USA
| | - Luis Nivelo
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Jillian Perry
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Ilin Joshi
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Nicolas Pereira
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Fabrício Marcus Silva Oliveira
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Science, George Washington University, Washington, DC, USA
| | - Anthony C Cruz
- Lymphocyte Cell Biology Section, NIAMS, NIH, Bethesda, MD, USA
| | - Kirk M Druey
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Justin B Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), NIAID, NIH, Bethesda, MD, USA
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Alejandro V Villarino
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - John J O'Shea
- Lymphocyte Cell Biology Section, NIAMS, NIH, Bethesda, MD, USA
| | - Behdad Afzali
- Immunoregulation Section, NIDDK, NIH, Bethesda, MD, USA
| | | |
Collapse
|
5
|
刘 清, 彭 力, 黄 寒, 邓 亮, 钟 礼. [Activated phosphoinositide 3-kinase delta syndrome: report of seven cases]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:499-505. [PMID: 38802911 PMCID: PMC11135056 DOI: 10.7499/j.issn.1008-8830.2312065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/14/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVES To summarize the clinical data of 7 children with activated phosphoinositide 3-kinase delta syndrome (APDS) and enhance understanding of the disease. METHODS A retrospective analysis was conducted on clinical data of 7 APDS children admitted to Hunan Provincial People's Hospital from January 2019 to August 2023. RESULTS Among the 7 children (4 males, 3 females), the median age of onset was 30 months, and the median age at diagnosis was 101 months. Recurrent respiratory tract infections, hepatosplenomegaly, and multiple lymphadenopathy were observed in all 7 cases. Sepsis was observed in 5 cases, otitis media and multiple caries were observed in 3 cases, and diarrhea and joint pain were observed in 2 cases. Lymphoma and systemic lupus erythematosus were observed in 1 case each. Fiberoptic bronchoscopy was performed in 4 cases, revealing scattered nodular protrusions in the bronchial lumen. The most common respiratory pathogen was Streptococcus pneumoniae (4 cases). Six patients had a p.E1021K missense mutation, and one had a p.434-475del splice site mutation. CONCLUSIONS p.E1021K is the most common mutation site in APDS children. Children who present with one or more of the following symptoms: recurrent respiratory tract infections, hepatosplenomegaly, multiple lymphadenopathy, otitis media, and caries, and exhibit scattered nodular protrusions on fiberoptic bronchoscopy, should be vigilant for APDS. Citation:Chinese Journal of Contemporary Pediatrics, 2024, 26(5): 499-505.
Collapse
|
6
|
Li Q, Wang W, Wu Q, Zhou Q, Ying W, Hui X, Sun B, Hou J, Qian F, Wang X, Sun J. Phenotypic and Immunological Characterization of Patients with Activated PI3Kδ Syndrome 1 Presenting with Autoimmunity. J Clin Immunol 2024; 44:102. [PMID: 38634985 PMCID: PMC11026262 DOI: 10.1007/s10875-024-01705-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Autoimmunity is a significant feature of APDS1 patients. We aimed to explore the pathogenic immune phenotype and possible mechanisms of autoimmunity in APDS1 patients. METHODS The clinical records and laboratory data of 42 APDS1 patients were reviewed. Immunophenotypes were evaluated by multiparametric flow cytometry. Autoantibodies were detected via antigen microarray analysis. RESULTS A total of 42 children with PIK3CD gene mutations were enrolled. Immunological tests revealed increased proportions of effector memory cells (86%) and central memory cells (59%) among CD4+ T cells; increased proportions of effector memory cells (83%) and terminally differentiated effector memory T cells (38%) among CD8+ T cells. Fewer CD3+ T cells and B cells and higher IgG levels were reported in patients with autoimmunity. The proportion of Tregs was decreased, and the proportions of Th9, Tfh, and Tfr cells were increased in APDS1 patients. Among APDS1 patients, higher proportion of Th2 and Tfr cells were found in those with autoimmunity. The proportions of CD11c+ B and CD21lo B cells in patients with autoimmunity were significantly increased. Antigen microarray analysis revealed a wide range of IgG/IgM autoantibodies in patients with APDS1. In patients with autoimmunity, the proportion of Tfr might be positively correlated with autoantibodies. CONCLUSIONS The pathogenic immune phenotype of APDS1 patients included (1) deceased CD3+ T-cell and B-cell counts and increased IgG levels in patients with autoimmunity, (2) an imbalanced T helper cell subset, (3) increased proportions of autoreactive B cells, and (4) distinct autoantibody reactivities in patients with autoimmunity.
Collapse
Affiliation(s)
- Qifan Li
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Wenjie Wang
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Qi Wu
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Qinhua Zhou
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Wenjing Ying
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Xiaoying Hui
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Bijun Sun
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Jia Hou
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Feng Qian
- Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xiaochuan Wang
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 200032, China.
| | - Jinqiao Sun
- Department of Clinical Immunology, National Children Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
7
|
Abad MR, Alerany C, Alsina L, Granados EL, Neth O, Poveda JL, Rivière JG, Rodríguez-Gallego C, Tutein Nolthenius JB, Figueiredo R, Labazuy SS, Gil A. Determining value in the treatment of activated PI3Kδ syndrome in Spain: a multicriteria decision analysis from the perspective of key stakeholders. GLOBAL & REGIONAL HEALTH TECHNOLOGY ASSESSMENT 2024; 11:124-130. [PMID: 38784663 PMCID: PMC11113520 DOI: 10.33393/grhta.2024.3041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Activated phosphoinositide 3-kinase (PI3K)δ syndrome (APDS) is an ultra-rare inborn error of immunity (IEI) combining immunodeficiency and immune dysregulation. This study determined what represents value in APDS in Spain from a multidisciplinary perspective applying multicriteria decision analysis (MCDA) methodology. Methods A multidisciplinary committee of nine experts scored the evidence matrix. A specific framework for orphan drug evaluation in Spain and the weights assigned by a panel of 98 evaluators and decision-makers was used. Re-evaluation of scores was performed. Results APDS is considered a very severe disease with important unmet needs, including misdiagnosis and diagnostic delay. Current management is limited to treatment of symptoms with off-label use of therapies supported by limited evidence. Therapeutic benefit is partial, resulting in limited disease control. Haematopoietic stem cell transplantation (HSCT), the only potential curative alternative, is restricted to a reduced patient population and without evidence of long-term efficacy or safety. All options present a limited safety profile. Data on patients' quality of life are lacking. APDS is associated with high pharmacological, medical and indirect costs. Conclusions APDS is considered a severe disease, with limited understanding by key stakeholders of how treatment success is assessed in clinical practice, the serious impact that has on patients and the associated high economic burden. This study brings to light how MCDA methodology could represent a useful tool to complement current clinical and decision-making methods used by APDS experts and evaluators.
Collapse
Affiliation(s)
| | - Carmen Alerany
- Pharmacy Department, H.U. Vall d’ Hebron, Barcelona - Spain
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, H. Sant Joan de Déu. Institut de Recerca Sant Joan de Déu. Universitat de Barcelona, Barcelona - Spain
| | | | - Olaf Neth
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit H.U. Virgen del Rocío, Seville - Spain
| | | | - Jacques G. Rivière
- Paediatric Infectious Diseases and Immunodeficiencies Unit, H.U. Vall d’Hebron University Hospital, Barcelona - Spain
- Infection and Immunity in Pediatric Patients Research Group, Vall d’Hebron Institut de Recerca (VHIR), H.U. Vall d’Hebron, Barcelona - Spain
- Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Medical University of South Carolina, Barcelona - Spain
| | - Carlos Rodríguez-Gallego
- Department of Immunology, H.U. of Gran Canaria Dr. Negrin, Las Palmas de Gran Canaria, Gran Canaria - Spain
| | | | | | | | - Alicia Gil
- Omakase Consulting S.L., Barcelona - Spain
| |
Collapse
|
8
|
Sullivan NP, Maniam N, Maglione PJ. Interstitial lung diseases in inborn errors of immunity. Curr Opin Allergy Clin Immunol 2023; 23:500-506. [PMID: 37823528 DOI: 10.1097/aci.0000000000000951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
PURPOSE OF REVIEW Our goal is to review current understanding of interstitial lung disease (ILD) affecting patients with inborn errors of immunity (IEI). This includes understanding how IEI might predispose to and promote development or progression of ILD as well as how our growing understanding of IEI can help shape treatment of ILD in these patients. Additionally, by examining current knowledge of ILD in IEI, we hope to identify key knowledge gaps that can become focus of future investigative efforts. RECENT FINDINGS Recent identification of novel IEI associated with ILD and the latest reports examining treatment of ILD in IEI are included. Of noted interest, are recent clinical studies of immunomodulatory therapy for ILD in common variable immunodeficiency. SUMMARY ILD is a frequent complication found in many IEI. This article provides a guide to identifying manifestations of ILD in IEI. We review a broad spectrum of IEI that develop ILD, including antibody deficiency and immune dysregulation disorders that promote autoimmunity and autoinflammation. This work integrates clinical information with molecular mechanisms of disease and diagnostic assessments to provide an expedient overview of a clinically relevant and expanding topic.
Collapse
Affiliation(s)
| | - Nivethietha Maniam
- Section of Pulmonary, Allergy, Sleep and Critical Care Medicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Paul J Maglione
- Section of Pulmonary, Allergy, Sleep and Critical Care Medicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Vanselow S, Hanitsch L, Hauck F, Körholz J, Maccari ME, Meinhardt A, Sogkas G, Schuetz C, Grimbacher B. Future Directions in the Diagnosis and Treatment of APDS and IEI: a Survey of German IEI Centers. Front Immunol 2023; 14:1279652. [PMID: 37868971 PMCID: PMC10588788 DOI: 10.3389/fimmu.2023.1279652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction The diagnosis and treatment of inborn errors of immunity (IEI) is a major challenge as the individual conditions are rare and often characterized by a variety of symptoms, which are often non disease-specific. Ideally, patients are treated in dedicated centers by physicians who specialize in the management of primary immune disorders. In this study, we used the example of Activated PI3Kδ syndrome (APDS), a rare IEI with an estimated prevalence of 1:1,000,000. We conducted surveys by questionnaire and interviewed physicians at different IEI centers in Germany. Methods We queried structural aspects of IEI care in Germany, diagnostic procedures in IEI care (including molecular diagnostics), distribution of APDS patients, APDS symptoms and severity, treatment algorithms in APDS, the role of stem cell transplantation and targeted therapies in IEI with focus on APDS. We were especially interested in how genetic diagnostics may influence treatment decisions, e.g. with regard to targeted therapies. Results/discussion Most centers care for both pediatric and adult patients. A total of 28 APDS patients are currently being treated at the centers we surveyed. Patient journeys vary considerably, as does severity of disease. Genetic diagnosis continues to gain importance - whole genome sequencing is likely to become routine in IEI in the next few years. According to the experts interviewed, stem cell transplantation and - with new molecules being approved - targeted therapies, will gain in importance for the treatment of APDS and IEI in general.
Collapse
Affiliation(s)
- Sven Vanselow
- Infill Healthcare Communication, Königswinter, Germany
| | - Leif Hanitsch
- Institute of Medical Immunology, Institute of Occupational Medicine, Charité – University Medicine Berlin, corporate member of Freie University, Berlin and Humboldt-University of Berlin, Berlin, Germany
| | - Fabian Hauck
- Department of Pediatric Immunology and Rheumatology, Dr. Von Hauner Children’s Hospital, Ludwig-Maximilians-Universität (LMU) Munich University Hospital, Munich, Germany
| | - Julia Körholz
- Department of Pediatrics, University Hospital Carl Gustav Carus, Dresden, Germany
- University Center for Rare Diseases, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Maria-Elena Maccari
- Center for Chronic Immunodeficiency, University of Freiburg Medical Center, Freiburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Andrea Meinhardt
- Center for Pediatrics and Adolescent Medicine, Department of Pediatric Oncology, Hematology and Immunodeficiencies, University Hospital Giessen, Giessen, Germany
| | - Georgios Sogkas
- Clinic for Rheumatology and Immunology, Center for Internal Medicine, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Catharina Schuetz
- Department of Pediatrics, University Hospital Carl Gustav Carus, Dresden, Germany
- University Center for Rare Diseases, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Bodo Grimbacher
- Center for Chronic Immunodeficiency, University of Freiburg Medical Center, Freiburg, Germany
| |
Collapse
|
10
|
Maccari ME, Wolkewitz M, Schwab C, Lorenzini T, Leiding JW, Aladjdi N, Abolhassani H, Abou-Chahla W, Aiuti A, Azarnoush S, Baris S, Barlogis V, Barzaghi F, Baumann U, Bloomfield M, Bohynikova N, Bodet D, Boutboul D, Bucciol G, Buckland MS, Burns SO, Cancrini C, Cathébras P, Cavazzana M, Cheminant M, Chinello M, Ciznar P, Coulter TI, D'Aveni M, Ekwall O, Eric Z, Eren E, Fasth A, Frange P, Fournier B, Garcia-Prat M, Gardembas M, Geier C, Ghosh S, Goda V, Hammarström L, Hauck F, Heeg M, Heropolitanska-Pliszka E, Hilfanova A, Jolles S, Karakoc-Aydiner E, Kindle GR, Kiykim A, Klemann C, Koletsi P, Koltan S, Kondratenko I, Körholz J, Krüger R, Jeziorski E, Levy R, Le Guenno G, Lefevre G, Lougaris V, Marzollo A, Mahlaoui N, Malphettes M, Meinhardt A, Merlin E, Meyts I, Milota T, Moreira F, Moshous D, Mukhina A, Neth O, Neubert J, Neven B, Nieters A, Nove-Josserand R, Oksenhendler E, Ozen A, Olbrich P, Perlat A, Pac M, Schmid JP, Pacillo L, Parra-Martinez A, Paschenko O, Pellier I, Sefer AP, Plebani A, Plantaz D, Prader S, Raffray L, Ritterbusch H, Riviere JG, Rivalta B, Rusch S, Sakovich I, Savic S, Scheible R, Schleinitz N, Schuetz C, Schulz A, et alMaccari ME, Wolkewitz M, Schwab C, Lorenzini T, Leiding JW, Aladjdi N, Abolhassani H, Abou-Chahla W, Aiuti A, Azarnoush S, Baris S, Barlogis V, Barzaghi F, Baumann U, Bloomfield M, Bohynikova N, Bodet D, Boutboul D, Bucciol G, Buckland MS, Burns SO, Cancrini C, Cathébras P, Cavazzana M, Cheminant M, Chinello M, Ciznar P, Coulter TI, D'Aveni M, Ekwall O, Eric Z, Eren E, Fasth A, Frange P, Fournier B, Garcia-Prat M, Gardembas M, Geier C, Ghosh S, Goda V, Hammarström L, Hauck F, Heeg M, Heropolitanska-Pliszka E, Hilfanova A, Jolles S, Karakoc-Aydiner E, Kindle GR, Kiykim A, Klemann C, Koletsi P, Koltan S, Kondratenko I, Körholz J, Krüger R, Jeziorski E, Levy R, Le Guenno G, Lefevre G, Lougaris V, Marzollo A, Mahlaoui N, Malphettes M, Meinhardt A, Merlin E, Meyts I, Milota T, Moreira F, Moshous D, Mukhina A, Neth O, Neubert J, Neven B, Nieters A, Nove-Josserand R, Oksenhendler E, Ozen A, Olbrich P, Perlat A, Pac M, Schmid JP, Pacillo L, Parra-Martinez A, Paschenko O, Pellier I, Sefer AP, Plebani A, Plantaz D, Prader S, Raffray L, Ritterbusch H, Riviere JG, Rivalta B, Rusch S, Sakovich I, Savic S, Scheible R, Schleinitz N, Schuetz C, Schulz A, Sediva A, Semeraro M, Sharapova SO, Shcherbina A, Slatter MA, Sogkas G, Soler-Palacin P, Speckmann C, Stephan JL, Suarez F, Tommasini A, Trück J, Uhlmann A, van Aerde KJ, van Montfrans J, von Bernuth H, Warnatz K, Williams T, Worth AJJ, Ip W, Picard C, Catherinot E, Nademi Z, Grimbacher B, Forbes Satter LR, Kracker S, Chandra A, Condliffe AM, Ehl S. Activated phosphoinositide 3-kinase δ syndrome: Update from the ESID Registry and comparison with other autoimmune-lymphoproliferative inborn errors of immunity. J Allergy Clin Immunol 2023; 152:984-996.e10. [PMID: 37390899 DOI: 10.1016/j.jaci.2023.06.015] [Show More Authors] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND Activated phosphoinositide-3-kinase δ syndrome (APDS) is an inborn error of immunity (IEI) with infection susceptibility and immune dysregulation, clinically overlapping with other conditions. Management depends on disease evolution, but predictors of severe disease are lacking. OBJECTIVES This study sought to report the extended spectrum of disease manifestations in APDS1 versus APDS2; compare these to CTLA4 deficiency, NFKB1 deficiency, and STAT3 gain-of-function (GOF) disease; and identify predictors of severity in APDS. METHODS Data was collected from the ESID (European Society for Immunodeficiencies)-APDS registry and was compared with published cohorts of the other IEIs. RESULTS The analysis of 170 patients with APDS outlines high penetrance and early onset of APDS compared to the other IEIs. The large clinical heterogeneity even in individuals with the same PIK3CD variant E1021K illustrates how poorly the genotype predicts the disease phenotype and course. The high clinical overlap between APDS and the other investigated IEIs suggests relevant pathophysiological convergence of the affected pathways. Preferentially affected organ systems indicate specific pathophysiology: bronchiectasis is typical of APDS1; interstitial lung disease and enteropathy are more common in STAT3 GOF and CTLA4 deficiency. Endocrinopathies are most frequent in STAT3 GOF, but growth impairment is also common, particularly in APDS2. Early clinical presentation is a risk factor for severe disease in APDS. CONCLUSIONS APDS illustrates how a single genetic variant can result in a diverse autoimmune-lymphoproliferative phenotype. Overlap with other IEIs is substantial. Some specific features distinguish APDS1 from APDS2. Early onset is a risk factor for severe disease course calling for specific treatment studies in younger patients.
Collapse
Affiliation(s)
- Maria Elena Maccari
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Martin Wolkewitz
- Institute of Medical Biometry and Statistics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Schwab
- Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tiziana Lorenzini
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Pediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, University of Brescia and ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Nathalie Aladjdi
- Pediatric Haemato-Immunology, Clinical Investigation Center (CIC) 1401, Institut National de la Santé et de la Recherche Médicale (INSERM) Centre d'Investigation Clinique Pluridisciplinaire (CICP), Bordeaux University Hospital and Centre de Reference National des Cytopenies Auto-immunoes de l'Enfant (CEREVANCE), Bordeaux, France
| | - Hassan Abolhassani
- Division of Clinical Immunology, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Wadih Abou-Chahla
- Department of Pediatric Hematology, Jeanne de Flandre Hospital, Centre Hospitalier Universitaire (CHU), Lille, France
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (Sr-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Saba Azarnoush
- Pediatric Hematology and Immunology Unit, Robert Debré Hospital, Paris, France
| | - Safa Baris
- Pediatric Allergy and Immunology, Faculty of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Vincent Barlogis
- Pediatric Hematology, Immunology and Oncology, Aix-Marseille Université, Marseille, France
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (Sr-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Ulrich Baumann
- Pediatric Pulmonology, Allergy, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Marketa Bloomfield
- Department of Immunology, Motol University Hospital, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nadezda Bohynikova
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
| | - Damien Bodet
- Department of Pediatric Hematology and Oncology, University Hospital of Caen, Caen, France
| | - David Boutboul
- Clinical Immunology Department, Hôpital Saint-Louis, Paris, France
| | - Giorgia Bucciol
- Departments of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Microbiology, Immunology, and Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Matthew S Buckland
- Barts Health National Health Service Trust, London, United Kingdom; Molecular and Cellular Immunology Section, Immunity and Inflammation Department, Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, London, United Kingdom; Department of Immunology, Royal Free London National Health Service Foundation Trust, London, United Kingdom
| | - Caterina Cancrini
- Department of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy; Research and Clinical Unit of Primary Immunodeficiencies, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | | | - Marina Cavazzana
- Imagine Institute, INSERM U1163, Institut Imagine, Université Paris Cité, Paris, France; Biotherapy Department, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France; Biotherapy Clinical Investigation Center Groupe Hospitalier Centre, AP-HP, INSERM, Paris, France
| | - Morgane Cheminant
- Imagine Institute, INSERM U1163, Institut Imagine, Université Paris Cité, Paris, France; Service d'Hématologie Adulte, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France
| | - Matteo Chinello
- Pediatric Hematology Oncology, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Peter Ciznar
- Pediatric Department, Comenius University Medical Faculty, Bratislava, Slovakia
| | - Tanya I Coulter
- Belfast Health and Social Care Trust, Ireland, United Kingdom
| | - Maud D'Aveni
- Department of Hematology, Nancy University Hospital, Université de Lorraine, Nancy, France; UMR 7365, Centre National de la Recherche Scientifique, Ingénierie Moléculaire et Physiopathologie Articulaire, Université de Lorraine, Nancy, France
| | - Olov Ekwall
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Rheumatology and Inflammation Research, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Zelimir Eric
- University Clinical Centre of the Republic of Srpska, Republic of Srpska, Bosnia and Herzegovina
| | - Efrem Eren
- University Hospital Southampton, Southampton, United Kingdom
| | - Anders Fasth
- Department of Pediatrics, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Medicine, Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Pierre Frange
- Unité de Recherche Propre 7328, Fédération pour l'Étude et évaluation des Thérapeutiques intra-UtérineS (FETUS), Institut Imagine, Université Paris Cité, Paris, France; Laboratory of Clinical Microbiology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France
| | - Benjamin Fournier
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France
| | - Marina Garcia-Prat
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Christoph Geier
- Department of Rheumatology and Clinical Immunology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sujal Ghosh
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University-University Hospital Düsseldorf, Düsseldorf, Germany
| | - Vera Goda
- Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Lennart Hammarström
- Division of Clinical Immunology, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Anna Hilfanova
- Department of Pediatrics, Immunology, Infectious and Rare Diseases, European Medical School, International European University, Kyiv, Ukraine
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, United Kingdom
| | - Elif Karakoc-Aydiner
- Pediatric Allergy and Immunology, Faculty of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Gerhard R Kindle
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Centre for Biobanking FREEZE, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ayca Kiykim
- Pediatric Allergy and Immunology, Istanbul University Cerrahpasa Medical Faculty, Istanbul, Turkey
| | - Christian Klemann
- Departments of Human Genetics, Hannover Medical School, Hannover, Germany; Department of Pediatric Immunology, Rheumatology, & Infectiology, Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Patra Koletsi
- Department of Pediatrics, Penteli Children's Hospital, Athens, Greece
| | - Sylwia Koltan
- Department of Paediatric Haematology and Oncology, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Irina Kondratenko
- Russian Clinical Childrens Hospital, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Julia Körholz
- Department of Pediatrics, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Renate Krüger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Berlin Institute of Health, Berlin, Germany
| | - Eric Jeziorski
- General Pediatrics, CHU Montpellier, Montpellier, France; Pathogenesis and Control of Chronic Infections, INSERM, Université de Montpellier, Montpellier, France
| | - Romain Levy
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France
| | - Guillaume Le Guenno
- Department of Internal Medicine, Hôpital d'Estaing, Clermont-Ferrand, France
| | - Guillaume Lefevre
- CHU Lille, Institut d'Immunologie and University of Lille, Lille, France; Inserm U995, LIRIC-Lille Inflammation Research International Center, Lille, France
| | - Vassilios Lougaris
- Pediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, University of Brescia and ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology, and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Nizar Mahlaoui
- Pediatric Immunology-Hematology and Rheumatology Unit, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France; Necker Enfants Malades University Hospital, AP-HP, French National Reference Center for Primary Immune Deficiencies (CEREDIH), Paris Université Cité, Paris, France
| | | | - Andrea Meinhardt
- Center for Pediatrics and Adolescent Medicine, Department of Pediatric Hematology and Oncology, Medical Center, University Hospital Giessen, Giessen, Germany
| | - Etienne Merlin
- Department of Pediatrics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Isabelle Meyts
- Departments of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Microbiology, Immunology, and Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Tomas Milota
- Department of Immunology, Motol University Hospital, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Fernando Moreira
- Department of Immunology, Royal Free London National Health Service Foundation Trust, London, United Kingdom
| | - Despina Moshous
- Laboratories of Dynamique du Génome et Système Immunitaire, Institut Imagine, Université Paris Cité, Paris, France; Pediatric Immunology-Hematology and Rheumatology Unit, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France; Necker Enfants Malades University Hospital, AP-HP, French National Reference Center for Primary Immune Deficiencies (CEREDIH), Paris Université Cité, Paris, France
| | - Anna Mukhina
- Department of Immunology, Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Olaf Neth
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Red de Investigación Translacional en Infectología Pediátrica, Seville, Spain
| | - Jennifer Neubert
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University-University Hospital Düsseldorf, Düsseldorf, Germany
| | - Benedicte Neven
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Institut Imagine, Université Paris Cité, Paris, France; Pediatric Immunology-Hematology and Rheumatology Unit, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France
| | - Alexandra Nieters
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Centre for Biobanking FREEZE, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | - Ahmet Ozen
- Pediatric Allergy and Immunology, Faculty of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey; Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Peter Olbrich
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Consejo Superior de Investigaciones Cientificas, Red de Investigación Translacional en Infectología Pediátrica, Seville, Spain
| | | | - Malgorzata Pac
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
| | - Jana Pachlopnik Schmid
- Division of Immunology, University Children's Hospital Zurich, Zurich, Switzerland; Children's Research Center, Zurich, Switzerland
| | - Lucia Pacillo
- Department of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy; Research and Clinical Unit of Primary Immunodeficiencies, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Alba Parra-Martinez
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Olga Paschenko
- Russian Clinical Childrens Hospital, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Asena Pinar Sefer
- Pediatric Allergy and Immunology, Faculty of Medicine, Marmara University, Istanbul, Turkey; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Alessandro Plebani
- Pediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, University of Brescia and ASST-Spedali Civili of Brescia, Brescia, Italy
| | - Dominique Plantaz
- Unit of Pediatric Immuno Hemato and Oncology, University Hospital Centre of Grenoble, Grenoble, France
| | - Seraina Prader
- Division of Immunology, University Children's Hospital Zurich, Zurich, Switzerland; Children's Research Center, Zurich, Switzerland
| | - Loic Raffray
- Internal Medicine Department, Felix Guyon University Hospital, Saint Denis, La Réunion, France; Mixed Research Unit (UMR) "Infectious Processes in Tropical Island Environments", La Réunion, France
| | - Henrike Ritterbusch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jacques G Riviere
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Beatrice Rivalta
- Department of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy; Research and Clinical Unit of Primary Immunodeficiencies, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Stephan Rusch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Inga Sakovich
- Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology, Minsk, Belarus
| | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom; Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, United Kingdom
| | - Raphael Scheible
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for AI and Informatics in Medicine, University Hospital Rechts der Isar, Technical University Munich, Munich, Germany
| | - Nicolas Schleinitz
- Département de Médecine Interne, Timone Hospital, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille Université, Marseille, France
| | - Catharina Schuetz
- Department of Pediatrics, Universitätsklinikum Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Anna Sediva
- Department of Immunology, Motol University Hospital, Prague, Czech Republic; Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michaela Semeraro
- Clinical Investigation Center (CIC) 1419, Necker-Enfants Malades Hospital, AP-HP, Groupe Hospitalier Paris Centre, Paris, France; EA7323 Pediatric and Perinatal Drug Evaluation and Pharmacology Research Unit, Université Paris Cité, Paris, France
| | - Svetlana O Sharapova
- Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology, Minsk, Belarus
| | - Anna Shcherbina
- Department of Immunology, Research and Clinical Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mary A Slatter
- Great North Children' s Hospital, Newcastle upon Tyne, United Kingdom; Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Georgios Sogkas
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Carsten Speckmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jean-Louis Stephan
- Department of Pediatrics, North Hospital, University Hospital of Saint Etienne, Saint-Etienne, France; University Jean Monnet, Saint-Etienne, France
| | - Felipe Suarez
- Imagine Institute, INSERM U1163, Institut Imagine, Université Paris Cité, Paris, France; Service d'Hématologie Adulte, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France
| | - Alberto Tommasini
- Department of Medical Sciences, University of Trieste, Trieste, Italy; Institute for Maternal and Child Health, IRCCS Burlo Garofalo, Trieste, Italy
| | - Johannes Trück
- Division of Immunology, University Children's Hospital Zurich, Zurich, Switzerland; Children's Research Center, Zurich, Switzerland
| | - Annette Uhlmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Clinical Trials Unit, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Koen J van Aerde
- Amalia Children's Hospital, Radboudumc, Nijmegen, The Netherlands
| | - Joris van Montfrans
- Department of Pediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Horst von Bernuth
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Berlin Institute of Health, Berlin, Germany
| | - Klaus Warnatz
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Rheumatology and Clinical Immunology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Tony Williams
- University Hospital Southampton, Southampton, United Kingdom
| | - Austen J J Worth
- Great Ormond Street Hospital for Children, University College London, London, United Kingdom
| | - Winnie Ip
- Great Ormond Street Institute of Child Health, London, United Kingdom; Great Ormond Street Hospital for Children, University College London, London, United Kingdom
| | - Capucine Picard
- Lymphocyte Activation and Susceptibility to EBV Infection, Institut Imagine, Université Paris Cité, Paris, France; Pediatric Immunology-Hematology and Rheumatology Unit, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France; Study Center for Primary Immunodeficiencies, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Centre, Paris, France; Necker Enfants Malades University Hospital, AP-HP, French National Reference Center for Primary Immune Deficiencies (CEREDIH), Paris Université Cité, Paris, France
| | | | - Zohreh Nademi
- Great North Children' s Hospital, Newcastle upon Tyne, United Kingdom; Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Rheumatology and Clinical Immunology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; DZIF-German Center for Infection Research, Satellite Center Freiburg, Freiburg, Germany; CIBSS-Centre for Integrative Biological Signalling Studies, Albert-Ludwigs University, Freiburg, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Lisa R Forbes Satter
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex; William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, Tex
| | - Sven Kracker
- Human Lymphohematopoiesis, INSERM Unité Mixte de Recherche (UMR) 1163, Institut Imagine, Université Paris Cité, Paris, France; Université Paris Cité, Paris, France
| | - Anita Chandra
- Department of Clinical Immunology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Sheffield, United Kingdom
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Lam KF, Chan S, Kam LS, Kwok J, Lee PPW, Lam DSY. Endobronchial mucosal nodules and actinomycosis in a child with activated phosphatidylinositol 3-kinase delta syndrome (APDS). Respir Med Case Rep 2023; 45:101909. [PMID: 37635731 PMCID: PMC10458975 DOI: 10.1016/j.rmcr.2023.101909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/11/2023] [Indexed: 08/29/2023] Open
Abstract
In this report, we describe a case of a 5-year-old girl with poor growth and unresolving pneumonia. Bronchoscopy showed numerous endobronchial mucosal nodules, consisting of dense lymphoid infiltrates. Bacterial culture of the nodule biopsy suggested endobronchial actinomycosis. Genetic test confirmed the diagnosis of APDS.
Collapse
Affiliation(s)
- Kwan Fung Lam
- Department of Paediatrics and Adolescent Medicine, Caritas Medical Centre, Hong Kong, China
| | - Shun Chan
- Department of Surgery, Tuen Mun Hospital, Hong Kong, China
| | - Lok Sang Kam
- Department of Clinical Pathology, Tuen Mun Hospital, Hong Kong, China
| | - Janette Kwok
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - Pamela Pui-Wah Lee
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - David Shu Yan Lam
- Department of Paediatrics and Adolescent Medicine, Tuen Mun Hospital, Hong Kong, China
| |
Collapse
|
12
|
Vanselow S, Wahn V, Schuetz C. Activated PI3Kδ syndrome - reviewing challenges in diagnosis and treatment. Front Immunol 2023; 14:1208567. [PMID: 37600808 PMCID: PMC10432830 DOI: 10.3389/fimmu.2023.1208567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/04/2023] [Indexed: 08/22/2023] Open
Abstract
Activated PI3Kδ syndrome (APDS) is a rare inborn error of immunity (IEI) characterized primarily by frequent infections, lymphoproliferation and autoimmunity. Since its initial description in 2013, APDS has become part of the growing group of nearly 500 IEIs affecting various components of the immune system. The two subtypes of APDS - APDS1 and APDS2 - are caused by variants in the PIK3CD and PIK3R1 genes, respectively. Due to the rarity of the disease and the heterogeneous clinical picture, many patients are not diagnosed until years after symptom onset. Another challenge is the large number of PIK3CD and PIK3R1 variants whose functional significance for developing APDS is inconclusive. Treatment of APDS has so far been mostly symptom-oriented with immunoglobulin replacement therapy, immunosuppressive therapies and antibiotic or antiviral prophylaxes. Additionally, allogeneic stem cell transplantation as well as new targeted therapies are options targeting the root cause that may improve patients' quality of life and life expectancy. However, the clinical course of the disease is difficult to predict which complicates the choice of appropriate therapies. This review article discusses diagnostic procedures and current and future treatment options, and highlights the difficulties that physicians, patients and their caretakers face in managing this complex disease. This article is based on cohort studies, the German and US guidelines on the management of primary immunodeficiencies as well as on published experience with diagnosis and compiled treatment experience for APDS.
Collapse
Affiliation(s)
- Sven Vanselow
- Infill Healthcare Communication, Königswinter, Germany
| | - Volker Wahn
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine at Charité University Hospital Berlin, Berlin, Germany
| | - Catharina Schuetz
- Medical Faculty of The Technical University (TU) Dresden, Department of Pediatrics, University Hospital Carl Gustav Carus, Dresden, Germany
- University Center for Rare Diseases, University Hospital Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
13
|
Zhang Y, Ma Z, Wang Y, Feng X, An Z. Phosphatidylinositol 3 kinase inhibitor-related pneumonitis: a systematic review and meta-analysis. Expert Rev Clin Pharmacol 2023; 16:855-863. [PMID: 37489925 DOI: 10.1080/17512433.2023.2238602] [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: 04/16/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
INTRODUCTION Serious phosphatidylinositol 3 kinase (PI3K) inhibitor-related pneumonitis has raised clinical concerns, and integrated data for this condition are lacking. METHODS Randomized controlled trials (RCTs) comparing PI3K inhibitor therapy with control treatments from electronic databases and registrations were searched from inception to 1 April 20231 April 2023seven1 April 2023. The outcomes of our study were the incidence and risk of all-grade and grade ≥ 3 PI3K inhibitor-associated pneumonitis compared with controls. RESULTS The meta-analysis included 13 studies comprising 3916 patients. The incidence of all-grade and grade ≥ 3 pneumonitis was 3.7% (82/2210) and 3.0% (35/1162) in patients treated with PI3K inhibitors. PI3K inhibitors significantly increased the risk of all-grade and grade ≥ 3 pneumonitis compared with controls (RR 5.63, 95% CI [2.97, 10.65], P < 0.00001; RR 6.85, 95% CI [2.45, 19.11], P = 0.0002, respectively) with no significant heterogeneity across studies. In terms of different PI3K inhibitors, copanlisib and idelalisib significantly increased the risk of pneumonitis compared to controls (RR 4.99, 95% CI [1.19, 21.01], P = 0.03; RR 5.53, 95% CI [2.35, 13.01], P < 0.0001, respectively). CONCLUSION PI3K inhibitors significantly increased the risk of pneumonitis compared with controls, and most cases are severe or even life-threatening. PROSPERO REGISTRATION NUMBER CRD42022318878.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Pharmacy, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Zhuo Ma
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yushu Wang
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Pharmacy, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xin Feng
- Department of Pharmacy, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Zhuoling An
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Mancuso G, Bechi Genzano C, Fierabracci A, Fousteri G. Type 1 diabetes and inborn errors of immunity: Complete strangers or 2 sides of the same coin? J Allergy Clin Immunol 2023; 151:1429-1447. [PMID: 37097271 DOI: 10.1016/j.jaci.2023.03.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/26/2023]
Abstract
Type 1 diabetes (T1D) is a polygenic disease and does not follow a mendelian pattern. Inborn errors of immunity (IEIs), on the other hand, are caused by damaging germline variants, suggesting that T1D and IEIs have nothing in common. Some IEIs, resulting from mutations in genes regulating regulatory T-cell homeostasis, are associated with elevated incidence of T1D. The genetic spectrum of IEIs is gradually being unraveled; consequently, molecular pathways underlying human monogenic autoimmunity are being identified. There is an appreciable overlap between some of these pathways and the genetic variants that determine T1D susceptibility, suggesting that after all, IEI and T1D are 2 sides of the same coin. The study of monogenic IEIs with a variable incidence of T1D has the potential to provide crucial insights into the mechanisms leading to T1D. These insights contribute to the definition of T1D endotypes and explain disease heterogeneity. In this review, we discuss the interconnected pathogenic pathways of autoimmunity, β-cell function, and primary immunodeficiency. We also examine the role of environmental factors in disease penetrance as well as the circumstantial evidence of IEI drugs in preventing and curing T1D in individuals with IEIs, suggesting the repositioning of these drugs also for T1D therapy.
Collapse
Affiliation(s)
- Gaia Mancuso
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camillo Bechi Genzano
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | | | - Georgia Fousteri
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
15
|
Begg M, Amour A, Jarvis E, Tang T, Franco SS, Want A, Beerahee M, Fernando D, Karkera Y, Sander C, Southworth T, Singh D, Clark J, Nejentsev S, Okkenhaug K, Condliffe A, Chandra A, Cahn A, Hall EB. An open label trial of nemiralisib, an inhaled PI3 kinase delta inhibitor for the treatment of Activated PI3 kinase Delta Syndrome. Pulm Pharmacol Ther 2023; 79:102201. [PMID: 36841351 DOI: 10.1016/j.pupt.2023.102201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/08/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023]
Abstract
Activated PI3Kδ Syndrome (APDS) is a rare inherited inborn error of immunity caused by mutations that constitutively activate the p110 delta isoform of phosphoinositide 3-kinase (PI3Kδ), resulting in recurring pulmonary infections. Currently no licensed therapies are available. Here we report the results of an open-label trial in which five subjects were treated for 12 weeks with nemiralisib, an inhaled inhibitor of PI3Kδ, to determine safety, systemic exposure, together with lung and systemic biomarker profiles (Clinicaltrial.gov: NCT02593539). Induced sputum was captured to measure changes in phospholipids and inflammatory mediators, and blood samples were collected to assess pharmacokinetics of nemiralisib, and systemic biomarkers. Nemiralisib was shown to have an acceptable safety and tolerability profile, with cough being the most common adverse event, and no severe adverse events reported during the study. No meaningful changes in phosphatidylinositol (3,4,5)-trisphosphate (PIP3; the enzyme product of PI3Kδ) or downstream inflammatory markers in induced sputum, were observed following nemiralisib treatment. Similarly, there were no meaningful changes in blood inflammatory markers, or lymphocytes subsets. Systemic levels of nemiralisib were higher in subjects in this study compared to previous observations. While nemiralisib had an acceptable safety profile, there was no convincing evidence of target engagement in the lung following inhaled dosing and no downstream effects observed in either the lung or blood compartments. We speculate that this could be explained by nemiralisib not being retained in the lung for sufficient duration, suggested by the increased systemic exposure, perhaps due to pre-existing structural lung damage. In this study investigating a small number of subjects with APDS, nemiralisib appeared to be safe and well-tolerated. However, data from this study do not support the hypothesis that inhaled treatment with nemiralisib would benefit patients with APDS.
Collapse
Affiliation(s)
- Malcolm Begg
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK.
| | - Augustin Amour
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Emily Jarvis
- Clinical Statistics, Development, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Teresa Tang
- Global Medical Safety, Development, GlaxoSmithKline, GSK House, London, UK
| | - Sara Santos Franco
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| | - Andrew Want
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| | - Misba Beerahee
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Disala Fernando
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| | - Yakshitha Karkera
- Clinical Statistics, Development, GlaxoSmithKline, Prestige Trade Tower, Palace Road, Bangalore, India
| | | | - Thomas Southworth
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK; Medicines Evaluation Unit, Manchester University NHS Hospital Trust, Manchester, UK
| | - Dave Singh
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK; Medicines Evaluation Unit, Manchester University NHS Hospital Trust, Manchester, UK
| | | | - Sergey Nejentsev
- Department of Medicine, University of Cambridge, Cambridge, UK; Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, Amsterdam, the Netherlands; Amsterdam Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Alison Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, UK
| | - Anita Chandra
- Department of Medicine, University of Cambridge, Cambridge, UK; Department of Clinical Immunology, Addenbrooke's Hospital, Cambridge, UK
| | - Anthony Cahn
- Research, GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | - Edward Banham Hall
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
16
|
Mohammadi F, Yadegar A, Mardani M, Ayati A, Abolhassani H, Rezaei N. Organ-based clues for diagnosis of inborn errors of immunity: A practical guide for clinicians. Immun Inflamm Dis 2023; 11:e833. [PMID: 37102642 PMCID: PMC10091206 DOI: 10.1002/iid3.833] [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: 02/16/2023] [Accepted: 03/24/2023] [Indexed: 04/28/2023] Open
Abstract
Inborn errors of immunity (IEI) comprise a group of about 490 genetic disorders that lead to aberrant functioning or the development of distinct immune system components. So far, a broad spectrum of IEI-related manifestations has been noted in the literature. Due to overlapping signs and symptoms of IEI, physicians face challenges in appropriately diagnosing and managing affected individuals. The last decade has witnesses improving in the molecular diagnosis of IEI patients. As a result, it can be the mainstay of diagnostic algorithms, prognosis, and possibly therapeutic interventions in patients with IEI. Furthermore, reviewing IEI clinical complications demonstrates that the manifestations and severity of the symptoms depend on the involved gene that causes the disease and its penetrance. Although several diagnostic criteria have been used for IEI, not every patient can be explored in the same way. As a result of the failure to consider IEI diagnosis and the variety of diagnostic capabilities and laboratory facilities in different regions, undiagnosed patients are increasing. On the other hand, early diagnosis is an almost essential element in improving the quality of life in IEI patients. Since there is no appropriate guideline for IEI diagnosis in different organs, focusing on the clues in the patient's chief complaint and physical exams can help physicians narrow their differential diagnosis. This article aims to provide a practical guide for IEI diagnosis based on the involved organ. We hope to assist clinicians in keeping IEI diagnosis in mind and minimizing possible related complications due to delayed diagnosis.
Collapse
Affiliation(s)
- Fatemeh Mohammadi
- School of MedicineTehran University of Medical SciencesTehranIran
- Universal Scientific Education and Research Network (USERN)Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)TehranIran
| | - Amirhossein Yadegar
- School of MedicineTehran University of Medical SciencesTehranIran
- Universal Scientific Education and Research Network (USERN)Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)TehranIran
| | - Mahta Mardani
- School of MedicineTehran University of Medical SciencesTehranIran
- Universal Scientific Education and Research Network (USERN)Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)TehranIran
| | - Aryan Ayati
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart CenterTehran University of Medical ScienceTehranIran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical CenterTehran University of Medical SciencesTehranIran
| | - Nima Rezaei
- Universal Scientific Education and Research Network (USERN)Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA)TehranIran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical CenterTehran University of Medical SciencesTehranIran
- Primary Immunodeficiency Diseases Network (PIDNet)TehranIran
- Children's Medical CenterTehranIran
| |
Collapse
|
17
|
Elworthy S, Rutherford HA, Prajsnar TK, Hamilton NM, Vogt K, Renshaw SA, Condliffe AM. Activated PI3K delta syndrome 1 mutations cause neutrophilia in zebrafish larvae. Dis Model Mech 2023; 16:dmm049841. [PMID: 36805642 PMCID: PMC10655814 DOI: 10.1242/dmm.049841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
People with activated PI3 kinase delta syndrome 1 (APDS1) suffer from immune deficiency and severe bronchiectasis. APDS1 is caused by dominant activating mutations of the PIK3CD gene that encodes the PI3 kinase delta (PI3Kδ) catalytic subunit. Despite the importance of innate immunity defects in bronchiectasis, there has been limited investigation of neutrophils or macrophages in APDS1 patients or mouse models. Zebrafish embryos provide an ideal system to study neutrophils and macrophages. We used CRISPR-Cas9 and CRISPR-Cpf1, with oligonucleotide-directed homologous repair, to engineer zebrafish equivalents of the two most prevalent human APDS1 disease mutations. These zebrafish pik3cd alleles dominantly caused excessive neutrophilic inflammation in a tail-fin injury model. They also resulted in total body neutrophilia in the absence of any inflammatory stimulus but normal numbers of macrophages. Exposure of zebrafish to the PI3Kδ inhibitor CAL-101 reversed the total body neutrophilia. There was no apparent defect in neutrophil maturation or migration, and tail-fin regeneration was unimpaired. Overall, the finding is of enhanced granulopoeisis, in the absence of notable phenotypic change in neutrophils and macrophages.
Collapse
Affiliation(s)
- Stone Elworthy
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Holly A. Rutherford
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Tomasz K. Prajsnar
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - Noémie M. Hamilton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Katja Vogt
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Alison M. Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
18
|
Gutierrez MJ, Nino G, Sun D, Restrepo-Gualteros S, Sadreameli SC, Fiorino EK, Wu E, Vece T, Hagood JS, Maglione PJ, Kurland G, Koumbourlis A, Sullivan KE. The lung in inborn errors of immunity: From clinical disease patterns to molecular pathogenesis. J Allergy Clin Immunol 2022; 150:1314-1324. [PMID: 36244852 PMCID: PMC9826631 DOI: 10.1016/j.jaci.2022.08.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022]
Abstract
In addition to being a vital organ for gas exchange, the lung is a crucial immune organ continuously exposed to the external environment. Genetic defects that impair immune function, called inborn errors of immunity (IEI), often have lung disease as the initial and/or primary manifestation. Common types of lung disease seen in IEI include infectious complications and a diverse group of diffuse interstitial lung diseases. Although lung damage in IEI has been historically ascribed to recurrent infections, contributions from potentially targetable autoimmune and inflammatory pathways are now increasingly recognized. This article provides a practical guide to identifying the diverse pulmonary disease patterns in IEI based on lung imaging and respiratory manifestations, and integrates this clinical information with molecular mechanisms of disease and diagnostic assessments in IEI. We cover the entire IEI spectrum, including immunodeficiencies and immune dysregulation with monogenic autoimmunity and autoinflammation, as well as recently described IEI with pulmonary manifestations. Although the pulmonary manifestations of IEI are highly relevant for all age groups, special emphasis is placed on the pediatric population, because initial presentations often occur during childhood. We also highlight the pivotal role of genetic testing in the diagnosis of IEI involving the lungs and the critical need to develop multidisciplinary teams for the challenging evaluation of these rare but potentially life-threatening disorders.
Collapse
Affiliation(s)
- Maria J Gutierrez
- Division of Pediatric Allergy, Immunology and Rheumatology, Johns Hopkins University, Baltimore, Md.
| | - Gustavo Nino
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, Washington, DC; Department of Pediatrics, George Washington University School of Medicine, Washington, DC
| | - Di Sun
- Division of Pediatric Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Sonia Restrepo-Gualteros
- Department of Pediatrics, School of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia; Division of Pediatric Pulmonology, Fundacion Hospital La Misericordia, Bogotá, Colombia
| | - Sarah C Sadreameli
- Division of Pediatric Pulmonology and Sleep Medicine, Johns Hopkins University, Baltimore, Md
| | - Elizabeth K Fiorino
- Departments of Science Education and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| | - Eveline Wu
- Division of Pediatric Allergy, Immunology and Rheumatology, University of North Carolina, Chapel Hill, NC
| | - Timothy Vece
- Division of Pediatric Pulmonology, University of North Carolina, Chapel Hill, NC
| | - James S Hagood
- Division of Pediatric Pulmonology, University of North Carolina, Chapel Hill, NC
| | - Paul J Maglione
- Division of Allergy and Immunology, Boston University, Boston, Mass
| | - Geoffrey Kurland
- Division of Pediatric Pulmonology and Sleep Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Anastassios Koumbourlis
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, Washington, DC; Department of Pediatrics, George Washington University School of Medicine, Washington, DC
| | - Kathleen E Sullivan
- Division of Pediatric Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pa
| |
Collapse
|
19
|
Fekrvand S, Khanmohammadi S, Abolhassani H, Yazdani R. B- and T-Cell Subset Abnormalities in Monogenic Common Variable Immunodeficiency. Front Immunol 2022; 13:912826. [PMID: 35784324 PMCID: PMC9241517 DOI: 10.3389/fimmu.2022.912826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
Common variable immunodeficiency (CVID) is a heterogeneous group of inborn errors of immunity characterized by reduced serum concentrations of different immunoglobulin isotypes. CVID is the most prevalent symptomatic antibody deficiency with a broad range of infectious and non-infectious clinical manifestations. Various genetic and immunological defects are known to be involved in the pathogenesis of CVID. Monogenic defects account for the pathogenesis of about 20-50% of CVID patients, while a variety of cases do not have a defined genetic background. Deficiencies in molecules of B cell receptor signaling or other pathways involving B-cell development, activation, and proliferation could be associated with monogenetic defects of CVID. Genetic defects damping different B cell developmental stages can alter B- and even other lymphocytes’ differentiation and might be involved in the clinical and immunologic presentations of the disorder. Reports concerning T and B cell abnormalities have been published in CVID patients, but such comprehensive data on monogenic CVID patients is few and no review article exists to describe the abrogation of lymphocyte subsets in these disorders. Hence, we aimed to review the role of altered B- and T-cell differentiation in the pathogenesis of CVID patients with monogenic defects.
Collapse
Affiliation(s)
- Saba Fekrvand
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Shaghayegh Khanmohammadi
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Science, Tehran, Iran
- Division of Clinical Immunology, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Science, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Reza Yazdani, ;
| |
Collapse
|
20
|
Zhang J, Jiang H, Lin S, Wu D, Tian H, Jiang L, Cui Y, Jin J, Chen X, Xu H. Design and Optimization of Thienopyrimidine Derivatives as Potent and Selective PI3Kδ Inhibitors for the Treatment of B-Cell Malignancies. J Med Chem 2022; 65:8011-8028. [PMID: 35609190 DOI: 10.1021/acs.jmedchem.2c00530] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Phosphoinositide 3-kinase δ (PI3Kδ) plays a critical role in B lymphocyte (B-cell) development and activation and has been a validated target for the treatment of B-cell malignancies. Herein, we report a series of thienopyrimidine derivatives as novel potent and selective PI3Kδ inhibitors based on a scaffold hopping design strategy. Among them, compound 6 exhibited nanomolar PI3Kδ potency and a favorable selectivity profile compared to other class I PI3K isoforms. In cellular assays, compound 6 showed antiproliferative activity against a panel of B-cell lymphoma cell lines in a low micromolar range, caused cell cycle arrest, and induced apoptosis in Pfeiffer and SU-DHL-6 cells. Further, compound 6 inhibited the activation of mouse B-cells. With support from in vivo pharmacokinetic studies, compound 6 demonstrated significant anticancer efficacy in a Pfeiffer xenograft mouse model. Overall, compound 6 is a promising PI3Kδ inhibitor worthy of further preclinical investigation for the treatment of B-cell malignancies.
Collapse
Affiliation(s)
- Jingbo Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Huimin Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Songwen Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Deyu Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hua Tian
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lin Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yiman Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jing Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Heng Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
21
|
Oh J, Garabedian E, Fuleihan R, Cunningham-Rundles C. Clinical Manifestations and Outcomes of Activated Phosphoinositide 3-Kinase δ Syndrome from the USIDNET Cohort. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:4095-4102. [PMID: 34352450 DOI: 10.1016/j.jaip.2021.07.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/01/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Activated phosphoinosidtide 3-kinase δ syndrome (APDS) is a combined primary immunodeficiency characterized by gain-of-function mutations in PIK3CD and PIK3R1. APDS demonstrates a large range of phenotypes including respiratory and herpesvirus infections, lymphadenopathy, autoimmunity, and developmental delay. OBJECTIVE This study describes clinical phenotypes and disease outcomes of a large APDS cohort from the USIDNET Registry. METHODS 38 patients were enrolled in USIDNET and 2 additional patients were obtained from the Clinical Immunology division at Mount Sinai Hospital. Each patient's demographics, disease complications, genetic studies, laboratory data, therapeutic interventions, and clinical outcomes were reviewed. RESULTS There was a high frequency of respiratory infections (70.0% pneumonia) and herpesvirus infections (37.5%). Bronchiectasis was observed in 45.0% of patients. Lymphadenopathy was common (52.5%) and 12.5% of patients developed lymphoma. Neurological and developmental findings were common: 20.0% had developmental delay, 15.0% had seizures, and 10.0% had dysmorphic features. Asthma was more common in PIK3CD compared to PIK3R1 patients (63.6% vs 14.3%). More subjects with PIK3CD had CD3 lymphopenia compared to the PIK3R1 cohort. Seven patients underwent hematopietic stem cell transplantation. One patient died from infectious complications. CONCLUSION This is the first cohort comparing clinical manifestations in PIK3CD and PIK3R1 patients from the USIDNET Registry. Similar frequencies of respiratory and herpesvirus infections, lymphadenopathy, and developmental delay were observed compared to prior cohort studies. However, a higher frequency of asthma and CD3 lymphopenia in the PIK3CD cohort compared to the PIK3R1 cohort was observed.
Collapse
Affiliation(s)
- Jessica Oh
- Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai.
| | - Elizabeth Garabedian
- National Institute of Allergy and Infectious Diseases, National Institute of Health
| | - Ramsay Fuleihan
- Division of Pediatric Allergy, Immunology, and Rheumatology, Columbia University Irving Medical Center, New York, NY 10032
| | | |
Collapse
|
22
|
Nguyen T, Deenick EK, Tangye SG. Phosphatidylinositol 3-kinase signaling and immune regulation: insights into disease pathogenesis and clinical implications. Expert Rev Clin Immunol 2021; 17:905-914. [PMID: 34157234 DOI: 10.1080/1744666x.2021.1945443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Phosphatidylinositol 3-kinase (PI3K) is a lipid kinase that plays a fundamental role in cell survival, metabolism, proliferation and differentiation. Thus, balanced PI3K signalling is critical for multiple aspects of human health. The discovery that germline variants in genes in the PI3K pathway caused inborn errors of immunity highlighted the non-redundant role of these signalling proteins in the human immune system. The subsequent identification and characterisation of >300 individuals with a novel immune dysregulatory disorder, termed activated PI3K-delta syndrome (APDS), has reinforced the status of PI3K as a key pathway regulating immune function. Studies of APDS have demonstrated that dysregulated PI3K function is disruptive for immune cell development, activation, differentiation, effector function and self-tolerance, which are all important in supporting effective, long-term immune responses. AREAS COVERED In this review, we recount recent findings regarding humans with germline variants in PI3K genes and discuss the underlying cellular and molecular pathologies, with a focus on implications for therapy in APDS patients. EXPERT OPINION Modulating PI3K immune cell signalling by offers opportunities for therapeutic interventions in settings of immunodeficiency, autoimmunity and malignancy, but also highlights potential adverse events that may result from overt pharmacological or intrinsic inhibition of PI3K function.
Collapse
Affiliation(s)
- Tina Nguyen
- Immunity & Inflammation Theme, Garvan Institute of Medical Research, Darlinghurst, Australia.,St Vincent's Clinical Clinical School, University of NSW, Kensington, NSW, Australia
| | - Elissa K Deenick
- Immunity & Inflammation Theme, Garvan Institute of Medical Research, Darlinghurst, Australia.,St Vincent's Clinical Clinical School, University of NSW, Kensington, NSW, Australia
| | - Stuart G Tangye
- Immunity & Inflammation Theme, Garvan Institute of Medical Research, Darlinghurst, Australia.,St Vincent's Clinical Clinical School, University of NSW, Kensington, NSW, Australia
| |
Collapse
|
23
|
Zhang X, Wang J, Zhu K, Jin Y, Fu H, Mao J. Activated phosphoinositide 3-kinase delta syndrome misdiagnosed as anti-neutrophil cytoplasmic antibody-associated vasculitis: a case report. J Int Med Res 2021; 49:3000605211013222. [PMID: 34039074 PMCID: PMC8755648 DOI: 10.1177/03000605211013222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Activated phosphoinositide 3-kinase delta syndrome (APDS) is a combined inborn error of immunity mainly caused by PIK3CD mutations. We herein describe a 4-year-old Chinese boy who was admitted for recurrent pneumonia and persistent hematuria and exhibited multisystem involvement and anti-neutrophil cytoplasmic antibody (ANCA) positivity. He was initially diagnosed with ANCA-associated vasculitis. However, genetic testing revealed a c.1574A>G PIK3CD mutation, resulting in a diagnosis of APDS1.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jingjing Wang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Kun Zhu
- Department of Pathology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yanyan Jin
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Haidong Fu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
24
|
Brodsky NN, Lucas CL. Infections in activated PI3K delta syndrome (APDS). Curr Opin Immunol 2021; 72:146-157. [PMID: 34052541 DOI: 10.1016/j.coi.2021.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/25/2021] [Accepted: 04/27/2021] [Indexed: 01/07/2023]
Abstract
Activated PI3K-delta Syndrome (APDS), also called PI3K-delta activating mutation causing senescent T cells, lymphadenopathy, and immunodeficiency (PASLI), is an autosomal dominant disorder caused by inherited or de novo gain-of-function mutations in one of two genes encoding subunits of the phosphoinositide-3-kinase delta (PI3Kδ) complex. This largely leukocyte-restricted protein complex regulates cell growth, activation, proliferation, and survival. Patients who harbor these mutations have early onset immunodeficiency with recurrent infections, lymphadenopathy, and autoimmunity. The most common infection susceptibilities are sinopulmonary (encapsulated bacteria) and herpesviruses. Multiple defects in both innate and adaptive immune function are responsible for this phenotype. Apart from anti-microbial prophylaxis and immunoglobulin replacement, patients are treated with a variety of immunomodulatory agents and some have needed hematopoietic stem cell transplants. Here, we highlight the spectrum of infections, immune defects, and therapy options in this inborn error of immunity.
Collapse
Affiliation(s)
- Nina N Brodsky
- Department of Immunobiology, Yale University School of Medicine, 300 George Street 353G, New Haven, CT, 06511, USA; Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, P.O. Box 208064, New Haven, CT 06520, USA
| | - Carrie L Lucas
- Department of Immunobiology, Yale University School of Medicine, 300 George Street 353G, New Haven, CT, 06511, USA.
| |
Collapse
|
25
|
Adefemi F, Fruman DA, Marshall AJ. A Case for Phosphoinositide 3-Kinase-Targeted Therapy for Infectious Disease. THE JOURNAL OF IMMUNOLOGY 2021; 205:3237-3245. [PMID: 33288538 DOI: 10.4049/jimmunol.2000599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/22/2020] [Indexed: 12/19/2022]
Abstract
PI3Ks activate critical signaling cascades and have multifaceted regulatory functions in the immune system. Loss-of-function and gain-of-function mutations in the PI3Kδ isoform have revealed that this enzyme can substantially impact immune responses to infectious agents and their products. Moreover, reports garnered from decades of infectious disease studies indicate that pharmacologic inhibition of the PI3K pathway could potentially be effective in limiting the growth of certain microbes via modulation of the immune system. In this review, we briefly highlight the development and applications of PI3K inhibitors and summarize data supporting the concept that PI3Kδ inhibitors initially developed for oncology have immune regulatory potential that could be exploited to improve the control of some infectious diseases. This repurposing of existing kinase inhibitors could lay the foundation for alternative infectious disease therapy using available therapeutic agents.
Collapse
Affiliation(s)
- Folayemi Adefemi
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, R3E-0T5 Winnipeg, Manitoba, Canada
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697; and.,Institute for Immunology, University of California, Irvine, CA 92697
| | - Aaron J Marshall
- Department of Immunology, Rady Faculty of Health Sciences, University of Manitoba, R3E-0T5 Winnipeg, Manitoba, Canada;
| |
Collapse
|
26
|
Methot JL, Zhou H, McGowan MA, Anthony NJ, Christopher M, Garcia Y, Achab A, Lipford K, Trotter BW, Altman MD, Fradera X, Lesburg CA, Li C, Alves S, Chappell CP, Jain R, Mangado R, Pinheiro E, Williams SMG, Goldenblatt P, Hill A, Shaffer L, Chen D, Tong V, McLeod RL, Lee HH, Yu H, Shah S, Katz JD. Projected Dose Optimization of Amino- and Hydroxypyrrolidine Purine PI3Kδ Immunomodulators. J Med Chem 2021; 64:5137-5156. [PMID: 33797901 DOI: 10.1021/acs.jmedchem.1c00237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The approvals of idelalisib and duvelisib have validated PI3Kδ inhibitors for the treatment for hematological malignancies driven by the PI3K/AKT pathway. Our program led to the identification of structurally distinct heterocycloalkyl purine inhibitors with excellent isoform and kinome selectivity; however, they had high projected human doses. Improved ligand contacts gave potency enhancements, while replacement of metabolic liabilities led to extended half-lives in preclinical species, affording PI3Kδ inhibitors with low once-daily predicted human doses. Treatment of C57BL/6-Foxp3-GDL reporter mice with 30 and 100 mg/kg/day of 3c (MSD-496486311) led to a 70% reduction in Foxp3-expressing regulatory T cells as observed through bioluminescence imaging with luciferin, consistent with the role of PI3K/AKT signaling in Treg cell proliferation. As a model for allergic rhinitis and asthma, treatment of ovalbumin-challenged Brown Norway rats with 0.3 to 30 mg/kg/day of 3c gave a dose-dependent reduction in pulmonary bronchoalveolar lavage inflammation eosinophil cell count.
Collapse
Affiliation(s)
- Joey L Methot
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Hua Zhou
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Meredeth A McGowan
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Neville John Anthony
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Matthew Christopher
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Yudith Garcia
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Abdelghani Achab
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Kathryn Lipford
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Benjamin Wesley Trotter
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Michael D Altman
- Computational and Structural Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Xavier Fradera
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Charles A Lesburg
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Chaomin Li
- Process Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Stephen Alves
- Discovery Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Craig P Chappell
- Discovery Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Renu Jain
- Discovery Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Ruban Mangado
- Discovery Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Elaine Pinheiro
- Discovery Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Sybill M G Williams
- Discovery Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Peter Goldenblatt
- In Vitro Pharmacology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Armetta Hill
- In Vitro Pharmacology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Lynsey Shaffer
- In Vitro Pharmacology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Dapeng Chen
- Preclinical Pharmacokinetics and Drug Metabolism, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Vincent Tong
- Preclinical Pharmacokinetics and Drug Metabolism, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Robbie L McLeod
- In Vivo Pharmacology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Hyun-Hee Lee
- In Vivo Pharmacology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Hongshi Yu
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Sanjiv Shah
- In Vitro Pharmacology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| | - Jason D Katz
- Discovery Chemistry, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115 United States
| |
Collapse
|
27
|
Rivalta B, Amodio D, Milito C, Chiriaco M, Di Cesare S, Giancotta C, Conti F, Santilli V, Pacillo L, Cifaldi C, Desimio MG, Doria M, Quinti I, De Vito R, Di Matteo G, Finocchi A, Palma P, Trizzino A, Tommasini A, Cancrini C. Case Report: EBV Chronic Infection and Lymphoproliferation in Four APDS Patients: The Challenge of Proper Characterization, Therapy, and Follow-Up. Front Pediatr 2021; 9:703853. [PMID: 34540765 PMCID: PMC8448282 DOI: 10.3389/fped.2021.703853] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Activated PI3K-kinase Delta Syndrome (APDS) is an autosomal-dominant primary immunodeficiency (PID) caused by the constitutive activation of the PI3Kδ kinase. The consequent hyperactivation of the PI3K-Akt-mTOR pathway leads to an impaired T- and B-cells differentiation and function, causing progressive lymphopenia, hypogammaglobulinemia and hyper IgM. Patients with APDS show recurrent sinopulmonary and chronic herpes virus infections, immune dysregulation manifestations, including cytopenia, arthritis, inflammatory enteropathy, and a predisposition to persistent non-neoplastic splenomegaly/lymphoproliferation and lymphoma. The recurrence of the lymphoproliferative disorder and the difficulties in the proper definition of malignancy on histological examination represents the main challenge in the clinical management of APDS patients, since a prompt and correct diagnosis is needed to avoid major complications. Targeted therapies with PI3Kδ-Akt-mTOR pathway pharmacologic inhibitors (i.e., Rapamycin, Theophylline, PI3K inhibitors) represent a good therapeutic strategy. They can also be used as bridge therapies when HSCT is required in order to control refractory symptoms. Indeed, treated patients showed a good tolerance, improved immunologic phenotype and reduced incidence/severity of immune dysregulation manifestations. Here, we describe our experience in the management of four patients, one male affected with APDS1 (P1) and the other three, a male and two females, with APDS2 (P2, P3, P4) presenting with chronic EBV replication, recurrent episodes of immune dysregulation manifestations and lymphomas. These cases highlighted the importance of a tailored and close follow-up, including serial endoscopic and lymph nodes biopsies control to detect a prompt and correct diagnosis and offer the best therapeutic strategy.
Collapse
Affiliation(s)
- Beatrice Rivalta
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Donato Amodio
- Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Chiriaco
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Silvia Di Cesare
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Carmela Giancotta
- Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Bologna, Italy
| | - Veronica Santilli
- Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lucia Pacillo
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Cristina Cifaldi
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Giovanna Desimio
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Margherita Doria
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Rita De Vito
- Pathology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, Rome, Italy
| | - Gigliola Di Matteo
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Andrea Finocchi
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paolo Palma
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonino Trizzino
- Department of Pediatric Hematology and Oncology, ARNAS Civico Di Cristina and Benfratelli Hospital, Palermo, Italy
| | - Alberto Tommasini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Caterina Cancrini
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
28
|
Thouenon R, Moreno-Corona N, Poggi L, Durandy A, Kracker S. Activated PI3Kinase Delta Syndrome-A Multifaceted Disease. Front Pediatr 2021; 9:652405. [PMID: 34249806 PMCID: PMC8267809 DOI: 10.3389/fped.2021.652405] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
Autosomal dominant gain-of-function mutations in the PIK3CD gene encoding the catalytic subunit p110δ of phosphoinositide 3-kinase-δ (PI3K-δ) or autosomal dominant loss-of-function mutations in the PIK3R1 gene encoding the p85α, p55α and p50α regulatory subunits cause Activated PI3-kinase-δ syndrome (APDS; referred as type 1 APDS and type 2 APDS, respectively). Consequences of these mutations are PI3K-δ hyperactivity. Clinical presentation described for both types of APDS patients is very variable, ranging from mild or asymptomatic features to profound combined immunodeficiency. Massive lymphoproliferation, bronchiectasis, increased susceptibility to bacterial and viral infections and, at a lesser extent, auto-immune manifestations and occurrence of cancer, especially B cell lymphoma, have been described for both types of APDS patients. Here, we review clinical presentation and treatment options as well as fundamental immunological and biological features associated to PI3K-δ increased signaling.
Collapse
Affiliation(s)
- Romane Thouenon
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Nidia Moreno-Corona
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Lucie Poggi
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Anne Durandy
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Sven Kracker
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| |
Collapse
|
29
|
Redenbaugh V, Coulter T. Disorders Related to PI3Kδ Hyperactivation: Characterizing the Clinical and Immunological Features of Activated PI3-Kinase Delta Syndromes. Front Pediatr 2021; 9:702872. [PMID: 34422726 PMCID: PMC8374435 DOI: 10.3389/fped.2021.702872] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/28/2021] [Indexed: 11/28/2022] Open
Abstract
Phosphoinositide-3-kinase δ (PI3Kδ) is found in immune cells and is part of the PI3K/AKT/mTOR/S6K signalling pathway essential to cell survival, growth and differentiation. Hyperactivation of PI3Kδ enzyme results in Activated PI3-kinase delta syndrome (APDS). This childhood onset, autosomal dominant, combined immunodeficiency, is caused by heterozygous gain of function (GOF) mutations in PIK3CD (encodes PI3Kδ catalytic subunit p110δ), mutations in PIK3R1 (encodes PI3Kδ regulatory subunit p85α) or LOF mutations in PTEN (terminates PI3Kδ signalling) leading to APDS1, APDS2 and APDS-Like (APDS-L), respectively. APDS was initially described in 2013 and over 285 cases have now been reported. Prompt diagnosis of APDS is beneficial as targeted pharmacological therapies such as sirolimus and potentially PI3Kδ inhibitors can be administered. In this review, we provide an update on the clinical and laboratory features of this primary immunodeficiency. We discuss the common manifestations such as sinopulmonary infections, bronchiectasis, lymphoproliferation, susceptibility to herpesvirus, malignancy, as well as more rare non-immune features such as short stature and neurodevelopmental abnormalities. Laboratory characteristics, such as antibody deficiency and B cell and T cell, phenotypes are also summarised.
Collapse
Affiliation(s)
- Vyanka Redenbaugh
- Regional Immunology Services of Northern Ireland, Belfast Health and Social Care Trust, Belfast, United Kingdom.,Mayo Clinic, Rochester, MN, United States
| | - Tanya Coulter
- Regional Immunology Services of Northern Ireland, Belfast Health and Social Care Trust, Belfast, United Kingdom
| |
Collapse
|
30
|
Williams SN. Endoscopic airway manifestations in a pediatric patient with activated PI3K-delta syndrome. Pediatr Pulmonol 2020; 55:2836-2837. [PMID: 32969594 DOI: 10.1002/ppul.25021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 11/12/2022]
Affiliation(s)
- Sophia N Williams
- Division of Pulmonology, Department of Pediatrics, Phoenix Children's Hospital, Phoenix, Arizona
| |
Collapse
|
31
|
Nunes-Santos CJ, Uzel G, Rosenzweig SD. PI3K pathway defects leading to immunodeficiency and immune dysregulation. J Allergy Clin Immunol 2020; 143:1676-1687. [PMID: 31060715 DOI: 10.1016/j.jaci.2019.03.017] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 12/16/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway is involved in a broad range of cellular processes, including growth, metabolism, differentiation, proliferation, motility, and survival. The PI3Kδ enzyme complex is primarily present in the immune system and comprises a catalytic (p110δ) and regulatory (p85α) subunit. Dynamic regulation of PI3Kδ activity is required to ensure normal function and differentiation of immune cells. In the last decade, discovery of germline mutations in genes involved in the PI3Kδ pathway (PIK3CD, PIK3R1, or phosphatase and tensin homolog [PTEN]) proved that both overactivation and underactivation (gain of function and loss of function, respectively) of PI3Kδ lead to impaired and dysregulated immunity. Although a small group of patients reported to underactivate PI3Kδ show predominantly humoral defects and autoimmune features, more than 200 patients have been described with overactivation of PI3Kδ, presenting with a much more complex phenotype of combined immunodeficiency and immune dysregulation. The clinical and immunologic characterization, as well as current pathophysiologic understanding and specific therapies for PI3K pathway defects leading to immunodeficiency and immune dysregulation, are reviewed here.
Collapse
Affiliation(s)
- Cristiane J Nunes-Santos
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health (NIH) Clinical Center, Bethesda, Md; Faculdade de Medicina, Instituto da Crianca, Universidade de São Paulo, São Paulo, Brazil
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Md
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health (NIH) Clinical Center, Bethesda, Md.
| |
Collapse
|
32
|
Yazdani R, Hamidi Z, Babaha F, Azizi G, Fekrvand S, Abolhassani H, Aghamohammadi A. PIK3R1 Mutation Associated with Hyper IgM (APDS2 Syndrome): A Case Report and Review of the Literature. Endocr Metab Immune Disord Drug Targets 2020; 19:941-958. [PMID: 30799802 DOI: 10.2174/1871530319666190225114739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/08/2019] [Accepted: 02/07/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE APDS [Activated phosphoinositide 3-kinase (PI3K) δ Syndrome] is a newly found special form of primary immunodeficiency caused by mutations in genes encoding PI3Kδ subunits and over-activation of the PI3K signaling pathway. Gain-of-function and loss-of-function mutations in PIK3CD (encoding P110δ) and PIK3R1 (encoding p85α, p55α and p50α) lead to APDS1 and APDS2, respectively. The subsequent irregular PI3K downstream signaling cascade is associated with abnormalities in B cells and T cells and the consequent heterogeneous clinical manifestations including respiratory tract infections, autoimmunity, lymphoproliferation and not to mention primary antibody deficiency. In this study, we report a 12-year-old girl with a mutation in the PIK3R1 gene who manifested immunological phenotypes resembling hyper IgM syndrome along with a review of the literature of the previously reported patients. METHODS Whole exome sequencing was performed to detect the underlying genetic mutation in this patient. RESULTS A de novo heterozygous splice site mutation in the hot spot of the PIK3R1 gene within the intron 10 was found (c.1425+1G>A). CONCLUSION Further investigations are required for evaluation of the underlying genetic defects and the possible associations between genetic underpinning and heterogeneous severity and features of the disease.
Collapse
Affiliation(s)
- Reza Yazdani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran, and the University of Medical Science, Tehran, Iran
| | - Zahra Hamidi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fateme Babaha
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran, and the University of Medical Science, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran, and the University of Medical Science, Tehran, Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran, and the University of Medical Science, Tehran, Iran
| |
Collapse
|
33
|
Ramzi N, Jamee M, Bakhtiyari M, Rafiemanesh H, Zainaldain H, Tavakol M, Rezaei A, Kalvandi M, Zian Z, Mohammadi H, Jadidi-Niaragh F, Yazdani R, Abolhassani H, Aghamohammadi A, Azizi G. Bronchiectasis in common variable immunodeficiency: A systematic review and meta-analysis. Pediatr Pulmonol 2020; 55:292-299. [PMID: 31833673 DOI: 10.1002/ppul.24599] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is the most prevalent symptomatic primary immunodeficiency disorder characterized by infectious and noninfectious complications. Bronchiectasis continues to be a common respiratory problem and therapeutic challenge in CVID. The aim of this study is to estimate the overall prevalence of bronchiectasis and its associated phenotype in patients with CVID. METHODS A systematic literature search was performed in Web of Science, PubMed, and Scopus from the earliest available date to February 2019 with standard keywords. All pooled analyses of bronchiectasis prevalence and the corresponding 95% confidence intervals (CIs) were based on random-effects models. RESULTS Fifty-five studies comprising 8535 patients with CVID were included in the meta-analysis. Overall prevalence of bronchiectasis was 34% (95% CI: 30-38; I2 = 90.19%). CVID patients with bronchiectasis had significantly lower serum immunoglobulin A (IgA) and IgM levels at the time of diagnosis compared with those without bronchiectasis. Among the clinical features, the frequencies of splenomegaly, pneumonia, otitis media, and lymphocytic interstitial pneumonia were significantly higher in CVID patients with bronchiectasis compared with those without bronchiectasis, respectively. CONCLUSION A higher prevalence of bronchiectasis in patients with CVID should be managed by controlling recurrent and severe pneumonia episodes which are immune dysregulation since this complication is associated with poor prognosis in these patients.
Collapse
Affiliation(s)
- Nasim Ramzi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mahnaz Jamee
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mahmood Bakhtiyari
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Community Medicine, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Hosein Rafiemanesh
- Student Research Committee, Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Zainaldain
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Tavakol
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Amir Rezaei
- Department of Pediatrics, Imam Ali Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Mustafa Kalvandi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Zeineb Zian
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Abolhassani
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
34
|
Gereige JD, Maglione PJ. Current Understanding and Recent Developments in Common Variable Immunodeficiency Associated Autoimmunity. Front Immunol 2019; 10:2753. [PMID: 31921101 PMCID: PMC6914703 DOI: 10.3389/fimmu.2019.02753] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Common variable immunodeficiency (CVID) is the most prevalent symptomatic primary immunodeficiency and comprises a group of disorders with similar antibody deficiency but a myriad of different etiologies, most of which remain undefined. The variable aspect of CVID refers to the approximately half of patients who develop non-infectious complications in addition to heightened susceptibility to infection. The pathogenesis of these complications is poorly understood and somewhat counterintuitive because these patients that are defined by their immune futility simultaneously have elevated propensity for autoimmune disease. There are numerous aspects of immune dysregulation associated with autoimmunity in CVID that have only begun to be studied. These findings include elevations of T helper type 1 and follicular helper T cells and B cells expressing low levels of CD21 as well as reciprocal decreases in regulatory T cells and isotype-switched memory B cells. Recently, advances in genomics have furthered our understanding of the fundamental biology underlying autoimmunity in CVID and led to precision therapeutic approaches. However, these genetic etiologies are also associated with clinical heterogeneity and incomplete penetrance, highlighting the fact that continued research efforts remain necessary to optimize treatment. Additional factors, such as commensal microbial dysbiosis, remain to be better elucidated. Thus, while recent advances in our understanding of CVID-associated autoimmunity have been exciting and substantial, these current scientific advances must now serve as building blocks for the next stages of discovery.
Collapse
Affiliation(s)
- Jessica D Gereige
- Department of Pulmonary, Allergy, Sleep & Critical Care Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| | - Paul J Maglione
- Department of Pulmonary, Allergy, Sleep & Critical Care Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
35
|
Delmonte OM, Notarangelo LD. Targeted Therapy with Biologicals and Small Molecules in Primary Immunodeficiencies. Med Princ Pract 2019; 29:101-112. [PMID: 31597133 PMCID: PMC7098309 DOI: 10.1159/000503997] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 10/09/2019] [Indexed: 01/14/2023] Open
Abstract
Primary immunodeficiencies are disorders resulting from mutations in genes involved in immune defense and immune regulation. These conditions are characterized by various combinations of recurrent infections, autoimmunity, lymphoproliferation, inflammatory manifestations, and malignancy. In the last 20 years, newborn screening programs and next generation sequencing techniques have increased the ability to diagnose primary immunodeficiencies. Furthermore, an advanced understanding of the molecular basis of these inherited disorders has led to the implementation of targeted therapies that utilize small molecules and biologics to modulate the activity of impaired intracellular pathways. This article will discuss selected primary immunodeficiencies, the genetic defects of which have been recently studied and are amenable to targeted therapy as a reflection of the potential of precision medicine in the future.
Collapse
Affiliation(s)
- Ottavia Maria Delmonte
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Luigi Daniele Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA,
| |
Collapse
|
36
|
Delmonte OM, Castagnoli R, Calzoni E, Notarangelo LD. Inborn Errors of Immunity With Immune Dysregulation: From Bench to Bedside. Front Pediatr 2019; 7:353. [PMID: 31508401 PMCID: PMC6718615 DOI: 10.3389/fped.2019.00353] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
Inborn errors of immunity are genetic disorders with broad clinical manifestations, ranging from increased susceptibility to infections to significant immune dysregulation, often leading to multiple autoimmune phenomena, lymphoproliferation, and malignancy. The treatment is challenging as it requires careful balancing of immunosuppression in subjects at increased risk of infections. Recently, the improved ability to define inborn errors of immunity pathophysiology at the molecular level has set the basis for the development of targeted therapeutic interventions. Such a "precision medicine" approach is mainly bases on the use of available small molecules and biologics to target a specific cell function. In this article, we summarize the clinical and laboratory features of various recently described inborn errors of immunity associated with immune dysregulation and hyperinflammation in which mechanism-based therapeutic approaches have been implemented.
Collapse
Affiliation(s)
- Ottavia Maria Delmonte
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- Foundation IRCCS Policlinico San Matteo, Department of Pediatrics, University of Pavia, Pavia, Italy
| | - Enrica Calzoni
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- Department of Molecular and Translational Medicine, A. Nocivelli Institute for Molecular Medicine, University of Brescia, Brescia, Italy
| | - Luigi Daniele Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
37
|
Hart M, Walch-Rückheim B, Krammes L, Kehl T, Rheinheimer S, Tänzer T, Glombitza B, Sester M, Lenhof HP, Keller A, Meese E. miR-34a as hub of T cell regulation networks. J Immunother Cancer 2019; 7:187. [PMID: 31311583 PMCID: PMC6636054 DOI: 10.1186/s40425-019-0670-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/08/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Micro(mi)RNAs are increasingly recognized as central regulators of immune cell function. While it has been predicted that miRNAs have multiple targets, the majority of these predictions still await experimental confirmation. Here, miR-34a, a well-known tumor suppressor, is analyzed for targeting genes involved in immune system processes of leucocytes. METHODS Using an in-silico approach, we combined miRNA target prediction with GeneTrail2, a web tool for Multi-omics enrichment analysis, to identify miR-34a target genes, which are involved in the immune system process subcategory of Gene Ontology. RESULTS Out of the 193 predicted target genes in this subcategory we experimentally tested 22 target genes and confirmed binding of miR-34a to 14 target genes including VAMP2, IKBKE, MYH9, MARCH8, KLRK1, CD11A, TRAFD1, CCR1, PYDC1, PRF1, PIK3R2, PIK3CD, AP1B1, and ADAM10 by dual luciferase assays. By transfecting Jurkat, primary CD4+ and CD8+ T cells with miR-34a, we demonstrated that ectopic expression of miR-34a leads to reduced levels of endogenous VAMP2 and CD11A, which are central to the analyzed subcategories. Functional downstream analysis of miR-34a over-expression in activated CD8+ T cells exhibits a distinct decrease of PRF1 secretion. CONCLUSIONS By simultaneous targeting of 14 mRNAs miR-34a acts as major hub of T cell regulatory networks suggesting to utilize miR-34a as target of intervention towards a modulation of the immune responsiveness of T-cells in a broad tumor context.
Collapse
Affiliation(s)
- Martin Hart
- Institute of Human Genetics, Saarland University, Building 60, 66421, Homburg, Germany.
| | - Barbara Walch-Rückheim
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Lena Krammes
- Institute of Human Genetics, Saarland University, Building 60, 66421, Homburg, Germany
| | - Tim Kehl
- Center for Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Stefanie Rheinheimer
- Institute of Human Genetics, Saarland University, Building 60, 66421, Homburg, Germany
| | - Tanja Tänzer
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Birgit Glombitza
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, Building 60, 66421, Homburg, Germany
| |
Collapse
|
38
|
Methot JL, Zhou H, Kattar SD, McGowan MA, Wilson K, Garcia Y, Deng Y, Altman M, Fradera X, Lesburg C, Fischmann T, Li C, Alves S, Shah S, Fernandez R, Goldenblatt P, Hill A, Shaffer L, Chen D, Tong V, McLeod RL, Yu H, Bass A, Kemper R, Gatto NT, LaFranco-Scheuch L, Trotter BW, Guzi T, Katz JD. Structure Overhaul Affords a Potent Purine PI3Kδ Inhibitor with Improved Tolerability. J Med Chem 2019; 62:4370-4382. [DOI: 10.1021/acs.jmedchem.8b01818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
39
|
Benavides-Nieto M, Méndez-Echevarría A, Del Rosal T, García-García ML, Casas I, Pozo F, de la Serna O, Lopez-Granados E, Rodriguez-Pena R, Calvo C. The role of respiratory viruses in children with humoral immunodeficiency on immunoglobulin replacement therapy. Pediatr Pulmonol 2019; 54:194-199. [PMID: 30575324 PMCID: PMC7167964 DOI: 10.1002/ppul.24214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/14/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND The role of viruses in children with respiratory tract infections and humoral immunodeficiencies has hardly been studied. We have evaluated these infections in children with humoral immunodeficiencies who required immunoglobulin replacement therapy, considering their relationship with symptoms, lung function, bacterial co-infection, and outcomes. METHODS We conducted a prospective case-control study during a 1-year period, including children with humoral immunodeficiencies receiving immunoglobulin replacement therapy. For each patient, at least one healthy family member was included. Respiratory samples for viral detection were taken every 1-3 months, and in case of respiratory tract infections. Symptoms questionnaires were filled biweekly. Spirometry and sputum culture were performed in every episode. RESULTS Sixty-six episodes were analyzed in 14 patients (median age 12 years; IQR 7-17), identifying 18 respiratory viruses (27.3%), being rhinovirus the most frequently isolated one (12/18; 66%). Positive viral episodes were associated with clinical symptoms (89% vs 43%), more frequent antibiotic treatment (44% vs 15%) or hospital admission (22% vs 0%) than negative ones. Patients with positive viral detection showed impaired lung function, with lower FEV1 and FVC values. CONCLUSIONS In our experience, viral respiratory tract infections can cause significant respiratory symptoms and impaired lung function, in children with HID, despite immunoglobulin replacement therapy. These patients could benefit from the monitoring of viral infections, as these may be a gateway for ongoing lung damage.
Collapse
Affiliation(s)
- Marta Benavides-Nieto
- Pediatric Infectious Diseases Department, Hospital Universitario La Paz, Madrid, Fundación IdiPaz, Madrid, Spain
| | - Ana Méndez-Echevarría
- Pediatric Infectious Diseases Department, Hospital Universitario La Paz, Madrid, Fundación IdiPaz, Madrid, Spain.,Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain
| | - Teresa Del Rosal
- Pediatric Infectious Diseases Department, Hospital Universitario La Paz, Madrid, Fundación IdiPaz, Madrid, Spain.,Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain
| | - María Luz García-García
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain.,Pediatrics and Pneumology Department, Severo Ochoa Hospital, Leganés, Madrid, Spain
| | - Inmaculada Casas
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain.,Respiratory Virus and Influenza Unit, CNM, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Francisco Pozo
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain.,Respiratory Virus and Influenza Unit, CNM, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Olga de la Serna
- Paediatric Pneumology Department, Hospital La Paz, Madrid, Spain
| | | | - Rebeca Rodriguez-Pena
- Immunology Department, Hospital Universitario La Paz, Fundación IdiPaz, Madrid, Spain
| | - Cristina Calvo
- Pediatric Infectious Diseases Department, Hospital Universitario La Paz, Madrid, Fundación IdiPaz, Madrid, Spain.,Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain.,TEDDY Network (European Network of Excellence for Pediatric Clinical Research), Pavia, Italy
| |
Collapse
|
40
|
Castagnoli R, Delmonte OM, Calzoni E, Notarangelo LD. Hematopoietic Stem Cell Transplantation in Primary Immunodeficiency Diseases: Current Status and Future Perspectives. Front Pediatr 2019; 7:295. [PMID: 31440487 PMCID: PMC6694735 DOI: 10.3389/fped.2019.00295] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/03/2019] [Indexed: 12/29/2022] Open
Abstract
Primary immunodeficiencies (PID) are disorders that for the most part result from mutations in genes involved in immune host defense and immunoregulation. These conditions are characterized by various combinations of recurrent infections, autoimmunity, lymphoproliferation, inflammatory manifestations, atopy, and malignancy. Most PID are due to genetic defects that are intrinsic to hematopoietic cells. Therefore, replacement of mutant cells by healthy donor hematopoietic stem cells (HSC) represents a rational therapeutic approach. Full or partial ablation of the recipient's marrow with chemotherapy is often used to allow stable engraftment of donor-derived HSCs, and serotherapy may be added to the conditioning regimen to reduce the risks of graft rejection and graft versus host disease (GVHD). Initially, hematopoietic stem cell transplantation (HSCT) was attempted in patients with severe combined immunodeficiency (SCID) as the only available curative treatment. It was a challenging procedure, associated with elevated rates of morbidity and mortality. Overtime, outcome of HSCT for PID has significantly improved due to availability of high-resolution HLA typing, increased use of alternative donors and new stem cell sources, development of less toxic, reduced-intensity conditioning (RIC) regimens, and cellular engineering techniques for graft manipulation. Early identification of infants affected by SCID, prior to infectious complication, through newborn screening (NBS) programs and prompt genetic diagnosis with Next Generation Sequencing (NGS) techniques, have also ameliorated the outcome of HSCT. In addition, HSCT has been applied to treat a broader range of PID, including disorders of immune dysregulation. Yet, the broad spectrum of clinical and immunological phenotypes associated with PID makes it difficult to define a universal transplant regimen. As such, integration of knowledge between immunologists and transplant specialists is necessary for the development of innovative transplant protocols and to monitor their results during follow-up. Despite the improved outcome observed after HSCT, patients with severe forms of PID still face significant challenges of short and long-term transplant-related complications. To address this issue, novel HSCT strategies are being implemented aiming to improve both survival and long-term quality of life. This article will discuss the current status and latest developments in HSCT for PID, and present data regarding approach and outcome of HSCT in recently described PID, including disorders associated with immune dysregulation.
Collapse
Affiliation(s)
- Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Ottavia Maria Delmonte
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Enrica Calzoni
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Molecular and Translational Medicine, A. Nocivelli Institute for Molecular Medicine, University of Brescia, Brescia, Italy
| | - Luigi Daniele Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
41
|
Stark AK, Chandra A, Chakraborty K, Alam R, Carbonaro V, Clark J, Sriskantharajah S, Bradley G, Richter AG, Banham-Hall E, Clatworthy MR, Nejentsev S, Hamblin JN, Hessel EM, Condliffe AM, Okkenhaug K. PI3Kδ hyper-activation promotes development of B cells that exacerbate Streptococcus pneumoniae infection in an antibody-independent manner. Nat Commun 2018; 9:3174. [PMID: 30093657 PMCID: PMC6085315 DOI: 10.1038/s41467-018-05674-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/17/2018] [Indexed: 02/02/2023] Open
Abstract
Streptococcus pneumoniae is a major cause of pneumonia and a leading cause of death world-wide. Antibody-mediated immune responses can confer protection against repeated exposure to S. pneumoniae, yet vaccines offer only partial protection. Patients with Activated PI3Kδ Syndrome (APDS) are highly susceptible to S. pneumoniae. We generated a conditional knock-in mouse model of this disease and identify a CD19+B220- B cell subset that is induced by PI3Kδ signaling, resides in the lungs, and is correlated with increased susceptibility to S. pneumoniae during early phases of infection via an antibody-independent mechanism. We show that an inhaled PI3Kδ inhibitor improves survival rates following S. pneumoniae infection in wild-type mice and in mice with activated PI3Kδ. These results suggest that a subset of B cells in the lung can promote the severity of S. pneumoniae infection, representing a potential therapeutic target.
Collapse
Affiliation(s)
- Anne-Katrien Stark
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB21 3AT, UK
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Anita Chandra
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB21 3AT, UK
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Department of Medicine, University of Cambridge, Cambridge, CB2 OQQ, UK
- Cambridge University Hospitals NHS Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Krishnendu Chakraborty
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB21 3AT, UK
- Department of Medicine, University of Cambridge, Cambridge, CB2 OQQ, UK
| | - Rafeah Alam
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB21 3AT, UK
| | - Valentina Carbonaro
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB21 3AT, UK
| | - Jonathan Clark
- Biological Chemistry Laboratory, Babraham Institute, Cambridge, CB21 3AT, UK
| | - Srividya Sriskantharajah
- Refractory Respiratory Inflammation Discovery Performance Unit, Respiratory Therapy Area, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Glyn Bradley
- Computational Biology and Statistics, Target Sciences, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Alex G Richter
- Department of Immunology, Queen Elizabeth Hospital, Birmingham, B15 2TH, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Edward Banham-Hall
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB21 3AT, UK
- Department of Medicine, University of Cambridge, Cambridge, CB2 OQQ, UK
- Cambridge University Hospitals NHS Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Menna R Clatworthy
- Molecular Immunity Unit, MRC Laboratory of Molecular Biology, University of Cambridge Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, CB2 OQQ, UK
| | - Sergey Nejentsev
- Department of Medicine, University of Cambridge, Cambridge, CB2 OQQ, UK
| | - J Nicole Hamblin
- Refractory Respiratory Inflammation Discovery Performance Unit, Respiratory Therapy Area, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Edith M Hessel
- Refractory Respiratory Inflammation Discovery Performance Unit, Respiratory Therapy Area, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Sheffield, S10 2RX, UK
| | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB21 3AT, UK.
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.
| |
Collapse
|
42
|
Cannons JL, Preite S, Kapnick SM, Uzel G, Schwartzberg PL. Genetic Defects in Phosphoinositide 3-Kinase δ Influence CD8 + T Cell Survival, Differentiation, and Function. Front Immunol 2018; 9:1758. [PMID: 30116245 PMCID: PMC6082933 DOI: 10.3389/fimmu.2018.01758] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/16/2018] [Indexed: 12/19/2022] Open
Abstract
Activated phosphoinositide 3-kinase delta syndrome (APDS), also known as p110 delta-activating mutation causing senescent T cells, lymphadenopathy and immunodeficiency (PASLI), is an autosomal dominant primary human immunodeficiency (PID) caused by heterozygous gain-of-function mutations in PIK3CD, which encodes the p110δ catalytic subunit of PI3K. This recently described PID is characterized by diverse and heterogeneous clinical manifestations that include recurrent respiratory infections, lymphoproliferation, progressive lymphopenia, and defective antibody responses. A major clinical manifestation observed in the NIH cohort of patients with PIK3CD mutations is chronic Epstein-Barr virus (EBV) and/or cytomegalovirus viremia. Despite uncontrolled EBV infection, many APDS/PASLI patients had normal or higher frequencies of EBV-specific CD8+ T cells. In this review, we discuss data pertaining to CD8+ T cell function in APDS/PASLI, including increased cell death, expression of exhaustion markers, and altered killing of autologous EBV-infected B cells, and how these and other data on PI3K provide insight into potential cellular defects that prevent clearance of chronic infections.
Collapse
Affiliation(s)
- Jennifer L Cannons
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States.,National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Silvia Preite
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States.,National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Senta M Kapnick
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Gulbu Uzel
- National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| | - Pamela L Schwartzberg
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States.,National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|