1. Academic Validation
  2. Clinical and immunologic phenotype associated with activated phosphoinositide 3-kinase δ syndrome 2: A cohort study

Clinical and immunologic phenotype associated with activated phosphoinositide 3-kinase δ syndrome 2: A cohort study

  • J Allergy Clin Immunol. 2016 Jul;138(1):210-218.e9. doi: 10.1016/j.jaci.2016.03.022.
Elodie Elkaim 1 Benedicte Neven 2 Julie Bruneau 3 Kanako Mitsui-Sekinaka 4 Aurelie Stanislas 5 Lucie Heurtier 6 Carrie L Lucas 7 Helen Matthews 7 Marie-Céline Deau 6 Svetlana Sharapova 8 James Curtis 9 Janine Reichenbach 10 Catherine Glastre 11 David A Parry 12 Gururaj Arumugakani 13 Elizabeth McDermott 14 Sara Sebnem Kilic 15 Motoi Yamashita 16 Despina Moshous 2 Hicham Lamrini 6 Burkhard Otremba 17 Andrew Gennery 18 Tanya Coulter 19 Isabella Quinti 20 Jean-Louis Stephan 21 Vassilios Lougaris 22 Nicholas Brodszki 23 Vincent Barlogis 24 Takaki Asano 25 Lionel Galicier 26 David Boutboul 26 Shigeaki Nonoyama 4 Andrew Cant 18 Kohsuke Imai 27 Capucine Picard 28 Sergey Nejentsev 9 Thierry Jo Molina 3 Michael Lenardo 7 Sinisa Savic 29 Marina Cavazzana 30 Alain Fischer 31 Anne Durandy 6 Sven Kracker 32
Affiliations

Affiliations

  • 1 Department of Pediatric Immunology, Hematology and Rheumatology, AP-HP, Necker Children's Hospital, Paris, France; INSERM UMR1163, Paris, France.
  • 2 Department of Pediatric Immunology, Hematology and Rheumatology, AP-HP, Necker Children's Hospital, Paris, France; INSERM UMR1163, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France.
  • 3 Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Department of Pathology, Hôpital Necker-Enfants Malades, Assistance Publique des Hôpitaux de Paris, Paris, France.
  • 4 Department of Pediatrics, National Defense Medical College, Saitama, Japan.
  • 5 Départment de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
  • 6 INSERM UMR1163, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France.
  • 7 Laboratory of Immunology, Molecular Development of the Immune System Section, NIAID Clinical Genomics Program, NIAID, NIH, Bethesda, Md.
  • 8 Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus.
  • 9 Department of Medicine, University of Cambridge, Cambridge, United Kingdom.
  • 10 Division of Immunology, University Children's Hospital Zurich, Children's Research Center, Competence Center for Applied Biotechnology and Molecular Medicine and the Swiss Center for Regenerative Medicine, University Zurich, Zurich, Switzerland.
  • 11 Service de Pédiatrie, Centre Hospitalier Annecy Genevois, Metz-Tessy, France.
  • 12 Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom.
  • 13 Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, United Kingdom.
  • 14 Nottingham University Hospitals, Nottingham, United Kingdom.
  • 15 Pediatric Immunology Division, Uludag University Medical Faculty, Department of Pediatrics, Bursa, Turkey.
  • 16 Department of Pediatrics and Developmental Biology, School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
  • 17 Oncological Practice Oldenburg/Delmenhorst, Oldenburg, Germany.
  • 18 Institute of Cellular Medicine, Paediatric Immunology Department, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom.
  • 19 Department of Immunology, School of Medicine, Trinity College Dublin, St James's Hospital, Dublin, Ireland; Department of Pediatric Immunology and Infectious Diseases, Our Lady's Children's Hospital Crumlin, Dublin, Ireland.
  • 20 Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
  • 21 Department of Pediatric, Hôpital Nord, Saint-Etienne, France.
  • 22 Pediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, Department of Clinical and Experimental Sciences, University of Brescia, Spedali Civili di Brescia, Brescia, Italy.
  • 23 Children's Hospital, Skåne University Hospital, Lund, Sweden.
  • 24 Service d'Hématologie Pédiatrique, Marseille, France.
  • 25 Department of Pediatrics, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan.
  • 26 Clinical Immunology Department, Hôpital Saint Louis, Assistance Publique Hôpitaux de Paris, Université Paris Diderot, Paris, France.
  • 27 Department of Pediatrics, National Defense Medical College, Saitama, Japan; Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan.
  • 28 INSERM UMR1163, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Study Center for Primary Immunodeficiencies, Assistance Publique Hôpitaux de Paris, Necker Hospital, Paris, France.
  • 29 Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom; National Institute for Health Research-Leeds Musculoskeletal Biomedical Research Unit (NIHR-LMBRU) and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), St James's University Hospital, Leeds, United Kingdom.
  • 30 INSERM UMR1163, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Départment de Biothérapie, Centre d'Investigation Clinique intégré en Biothérapies, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
  • 31 Department of Pediatric Immunology, Hematology and Rheumatology, AP-HP, Necker Children's Hospital, Paris, France; INSERM UMR1163, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France; Collège de France, Paris, France.
  • 32 INSERM UMR1163, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France. Electronic address: [email protected].
Abstract

Background: Activated phosphoinositide 3-kinase δ syndrome (APDS) 2 (p110δ-activating mutations causing senescent T cells, lymphadenopathy, and immunodeficiency [PASLI]-R1), a recently described primary immunodeficiency, results from autosomal dominant mutations in PIK3R1, the gene encoding the regulatory subunit (p85α, p55α, and p50α) of class IA phosphoinositide 3-kinases.

Objectives: We sought to review the clinical, immunologic, and histopathologic phenotypes of APDS2 in a genetically defined international patient cohort.

Methods: The medical and biological records of 36 patients with genetically diagnosed APDS2 were collected and reviewed.

Results: Mutations within splice acceptor and donor sites of exon 11 of the PIK3R1 gene lead to APDS2. Recurrent upper respiratory tract infections (100%), pneumonitis (71%), and chronic lymphoproliferation (89%, including adenopathy [75%], splenomegaly [43%], and upper respiratory tract lymphoid hyperplasia [48%]) were the most common features. Growth retardation was frequently noticed (45%). Other complications were mild neurodevelopmental delay (31%); malignant diseases (28%), most of them being B-cell lymphomas; autoimmunity (17%); bronchiectasis (18%); and chronic diarrhea (24%). Decreased serum IgA and IgG levels (87%), increased IgM levels (58%), B-cell lymphopenia (88%) associated with an increased frequency of transitional B cells (93%), and decreased numbers of naive CD4 and naive CD8 cells but increased numbers of CD8 effector/memory T cells were predominant immunologic features. The majority of patients (89%) received immunoglobulin replacement; 3 patients were treated with rituximab, and 6 were treated with rapamycin initiated after diagnosis of APDS2. Five patients died from APDS2-related complications.

Conclusion: APDS2 is a combined immunodeficiency with a variable clinical phenotype. Complications are frequent, such as severe Bacterial and viral infections, lymphoproliferation, and lymphoma similar to APDS1/PASLI-CD. Immunoglobulin replacement therapy, rapamycin, and, likely in the near future, selective phosphoinositide 3-kinase δ inhibitors are possible treatment options.

Keywords

Primary immunodeficiency; activated phosphoinositide 3-kinase δ syndrome; adenopathy; and immunodeficiency; antibody deficiency; hyper-IgM; immunodeficiency; lymphadenopathy; p110δ; p110δ-activating mutations causing senescent T cells; p85α; phosphoinositide 3-kinase.

Figures