1
|
Adelon J, Abolhassani H, Esenboga S, Fouyssac F, Cagdas D, Tezcan I, Kuskonmaz B, Cetinkaya D, Suarez F, Mahdaviani SA, Plassart S, Mathieu AL, Fabien N, Malcus C, Morfin-Sherpa F, Billaud G, Tusseau M, Benezech S, Walzer T, De Villartay JP, Bertrand Y, Belot A. Human DNA-dependent protein kinase catalytic subunit deficiency: A comprehensive review and update. J Allergy Clin Immunol 2024:S0091-6749(24)00677-8. [PMID: 38977084 DOI: 10.1016/j.jaci.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND DNA-dependent protein kinase catalytic subunit (DNA-PKcs) has an essential role in the non-homologous end-joining pathway that repairs DNA double-strand breaks in V(D)J recombination involved in the expression of T- and B-cell receptors. Whereas homozygous mutations in Prkdc define the Scid mouse, a model that has been widely used in biology, human mutations in PRKDC are extremely rare and the disease spectrum has not been described so far. OBJECTIVES To provide an update on the genetics, clinical spectrum, immunological profile, and therapy of DNA-PKcs deficiency in human. METHODS The clinical, biological, and treatment data from the 6 cases published to date and from 1 new patient were obtained and analyzed. Rubella PCR was performed on available granuloma material. RESULTS We report on 7 patients; 6 patients displayed the autosomal recessive p.L3062R mutation in PRKDC-encoding DNA-PKcs. Atypical severe combined immunodeficiency with inflammatory lesions, granulomas, and autoimmunity was the predominant clinical manifestation (n = 5 of 7). Rubella viral strain was detected in the granuloma of 1 patient over the 2 tested. T-cell counts, including naive CD4+CD45RA+ T cells and T-cell function were low at diagnosis for 6 patients. For most patients with available values, naive CD4+CD45RA+ T cells decreased over time (n = 5 of 6). Hematopoietic stem cell transplantation was performed in 5 patients, of whom 4 are still alive without transplant-related morbidity. Sustained T- and B-cell reconstitution was observed, respectively, for 4 and 3 patients, after a median follow-up of 8 years (range 3-16 years). CONCLUSIONS DNA-PKcs deficiency mainly manifests as an inflammatory disease with granuloma and autoimmune features, along with severe infections.
Collapse
Affiliation(s)
- Jihane Adelon
- Department of Pediatric Immunology and Hematology, Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Hospices Civils de Lyon, Lyon, France.
| | - Hassan Abolhassani
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Saliha Esenboga
- Department of Pediatric Immunology, Ihsan Dogramacı Children's Hospital, Hacettepe University, Ankara, Turkey
| | - Fanny Fouyssac
- Department of Pediatric Oncology, Children's Hospital, Nancy, France
| | - Deniz Cagdas
- Department of Pediatric Immunology, Ihsan Dogramacı Children's Hospital, Hacettepe University, Ankara, Turkey
| | - Ilhan Tezcan
- Department of Pediatric Immunology, Ihsan Dogramacı Children's Hospital, Hacettepe University, Ankara, Turkey
| | - Barıs Kuskonmaz
- Department of Pediatric Hematology, Ihsan Dogramacı Children's Hospital, Hacettepe University, Ankara, Turkey
| | - Duygu Cetinkaya
- Department of Pediatric Hematology, Ihsan Dogramacı Children's Hospital, Hacettepe University, Ankara, Turkey
| | - Felipe Suarez
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutical Implications, INSERM UMR1163/CNRS URL 8254, Paris, France; French National Center for Primary Immunodeficiencies, Necker University Hospital, AP-HP, Paris, France; INSERM UMR1163, Imagine Institut, Sorbonne Paris Cité, Paris, France; Department of Hematology, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique des Hôpitaux de Paris, Paris, France; Université Paris Cité, Paris, France
| | - Seyed Alireza Mahdaviani
- Pediatric Respiratory Diseases Research Centre, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Plassart
- Centre de Références Maladies Rares, Rhumatismes inflammatoires et les maladies Auto-Immunes Systémiques rares de l'Enfant (RAISE), Lyon, France
| | - Anne-Laure Mathieu
- Centre de Références Maladies Rares, Rhumatismes inflammatoires et les maladies Auto-Immunes Systémiques rares de l'Enfant (RAISE), Lyon, France; Centre International de Recherche en Infectiologie (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon, Lyon, France
| | - Nicole Fabien
- Department of Immunology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Benite, France
| | - Christophe Malcus
- Department of Immunology, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Florence Morfin-Sherpa
- Laboratoire de Virologie, Institut des Agents Infectieux, Hospices Civils de Lyon, Groupement Hospitalier Nord, Lyon, France; Laboratoire Virologie et Pathologies humaines (VirPath),Centre International de Recherche en Infectiologie (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon, Lyon, France
| | - Geneviève Billaud
- Laboratoire de Virologie, Institut des Agents Infectieux, Hospices Civils de Lyon, Groupement Hospitalier Nord, Lyon, France
| | - Maud Tusseau
- Hospices Civils de Lyon, Lyon, France; Centre International de Recherche en Infectiologie (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon, Lyon, France; Service de Génétique, Groupe Hospitalier Est, Hospices Civils de Lyon, Bron, France
| | - Sarah Benezech
- Department of Pediatric Immunology and Hematology, Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Hospices Civils de Lyon, Lyon, France; Centre International de Recherche en Infectiologie (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon, Lyon, France
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon, Lyon, France
| | - Jean-Pierre De Villartay
- Laboratory "Genome Dynamics in the Immune System" INSERM UMR 1163, Imagine Institute, Université de Paris Cité, Paris, France
| | - Yves Bertrand
- Department of Pediatric Immunology and Hematology, Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Alexandre Belot
- Hospices Civils de Lyon, Lyon, France; Centre de Références Maladies Rares, Rhumatismes inflammatoires et les maladies Auto-Immunes Systémiques rares de l'Enfant (RAISE), Lyon, France; Centre International de Recherche en Infectiologie (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique Unité Mixte de Recherche (UMR) 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon, Lyon, France; Department of Pediatrics Nephrology, Rheumatology, and Dermatology, Hôpital Femme-Mère-Enfant, Bron, France.
| |
Collapse
|
2
|
Pavel-Dinu M, Gardner CL, Nakauchi Y, Kawai T, Delmonte OM, Palterer B, Bosticardo M, Pala F, Viel S, Malech HL, Ghanim HY, Bode NM, Kurgan GL, Detweiler AM, Vakulskas CA, Neff NF, Sheikali A, Menezes ST, Chrobok J, Hernández González EM, Majeti R, Notarangelo LD, Porteus MH. Genetically corrected RAG2-SCID human hematopoietic stem cells restore V(D)J-recombinase and rescue lymphoid deficiency. Blood Adv 2024; 8:1820-1833. [PMID: 38096800 PMCID: PMC11006817 DOI: 10.1182/bloodadvances.2023011766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/23/2023] [Indexed: 04/10/2024] Open
Abstract
ABSTRACT Recombination-activating genes (RAG1 and RAG2) are critical for lymphoid cell development and function by initiating the variable (V), diversity (D), and joining (J) (V(D)J)-recombination process to generate polyclonal lymphocytes with broad antigen specificity. The clinical manifestations of defective RAG1/2 genes range from immune dysregulation to severe combined immunodeficiencies (SCIDs), causing life-threatening infections and death early in life without hematopoietic cell transplantation (HCT). Despite improvements, haploidentical HCT without myeloablative conditioning carries a high risk of graft failure and incomplete immune reconstitution. The RAG complex is only expressed during the G0-G1 phase of the cell cycle in the early stages of T- and B-cell development, underscoring that a direct gene correction might capture the precise temporal expression of the endogenous gene. Here, we report a feasibility study using the CRISPR/Cas9-based "universal gene-correction" approach for the RAG2 locus in human hematopoietic stem/progenitor cells (HSPCs) from healthy donors and RAG2-SCID patient. V(D)J-recombinase activity was restored after gene correction of RAG2-SCID-derived HSPCs, resulting in the development of T-cell receptor (TCR) αβ and γδ CD3+ cells and single-positive CD4+ and CD8+ lymphocytes. TCR repertoire analysis indicated a normal distribution of CDR3 length and preserved usage of the distal TRAV genes. We confirmed the in vivo rescue of B-cell development with normal immunoglobulin M surface expression and a significant decrease in CD56bright natural killer cells. Together, we provide specificity, toxicity, and efficacy data supporting the development of a gene-correction therapy to benefit RAG2-deficient patients.
Collapse
Affiliation(s)
- Mara Pavel-Dinu
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
| | - Cameron L. Gardner
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Yusuke Nakauchi
- Division of Hematology, Department of Medicine, Cancer Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA
| | - Tomoki Kawai
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ottavia M. Delmonte
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Boaz Palterer
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Marita Bosticardo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Francesca Pala
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sebastien Viel
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
- Service d’immunologie biologique, Hospices Civils de Lyon, Centre International de Recherche en Infectivologie, Centre International de Recheerche in Infectivalogie, INSERM U1111, Université Claude Bernard Lyon 1, Centre National de la Recherge Scientifique, UMR5308, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Harry L. Malech
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Hana Y. Ghanim
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
| | | | | | | | | | | | - Adam Sheikali
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
| | - Sherah T. Menezes
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
| | - Jade Chrobok
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
| | - Elaine M. Hernández González
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Cancer Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA
| | - Luigi D. Notarangelo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Matthew H. Porteus
- Division of Oncology, Hematology, Stem Cell Transplantation, Department of Pediatrics, Stanford University, Stanford, CA
| |
Collapse
|
3
|
Oftedal BE, Sjøgren T, Wolff ASB. Interferon autoantibodies as signals of a sick thymus. Front Immunol 2024; 15:1327784. [PMID: 38455040 PMCID: PMC10917889 DOI: 10.3389/fimmu.2024.1327784] [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: 10/25/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Type I interferons (IFN-I) are key immune messenger molecules that play an important role in viral defense. They act as a bridge between microbe sensing, immune function magnitude, and adaptive immunity to fight infections, and they must therefore be tightly regulated. It has become increasingly evident that thymic irregularities and mutations in immune genes affecting thymic tolerance can lead to the production of IFN-I autoantibodies (autoAbs). Whether these biomarkers affect the immune system or tissue integrity of the host is still controversial, but new data show that IFN-I autoAbs may increase susceptibility to severe disease caused by certain viruses, including SARS-CoV-2, herpes zoster, and varicella pneumonia. In this article, we will elaborate on disorders that have been identified with IFN-I autoAbs, discuss models of how tolerance to IFN-Is is lost, and explain the consequences for the host.
Collapse
Affiliation(s)
- Bergithe E. Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Thea Sjøgren
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S. B. Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
4
|
Stephan C, Grossman ME, Magro CM. Primary cutaneous acral CD8-positive T-cell lymphoproliferative disorder: A clinical and histologic retrospective cohort study. Clin Dermatol 2023; 41:666-679. [PMID: 37716580 DOI: 10.1016/j.clindermatol.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2023]
Abstract
Clonally restricted, non-epidermotropic, low-grade, CD8-positive T-cell infiltrates of the skin was recognized as a unique form of indolent CD8-positive lymphoproliferative disease in 2007 when it was first called primary cutaneous indolent CD8-positive lymphoid proliferation. More recently, the designation of primary cutaneous acral CD8-positive T-cell lymphoproliferative disorder has been used. It is unique as a cutaneous lymphoproliferative disorder because of relative uniformity in its clinical presentation and histomorphology. It has been recognized as having an interesting predilection for the ear and acral sites, characteristically presenting as a solitary lesion. The basic morphology is one characterized by a non-epidermotropic, tumefactive infiltrate of well-differentiated, noncerebriform, atypical, small- to intermediate-sized lymphocytes that exhibit a specific phenotype characterized by CD8 and TIA positivity in concert with a distinct perinuclear Golgi staining pattern for CD68. The typical presentation is in the context of a solitary lesion, which can be treated surgically or with local irradiation. We describe in detail two very unusual cases that expand the clinical spectrum of this condition given the non-acral localization, the multiplicity of lesions to involve the trunk and extremities, and, in one case, the stable but recalcitrant course over 30 years. In addition, the second patient developed paraneoplastic dermatomyositis. We also retrospectively review our database for other cases that represent the entity of primary cutaneous acral CD8-positive T-cell lymphoproliferative disorder and review the literature focusing on non-acral cases. Nomenclature evolution from its first recognition in 2007 to the present is discussed.
Collapse
Affiliation(s)
- Carla Stephan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/New York-Presbyterian, New York, USA
| | - Marc E Grossman
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Cynthia M Magro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/New York-Presbyterian, New York, USA.
| |
Collapse
|
5
|
Braams M, Pike-Overzet K, Staal FJT. The recombinase activating genes: architects of immune diversity during lymphocyte development. Front Immunol 2023; 14:1210818. [PMID: 37497222 PMCID: PMC10367010 DOI: 10.3389/fimmu.2023.1210818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
The mature lymphocyte population of a healthy individual has the remarkable ability to recognise an immense variety of antigens. Instead of encoding a unique gene for each potential antigen receptor, evolution has used gene rearrangements, also known as variable, diversity, and joining gene segment (V(D)J) recombination. This process is critical for lymphocyte development and relies on recombination-activating genes-1 (RAG1) and RAG2, here collectively referred to as RAG. RAG serves as powerful genome editing tools for lymphocytes and is strictly regulated to prevent dysregulation. However, in the case of dysregulation, RAG has been implicated in cases of cancer, autoimmunity and severe combined immunodeficiency (SCID). This review examines functional protein domains and motifs of RAG, describes advances in our understanding of the function and (dys)regulation of RAG, discuss new therapeutic options, such as gene therapy, for RAG deficiencies, and explore in vitro and in vivo methods for determining RAG activity and target specificity.
Collapse
Affiliation(s)
- Merijn Braams
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Karin Pike-Overzet
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Frank J. T. Staal
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Leiden University Medical Centre, Leiden, Netherlands
- Department of Paediatrics, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
6
|
Min Q, Csomos K, Li Y, Dong L, Hu Z, Meng X, Yu M, Walter JE, Wang JY. B cell abnormalities and autoantibody production in patients with partial RAG deficiency. Front Immunol 2023; 14:1155380. [PMID: 37475856 PMCID: PMC10354446 DOI: 10.3389/fimmu.2023.1155380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/15/2023] [Indexed: 07/22/2023] Open
Abstract
Mutations in the recombination activating gene 1 (RAG1) and RAG2 in humans are associated with a broad spectrum of clinical phenotypes, from severe combined immunodeficiency to immune dysregulation. Partial (hypomorphic) RAG deficiency (pRD) in particular, frequently leads to hyperinflammation and autoimmunity, with several underlying intrinsic and extrinsic mechanisms causing a break in tolerance centrally and peripherally during T and B cell development. However, the relative contributions of these processes to immune dysregulation remain unclear. In this review, we specifically focus on the recently described tolerance break and B cell abnormalities, as well as consequent molecular and cellular mechanisms of autoantibody production in patients with pRD.
Collapse
Affiliation(s)
- Qing Min
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Krisztian Csomos
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Yaxuan Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lulu Dong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziying Hu
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xin Meng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meiping Yu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jolan E Walter
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
- Division of Pediatric Allergy/Immunology, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Ji-Yang Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Shanghai Huashen Institute of Microbes and Infections, Shanghai, China
| |
Collapse
|
7
|
Ruan Y, Zhao Q, Liu Q, Zhao HY, Zhang ZY, Ding Y, Zhao XD. A novel homozygous RAG1 mutation in a girl presenting with granulomas and alopecia capitis totalis. World J Pediatr 2022; 18:294-299. [PMID: 35157248 DOI: 10.1007/s12519-021-00503-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/12/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Yu Ruan
- Growth, Development, and Mental Health Center of Children and Adolescents, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Qin Zhao
- Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Qing Liu
- Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Hong-Yi Zhao
- Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Zhi-Yong Zhang
- Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China.,Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yuan Ding
- Growth, Development, and Mental Health Center of Children and Adolescents, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Xiao-Dong Zhao
- Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China. .,Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
8
|
Picado C, de Landazuri IO, Vlagea A, Bobolea I, Arismendi E, Amaro R, Sellarés J, Bartra J, Sanmarti R, Hernandez-Rodriguez J, Mascaró JM, Colmenero J, Vaquero EC, Pascal M. Spectrum of Disease Manifestations in Patients with Selective Immunoglobulin E Deficiency. J Clin Med 2021; 10:jcm10184160. [PMID: 34575269 PMCID: PMC8466644 DOI: 10.3390/jcm10184160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Selective IgE deficiency (SIgED) has been previously evaluated in selected patients from allergy units. This study investigates the effects of SIgED on the entire population in a hospital setting and sought to delineate in detail the clinical aspects of SIgED. METHODS A retrospective study of the data obtained from electronic medical records of 52 adult patients (56% female) with a mean age of 43 years and IgE levels of <2.0 kU/L with normal immunoglobulin (Ig) IgG, IgA, and IgM levels, seen at our hospital, without selection bias, from 2010 to 2019. RESULTS Recurrent upper respiratory infections were recorded in 18 (34.6%) patients, pneumonia was recorded in 16 (30.7%) patients, bronchiectasis was recorded in 16 (30.7%) patients, and asthma was recorded in 10 (19.2%) patients. Eighteen patients (34.6%) suffered autoimmune clinical manifestations either isolated (19%) or combining two or more diseases (15%), Hashimoto's thyroiditis being the most frequent (19%), which was followed by arthritis (10%) and thrombocytopenia and/or neutropenia (5.7%). Other less frequent associations were Graves' disease, primary sclerosing cholangitis, Sjögren's syndrome, and autoimmune hepatitis. Eczematous dermatitis (15.3%), chronic spontaneous urticaria (17.3%), and symptoms of enteropathy (21%) were also highly prevalent. Thirty percent of patients developed malignancies, with non-Hodgkin lymphomas (13.4%) being the most prevalent. CONCLUSIONS The clinical manifestations of SIgED encompass a variety of infectious, non-infectious complications, and malignancy. Since it cannot be ruled out that some type of selection bias occurred in the routine assessment of IgE serum Ievels, prospective studies are required to better characterize SIgED and to determine whether it should be added to the list of antibody deficiencies.
Collapse
Affiliation(s)
- César Picado
- Institut Clinic Respiratory, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.B.); (E.A.); (R.A.); (J.S.); (J.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Correspondence:
| | - Iñaki Ortiz de Landazuri
- Immunology Department, CDB. Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.O.d.L.); (A.V.)
| | - Alexandru Vlagea
- Immunology Department, CDB. Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.O.d.L.); (A.V.)
| | - Irina Bobolea
- Institut Clinic Respiratory, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.B.); (E.A.); (R.A.); (J.S.); (J.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Ebymar Arismendi
- Institut Clinic Respiratory, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.B.); (E.A.); (R.A.); (J.S.); (J.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Rosanel Amaro
- Institut Clinic Respiratory, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.B.); (E.A.); (R.A.); (J.S.); (J.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Jacobo Sellarés
- Institut Clinic Respiratory, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.B.); (E.A.); (R.A.); (J.S.); (J.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Joan Bartra
- Institut Clinic Respiratory, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.B.); (E.A.); (R.A.); (J.S.); (J.B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
| | - Raimon Sanmarti
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Department of Rheumatology, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - José Hernandez-Rodriguez
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Department of Autoimmune Diseases, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - José-Manuel Mascaró
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Department of Dermatology, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Jordi Colmenero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Liver Unit, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain
- Centro de Investigaciones en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain
| | - Eva C. Vaquero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Centro de Investigaciones en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain
- Department of Gastroenterology, Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Mariona Pascal
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.S.); (J.H.-R.); (J.-M.M.); (J.C.); (E.C.V.); (M.P.)
- Immunology Department, CDB. Hospital Clinic, Universitat de Barcelona, 08036 Barcelona, Spain; (I.O.d.L.); (A.V.)
| |
Collapse
|
9
|
Bosticardo M, Pala F, Notarangelo LD. RAG deficiencies: Recent advances in disease pathogenesis and novel therapeutic approaches. Eur J Immunol 2021; 51:1028-1038. [PMID: 33682138 PMCID: PMC8325549 DOI: 10.1002/eji.202048880] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/13/2021] [Accepted: 03/03/2021] [Indexed: 12/26/2022]
Abstract
The RAG1 and RAG2 proteins initiate the process of V(D)J recombination and therefore play an essential role in adaptive immunity. While null mutations in the RAG genes cause severe combined immune deficiency with lack of T and B cells (T- B- SCID) and susceptibility to life-threatening, early-onset infections, studies in humans and mice have demonstrated that hypomorphic RAG mutations are associated with defects of central and peripheral tolerance resulting in immune dysregulation. In this review, we provide an overview of the extended spectrum of RAG deficiencies and their associated clinical and immunological phenotypes in humans. We discuss recent advances in the mechanisms that control RAG expression and function, the effects of perturbed RAG activity on lymphoid development and immune homeostasis, and propose novel approaches to correct this group of disorders.
Collapse
Affiliation(s)
- Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Fieschi C, Viallard JF. [Common variable immunodeficiency disorders: Updated diagnostic criteria and genetics]. Rev Med Interne 2021; 42:465-472. [PMID: 33875312 DOI: 10.1016/j.revmed.2021.03.328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 02/26/2021] [Accepted: 03/21/2021] [Indexed: 12/24/2022]
Abstract
Common variable immunodeficiency disorders (CVID) are a heterogeneous group of conditions with hypogammaglobulinemia as the common denominator. These are the most common symptomatic primary immunodeficiency disorder in adults. Two different clinical forms are described: one group only develops infections, while a second includes (sometimes without infections, at least at the onset of disease course) a variety of non-infectious autoimmune, inflammatory, granulomatous and/or lymphoproliferative manifestations, sometimes revealing the disease and often observed in Internal Medicine. The international diagnostic criteria for CVID were updated in 2016 and are the subject of several comments in this general review. The recent use of new sequencing techniques makes it possible to better genetically define CVID. The identification of such a genetic disease makes it possible to treat pathophysiologically, in particular autoimmune and lymphoproliferative complications, with targeted treatments, sometimes used in other diseases. Determining a genetic disease in these patients also makes it possible to provide appropriate genetic counseling, and therefore to monitor mutated individuals, symptomatic or not.
Collapse
Affiliation(s)
- C Fieschi
- Département d'immunologie, Assistance Publique hôpitaux de Paris (AP-HP), Université de Paris, Paris, France; Inserm U976, institut de recherche Saint-Louis, hôpital Saint-Louis, centre constitutif déficit immunitaire chez l'adulte, CEREDIH, Paris, France
| | - J-F Viallard
- Service de médecine interne et maladies infectieuses, hôpital Haut-Lévêque, CHU de Bordeaux, 5, avenue de Magellan, 33604 Pessac, France; Université de Bordeaux, Bordeaux, France.
| |
Collapse
|
11
|
van Stigt AC, Dik WA, Kamphuis LSJ, Smits BM, van Montfrans JM, van Hagen PM, Dalm VASH, IJspeert H. What Works When Treating Granulomatous Disease in Genetically Undefined CVID? A Systematic Review. Front Immunol 2021; 11:606389. [PMID: 33391274 PMCID: PMC7773704 DOI: 10.3389/fimmu.2020.606389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022] Open
Abstract
Background Granulomatous disease is reported in at least 8–20% of patients with common variable immunodeficiency (CVID). Granulomatous disease mainly affects the lungs, and is associated with significantly higher morbidity and mortality. In half of patients with granulomatous disease, extrapulmonary manifestations are found, affecting e.g. skin, liver, and lymph nodes. In literature various therapies have been reported, with varying effects on remission of granulomas and related clinical symptoms. However, consensus recommendations for optimal management of extrapulmonary granulomatous disease are lacking. Objective To present a literature overview of the efficacy of currently described therapies for extrapulmonary granulomatous disease in CVID (CVID+EGD), compared to known treatment regimens for pulmonary granulomatous disease in CVID (CVID+PGD). Methods The following databases were searched: Embase, Medline (Ovid), Web-of-Science Core Collection, Cochrane Central, and Google Scholar. Inclusion criteria were 1) CVID patients with granulomatous disease, 2) treatment for granulomatous disease reported, and 3) outcome of treatment reported. Patient characteristics, localization of granuloma, treatment, and association with remission of granulomatous disease were extracted from articles. Results We identified 64 articles presenting 95 CVID patients with granulomatous disease, wherein 117 different treatment courses were described. Steroid monotherapy was most frequently described in CVID+EGD (21 out of 53 treatment courses) and resulted in remission in 85.7% of cases. In CVID+PGD steroid monotherapy was described in 15 out of 64 treatment courses, and was associated with remission in 66.7% of cases. Infliximab was reported in CVID+EGD in six out of 53 treatment courses and was mostly used in granulomatous disease affecting the skin (four out of six cases). All patients (n = 9) treated with anti-TNF-α therapies (infliximab and etanercept) showed remission of extrapulmonary granulomatous disease. Rituximab with or without azathioprine was rarely used for CVID+EGD, but frequently used in CVID+PGD where it was associated with remission of granulomatous disease in 94.4% (17 of 18 treatment courses). Conclusion Although the number of CVID+EGD patients was limited, data indicate that steroid monotherapy often results in remission, and that anti-TNF-α treatment is effective for granulomatous disease affecting the skin. Also, rituximab with or without azathioprine was mainly described in CVID+PGD, and only in few cases of CVID+EGD.
Collapse
Affiliation(s)
- Astrid C van Stigt
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Division of Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center, Rotterdam, Netherlands
| | - Willem A Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center, Rotterdam, Netherlands
| | - Lieke S J Kamphuis
- Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Bas M Smits
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Centre (UMC), Utrecht, Netherlands
| | - Joris M van Montfrans
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Centre (UMC), Utrecht, Netherlands
| | - P Martin van Hagen
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Division of Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center, Rotterdam, Netherlands
| | - Virgil A S H Dalm
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Division of Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hanna IJspeert
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands.,Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
12
|
Dhalla F, Lochlainn DJM, Chapel H, Patel SY. Histology of Interstitial Lung Disease in Common Variable Immune Deficiency. Front Immunol 2020; 11:605187. [PMID: 33329602 PMCID: PMC7718002 DOI: 10.3389/fimmu.2020.605187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022] Open
Abstract
Interstitial lung disease (ILD) is an important non-infectious complication in several primary immune deficiencies. In common variable immune deficiency (CVID) it is associated with complex clinical phenotypes and adverse outcomes. The histology of ILD in CVID is heterogeneous and mixed patterns are frequently observed within a single biopsy, including non-necrotising granulomatous inflammation, lymphoid interstitial pneumonitis, lymphoid hyperplasia, follicular bronchiolitis, organizing pneumonia, and interstitial fibrosis; ILD has to be differentiated from lymphoma. The term granulomatous-lymphocytic interstitial lung disease (GLILD), coined to describe the histopathological findings within the lungs of patients with CVID with or without multisystem granulomata, is somewhat controversial as pulmonary granulomata are not always present on histology and the nature of infiltrating lymphocytes is variable. In this mini review we summarize the literature on the histology of CVID-related ILD and discuss some of the factors that may contribute to the inter- and intra- patient variability in the histological patterns reported. Finally, we highlight areas for future development. In particular, there is a need for standardization of histological assessments and reporting, together with a better understanding of the immunopathogenesis of CVID-related ILD to resolve the apparent heterogeneity of ILD in this setting and guide the selection of rational targeted therapies in different patients.
Collapse
Affiliation(s)
- Fatima Dhalla
- Department of Clinical Immunology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom.,Developmental Immunology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Dylan J Mac Lochlainn
- Department of Clinical Immunology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Helen Chapel
- Department of Clinical Immunology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom.,Primary Immunodeficiency Unit, Nuffield Department of Medicine and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Smita Y Patel
- Department of Clinical Immunology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom.,Primary Immunodeficiency Unit, Nuffield Department of Medicine and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Cirillo E, Giardino G, Ricci S, Moschese V, Lougaris V, Conti F, Azzari C, Barzaghi F, Canessa C, Martire B, Badolato R, Dotta L, Soresina A, Cancrini C, Finocchi A, Montin D, Romano R, Amodio D, Ferrua F, Tommasini A, Baselli LA, Dellepiane RM, Polizzi A, Chessa L, Marzollo A, Cicalese MP, Putti MC, Pession A, Aiuti A, Locatelli F, Plebani A, Pignata C. Consensus of the Italian Primary Immunodeficiency Network on transition management from pediatric to adult care in patients affected with childhood-onset inborn errors of immunity. J Allergy Clin Immunol 2020; 146:967-983. [PMID: 32827505 DOI: 10.1016/j.jaci.2020.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/17/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023]
Abstract
Medical advances have dramatically improved the long-term prognosis of children and adolescents with inborn errors of immunity (IEIs). Transfer of the medical care of individuals with pediatric IEIs to adult facilities is also a complex task because of the large number of distinct disorders, which requires involvement of patients and both pediatric and adult care providers. To date, there is no consensus on the optimal pathway of the transitional care process and no specific data are available in the literature regarding patients with IEIs. We aimed to develop a consensus statement on the transition process to adult health care services for patients with IEIs. Physicians from major Italian Primary Immunodeficiency Network centers formulated and answered questions after examining the currently published literature on the transition from childhood to adulthood. The authors voted on each recommendation. The most frequent IEIs sharing common main clinical problems requiring full attention during the transitional phase were categorized into different groups of clinically related disorders. For each group of clinically related disorders, physicians from major Italian Primary Immunodeficiency Network institutions focused on selected clinical issues representing the clinical hallmark during early adulthood.
Collapse
Affiliation(s)
- Emilia Cirillo
- Department of Translational Medical Sciences, Pediatric Section, Federico II University, Naples, Italy
| | - Giuliana Giardino
- Department of Translational Medical Sciences, Pediatric Section, Federico II University, Naples, Italy
| | - Silvia Ricci
- Division of Pediatric Immunology, Department of Health Sciences, University of Florence and Meyer Children's Hospital, Florence, Italy
| | - Viviana Moschese
- Pediatric Immunopathology and Allergology Unit, University of Rome Tor Vergata, Rome, Italy
| | - Vassilios Lougaris
- Department of Clinical and Experimental Sciences, University of Brescia and Department of Pediatrics, ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Francesca Conti
- Unit of Pediatrics, University of Bologna, St. Orsola University Hospital, Bologna, Italy
| | - Chiara Azzari
- Division of Pediatric Immunology, Department of Health Sciences, University of Florence and Meyer Children's Hospital, Florence, Italy
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Clementina Canessa
- Division of Pediatric Immunology, Department of Health Sciences, University of Florence and Meyer Children's Hospital, Florence, Italy
| | - Baldassarre Martire
- Unit of Pediatric and Neonatology, Maternal-Infant Department, Mons A. R. Dimiccoli Hospital, Barletta, Italy
| | - Raffaele Badolato
- Department of Clinical and Experimental Sciences, University of Brescia and Department of Pediatrics, ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Laura Dotta
- Department of Clinical and Experimental Sciences, University of Brescia and Department of Pediatrics, ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Annarosa Soresina
- Department of Clinical and Experimental Sciences, University of Brescia and Department of Pediatrics, ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Caterina Cancrini
- Unit of Immunology and Infectious Diseases, Academic Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Finocchi
- Unit of Immunology and Infectious Diseases, Academic Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Davide Montin
- Division of Pediatric Immunology and Rheumatology, Department of Public Health and Pediatrics, Regina Margherita Children Hospital, University of Turin, Turin, Italy
| | - Roberta Romano
- Department of Translational Medical Sciences, Pediatric Section, Federico II University, Naples, Italy
| | - Donato Amodio
- Unit of Immunology and Infectious Diseases, Academic Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Ferrua
- San Raffaele Telethon Institute for Gene Therapy and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Tommasini
- Department of Pediatrics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Lucia Augusta Baselli
- Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Department of Pediatrics, Milan, Italy
| | - Rosa Maria Dellepiane
- Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Department of Pediatrics, Milan, Italy
| | - Agata Polizzi
- Department of Educational Sciences, University of Catania, Catania, Italy
| | - Luciana Chessa
- Department of Clinical and Molecular Medicine, Sapienza, University of Rome, Rome, Italy
| | - Antonio Marzollo
- Department of Women's and Children's Health, Pediatric Hematology-Oncology Unit, University of Padua, Padua, Italy
| | - Maria Pia Cicalese
- San Raffaele Telethon Institute for Gene Therapy and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Caterina Putti
- Department of Women's and Children's Health, Pediatric Hematology-Oncology Unit, University of Padua, Padua, Italy
| | - Andrea Pession
- Unit of Pediatrics, University of Bologna, St. Orsola University Hospital, Bologna, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Childrens' Hospital, Sapienza, University of Rome, Rome Italy
| | - Alessandro Plebani
- Department of Clinical and Experimental Sciences, University of Brescia and Department of Pediatrics, ASST-Spedali Civili di Brescia, Brescia, Italy
| | - Claudio Pignata
- Department of Translational Medical Sciences, Pediatric Section, Federico II University, Naples, Italy.
| |
Collapse
|
14
|
Verbsky JW, Hintermeyer MK, Simpson PM, Feng M, Barbeau J, Rao N, Cool CD, Sosa-Lozano LA, Baruah D, Hammelev E, Busalacchi A, Rymaszewski A, Woodliff J, Chen S, Bausch-Jurken M, Routes JM. Rituximab and antimetabolite treatment of granulomatous and lymphocytic interstitial lung disease in common variable immunodeficiency. J Allergy Clin Immunol 2020; 147:704-712.e17. [PMID: 32745555 DOI: 10.1016/j.jaci.2020.07.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Granulomatous and lymphocytic interstitial lung disease (GLILD) is a life-threatening complication in patients with common variable immunodeficiency (CVID), but the optimal treatment is unknown. OBJECTIVE Our aim was to determine whether rituximab with azathioprine or mycophenolate mofetil improves the high-resolution computed tomography (HRCT) chest scans and/or pulmonary function test results in patients with CVID and GLILD. METHODS A retrospective chart review of clinical and laboratory data on 39 patients with CVID and GLILD who completed immunosuppressive therapy was performed. Chest HRCT scans, performed before therapy and after the conclusion of therapy, were blinded, randomized, and scored independently by 2 radiologists. Differences between pretreatment and posttreatment HRCT scan scores, pulmonary function test results, and lymphocyte subsets were analyzed. Whole exome sequencing was performed on all patients. RESULTS Immunosuppressive therapy improved patients' HRCT scan scores (P < .0001), forced vital capacity (P = .0017), FEV1 (P = .037), and total lung capacity (P = .013) but not their lung carbon monoxide diffusion capacity (P = .12). Nine patients relapsed and 6 completed retreatment, with 5 of 6 of these patients (83%) having improved HRCT scan scores (P = .063). Relapse was associated with an increased number of B cells (P = .016) and activated CD4 T cells (P = .016). Four patients (10%) had pneumonia while undergoing active treatment, and 2 patients (5%) died after completion of therapy. Eight patients (21%) had a damaging mutation in a gene known to predispose (TNFRSF13B [n = 3]) or cause a CVID-like primary immunodeficiency (CTLA4 [n = 2], KMT2D [n = 2], or BIRC4 [n = 1]). Immunosuppression improved the HRCT scan scores in patients with (P = .0078) and without (P < .0001) a damaging mutation. CONCLUSIONS Immunosuppressive therapy improved the radiographic abnormalities and pulmonary function of patients with GLILD. A majority of patients had sustained remissions.
Collapse
Affiliation(s)
- James W Verbsky
- Division of Pediatric Rheumatology, Medical College Wisconsin, Milwaukee, Wis; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis
| | - Mary K Hintermeyer
- Asthma, Allergy and Clinical Immunology, Children's Wisconsin, Milwaukee, Wis
| | - Pippa M Simpson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Department of Quantitative Health Sciences, Medical College Wisconsin, Milwaukee, Wis
| | - Mingen Feng
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Department of Quantitative Health Sciences, Medical College Wisconsin, Milwaukee, Wis
| | - Jody Barbeau
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Department of Quantitative Health Sciences, Medical College Wisconsin, Milwaukee, Wis
| | - Nagarjun Rao
- Department of Pathology, Aurora Clinical Laboratories/Great Lakes Pathologists, Aurora West Allis Medical Center, West Allis, Wis
| | - Carlyne D Cool
- Department of Pathology and Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo; National Jewish Health, Denver, Colo
| | - Luis A Sosa-Lozano
- Division of Diagnostic Radiology, Medical College of Wisconsin, Milwaukee, Wis
| | - Dhiraj Baruah
- Division of Thoracic Radiology, Medical University of South Carolina, Charleston, SC
| | - Erin Hammelev
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Division of Asthma, Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, Wis
| | - Alyssa Busalacchi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Division of Asthma, Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, Wis
| | - Amy Rymaszewski
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Division of Asthma, Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, Wis
| | - Jeff Woodliff
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Division of Asthma, Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, Wis
| | - Shaoying Chen
- Division of Pediatric Rheumatology, Medical College Wisconsin, Milwaukee, Wis; Division of Asthma, Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, Wis
| | - Mary Bausch-Jurken
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Division of Asthma, Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, Wis
| | - John M Routes
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis; Division of Asthma, Allergy and Clinical Immunology, Medical College of Wisconsin, Milwaukee, Wis.
| |
Collapse
|
15
|
Goldberg L, Simon AJ, Rechavi G, Lev A, Barel O, Kunik V, Toren A, Schiby G, Tamary H, Steinberg-Shemer O, Somech R. Congenital neutropenia with variable clinical presentation in novel mutation of the SRP54 gene. Pediatr Blood Cancer 2020; 67:e28237. [PMID: 32277798 DOI: 10.1002/pbc.28237] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 02/08/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND The SRP54 (signal recognition protein 54) is a conserved component of the ribonucleoprotein complex that mediates cotranslational targeting and translocation of proteins to the endoplasmic reticulum. In 2017, mutations in the gene have been described as a cause of congenital neutropenia with or without pancreatic insufficiency, and since then, only limited cases were added to the literature. METHODS Two patients with neutropenia underwent hematological, immunological, and genetic work-up, including lymphocyte phenotyping, immunoglobulins, and complement levels, antineutrophil and antinuclear antibodies, bone marrow FISH panel for myelodysplastic syndrome, whole-exome sequencing, and in silico proteomic analysis. RESULTS Clinical findings in the two families revealed a wide spectrum of immunological and clinical manifestations, ranging from mild asymptomatic neutropenia during febrile illnesses to severe neutropenia and life-threatening infection requiring leg amputation. Immunological and hematological work-up showed isolated neutropenia with normal lymphocyte subpopulations, immunoglobulin and complement levels, and negative autoimmune tests. Bone marrow aspirations showed variability ranging from normal myelopoiesis to myeloid maturation arrest at the promyelocytic stage, with normal FISH panel for myelodysplastic syndrome. Genetic analysis identified a novel, de novo, in-frame deletion in the SRP54 gene, c.342-344delAAC, p.T115del. In silico proteomic analysis suggested impaired SRP54 protein function due to reduced GTP activity and stability. CONCLUSIONS We describe congenital neutropenia with variable clinical presentation in novel mutation of the SRP54 gene.
Collapse
Affiliation(s)
- Lior Goldberg
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Department A, Pediatric Immunology Service, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation (JMF) Center, Tel HaShomer, Israel
| | - Amos J Simon
- Pediatric Department A, Pediatric Immunology Service, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation (JMF) Center, Tel HaShomer, Israel.,The Wohl Institute for Translational Medicine, Sheba Medical Center, Tel HaShomer, Israel
| | - Gideon Rechavi
- The Wohl Institute for Translational Medicine, Sheba Medical Center, Tel HaShomer, Israel.,Sheba Cancer Research Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Atar Lev
- Pediatric Department A, Pediatric Immunology Service, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation (JMF) Center, Tel HaShomer, Israel
| | - Ortal Barel
- The Wohl Institute for Translational Medicine, Sheba Medical Center, Tel HaShomer, Israel.,Sheba Cancer Research Center, Sheba Medical Center, Tel HaShomer, Israel
| | | | - Amos Toren
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Hemato/Oncology Division and Bone Marrow Transplantation Unit, Tel HaShomer, Israel
| | - Ginette Schiby
- The Wohl Institute for Translational Medicine, Sheba Medical Center, Tel HaShomer, Israel
| | - Hannah Tamary
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Departments of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tivka, Israel
| | - Orna Steinberg-Shemer
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Departments of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tivka, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Department A, Pediatric Immunology Service, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation (JMF) Center, Tel HaShomer, Israel.,The Wohl Institute for Translational Medicine, Sheba Medical Center, Tel HaShomer, Israel
| |
Collapse
|
16
|
Ho HE, Cunningham-Rundles C. Non-infectious Complications of Common Variable Immunodeficiency: Updated Clinical Spectrum, Sequelae, and Insights to Pathogenesis. Front Immunol 2020; 11:149. [PMID: 32117289 PMCID: PMC7025475 DOI: 10.3389/fimmu.2020.00149] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
Non-infectious complications in common variable immunodeficiency (CVID) have emerged as a major clinical challenge. Detailed clinical spectrum, organ-specific pathologies and associated sequelae from 623 CVID patients followed in New York since 1974 were analyzed, and recent insights to pathogenesis were reviewed. Non-infectious manifestations were present in 68.1% of patients, and they do not tend to be present in isolation. They include autoimmunity (33.2%), chronic lung disease (30.3%), lymphoid hyperplasia/splenomegaly (20.9%), liver disease (12.7%), granulomas (9.3%), gastrointestinal disease (7.3%), lymphoma (6.7%), and other malignancies (6.4%). In the lungs, interstitial disease and bronchiectasis were the most common findings, with lymphoma at this site being a rare (n = 6), but serious, manifestation. Bronchiectasis was not a prerequisite for the development of interstitial disease. In the liver, granulomas and nodular regenerative hyperplasia were the most common. Gastrointestinal disease may affect any segment of the intestinal tract, with lymphoid infiltrations and villous blunting being the leading histologic findings. With progression of organ-specific diseases, a wide spectrum of associated sequelae was observed. Lymphoma was more common in females (P = 0.036)—all B cell types except in one subject. Solid organ transplantations (liver, n = 5; lung, n = 4; combined lung and heart, n = 2) and hematopoietic stem cell transplantations (for B cell lymphoma, n = 1) have rarely been performed in this cohort, with mixed outcomes. Recent identification of monogenic defects, in ~10–30% of various CVID cohorts, has highlighted the molecular pathways that can affect both antibody production and broader immune regulation. In addition, cellular defects in both innate and adaptive immune systems are increasingly recognized in this syndrome.
Collapse
Affiliation(s)
- Hsi-En Ho
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charlotte Cunningham-Rundles
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
17
|
Greenberg-Kushnir N, Lee YN, Simon AJ, Lev A, Marcus N, Abuzaitoun O, Somech R, Stauber T. A Large Cohort of RAG1/2-Deficient SCID Patients—Clinical, Immunological, and Prognostic Analysis. J Clin Immunol 2019; 40:211-222. [DOI: 10.1007/s10875-019-00717-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022]
|
18
|
Ameratunga R, Lehnert K, Woon ST. All Patients With Common Variable Immunodeficiency Disorders (CVID) Should Be Routinely Offered Diagnostic Genetic Testing. Front Immunol 2019; 10:2678. [PMID: 31824486 PMCID: PMC6883368 DOI: 10.3389/fimmu.2019.02678] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/30/2019] [Indexed: 12/23/2022] Open
Affiliation(s)
- Rohan Ameratunga
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand.,Department of Clinical Immunology, Auckland City Hospital, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Klaus Lehnert
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - See-Tarn Woon
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
19
|
Aggarwal V, Banday AZ, Jindal AK, Das J, Rawat A. Recent advances in elucidating the genetics of common variable immunodeficiency. Genes Dis 2019; 7:26-37. [PMID: 32181273 PMCID: PMC7063417 DOI: 10.1016/j.gendis.2019.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/19/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Common variable immunodeficiency disorders (CVID), a heterogeneous group of inborn errors of immunity, is the most common symptomatic primary immunodeficiency disorder. Patients with CVID have highly variable clinical presentation. With the advent of whole genome sequencing and genome wide association studies (GWAS), there has been a remarkable improvement in understanding the genetics of CVID. This has also helped in understanding the pathogenesis of CVID and has drastically improved the management of these patients. A multi-omics approach integrating the DNA sequencing along with RNA sequencing, proteomics, epigenetic and metabolomics profile is the need of the hour to unravel specific CVID associated disease pathways and novel therapeutic targets. In this review, we elaborate various techniques that have helped in understanding the genetics of CVID.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aaqib Zaffar Banday
- Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankur Kumar Jindal
- Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jhumki Das
- Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
20
|
Bulkhi AA, Dasso JF, Schuetz C, Walter JE. Approaches to patients with variants in RAG genes: from diagnosis to timely treatment. Expert Rev Clin Immunol 2019; 15:1033-1046. [PMID: 31535575 DOI: 10.1080/1744666x.2020.1670060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Patients with primary immunodeficiency secondary to abnormal recombinase activating genes (RAG) can present with broad clinical phenotypes ranging from early severe infections to autoimmune complications and inflammation. Immunological phenotype may also vary from T-B- severe combined immunodeficiency to combined immunodeficiency or antibody deficiencies with near-normal T and B cell counts and even preserved specific antibody response to pathogens. It is not uncommon that RAG variants of uncertain significance are identified by serendipity during a broad genetic screening process and pathogenic RAG variants are increasingly recognized among all age groups, including adults. Establishing the pathogenicity and clinical relevance of novel RAG variants can be challenging since RAG genes are highly polymorphic. This review paper aims to summarize clinical phenotypes of RAG deficiencies and provide practical guidance for confirming the direct link between specific RAG variants and clinical disease. Lastly, we will review the current understanding of treatment option for patients with varying severity of RAG deficiencies. Area covered: This review discusses the different phenotypes and immunological aspects of RAG deficiencies, the diagnosis dilemma facing clinicians, and an overview of current and advancement in treatments. Expert opinion: A careful analysis of immunological and clinical data and their correlation with genetic findings helps to determine the significance of the genetic polymorphism. Advances in functional assays, as well as anti-cytokine antibodies, make it easier to resolve the diagnostic dilemma.
Collapse
Affiliation(s)
- Adeeb A Bulkhi
- Department of Internal Medicine, College of Medicine, Umm Al-Qura University , Makkah , Saudi Arabia.,Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa , FL , USA
| | - Joseph F Dasso
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| | - Catharina Schuetz
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| | - Jolan E Walter
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa , FL , USA.,Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children's Hospital , St. Petersburg , FL , USA.,Division of Allergy and Immunology, Massachusetts General Hospital for Children , Boston , MA , USA
| |
Collapse
|
21
|
Abstract
While widespread genome sequencing ushers in a new era of preventive medicine, the tools for predictive genomics are still lacking. Time and resource limitations mean that human diseases remain uncharacterized because of an inability to predict clinically relevant genetic variants. A strategy of targeting highly conserved protein regions is used commonly in functional studies. However, this benefit is lost for rare diseases where the attributable genes are mostly conserved. An immunological disorder exemplifying this challenge occurs through damaging mutations in RAG1 and RAG2 which presents at an early age with a distinct phenotype of life-threatening immunodeficiency or autoimmunity. Many tools exist for variant pathogenicity prediction, but these cannot account for the probability of variant occurrence. Here, we present a method that predicts the likelihood of mutation for every amino acid residue in the RAG1 and RAG2 proteins. Population genetics data from approximately 146,000 individuals was used for rare variant analysis. Forty-four known pathogenic variants reported in patients and recombination activity measurements from 110 RAG1/2 mutants were used to validate calculated scores. Probabilities were compared with 98 currently known human cases of disease. A genome sequence dataset of 558 patients who have primary immunodeficiency but that are negative for RAG deficiency were also used as validation controls. We compared the difference between mutation likelihood and pathogenicity prediction. Our method builds a map of most probable mutations allowing pre-emptive functional analysis. This method may be applied to other diseases with hopes of improving preparedness for clinical diagnosis.
Collapse
|
22
|
Cinetto F, Scarpa R, Pulvirenti F, Quinti I, Agostini C, Milito C. Appropriate lung management in patients with primary antibody deficiencies. Expert Rev Respir Med 2019; 13:823-838. [PMID: 31361157 DOI: 10.1080/17476348.2019.1641085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Human primary immunodeficiency diseases (PIDs) include a broad spectrum of more than 350 disorders, involving different branches of the immune system and classified as 'rare diseases.' Predominantly antibody deficiencies (PADs) represent more than half of the PIDs diagnosed in Europe and are often diagnosed in the adulthood. Areas covered: Although PAD could first present with autoimmune or neoplastic features, respiratory infections are frequent and respiratory disease represents a relevant cause of morbidity and mortality. Pulmonary complications may be classified as infection-related (acute and chronic), immune-mediated, and neoplastic. Expert opinion: At present, no consensus guidelines are available on how to monitor and manage lung complications in PAD patients. In this review, we will discuss the available diagnostic, prognostic and therapeutic instruments and we will suggest an appropriate and evidence-based approach to lung diseases in primary antibody deficiencies. We will also highlight the possible role of promising new tools and strategies in the management of pulmonary complications. However, future studies are needed to reduce of diagnostic delay of PAD and to better understand lung diseases mechanisms, with the final aim to ameliorate therapeutic options that will have a strong impact on Quality of Life and long-term prognosis of PAD patients.
Collapse
Affiliation(s)
- Francesco Cinetto
- Department of Medicine - DIMED, University of Padova , Padova , Italy.,Internal Medicine I, Ca' Foncello Hospital , Treviso , Italy
| | - Riccardo Scarpa
- Department of Medicine - DIMED, University of Padova , Padova , Italy.,Internal Medicine I, Ca' Foncello Hospital , Treviso , Italy
| | - Federica Pulvirenti
- Department of Molecular Medicine, "Sapienza" University of Roma , Roma , Italy
| | - Isabella Quinti
- Department of Molecular Medicine, "Sapienza" University of Roma , Roma , Italy
| | - Carlo Agostini
- Department of Medicine - DIMED, University of Padova , Padova , Italy.,Internal Medicine I, Ca' Foncello Hospital , Treviso , Italy
| | - Cinzia Milito
- Department of Molecular Medicine, "Sapienza" University of Roma , Roma , Italy
| |
Collapse
|
23
|
Farmer JR, Foldvari Z, Ujhazi B, De Ravin SS, Chen K, Bleesing JJH, Schuetz C, Al-Herz W, Abraham RS, Joshi AY, Costa-Carvalho BT, Buchbinder D, Booth C, Reiff A, Ferguson PJ, Aghamohammadi A, Abolhassani H, Puck JM, Adeli M, Cancrini C, Palma P, Bertaina A, Locatelli F, Di Matteo G, Geha RS, Kanariou MG, Lycopoulou L, Tzanoudaki M, Sleasman JW, Parikh S, Pinero G, Fischer BM, Dbaibo G, Unal E, Patiroglu T, Karakukcu M, Al-Saad KK, Dilley MA, Pai SY, Dutmer CM, Gelfand EW, Geier CB, Eibl MM, Wolf HM, Henderson LA, Hazen MM, Bonfim C, Wolska-Kuśnierz B, Butte MJ, Hernandez JD, Nicholas SK, Stepensky P, Chandrakasan S, Miano M, Westermann-Clark E, Goda V, Kriván G, Holland SM, Fadugba O, Henrickson SE, Ozen A, Karakoc-Aydiner E, Baris S, Kiykim A, Bredius R, Hoeger B, Boztug K, Pashchenko O, Neven B, Moshous D, Villartay JPD, Bousfiha AA, Hill HR, Notarangelo LD, Walter JE. Outcomes and Treatment Strategies for Autoimmunity and Hyperinflammation in Patients with RAG Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2019; 7:1970-1985.e4. [PMID: 30877075 PMCID: PMC6612449 DOI: 10.1016/j.jaip.2019.02.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Although autoimmunity and hyperinflammation secondary to recombination activating gene (RAG) deficiency have been associated with delayed diagnosis and even death, our current understanding is limited primarily to small case series. OBJECTIVE Understand the frequency, severity, and treatment responsiveness of autoimmunity and hyperinflammation in RAG deficiency. METHODS In reviewing the literature and our own database, we identified 85 patients with RAG deficiency, reported between 2001 and 2016, and compiled the largest case series to date of 63 patients with prominent autoimmune and/or hyperinflammatory pathology. RESULTS Diagnosis of RAG deficiency was delayed a median of 5 years from the first clinical signs of immune dysregulation. Most patients (55.6%) presented with more than 1 autoimmune or hyperinflammatory complication, with the most common etiologies being cytopenias (84.1%), granulomas (23.8%), and inflammatory skin disorders (19.0%). Infections, including live viral vaccinations, closely preceded the onset of autoimmunity in 28.6% of cases. Autoimmune cytopenias had early onset (median, 1.9, 2.1, and 2.6 years for autoimmune hemolytic anemia, immune thrombocytopenia, and autoimmune neutropenia, respectively) and were refractory to intravenous immunoglobulin, steroids, and rituximab in most cases (64.7%, 73.7%, and 71.4% for autoimmune hemolytic anemia, immune thrombocytopenia, and autoimmune neutropenia, respectively). Evans syndrome specifically was associated with lack of response to first-line therapy. Treatment-refractory autoimmunity/hyperinflammation prompted hematopoietic stem cell transplantation in 20 patients. CONCLUSIONS Autoimmunity/hyperinflammation can be a presenting sign of RAG deficiency and should prompt further evaluation. Multilineage cytopenias are often refractory to immunosuppressive treatment and may require hematopoietic cell transplantation for definitive management.
Collapse
Affiliation(s)
- Jocelyn R Farmer
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Zsofia Foldvari
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | - Boglarka Ujhazi
- University of South Florida and Johns Hopkins All Children's Hospital, Saint Petersburg, Fla
| | - Suk See De Ravin
- Laboratory of Host Defenses, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, Md
| | - Karin Chen
- Division of Allergy and Immunology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Jack J H Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Catharina Schuetz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Waleed Al-Herz
- Faculty of Medicine, Pediatrics Department, Kuwait University, Kuwait City, Kuwait; Allergy and Clinical Immunology Unit, Pediatrics Department, Alsabah Hospital, Kuwait City, Kuwait
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | - Avni Y Joshi
- Division of Pediatric Allergy/Immunology, Mayo Clinic Children's Center Rochester, Rochester, Minn
| | | | - David Buchbinder
- Pediatrics/Hematology, CHOC Children's Hospital - UC Irvine, Irvine, Calif
| | - Claire Booth
- Department of Paediatric Immunology, Great Ormond Street Hospital, London, United Kingdom
| | - Andreas Reiff
- Division of Rheumatology, Children's Hospital Los Angeles, Keck School of Medicine, USC, Los Angeles, Calif
| | - Polly J Ferguson
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jennifer M Puck
- Department of Pediatrics, University of California San Francisco and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Mehdi Adeli
- Sidra Medicine, Weill Cornell Medicine, and Hamad Medical Corporation, Doha, Qatar
| | - Caterina Cancrini
- Academic Department of Pediatrics (DPUO), Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Palma
- Academic Department of Pediatrics (DPUO), Research Unit in Congenital and Perinatal Infection, Children's Hospital Bambino Gesù, Rome, Italy
| | - Alice Bertaina
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Bambino Gesù, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Bambino Gesù, Rome, Italy; Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
| | - Gigliola Di Matteo
- Academic Department of Pediatrics (DPUO), Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Raif S Geha
- Immunology Division, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Maria G Kanariou
- Department of Immunology - Histocompatibility, Specialized Center & Referral Center for Primary Immunodeficiencies - Paediatric Immunology, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Lilia Lycopoulou
- 1st Department of Pediatrics, University of Athens, Aghia Sofia Children's Hospital, Athens, Greece
| | - Marianna Tzanoudaki
- Department of Immunology - Histocompatibility, Specialized Center & Referral Center for Primary Immunodeficiencies - Paediatric Immunology, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - John W Sleasman
- Division of Allergy, Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, NC
| | - Suhag Parikh
- Division of Pediatric Blood and Marrow Transplantation, Duke University School of Medicine, Durham, NC
| | - Gloria Pinero
- Division of Allergy, Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, NC
| | - Bernard M Fischer
- Division of Allergy, Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, NC
| | - Ghassan Dbaibo
- Department of Pediatrics and Adolescent Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Ekrem Unal
- Division of Pediatric Hematology and Oncology & HCST Unit, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Turkan Patiroglu
- Division of Pediatric Hematology and Oncology & HCST Unit, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey; Division of Pediatric Immunology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Musa Karakukcu
- Division of Pediatric Hematology and Oncology & HCST Unit, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Khulood Khalifa Al-Saad
- Salmanyia Medical Complex, Department of Pediatrics, Division of Pediatric Hematology and Oncology, Manama, Bahrain
| | - Meredith A Dilley
- Department of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Sung-Yun Pai
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, Mass; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Cullen M Dutmer
- Division of Allergy & Immunology, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colo
| | - Erwin W Gelfand
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | - Martha M Eibl
- Immunology Outpatient Clinic, Vienna, Austria; Biomedizinische Forschungs GmbH, Vienna, Austria
| | - Hermann M Wolf
- Immunology Outpatient Clinic, Vienna, Austria; Sigmund Freud Private University-Medical School, Vienna, Austria
| | - Lauren A Henderson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Melissa M Hazen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Carmem Bonfim
- Hospital Infantil Pequeno Principe, Curitiba, Brazil
| | | | - Manish J Butte
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics and Jeffrey Modell Diagnostic and Research Center, University of California, Los Angeles, Los Angeles, Calif
| | - Joseph D Hernandez
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, Stanford University, Stanford, Calif
| | - Sarah K Nicholas
- Section of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Polina Stepensky
- Department of Bone Marrow Transplantation, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Shanmuganathan Chandrakasan
- Division of Bone Marrow Transplant, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Ga
| | - Maurizio Miano
- Haematology Unit, Department of Pediatric Haematology-Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Emma Westermann-Clark
- Department of Internal Medicine, Division of Allergy/Immunology, University of South Florida Morsani College of Medicine, Tampa, Fla
| | - Vera Goda
- Department for Pediatric Hematology and Hemopoietic Stem Cell Transplantation, Central Hospital of Southern Pest- National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Gergely Kriván
- Department for Pediatric Hematology and Hemopoietic Stem Cell Transplantation, Central Hospital of Southern Pest- National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Md
| | - Olajumoke Fadugba
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Sarah E Henrickson
- Allergy Immunology Division, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pa; Institute for Immunology, the University of Pennsylvania, Philadelphia, Pa
| | - Ahmet Ozen
- Marmara University School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Marmara University School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkey
| | - Safa Baris
- Marmara University School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkey
| | - Ayca Kiykim
- Ministry of Health, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Robbert Bredius
- Department of Pediatrics, Section Pediatric Immunology, Infections and Stem Cell Transplantation, Leiden University Medical Center, Leiden, the Netherlands
| | - Birgit Hoeger
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; St Anna Kinderspital and Children's Cancer Research Institute, Department of Pediatrics, Medical University of Vienna, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Olga Pashchenko
- Department of Immunology, Pirogov Russian National Research Medical University, Russian Clinical Children's Hospital, Moscow, Russia
| | - Benedicte Neven
- Paris Descartes Sorbonne Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory "Immunogenetics of Pediatric Autoimmune Diseases", INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Despina Moshous
- Paris Descartes Sorbonne Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory "Genome Dynamics in The Immune System", INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Jean-Pierre de Villartay
- Laboratory "Genome Dynamics in The Immune System", INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Ahmed Aziz Bousfiha
- Laboratoire d'Immunologie Clinique, d'Inflammation et d'Allergie LICIA, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco; Clinical Immunology Unit, Casablanca Children's Hospital, Ibn Rochd Medical School, Hassan II University, Casablanca, Morocco
| | - Harry R Hill
- Division of Clinical Immunology, Departments of Pathology, Pediatrics and Medicine, University of Utah, Salt Lake City, Utah
| | - Luigi D Notarangelo
- Haematology Unit, Department of Pediatric Haematology-Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Jolan E Walter
- University of South Florida and Johns Hopkins All Children's Hospital, Saint Petersburg, Fla; Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, Mass.
| |
Collapse
|
24
|
Chandrakasan S, Chandra S, Davila Saldana BJ, Torgerson TR, Buchbinder D. Primary immune regulatory disorders for the pediatric hematologist and oncologist: A case-based review. Pediatr Blood Cancer 2019; 66:e27619. [PMID: 30697957 DOI: 10.1002/pbc.27619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/20/2022]
Abstract
An array of monogenic immune defects marked by autoimmunity, lymphoproliferation, and hyperinflammation rather than infections have been described. Primary immune regulatory disorders pose a challenge to pediatric hematologists and oncologists. This paper focuses on primary immune regulatory disorders including autoimmune lymphoproliferative syndrome (ALPS) and ALPS-like syndromes, immunodysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) and IPEX-like disorders, common variable immunodeficiency (CVID), CVID-like, and late-onset combined immunodeficiency (CID) disorders. Hyperinflammatory disorders and those associated with increased susceptibility to lymphoid malignancies are also discussed. Using a case-based approach, a review of clinical pearls germane to the clinical and laboratory evaluation as well as the treatment of these disorders is provided.
Collapse
Affiliation(s)
- Shanmuganathan Chandrakasan
- Division of Bone Marrow Transplant, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Blachy J Davila Saldana
- Division of Blood and Marrow Transplantation, Children's National Medical Center, Washington, District of Columbia.,Department of Pediatrics, The George Washington University, Washington, District of Columbia
| | - Troy R Torgerson
- Department of Pediatrics, Divisions of Immunology/Rheumatology University of Washington and Seattle Children's Hospital, Seattle, Washington
| | - David Buchbinder
- Department of Hematology, Children's Hospital of Orange County, Orange, California.,Department of Pediatrics, University of California at Irvine, Orange, California
| |
Collapse
|
25
|
Abstract
Recombination-activating genes (
RAG)
1 and
RAG2 initiate the molecular processes that lead to lymphocyte receptor formation through VDJ recombination. Nonsense mutations in
RAG1/
RAG2 cause the most profound immunodeficiency syndrome, severe combined immunodeficiency (SCID). Other severe and less-severe clinical phenotypes due to mutations in
RAG genes are now recognized. The degree of residual protein function may permit some lymphocyte receptor formation, which confers a less-severe clinical phenotype. Many of the non-SCID phenotypes are associated with autoimmunity. New findings into the effect of mutations in
RAG1/2 on the developing T- and B-lymphocyte receptor give insight into the development of autoimmunity. This article summarizes recent findings and places the genetic and molecular findings in a clinical context.
Collapse
Affiliation(s)
- Andrew Gennery
- Paediatric Immunology and Haematopoietic Stem Cell Transplantation, Great North Childrens' Hospital, Newcastle upon Tyne, UK.,Primary Immunodeficiency Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
26
|
Meshaal SS, El Hawary RE, Abd Elaziz DS, Eldash A, Alkady R, Lotfy S, Mauracher AA, Opitz L, Pachlopnik Schmid J, van der Burg M, Chou J, Galal NM, Boutros JA, Geha R, Elmarsafy AM. Phenotypical heterogeneity in RAG-deficient patients from a highly consanguineous population. Clin Exp Immunol 2019; 195:202-212. [PMID: 30307608 PMCID: PMC6330646 DOI: 10.1111/cei.13222] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2018] [Indexed: 12/16/2022] Open
Abstract
Mutations affecting recombination activation genes RAG1 and RAG2 are associated with variable phenotypes, depending on the residual recombinase activity. The aim of this study is to describe a variety of clinical phenotypes in RAG-deficient patients from the highly consanguineous Egyptian population. Thirty-one patients with RAG mutations (from 28 families) were included from 2013 to 2017. On the basis of clinical, immunological and genetic data, patients were subdivided into three groups; classical T- B- severe combined immunodeficiency (SCID), Omenn syndrome (OS) and atypical SCID. Nineteen patients presented with typical T- B- SCID; among these, five patients carried a homozygous RAG2 mutation G35V and five others carried two homozygous RAG2 mutations (T215I and R229Q) that were detected together. Four novel mutations were reported in the T- B- SCID group; three in RAG1 (A565P, N591Pfs*14 and K621E) and one in RAG2 (F29S). Seven patients presented with OS and a novel RAG2 mutation (C419W) was documented in one patient. The atypical SCID group comprised five patients. Two had normal B cell counts; one had a previously undescribed RAG2 mutation (V327D). The other three patients presented with autoimmune cytopaenias and features of combined immunodeficiency and were diagnosed at a relatively late age and with a substantial diagnostic delay; one patient had a novel RAG1 mutation (C335R). PID disorders are frequent among Egyptian children because of the high consanguinity. RAG mutations stand behind several variable phenotypes, including classical SCID, OS, atypical SCID with autoimmunity and T- B+ CID.
Collapse
Affiliation(s)
- S. S. Meshaal
- Clinical Pathology Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - R. E. El Hawary
- Clinical Pathology Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - D. S. Abd Elaziz
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - A. Eldash
- Clinical Pathology Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - R. Alkady
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - S. Lotfy
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - A. A. Mauracher
- Division of ImmunologyUniversity Children’s Hospital ZurichZurichSwitzerland
| | - L. Opitz
- Functional Genomics Center ZürichUniversity of Zurich, ETH ZurichZurichSwitzerland
| | | | - M. van der Burg
- Department of ImmunologyErasmus MC, University Medical Center RotterdamRotterdamNetherlands
| | - J. Chou
- Division of ImmunologyBoston Children’s Hospital, Harvard Medical SchoolBostonMAUSA
| | - N. M. Galal
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - J. A. Boutros
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - R. Geha
- Division of ImmunologyBoston Children’s Hospital, Harvard Medical SchoolBostonMAUSA
| | - A. M. Elmarsafy
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| |
Collapse
|
27
|
Tillman R, Guillerman RP, Trojan T, Silva-Carmona M, Chinn IK. Treatment-Responsive Granulomatous-Lymphocytic Interstitial Lung Disease in a Pediatric Case of Common Variable Immunodeficiency. Front Pediatr 2019; 7:105. [PMID: 30984724 PMCID: PMC6449420 DOI: 10.3389/fped.2019.00105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/05/2019] [Indexed: 12/17/2022] Open
Abstract
Granulomatous-Lymphocytic Interstitial Lung disease (GLILD) is a granulomatous and lymphoproliferative condition occurring in ~25% of Common Variable Immunodeficiency (CVID) patients with the highest prevalence in the late teen to young adult years. GLILD was first described in adults and carries a poor prognosis with survival estimated to be reduced by half. Here we report a pediatric case of CVID-associated GLILD that presented with rapid deterioration over 3 months and responded to adult-based treatment with dual chemotherapeutic agents (rituximab and azathioprine), resulting in complete resolution of clinical findings and near complete resolution of radiologic findings. This case highlights the opportunity to achieve a favorable outcome in GLILD following appropriate diagnosis and therapy.
Collapse
Affiliation(s)
- Robert Tillman
- Pediatric Pulmonary, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - R Paul Guillerman
- Pediatric Radiology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Timothy Trojan
- Allergy Immunology, Allergy Partners of Oklahoma, Endid, OK, United States
| | - Manuel Silva-Carmona
- Pediatric Pulmonary, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States.,Pediatric Critical Care, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Ivan K Chinn
- Pediatric Allergy and Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
28
|
Dorna MB, Barbosa PFA, Rangel-Santos A, Csomos K, Ujhazi B, Dasso JF, Thwaites D, Boyes J, Savic S, Walter JE. Combined Immunodeficiency With Late-Onset Progressive Hypogammaglobulinemia and Normal B Cell Count in a Patient With RAG2 Deficiency. Front Pediatr 2019; 7:122. [PMID: 31058115 PMCID: PMC6477099 DOI: 10.3389/fped.2019.00122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/12/2019] [Indexed: 11/13/2022] Open
Abstract
Proteins expressed by recombination activating genes 1 and 2 (RAG1/2) are essential in the process of V(D)J recombination that leads to generation of the T and B cell repertoires. Clinical and immunological phenotypes of patients with RAG deficiencies correlate well to the degree of impaired RAG activity and this has been expanding to variants of combined immunodeficiency (CID) or even milder antibody deficiency syndromes. Pathogenic variants that severely impair recombinase activity of RAG1/2 determine a severe combined immunodeficiency (SCID) phenotype, whereas hypomorphic variants result in leaky (partial) SCID and other immunodeficiencies. We report a patient with novel pathogenic compound heterozygous RAG2 variants that result in a CID phenotype with two distinctive characteristics: late-onset progressive hypogammaglobulinemia and highly elevated B cell count. In addition, the patient had early onset of infections, T cell lymphopenia and expansion of lymphocytes after exposure to herpes family viruses. This case highlights the importance of considering pathogenic RAG variants among patients with preserved B cell count and CID phenotype.
Collapse
Affiliation(s)
- Mayra B Dorna
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Pamela F A Barbosa
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Andréia Rangel-Santos
- Laboratory of Medical Investigation (LIM 36), Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, São Paulo, Brazil
| | - Krisztian Csomos
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Boglarka Ujhazi
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Joseph F Dasso
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Biology, University of Tampa, Tampa, FL, United States
| | - Daniel Thwaites
- School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Joan Boyes
- School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Sinisa Savic
- Department of Clinical Immunology and Allergy, Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James's University Hospital, Leeds, United Kingdom
| | - Jolan E Walter
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States.,Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
29
|
Villa A, Notarangelo LD. RAG gene defects at the verge of immunodeficiency and immune dysregulation. Immunol Rev 2019; 287:73-90. [PMID: 30565244 PMCID: PMC6309314 DOI: 10.1111/imr.12713] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022]
Abstract
Mutations of the recombinase activating genes (RAG) in humans underlie a broad spectrum of clinical and immunological phenotypes that reflect different degrees of impairment of T- and B-cell development and alterations of mechanisms of central and peripheral tolerance. Recent studies have shown that this phenotypic heterogeneity correlates, albeit imperfectly, with different levels of recombination activity of the mutant RAG proteins. Furthermore, studies in patients and in newly developed animal models carrying hypomorphic RAG mutations have disclosed various mechanisms underlying immune dysregulation in this condition. Careful annotation of clinical outcome and immune reconstitution in RAG-deficient patients who have received hematopoietic stem cell transplantation has shown that progress has been made in the treatment of this disease, but new approaches remain to be tested to improve stem cell engraftment and durable immune reconstitution. Finally, initial attempts have been made to treat RAG deficiency with gene therapy.
Collapse
Affiliation(s)
- Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
30
|
Differentiation of Common Variable Immunodeficiency From IgG Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 7:1277-1284. [PMID: 30557717 DOI: 10.1016/j.jaip.2018.12.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/02/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) and IgG deficiency are 2 of the more prevalent primary humoral immune defects. The former is defined by consensus with criteria for quantitative and qualitative antibody defects, whereas the latter is used to describe patients with reduced IgG, who commonly have recurrent sinopulmonary infections but do not fulfill CVID criteria. However, these patients are often given this diagnosis. OBJECTIVE To compare immunologic findings and clinical manifestations of 2 large cohorts of patients with CVID or IgG deficiency to better delineate differences between these syndromes. METHODS We extracted clinical and laboratory data from electronic medical records of patients at our institution who had received International Classification of Disease codes for either CVID, or IgG deficiency. We gathered immunoglobulin levels, lymphocyte subpopulation counts, and serological vaccine responses. In some patients, we performed flow cytometry to determine percentages of memory and switched-memory B cells. We compiled and statistically compared clinical data related to infectious manifestations, bronchiectasis, autoimmune diseases, infiltrative inflammatory processes, and lymphoid malignancies. RESULTS In contrast to IgG-deficient patients, we found that patients with CVID had lower IgG levels, greater unresponsiveness to most vaccines, lower percentages of memory and isotype switched-memory B cells, and lower CD4 T-cell counts. Clinically, patients with CVID presented similar rates of sinusitis and pneumonias, but a significantly higher prevalence of bronchiectasis and especially noninfectious complications. CONCLUSIONS CVID and IgG deficiency do not share the same disease spectrum, the former being associated with immunodysregulative manifestations and markers of a more severe immune defect. These data may allow clinicians to distinguish these conditions and the management differences that these patients pose.
Collapse
|
31
|
Maffucci P, Chavez J, Jurkiw TJ, O’Brien PJ, Abbott JK, Reynolds PR, Worth A, Notarangelo LD, Felgentreff K, Cortes P, Boisson B, Radigan L, Cobat A, Dinakar C, Ehlayel M, Ben-Omran T, Gelfand EW, Casanova JL, Cunningham-Rundles C. Biallelic mutations in DNA ligase 1 underlie a spectrum of immune deficiencies. J Clin Invest 2018; 128:5489-5504. [PMID: 30395541 PMCID: PMC6264644 DOI: 10.1172/jci99629] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/04/2018] [Indexed: 12/30/2022] Open
Abstract
We report the molecular, cellular, and clinical features of 5 patients from 3 kindreds with biallelic mutations in the autosomal LIG1 gene encoding DNA ligase 1. The patients exhibited hypogammaglobulinemia, lymphopenia, increased proportions of circulating γδT cells, and erythrocyte macrocytosis. Clinical severity ranged from a mild antibody deficiency to a combined immunodeficiency requiring hematopoietic stem cell transplantation. Using engineered LIG1-deficient cell lines, we demonstrated chemical and radiation defects associated with the mutant alleles, which variably impaired the DNA repair pathway. We further showed that these LIG1 mutant alleles are amorphic or hypomorphic, and exhibited variably decreased enzymatic activities, which lead to premature release of unligated adenylated DNA. The variability of the LIG1 genotypes in the patients was consistent with that of their immunological and clinical phenotypes. These data suggest that different forms of autosomal recessive, partial DNA ligase 1 deficiency underlie an immunodeficiency of variable severity.
Collapse
Affiliation(s)
- Patrick Maffucci
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
- Graduate School of Biomedical Sciences, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jose Chavez
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
| | - Thomas J. Jurkiw
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick J. O’Brien
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jordan K. Abbott
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Paul R. Reynolds
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Austen Worth
- Department of Pediatric Medicine, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Luigi D. 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
| | - Kerstin Felgentreff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Patricia Cortes
- Department of Molecular, Cellular and Biomedical Science, CUNY School of Medicine, City College of New York, New York, New York, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Paris Descartes University, Imagine Institute, Paris, France
| | - Lin Radigan
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
| | - Aurélie Cobat
- Paris Descartes University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
| | - Chitra Dinakar
- Allergy, Asthma & Immunodeficiency, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Mohammad Ehlayel
- Section of Pediatric Allergy-Immunology, Department of Pediatrics, Weill Cornell Medical College, Hamad Medical Corporation, Doha, Qatar
| | - Tawfeg Ben-Omran
- Department of Clinical and Metabolic Genetics, Department of Pediatrics, Weill Cornell Medical College, Hamad Medical Corporation, Doha, Qatar
| | - Erwin W. Gelfand
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Paris Descartes University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, Paris, France
| | - Charlotte Cunningham-Rundles
- Division of Clinical Immunology, Departments of Medicine and Pediatrics, and
- Graduate School of Biomedical Sciences, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
32
|
Henrickson SE, Walter JE, Quinn C, Kanakry JA, Bardakjian T, Dimitrova D, Ujhazi B, Csomos K, Bosticardo M, Dobbs K, Nasrallah M, Notarangelo LD, Holland SM, Fadugba O. Adult-Onset Myopathy in a Patient with Hypomorphic RAG2 Mutations and Combined Immune Deficiency. J Clin Immunol 2018; 38:642-645. [PMID: 30159811 DOI: 10.1007/s10875-018-0538-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/30/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Sarah E Henrickson
- The Children's Hospital of Philadelphia, Division of Allergy and Immunology and Institute for Immunology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Jolan E Walter
- Morsani College of Medicine, Division of Allergy and Immunology, University of South Florida, Tampa, FL, 33620, USA
- Johns Hopkins All Children's Hospital, St. Petersburg, FL, 33701, USA
| | - Colin Quinn
- Perelman School of Medicine, Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jennifer A Kanakry
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tanya Bardakjian
- Perelman School of Medicine, Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dimana Dimitrova
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Boglarka Ujhazi
- Morsani College of Medicine, Division of Allergy and Immunology, University of South Florida, Tampa, FL, 33620, USA
- Johns Hopkins All Children's Hospital, St. Petersburg, FL, 33701, USA
| | - Krisztian Csomos
- Morsani College of Medicine, Division of Allergy and Immunology, University of South Florida, Tampa, FL, 33620, USA
- Johns Hopkins All Children's Hospital, St. Petersburg, FL, 33701, USA
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD, USA
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD, USA
| | - MacLean Nasrallah
- Perelman School of Medicine, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD, USA
| | - Olajumoke Fadugba
- Perelman School of Medicine, Division of Pulmonary, Allergy and Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
33
|
Cinetto F, Scarpa R, Rattazzi M, Agostini C. The broad spectrum of lung diseases in primary antibody deficiencies. Eur Respir Rev 2018; 27:27/149/180019. [PMID: 30158276 PMCID: PMC9488739 DOI: 10.1183/16000617.0019-2018] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022] Open
Abstract
Human primary immunodeficiency diseases (PIDs) represent a heterogeneous group of more than 350 disorders. They are rare diseases, but their global incidence is more relevant than generally thought. The underlying defect may involve different branches of the innate and/or adaptive immune response. Thus, the clinical picture may range from severe phenotypes characterised by a broad spectrum of infections to milder infectious phenotypes due to more selective (and frequent) immune defects. Moreover, infections may not be the main clinical features in some PIDs that might present with autoimmunity, auto-inflammation and/or cancer. Primary antibody deficiencies (PADs) represent a small percentage of the known PIDs but they are the most frequently diagnosed, particularly in adulthood. Common variable immunodeficiency (CVID) is the most prevalent symptomatic PAD. PAD patients share a significant susceptibility to respiratory diseases that represent a relevant cause of morbidity and mortality. Pulmonary complications include acute and chronic infection-related diseases, such as pneumonia and bronchiectasis. They also include immune-mediated interstitial lung diseases, such as granulomatous-lymphocytic interstitial lung disease (GLILD) and cancer. Herein we will discuss the main pulmonary manifestations of PADs, the associated functional and imaging findings, and the relevant role of pulmonologists and chest radiologists in diagnosis and surveillance. The spectrum of lung complications in primary antibody deficiency ranges from asthma or COPD to extremely rare and specific ILDs. Early diagnosis of the underlying immune defect might significantly improve patients' lung disease, QoL and long-term prognosis.http://ow.ly/5cP230kZvOB
Collapse
Affiliation(s)
- Francesco Cinetto
- Dept of Medicine - DIMED, University of Padova, Padova, Italy.,Medicina Interna I, Ca' Foncello Hospital, Treviso, Italy
| | - Riccardo Scarpa
- Dept of Medicine - DIMED, University of Padova, Padova, Italy.,Medicina Interna I, Ca' Foncello Hospital, Treviso, Italy
| | - Marcello Rattazzi
- Dept of Medicine - DIMED, University of Padova, Padova, Italy.,Medicina Interna I, Ca' Foncello Hospital, Treviso, Italy
| | - Carlo Agostini
- Dept of Medicine - DIMED, University of Padova, Padova, Italy.,Medicina Interna I, Ca' Foncello Hospital, Treviso, Italy
| |
Collapse
|
34
|
Baumann U, Routes JM, Soler-Palacín P, Jolles S. The Lung in Primary Immunodeficiencies: New Concepts in Infection and Inflammation. Front Immunol 2018; 9:1837. [PMID: 30147696 PMCID: PMC6096054 DOI: 10.3389/fimmu.2018.01837] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022] Open
Abstract
Immunoglobulin replacement therapy (IGRT) has contributed critically to the management of primary antibody deficiencies (PAD) and the decrease in pneumonia rate. However, despite adequate IGRT and improved prognosis, patients with PAD continue to experience recurrent respiratory tract infections, leading to bronchiectasis and continuing decline in lung function with a severe impact on their quality of life. Moreover, non-infectious inflammatory and interstitial lung complications, such as granulomatous-lymphocytic interstitial lung disease, contribute substantially to the overall morbidity of PAD. These conditions develop much more often than appreciated and represent a major therapeutic challenge. Therefore, a regular assessment of the structural and functional condition of the lung and the upper airways with appropriate treatment is required to minimize the deterioration of lung function. This work summarizes the knowledge on lung complications in PAD and discusses the currently available diagnostic tools and treatment options.
Collapse
Affiliation(s)
- Ulrich Baumann
- Department of Paediatric Pulmonology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| | - John M Routes
- Division of Asthma, Allergy and Clinical Immunology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Pere Soler-Palacín
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, United Kingdom
| |
Collapse
|
35
|
Delmonte OM, Schuetz C, Notarangelo LD. RAG Deficiency: Two Genes, Many Diseases. J Clin Immunol 2018; 38:646-655. [PMID: 30046960 PMCID: PMC6643099 DOI: 10.1007/s10875-018-0537-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE To review the clinical and laboratory spectrum of RAG gene defects in humans, and discuss the mechanisms underlying phenotypic heterogeneity, the basis of immune dysregulation, and the current and perspective treatment modalities. METHODS Literature review and analysis of medical records RESULTS: RAG gene defects in humans are associated with a surprisingly broad spectrum of clinical and immunological phenotypes. Correlation between in vitro recombination activity of the mutant RAG proteins and the clinical phenotype has been observed. Altered T and B cell development in this disease is associated with defects of immune tolerance. Hematopoietic cell transplantation is the treatment of choice for the most severe forms of the disease, but a high rate of graft failure has been observed. CONCLUSIONS Phenotypic heterogeneity of RAG gene defects in humans may represent a diagnostic challenge. There is a need to improve treatment for severe, early-onset forms of the disease. Optimal treatment modalities for patients with delayed-onset disease presenting with autoimmunity and/or inflammation remain to be defined.
Collapse
Affiliation(s)
- Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Catharina Schuetz
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
36
|
Ameratunga R, Ahn Y, Jordan A, Lehnert K, Brothers S, Woon ST. Keeping it in the family: the case for considering late-onset combined immunodeficiency a subset of common variable immunodeficiency disorders. Expert Rev Clin Immunol 2018; 14:549-556. [PMID: 29806948 DOI: 10.1080/1744666x.2018.1481750] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Common variable immunodeficiency disorders (CVID) are the most frequent symptomatic primary immune defect in adults. Within the broad spectrum of CVID, a proportion of patients present with a predominant T cell phenotype associated with increased mortality. These patients are termed late-onset combined immunodeficiency (LOCID) and are currently separated from patients suffering from CVID. Areas covered: We have recently codiscovered a new CVID-like disorder caused by mutations of the NFKB1 gene. Members of this non-consanguineous New Zealand kindred have a very diverse spectrum of phenotypes in spite of carrying the identical mutation. The proband appears to have the autoimmune variant. The proband's recently deceased sister best matched LOCID while other family members are less severely affected, including one asymptomatic adult brother, who has an affected daughter. Differences in genetics was one of the main arguments for separating these disorders in the past. Expert commentary: Given the recent advances in the understanding of the genetic basis of these conditions, we present the case that LOCID should now be considered a subset of CVID, rather than a separate disorder. At a clinical level, this distinction is less important but it is imperative these patients are carefully evaluated, the relevant complications are treated, and they are offered prognostic information.
Collapse
Affiliation(s)
- Rohan Ameratunga
- a Department of Virology and Immunology , Auckland City Hospital , Auckland , New Zealand.,b Department of Clinical Immunology , Auckland City Hospital , Auckland , New Zealand
| | - Yeri Ahn
- a Department of Virology and Immunology , Auckland City Hospital , Auckland , New Zealand.,b Department of Clinical Immunology , Auckland City Hospital , Auckland , New Zealand
| | - Anthony Jordan
- b Department of Clinical Immunology , Auckland City Hospital , Auckland , New Zealand
| | - Klaus Lehnert
- c School of Biological Sciences , University of Auckland , Auckland , New Zealand
| | | | - See-Tarn Woon
- a Department of Virology and Immunology , Auckland City Hospital , Auckland , New Zealand
| |
Collapse
|
37
|
Gennery AR. Advances in genetic and molecular understanding of Omenn syndrome - implications for the future. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1478287] [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] [Indexed: 10/16/2022]
Affiliation(s)
- Andrew R Gennery
- Clinical Resource Building, Floor 4, Block 2, Great North Children’s Hospital, Newcastle Upon Tyne, UK
| |
Collapse
|
38
|
Prevalence and clinical challenges among adults with primary immunodeficiency and recombination-activating gene deficiency. J Allergy Clin Immunol 2018; 141:2303-2306. [PMID: 29477728 DOI: 10.1016/j.jaci.2018.02.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/05/2018] [Accepted: 02/11/2018] [Indexed: 12/17/2022]
|
39
|
Sood AK, Funkhouser W, Handly B, Weston B, Wu EY. Granulomatous-Lymphocytic Interstitial Lung Disease in 22q11.2 Deletion Syndrome: a Case Report and Literature Review. Curr Allergy Asthma Rep 2018; 18:14. [PMID: 29470661 PMCID: PMC5935501 DOI: 10.1007/s11882-018-0769-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Granulomatous-lymphocytic interstitial lung disease (GLILD) has classically been associated with common variable immune deficiency (CVID), but is increasingly being reported in other immunodeficiencies. We describe the second reported case of GLILD in a patient with 22q11.2 deletion syndrome (22q11.2DS) and review the recent literature surrounding GLILD. RECENT FINDINGS GLILD is characterized by granulomata and lymphoproliferation. Consensus statements and retrospective and case-control studies have better elucidated the clinicopathological and radiographic manifestations of GLILD, allowing for its differentiation from similar conditions like sarcoidosis. Gaps of knowledge remain, however, particularly regarding optimal management strategies. Combination therapies targeting T and B cell populations have recently shown favorable results. GLILD is associated with poorer outcomes in CVID. Its recognition as a rare complication of 22q11.2DS and other immunodeficiencies therefore has important therapeutic and prognostic implications. Additional research is needed to better understand the natural history and pathogenesis of GLILD and to develop evidence-based practice guidelines.
Collapse
Affiliation(s)
- Amika K Sood
- Department of Pediatrics, Division of Allergy, Immunology, and Rheumatology, University of North Carolina, Chapel Hill, NC, USA.
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, 104 Mason Farm Road, CB #7310, Chapel Hill, NC, 27599-7310, USA.
| | - William Funkhouser
- Deparment of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Brian Handly
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Brent Weston
- Department of Pediatrics, Division of Hematology-Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Eveline Y Wu
- Department of Pediatrics, Division of Allergy, Immunology, and Rheumatology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
40
|
Clinical Predictors of a Diagnosis of Common Variable Immunodeficiency-related Granulomatous-Lymphocytic Interstitial Lung Disease. Ann Am Thorac Soc 2018; 13:1042-9. [PMID: 27064856 DOI: 10.1513/annalsats.201511-728oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE Granulomatous-lymphocytic interstitial lung disease (GLILD) has emerged as a major cause of morbidity in patients with common variable immunodeficiency (CVID). While GLILD is among the most serious noninfectious pulmonary complications of CVID, risk factors for this condition have not been reported. OBJECTIVES To identify clinical, physiologic, and serologic risk factors for GLILD in adults with CVID. METHODS Of 345 consecutive adult patients with CVID, we identified 34 in the National Jewish Health research database who had a radiographic-pathologic diagnosis of GLILD evaluated between 2002 and 2014. Each case was age and sex matched to 52 CVID control subjects. We used logistic regression to determine independent predictors of GLILD. A mixed effects model was used to estimate the longitudinal change in percent predicted FVC. MEASUREMENTS AND MAIN RESULTS The mean time from CVID diagnosis to GLILD detection was 7.8 years. Compared with matched control subjects, cases were more likely to have a history of autoimmune cytopenia, hypersplenism, polyarthritis, lower marginal zone and switched memory B cells, and restrictive lung function. Multivariate analysis revealed that hypersplenism (odds ratio [OR], 24; 95% confidence interval [CI], 4.5-179.1), polyarthritis (OR, 19; 95% CI, 2.3-206.8), and percent predicted FVC (OR, 0.93; 95% CI, 0.87-0.98) were independently associated with the development of GLILD. The rate of change of percent predicted FVC (slope, P = 0.48) did not vary significantly in patients with GLILD over a mean follow-up of 7 years after diagnosis. CONCLUSIONS Hypersplenism and polyarthritis are strong risk factors for GLILD in patients with CVID. Percent predicted FVC remained stable over time in patients with GLILD.
Collapse
|
41
|
Schmidt RE, Grimbacher B, Witte T. Autoimmunity and primary immunodeficiency: two sides of the same coin? Nat Rev Rheumatol 2017; 14:7-18. [PMID: 29255211 DOI: 10.1038/nrrheum.2017.198] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Autoimmunity and immunodeficiency were previously considered to be mutually exclusive conditions; however, increased understanding of the complex immune regulatory and signalling mechanisms involved, coupled with the application of genetic analysis, is revealing the complex relationships between primary immunodeficiency syndromes and autoimmune diseases. Single-gene defects can cause rare diseases that predominantly present with autoimmune symptoms. Such genetic defects also predispose individuals to recurrent infections (a hallmark of immunodeficiency) and can cause primary immunodeficiencies, which can also lead to immune dysregulation and autoimmunity. Moreover, risk factors for polygenic rheumatic diseases often exist in the same genes as the mutations that give rise to primary immunodeficiency syndromes. In this Review, various primary immunodeficiency syndromes are presented, along with their pathogenetic mechanisms and relationship to autoimmune diseases, in an effort to increase awareness of immunodeficiencies that occur concurrently with autoimmune diseases and to highlight the need to initiate appropriate genetic tests. The growing knowledge of various genetically determined pathologic mechanisms in patients with immunodeficiencies who have autoimmune symptoms opens up new avenues for personalized molecular therapies that could potentially treat immunodeficiency and autoimmunity at the same time, and that could be further explored in the context of autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Reinhold E Schmidt
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover (MHH), Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| | - Bodo Grimbacher
- Centre for Chronic Immunodeficiency, University Medical Centre, University of Freiburg, Faculty of Medicine, Breisacher Straße 115, D-79106 Freiburg, Germany
| | - Torsten Witte
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover (MHH), Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| |
Collapse
|
42
|
Capo V, Castiello MC, Fontana E, Penna S, Bosticardo M, Draghici E, Poliani LP, Sergi Sergi L, Rigoni R, Cassani B, Zanussi M, Carrera P, Uva P, Dobbs K, Sacchetti N, Notarangelo LD, van Til NP, Wagemaker G, Villa A. Efficacy of lentivirus-mediated gene therapy in an Omenn syndrome recombination-activating gene 2 mouse model is not hindered by inflammation and immune dysregulation. J Allergy Clin Immunol 2017; 142:928-941.e8. [PMID: 29241731 DOI: 10.1016/j.jaci.2017.11.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/02/2017] [Accepted: 11/01/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Omenn syndrome (OS) is a rare severe combined immunodeficiency associated with autoimmunity and caused by defects in lymphoid-specific V(D)J recombination. Most patients carry hypomorphic mutations in recombination-activating gene (RAG) 1 or 2. Hematopoietic stem cell transplantation is the standard treatment; however, gene therapy (GT) might represent a valid alternative, especially for patients lacking a matched donor. OBJECTIVE We sought to determine the efficacy of lentiviral vector (LV)-mediated GT in the murine model of OS (Rag2R229Q/R229Q) in correcting immunodeficiency and autoimmunity. METHODS Lineage-negative cells from mice with OS were transduced with an LV encoding the human RAG2 gene and injected into irradiated recipients with OS. Control mice underwent transplantation with wild-type or OS-untransduced lineage-negative cells. Immunophenotyping, T-dependent and T-independent antigen challenge, immune spectratyping, autoantibody detection, and detailed tissue immunohistochemical analyses were performed. RESULTS LV-mediated GT allowed immunologic reconstitution, although it was suboptimal compared with that seen in wild-type bone marrow (BM)-transplanted OS mice in peripheral blood and hematopoietic organs, such as the BM, thymus, and spleen. We observed in vivo variability in the efficacy of GT correlating with the levels of transduction achieved. Immunoglobulin levels and T-cell repertoire normalized, and gene-corrected mice responded properly to challenges in vivo. Autoimmune manifestations, such as skin infiltration and autoantibodies, dramatically improved in GT mice with a vector copy number/genome higher than 1 in the BM and 2 in the thymus. CONCLUSIONS Our data show that LV-mediated GT for patients with OS significantly ameliorates the immunodeficiency, even in an inflammatory environment.
Collapse
Affiliation(s)
- Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Elena Fontana
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Sara Penna
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Marita Bosticardo
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Elena Draghici
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Luigi P Poliani
- Institute of Molecular Medicine "A. Nocivelli," University Hospital "Spedali Civili," Brescia, Italy
| | - Lucia Sergi Sergi
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Rosita Rigoni
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Barbara Cassani
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Monica Zanussi
- Genomics for the Diagnosis of Human Pathologies, San Raffaele Scientific Institute, Milan, Italy
| | - Paola Carrera
- Genomics for the Diagnosis of Human Pathologies, San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Pula, Italy
| | - Kerry Dobbs
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Nicolò Sacchetti
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Luigi D Notarangelo
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Niek P van Til
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands; Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard Wagemaker
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands; Stem Cell Research and Development Center, Hacettepe University, Ankara, Turkey; Raisa Gorbacheva Memorial Research Institute for Pediatric Oncology and Hematology, Saint Petersburg, Russia
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy; Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy.
| |
Collapse
|
43
|
Walter JE, Farmer JR, Foldvari Z, Torgerson TR, Cooper MA. Mechanism-Based Strategies for the Management of Autoimmunity and Immune Dysregulation in Primary Immunodeficiencies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 4:1089-1100. [PMID: 27836058 DOI: 10.1016/j.jaip.2016.08.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/01/2016] [Accepted: 08/19/2016] [Indexed: 01/27/2023]
Abstract
A broad spectrum of autoimmunity is now well described in patients with primary immunodeficiencies (PIDs). Management of autoimmune disease in the background of PID is particularly challenging given the seemingly discordant goals of immune support and immune suppression. Our growing ability to define the molecular underpinnings of immune dysregulation has facilitated novel targeted therapeutics. This review focuses on mechanism-based treatment strategies for the most common autoimmune and inflammatory complications of PID including autoimmune cytopenias, rheumatologic disease, and gastrointestinal disease. We aim to provide guidance regarding the rational use of these agents in the complex PID patient population.
Collapse
Affiliation(s)
- Jolan E Walter
- Department of Pediatrics & Medicine, University of South Florida at Johns Hopkins All Children's Hospital, St Petersburg, Fla; Division of Pediatric Allergy & Immunology, Massachusetts General Hospital for Children, Boston, Mass; Division of Immunology, Boston Children's Hospital, Boston, Mass.
| | - Jocelyn R Farmer
- Department of Allergy & Immunology, Massachusetts General Hospital, Boston, Mass
| | - Zsofia Foldvari
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway; K. G. Jebsen Centers for Cancer Immunotherapy and for Inflammation Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Troy R Torgerson
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Wash
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St Louis, Mo
| |
Collapse
|
44
|
Primary Immune Deficiencies in the Adult: A Previously Underrecognized Common Condition. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 4:1101-1107. [PMID: 27836059 DOI: 10.1016/j.jaip.2016.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/01/2016] [Accepted: 09/09/2016] [Indexed: 12/21/2022]
Abstract
The large majority of classified primary immune deficiency (PID) diseases present in childhood. Yet, most patients with PID are adults, with a large proportion experiencing onset of symptoms beyond their childhood years. Most of these are diagnosed predominantly with antibody defects, but cellular and other disorders are increasingly being identified in older patients as well. Moreover, advances in clinical immunology are allowing pediatric patients, even those with severe disease, to reach adulthood. Because of differences in the physiology and pathophysiology of children and adults, the presentation, diagnosis, and management of a complex chronic disease could differ significantly between these patient populations and therefore require modifications in approach.
Collapse
|
45
|
Kumánovics A, Lee YN, Close DW, Coonrod EM, Ujhazi B, Chen K, MacArthur DG, Krivan G, Notarangelo LD, Walter JE. Estimated disease incidence of RAG1/2 mutations: A case report and querying the Exome Aggregation Consortium. J Allergy Clin Immunol 2017; 139:690-692.e3. [PMID: 27609655 PMCID: PMC5303162 DOI: 10.1016/j.jaci.2016.07.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 06/30/2016] [Accepted: 07/26/2016] [Indexed: 10/21/2022]
Abstract
RAG deficiency is emerging as one of the leading causes of SCID and leaky SCID with an estimated incidence of 1:336,000. Hypomorphic mutations in the RAG genes can also lead to highly variable delayed-onset combined immunodeficiency diseases. We estimate the population genetic frequency of these hypomorphic diseases as up to 1:181,000, suggesting that RAG1/2 mutations are likely to contribute to undiagnosed cases of combined immunodeficiencies.
Collapse
Affiliation(s)
- Attila Kumánovics
- Department of Pathology, University of Utah, Salt Lake City, Utah; ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah.
| | - Yu Nee Lee
- Division of Immunology, Children's Hospital, Harvard Medical School, Boston, Mass
| | - Devin W Close
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah
| | - Emily M Coonrod
- ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah
| | - Boglarka Ujhazi
- Division of Allergy and Immunology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, Mass
| | - Karin Chen
- Division of Allergy and Immunology, Department of Pediatrics, University of Utah, Salt Lake City, Utah
| | - Daniel G MacArthur
- Analytic and Translational Genetics Unit, Division of Allergy and Immunology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, Mass
| | - Gergely Krivan
- United St Laszlo and St Istvan Hospital, Budapest, Hungary
| | - Luigi D Notarangelo
- Division of Immunology, Children's Hospital, Harvard Medical School, Boston, Mass
| | - Jolan E Walter
- Division of Immunology, Children's Hospital, Harvard Medical School, Boston, Mass; Division of Allergy and Immunology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, Mass.
| |
Collapse
|
46
|
Abstract
Although primary immunodeficiencies typically present with recurrent, chronic, or severe infections, autoimmune manifestations frequently accompany these disorders and may be the initial clinical manifestations. The presence of 2 or more autoimmune disorders, unusual severe atopic disease, or a combination of these disorders should lead a clinician to consider primary immunodeficiency disorders.
Collapse
Affiliation(s)
- John M Routes
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin, Children's Clinics Building, Suite B440, 9000 West Wisconsin Avenue, Milwaukee, WI 53226-4874, USA.
| | - James W Verbsky
- Department of Pediatrics, Children's Corporate Center, Children's Research Institute, Medical College of Wisconsin, Suite C465, 9000 West Wisconsin Avenue, Milwaukee, WI 53226-4874, USA
| |
Collapse
|
47
|
Lee YN, Frugoni F, Dobbs K, Tirosh I, Du L, Ververs FA, Ru H, Ott de Bruin L, Adeli M, Bleesing JH, Buchbinder D, Butte MJ, Cancrini C, Chen K, Choo S, Elfeky RA, Finocchi A, Fuleihan RL, Gennery AR, El-Ghoneimy DH, Henderson LA, Al-Herz W, Hossny E, Nelson RP, Pai SY, Patel NC, Reda SM, Soler-Palacin P, Somech R, Palma P, Wu H, Giliani S, Walter JE, Notarangelo LD. Characterization of T and B cell repertoire diversity in patients with RAG deficiency. Sci Immunol 2016; 1:eaah6109. [PMID: 28783691 PMCID: PMC5586490 DOI: 10.1126/sciimmunol.aah6109] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/22/2016] [Indexed: 12/13/2022]
Abstract
Recombination-activating genes 1 and 2 (RAG1 and RAG2) play a critical role in T and B cell development by initiating the recombination process that controls the expression of T cell receptor (TCR) and immunoglobulin genes. Mutations in the RAG1 and RAG2 genes in humans cause a broad spectrum of phenotypes, including severe combined immunodeficiency (SCID) with lack of T and B cells, Omenn syndrome, leaky SCID, and combined immunodeficiency with granulomas or autoimmunity (CID-G/AI). Using next-generation sequencing, we analyzed the TCR and B cell receptor (BCR) repertoire in 12 patients with RAG mutations presenting with Omenn syndrome (n = 5), leaky SCID (n = 3), or CID-G/AI (n = 4). Restriction of repertoire diversity skewed usage of variable (V), diversity (D), and joining (J) segment genes, and abnormalities of CDR3 length distribution were progressively more prominent in patients with a more severe phenotype. Skewed usage of V, D, and J segment genes was present also within unique sequences, indicating a primary restriction of repertoire. Patients with Omenn syndrome had a high proportion of class-switched immunoglobulin heavy chain transcripts and increased somatic hypermutation rate, suggesting in vivo activation of these B cells. These data provide a framework to better understand the phenotypic heterogeneity of RAG deficiency.
Collapse
Affiliation(s)
- Yu Nee Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Pediatric Department A and the Immunology Service, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation Center, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Francesco Frugoni
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kerry Dobbs
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Irit Tirosh
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Likun Du
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Francesca A Ververs
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Heng Ru
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Lisa Ott de Bruin
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mehdi Adeli
- Pediatrics Department, Weill Cornell Medical College, Hamad Medical Corporation, Doha, Qatar
| | - Jacob H Bleesing
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - David Buchbinder
- Division of Hematology, Children's Hospital Orange County, Orange County, CA 92868, USA
| | - Manish J Butte
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Caterina Cancrini
- DPUO, University Department of Pediatrics, Bambino Gesù Children's Hospital and University of Tor Vergata School of Medicine, Rome, Italy
| | - Karin Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Sharon Choo
- Department of Immunology, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Reem A Elfeky
- Department of Pediatric Allergy and Immunology, Children's Hospital, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Andrea Finocchi
- DPUO, University Department of Pediatrics, Bambino Gesù Children's Hospital and University of Tor Vergata School of Medicine, Rome, Italy
| | - Ramsay L Fuleihan
- Division of Allergy and Immunology, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Andrew R Gennery
- Department of Paediatric Immunology, Great North Children's Hospital, Newcastle Upon Tyne, U.K
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, U.K
| | - Dalia H El-Ghoneimy
- Department of Pediatric Allergy and Immunology, Children's Hospital, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Lauren A Henderson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Elham Hossny
- Department of Pediatric Allergy and Immunology, Children's Hospital, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Robert P Nelson
- Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sung-Yun Pai
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Niraj C Patel
- Division of Infectious Disease and Immunology, Department of Pediatrics, Levine Children's Hospital, Carolinas Medical Center, Charlotte, NC 28203, USA
| | - Shereen M Reda
- Department of Pediatric Allergy and Immunology, Children's Hospital, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Pere Soler-Palacin
- Paediatric Infectious Diseases and Immunodeficiencies Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Raz Somech
- Pediatric Department A and the Immunology Service, "Edmond and Lily Safra" Children's Hospital, Jeffrey Modell Foundation Center, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Paolo Palma
- DPUO, University Department of Pediatrics, Bambino Gesù Children's Hospital and University of Tor Vergata School of Medicine, Rome, Italy
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Silvia Giliani
- A. Nocivelli Institute for Molecular Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Section of Medical Genetics, Department of Pathology, Spedali Civili di Bresia, Brescia, Italy
| | - Jolan E Walter
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Pediatric Allergy/Immunology, University of South Florida, and Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701, USA
| | - Luigi D Notarangelo
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
48
|
Cifaldi C, Scarselli A, Petricone D, Di Cesare S, Chiriaco M, Claps A, Rossi P, Calzoni E, Yamazaki Y, Notarangelo LD, Di Matteo G, Cancrini C, Finocchi A. Agammaglobulinemia associated to nasal polyposis due to a hypomorphic RAG1 mutation in a 12 years old boy. Clin Immunol 2016; 173:121-123. [PMID: 27713031 DOI: 10.1016/j.clim.2016.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 11/30/2022]
Abstract
Recombination-activating gene (RAG) 1 and 2 mutations in humans cause T- B- NK+ SCID and Omenn syndrome, but milder phenotypes associated with residual protein activity have been recently described. We report a male patient with a diagnosis of common variable immunodeficiency (CVID) born from non-consanguineous parents, whose immunological phenotype was characterized by severe reduction of B cells and agammaglobulinemia for which several candidate genes were excluded by targeted Sanger sequencing. Next Generation Sequencing revealed two compound heterozygous mutations in the RAG1 gene: the previously described p.R624H, and the novel p.Y728H mutation, as well as the known polymorphism p.H249R. This case reinforces the notion of large phenotypic spectrum in RAG deficiency and opens questions on the management and follow-up of these patients.
Collapse
Affiliation(s)
- Cristina Cifaldi
- Department of Systems Medicine, "University of Rome Tor Vergata", Rome, Italy
| | - Alessia Scarselli
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Italy
| | - Davide Petricone
- Department of Systems Medicine, "University of Rome Tor Vergata", Rome, Italy
| | - Silvia Di Cesare
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Italy
| | - Maria Chiriaco
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Italy
| | - Alessia Claps
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Italy
| | - Paolo Rossi
- Department of Systems Medicine, "University of Rome Tor Vergata", Rome, Italy; University Department of Pediatrics, Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Italy
| | - Enrica Calzoni
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | | | - Luigi Daniele Notarangelo
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Gigliola Di Matteo
- Department of Systems Medicine, "University of Rome Tor Vergata", Rome, Italy
| | - Caterina Cancrini
- Department of Systems Medicine, "University of Rome Tor Vergata", Rome, Italy; University Department of Pediatrics, Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Italy
| | - Andrea Finocchi
- Department of Systems Medicine, "University of Rome Tor Vergata", Rome, Italy; University Department of Pediatrics, Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Italy.
| |
Collapse
|
49
|
Pathak S, McDermott MF, Savic S. Autoinflammatory diseases: update on classification diagnosis and management. J Clin Pathol 2016; 70:1-8. [PMID: 27646526 DOI: 10.1136/jclinpath-2016-203810] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 01/18/2023]
Abstract
The spectrum of systemic autoinflammatory disorders broadens continually. In part, this is due to the more widespread application of massive parallel sequencing, helping with novel gene discovery in this and other areas of rare diseases. Some of the conditions that have been described fit neatly into a conventional idea of autoinflammation. Others, such as interferon-mediated autoinflammatory diseases, are broadening the concept which we consider to be autoinflammatory disorders. There is also a widening of the clinical phenotypes associated with certain genetic mutations, as genetic testing is used more regularly and increasing numbers of patients are screened. It is also increasingly evident that both autoinflammatory and autoimmune problems are frequently seen as complications of primary immunodeficiency disorders. The aim of this review is to provide an update on some recently discovered conditions and to discuss how these disorders help to define the concept of autoinflammation. The review will also cover recent discoveries in the biology of innate-immune-mediated inflammation and describe how this has provided the biological rationale for using anti-interleukin-1 therapies in the treatment of many such conditions. Finally, we discuss the importance of recognising somatic mutations as causes of autoinflammatory clinical phenotypes and provide practical advice on how this could be tackled in everyday clinical practice.
Collapse
Affiliation(s)
- Shelly Pathak
- National Institute for Health Research-Leeds Musculoskeletal Biomedical Research Unit (NIHR-LMBRU), Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, UK
| | - Michael F McDermott
- National Institute for Health Research-Leeds Musculoskeletal Biomedical Research Unit (NIHR-LMBRU), Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, UK
| | - Sinisa Savic
- National Institute for Health Research-Leeds Musculoskeletal Biomedical Research Unit (NIHR-LMBRU), Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, UK.,Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, UK
| |
Collapse
|
50
|
John T, Walter JE, Schuetz C, Chen K, Abraham RS, Bonfim C, Boyce TG, Joshi AY, Kang E, Carvalho BTC, Mahajerin A, Nugent D, Puthenveetil G, Soni A, Su H, Cowan MJ, Notarangelo L, Buchbinder D. Unrelated Hematopoietic Cell Transplantation in a Patient with Combined Immunodeficiency with Granulomatous Disease and Autoimmunity Secondary to RAG Deficiency. J Clin Immunol 2016; 36:725-32. [PMID: 27539235 DOI: 10.1007/s10875-016-0326-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/03/2016] [Indexed: 10/21/2022]
Abstract
The use of HLA-identical hematopoietic stem cell transplantation (HSCT) demonstrates overall survival rates greater than 75 % for T-B-NK+ severe combined immunodeficiency secondary to pathogenic mutation of recombinase activating genes 1 and 2 (RAG1/2). Limited data exist regarding the use of HSCT in patients with hypomorphic RAG variants marked by greater preservation of RAG activity and associated phenotypes such as granulomatous disease in combination with autoimmunity. We describe a 17-year-old with combined immunodeficiency and immune dysregulation characterized by granulomatous lung disease and autoimmunity secondary to compound heterozygous RAG mutations. A myeloablative reduced toxicity HSCT was completed using an unrelated bone marrow donor. With the increasing cases of immune dysregulation being discovered with hypomorphic RAG variants, the use of HSCT may advance to the forefront of treatment. This case serves to discuss indications of HSCT, approaches to preparative therapy, and the potential complications in this growing cohort of patients with immune dysregulation and RAG deficiency.
Collapse
Affiliation(s)
- Tami John
- Division of Hematology/Oncology, CHOC Children's Hospital, 1201 W. La Veta Avenue, Orange, CA, 92868, USA.
| | - Jolan E Walter
- Division of Immunology, MassGeneral Hospital for Children, 55 Fruit Street, Boston, MA, 02114, USA
| | - Catherina Schuetz
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Karin Chen
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, University of Utah School of Medicine, 81 Mario Capecchi Drive, Salt Lake City, UT, USA
| | - Roshini S Abraham
- Allergy and Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Carmem Bonfim
- Bone Marrow Transplantation Unit, Federal University of Paraná, Rua XV de Novembro, 1299 - Centro, Curitiba, PR, 80060-000, Brazil
| | - Thomas G Boyce
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Avni Y Joshi
- Allergy and Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Elizabeth Kang
- NIAID, National Institutes of Health, Building 10CRC, Room 5-3940, 10 Center Drive, MSC 1456, Bethesda, MD, 20892-9806, USA
| | | | - Arash Mahajerin
- Division of Hematology, CHOC Children's Hospital, 1201 W. La Veta Avenue, Orange, CA, 92868, USA
| | - Diane Nugent
- Division of Hematology, CHOC Children's Hospital, 1201 W. La Veta Avenue, Orange, CA, 92868, USA
| | - Geetha Puthenveetil
- Division of Hematology, CHOC Children's Hospital, 1201 W. La Veta Avenue, Orange, CA, 92868, USA
| | - Amit Soni
- Division of Hematology, CHOC Children's Hospital, 1201 W. La Veta Avenue, Orange, CA, 92868, USA
| | - Helen Su
- NIAID, National Institutes of Health, Building 10CRC, Room 5-3940, 10 Center Drive, MSC 1456, Bethesda, MD, 20892-9806, USA
| | - Morton J Cowan
- Department of Pediatrics, University of California, San Francisco, Box 1278, UCSF, San Francisco, CA, 94143, USA
| | - Luigi Notarangelo
- Division of Immunology, Children's Hospital Boston, Karp Building, Room 10217, 1 Blackfan Circle, Boston, MA, 02115, USA
| | - David Buchbinder
- Division of Hematology, CHOC Children's Hospital, 1201 W. La Veta Avenue, Orange, CA, 92868, USA
| |
Collapse
|