1
|
Toskov V, Bali P, Hershfield MS, Ehl S, Speckmann C. Successful Long-Term Enzyme Replacement Therapy in a Patient with Delayed-Onset ADA Deficiency. J Clin Immunol 2024; 45:8. [PMID: 39264481 PMCID: PMC11393103 DOI: 10.1007/s10875-024-01794-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Affiliation(s)
- Vasil Toskov
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Pawan Bali
- Department of Medicine and Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Michael S Hershfield
- Department of Medicine and Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Breisacher Str. 117, 79106, Freiburg, Germany
| | - Carsten Speckmann
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany.
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Breisacher Str. 117, 79106, Freiburg, Germany.
| |
Collapse
|
2
|
Freeman AF, Gonzalez CE, Yates B, Cole K, Little L, Flannelly E, Steinberg SM, Mo G, Piette N, Hughes TE, Cuellar-Rodriguez J, Gea-Banacloche J, Heller T, Hammoud DA, Holland SM, Kong HH, Young FD, Jing H, Kayaoglu B, Su HC, Pai SY, Hickstein DD, Shah NN. Hematopoietic cell transplantation for DOCK8 deficiency: Results from a prospective clinical trial. J Allergy Clin Immunol 2024:S0091-6749(24)00904-7. [PMID: 39233015 DOI: 10.1016/j.jaci.2024.08.021] [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: 02/22/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND DOCK8 deficiency is a primary immunodeficiency in which allogeneic hematopoietic cell transplantation (HCT) represents the only known cure. We tested the ability of a busulfan-based regimen to achieve reliable engraftment and high levels of donor chimerism with acceptable toxicity in a prospective clinical trial in DOCK8 deficiency. OBJECTIVES To both evaluate the ability of HCT to reverse the clinical phenotype and to correct the immunologic abnormalities by 1 year post HCT. METHODS We conducted a prospective HCT trial for recipients with DOCK8 deficiency. Subjects were recruited from October 5, 2010, to December 30, 2022. Donor sources included fully matched related and unrelated donors and haploidentical donors. The reduced toxicity, myeloablative conditioning regimen contained no serotherapy. Graft-versus-host disease (GVHD) prophylaxis included either a calcineurin inhibitor with methotrexate or post-HCT cyclophosphamide (PT/Cy) followed by tacrolimus and mycophenolate mofetil. The trial was later amended to study PT/Cy in all patients. (Pilot Study of Reduced-Intensity Hematopoietic Stem Cell Transplant of DOCK8 [NCT01176006].) RESULTS: Thirty-six subjects, both children and adults (median age 16.4 years), underwent HCT for DOCK8 deficiency. Most patients, 33 of 36 (92%), achieved full (≥98%) donor chimerism in whole blood as early as day +30. With a median potential follow-up of 7.4 years, 29 (80.6%) were alive with no evidence of new DOCK8 deficiency-related complications. PT/Cy was effective in reducing the risk of acute GVHD in patients who had received matched unrelated donor and haploidentical transplants, but it was associated with transient delays in immune-reconstitution and hemorrhagic cystitis. CONCLUSIONS A busulfan-based HCT regimen using PT/Cy for GVHD prophylaxis and a broad range of donor types and hematopoietic cell sources were well tolerated, leading to the reversal of the clinical immunophenotype.
Collapse
Affiliation(s)
- Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Corina E Gonzalez
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md.
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | - Kristen Cole
- Transplant and Cell Therapy Program, Clinical Center, Bethesda, Md
| | - Lauren Little
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | - Erin Flannelly
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Seth M Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, Md
| | - George Mo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | | | | | - Jennifer Cuellar-Rodriguez
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Juan Gea-Banacloche
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Theo Heller
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Md
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, Bethesda, Md
| | - Steve M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Fernanda D Young
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Basak Kayaoglu
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Sung-Yun Pai
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Dennis D Hickstein
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| |
Collapse
|
3
|
Alsaati N, Grier A, Ochfeld E, McClory S, Heimall J. Hematopoietic stem cell transplantation for primary immunodeficiency. Allergy Asthma Proc 2024; 45:371-383. [PMID: 39294909 DOI: 10.2500/aap.2024.45.240069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Primary immunodeficiencies, also commonly called inborn errors of immunity (IEI), are commonly due to developmental or functional defects in peripheral blood cells derived from hematopoietic stem cells. In light of this, for the past 50 years, hematopoietic stem cell transplantation (HSCT) has been used as a definitive therapy for IEI. The fields of both clinical immunology and transplantation medicine have had significant advances. This, in turn, has allowed for both an increasing ability to determine a monogenic etiology for many IEIs and an increasing ability to successfully treat these patients with HSCT. Therefore, it has become more common for the practicing allergist/immunologist to diagnose and manage a broad range of patients with IEI before and after HSCT. This review aims to provide practical guidance for the clinical allergist/immunologist on the basics of HSCT and known outcomes in selected forms of IEI, the importance of pre-HSCT supportive care, and the critical importance of and guidance for life-long immunologic and medical monitoring of these patients.
Collapse
Affiliation(s)
- Nouf Alsaati
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| | - Alexandra Grier
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| | - Elisa Ochfeld
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| | - Susan McClory
- Cell Therapy and Transplant Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania
| | - Jennifer Heimall
- From the Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia Pennsylvania; and
| |
Collapse
|
4
|
Lum SH, Albert MH, Gilbert P, Sirait T, Algeri M, Muratori R, Fournier B, Laberko A, Karakukcu M, Unal E, Ayas M, Yadav SP, Fisgin T, Elfeky R, Fernandes J, Faraci M, Cole T, Schulz A, Meisel R, Zecca M, Ifversen M, Biffi A, Diana JS, Vallée T, Giardino S, Ersoy GZ, Moshous D, Gennery AR, Balashov D, Bonfim C, Locatelli F, Lankester A, Neven B, Slatter M. Outcomes of HLA-mismatched HSCT with TCRαβ/CD19 depletion or post-HSCT cyclophosphamide for inborn errors of immunity. Blood 2024; 144:565-580. [PMID: 38669631 DOI: 10.1182/blood.2024024038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/27/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
ABSTRACT HLA-mismatched transplants with either in vitro depletion of CD3+ T-cell receptor (TCR)αβ/CD19 (TCRαβ) cells or in vivo T-cell depletion using posttransplant cyclophosphamide (PTCY) have been increasingly used for patients with inborn errors of immunity (IEIs). We performed a retrospective multicenter study via the EBMT registry on 306 children with IEIs undergoing their first transplant between 2010 and 2019 from an HLA-mismatched donor using TCRαβ (n = 167) or PTCY (n = 139). The median age for hematopoietic stem cell transplantation (HSCT) was 1.2 years (range, 0.03-19.6 years). The 3-year overall survival (OS) was 78% (95% confidence interval (CI), 71-84) after TCRαβ and 66% (57-74) after PTCY (P = .013). Pre-HSCT morbidity score (hazard ratio [HR], 2.27; 1.07-4.80, P = .032) and non-busulfan/treosulfan conditioning (HR, 3.12; 1.98-4.92, P < .001) were the only independent predictors of unfavorable OS. The 3-year event-free survival (EFS) was 58% (50%-66%) after TCRαβ and 57% (48%-66%) after PTCY (P = .804). The cumulative incidence of severe acute graft-versus-host disease (GvHD) was higher after PTCY (15%, 9%-21%) than TCRαβ (6%, 2%-9%, P = .007), with no difference in chronic GvHD (PTCY, 11%, 6%-17%; TCRαβ, 7%, 3%-11%, P = .173). The 3-year GvHD-free EFS was 53% (44%-61%) after TCRαβ and 41% (32%-50%) after PTCY (P = .080). PTCY had significantly higher rates of veno-occlusive disease (14.4% vs TCRαβ 4.9%, P = .009), acute kidney injury (12.7% vs 4.6%, P = .032), and pulmonary complications (38.2% vs 24.1%, P = .017). Adenoviremia (18.3% vs PTCY 8.0%, P = .015), primary graft failure (10% vs 5%, P = .048), and second HSCT (17.4% vs 7.9%, P = .023) were significantly higher in TCRαβ. In conclusion, this study demonstrates that both approaches are suitable options in patients with IEIs, although they are characterized by different advantages and outcomes.
Collapse
Affiliation(s)
- Su Han Lum
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Michael H Albert
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Mattia Algeri
- Department of Paediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Magna Graecia University, Catanzaro, Italy
| | - Rafaella Muratori
- Pediatric Hematology and Transplantation Unit, Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil
| | - Benjamin Fournier
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Alexandra Laberko
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Musa Karakukcu
- Erciyes University, KANKA Pediatric Hematology/Oncology and BMT Hospital, Kayseri, Turkey
| | - Elrem Unal
- Hasan KALYONCU University and Medicalpoint Hospital, Gaziantep, Turkey
| | - Mouhab Ayas
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Tunc Fisgin
- Pediatric Hematology/Oncology and BMT Unit, Altinbas University Faculty of Medicine Medical Park Bahcelievler Hospital, Istanbul, Turkey
| | - Reem Elfeky
- Department of Paediatric Immunology, Great Ormand Street Children's Hospital, London, United Kingdom
| | - Juliana Fernandes
- Stem Cell Transplantation Unit, ITACI-Instituto da Criança-Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
- Hematology and Stem Cell Transplantation Unit, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Hematology and Stem Cell Transplantation Unit, Hospital 9 de Julho, São Paulo, Brazil
| | | | - Theresa Cole
- Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Marco Zecca
- Paediatric Haematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marianne Ifversen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Alessandra Biffi
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Women and Child Health Department, University of Padua and Padua University Hospital, Padua, Italy
| | - Jean-Sebastien Diana
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Tanja Vallée
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Gizem Zengin Ersoy
- Pediatric Hematology/Oncology and BMT Unit, Altinbas University Faculty of Medicine Medical Park Bahcelievler Hospital, Istanbul, Turkey
| | - Despina Moshous
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Dmitry Balashov
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Carmem Bonfim
- Instituto de Pesquisa Pele Pequeno Príncipe/Faculdades Pequeno Príncipe, Pediatric Blood and Marrow Transplantation Service Hospital Pequeno Príncipe, Curitiba, Brazil
| | - Franco Locatelli
- Department of Paediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Arjan Lankester
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Bénédicte Neven
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mary Slatter
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
5
|
Alligon M, Mahlaoui N, Bouaziz O. Pitfalls in time-to-event analysis of registry data: a tutorial based on simulated and real cases. FRONTIERS IN EPIDEMIOLOGY 2024; 4:1386922. [PMID: 39188581 PMCID: PMC11345615 DOI: 10.3389/fepid.2024.1386922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/21/2024] [Indexed: 08/28/2024]
Abstract
Survival analysis (also referred to as time-to-event analysis) is the study of the time elapsed from a starting date to some event of interest. In practice, these analyses can be challenging and, if methodological errors are to be avoided, require the application of appropriate techniques. By using simulations and real-life data based on the French national registry of patients with primary immunodeficiencies (CEREDIH), we sought to highlight the basic elements that need to be handled correctly when performing the initial steps in a survival analysis. We focused on non-parametric methods to deal with right censoring, left truncation, competing risks, and recurrent events. Our simulations show that ignoring these aspects induces a bias in the results; we then explain how to analyze the data correctly in these situations using non-parametric methods. Rare disease registries are extremely valuable in medical research. We discuss the application of appropriate methods for the analysis of time-to-event from the CEREDIH registry. The objective of this tutorial article is to provide clinicians and healthcare professionals with better knowledge of the issues facing them when analyzing time-to-event data.
Collapse
Affiliation(s)
- Mickaël Alligon
- French National Reference Center for Primary Immune Deficiencies (CEREDIH), Necker Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Nizar Mahlaoui
- French National Reference Center for Primary Immune Deficiencies (CEREDIH), Necker Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
- Immuno-Haematology and Rheumatology Unit, Necker Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | | |
Collapse
|
6
|
Rao VK, Kulm E, Grossman J, Buchbinder D, Chong H, Bradt J, Webster S, Šedivá A, Dalm VA, Uzel G. Long-term treatment with selective PI3Kδ inhibitor leniolisib in adults with activated PI3Kδ syndrome. Blood Adv 2024; 8:3092-3108. [PMID: 38593221 PMCID: PMC11222951 DOI: 10.1182/bloodadvances.2023011000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
ABSTRACT Activated phosphoinositide 3-kinase delta (PI3Kδ) syndrome (APDS) is an inborn error of immunity that manifests as immune deficiency and dysregulation; symptoms include frequent infections and lymphoproliferation. In our dose-finding and phase 3 placebo-controlled trials, treatment with the selective PI3Kδ inhibitor leniolisib reduced lymphoproliferation and normalized lymphocyte subsets. Here, we present 6 years of follow-up from the 6 adult patients in the original dose-finding trial receiving leniolisib. We used data from the ongoing open-label extension study, which was supplemented at later time points by investigators, including health-related quality of life (HRQoL) assessed through a clinician-reported questionnaire. We observed improvements in HRQoL: 5 of 6 patients experienced an increase in physical capabilities and socialization, and a decrease in prescribed medications. Immune subsets improved in all patients: mean transitional B-cell levels decreased from 38.17% to 2.47% and the CD4:CD8 T-cell ratio normalized to 1.11. Manifestations seen before and within the first year of leniolisib exposure, such as infections and gastrointestinal conditions, attenuated after year 2, with few new conditions emerging out to year 6. Thrombocytopenia or lymphopenia remained present in half of patients at year 6. Of 83 adverse events through year 5, 90.36% were grade 1; none were grade 4/5 nor deemed leniolisib related. Collectively, we saw an enhancement in HRQoL as well as durable changes in lymphocyte subsets and clinical manifestations, further supporting the use of leniolisib as a long-term therapeutic option for the treatment of APDS. This trial was registered at www.ClinicalTrials.gov as #NCT02859727.
Collapse
Affiliation(s)
- V. Koneti Rao
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Elaine Kulm
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Bethesda, MD
| | | | - David Buchbinder
- Division of Hematology, Children’s Hospital of Orange County, Orange, CA
| | - Hey Chong
- Division of Allergy and Immunology, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | | | - Sharon Webster
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Anna Šedivá
- Department of Immunology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Virgil A. Dalm
- Division of Allergy and Clinical Immunology and Department of Immunology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Gulbu Uzel
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
7
|
Vallée TC, Glasmacher JS, Buchner H, Arkwright PD, Behrends U, Bondarenko A, Browning MJ, Buchbinder D, Cattoni A, Chernyshova L, Ciznar P, Cole T, Czogała W, Dueckers G, Edgar JDM, Erbey F, Fasth A, Ferrua F, Formankova R, Gambineri E, Gennery AR, Goldman FD, Gonzalez-Granado LI, Heilmann C, Heiskanen-Kosma T, Juntti H, Kainulainen L, Kanegane H, Karaca NE, Kilic SS, Klein C, Kołtan S, Kondratenko I, Meyts I, Nasrullayeva GM, Notarangelo LD, Pasic S, Pellier I, Pignata C, Misbah S, Schulz A, Segundo GR, Shcherbina A, Slatter M, Sokolic R, Soler-Palacin P, Stepensky P, van Montfrans JM, Ryhänen S, Wolska-Kuśnierz B, Ziegler JB, Zhao X, Aiuti A, Ochs HD, Albert MH. Wiskott-Aldrich syndrome: a study of 577 patients defines the genotype as a biomarker for disease severity and survival. Blood 2024; 143:2504-2516. [PMID: 38579284 DOI: 10.1182/blood.2023021411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 04/07/2024] Open
Abstract
ABSTRACT Wiskott-Aldrich syndrome (WAS) is a multifaceted monogenic disorder with a broad disease spectrum and variable disease severity and a variety of treatment options including allogeneic hematopoietic stem cell transplantation (HSCT) and gene therapy (GT). No reliable biomarker exists to predict disease course and outcome for individual patients. A total of 577 patients with a WAS variant from 26 countries and a median follow-up of 8.9 years (range, 0.3-71.1), totaling 6118 patient-years, were included in this international retrospective study. Overall survival (OS) of the cohort (censored at HSCT or GT) was 82% (95% confidence interval, 78-87) at age 15 years and 70% (61-80) at 30 years. The type of variant was predictive of outcome: patients with a missense variant in exons 1 or 2 or with the intronic hot spot variant c.559+5G>A (class I variants) had a 15-year OS of 93% (89-98) and a 30-year OS of 91% (86-97), compared with 71% (62-81) and 48% (34-68) in patients with any other variant (class II; P < .0001). The cumulative incidence rates of disease-related complications such as severe bleeding (P = .007), life-threatening infection (P < .0001), and autoimmunity (P = .004) occurred significantly later in patients with a class I variant. The cumulative incidence of malignancy (P = .6) was not different between classes I and II. It confirms the spectrum of disease severity and quantifies the risk for specific disease-related complications. The class of the variant is a biomarker to predict the outcome for patients with WAS.
Collapse
Affiliation(s)
- Tanja C Vallée
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| | - Jannik S Glasmacher
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| | | | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester & Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Uta Behrends
- Children's Hospital, School of Medicine, Technical University Munich, Munich, Germany
| | - Anastasia Bondarenko
- Department of Pediatrics, Immunology, Infectious and Rare Diseases and Allergology, European Medical School, International European University, Kyiv, Ukraine
| | - Michael J Browning
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - David Buchbinder
- Department of Hematology, Children's Hospital of Orange County, Orange, CA
| | - Alessandro Cattoni
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Liudmyla Chernyshova
- Department of Pediatrics, Pediatric Infectious Diseases, Immunology and Allergology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Peter Ciznar
- Department of Pediatrics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Theresa Cole
- Department of Allergy and Immunology, The Royal Children's Hospital, Melbourne, Australia
| | - Wojciech Czogała
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Gregor Dueckers
- Helios Kliniken Krefeld, Children's Hospital, Krefeld, Germany
| | - John David M Edgar
- St James's Hospital & School of Medicine, Trinity College, Dublin, Ireland
| | - Fatih Erbey
- Department of Pediatric Hematology/Oncology, Koç University School of Medicine, İstanbul, Turkey
| | - Anders Fasth
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francesca Ferrua
- Pediatric Immunohematology and Stem Cell Program, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Renata Formankova
- Department of Pediatric Hematology and Oncology, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Eleonora Gambineri
- Department of NEUROFARBA, Section of Child's Health, University of Florence, Florence, Italy
- Department of Haematology-Oncology, Anna Meyer University Children's Hospital (AOU Meyer IRCCS), Florence, Italy
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, and Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Frederick D Goldman
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL
| | - Luis I Gonzalez-Granado
- Department of Pediatrics, Primary Immunodeficiencies Unit, Research Institute, Hospital 12 Octubre, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Carsten Heilmann
- Department for Children and Adolescents, Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Copenhagen University Hospital Rigshospitalet, København, Denmark
| | | | - Hanna Juntti
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital and Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Leena Kainulainen
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Neslihan E Karaca
- Division of Pediatric Immunology, Department of Pediatrics, Ege University, The Medical School, Izmir, Turkey
| | - Sara S Kilic
- Pediatric Immunology and Rheumatology, Bursa Uludag University School of Medicine, Bursa, Turkey
| | - Christoph Klein
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| | - Sylwia Kołtan
- Department of Paediatrics, Haematology and Oncology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Irina Kondratenko
- Russian Children's Clinical Hospital, Pirogov National Research Medical University, Moscow, Russia
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Srdjan Pasic
- Department of Immunology, Mother and Child Health Care Institute of Serbia, Belgrade, Serbia
| | - Isabelle Pellier
- Centre de référence des déficits immunitaires primitifs CEREDIH, CHU d'Angers, Angers, France
| | - Claudio Pignata
- Department of Translational Medical Science, Section of Pediatrics, Federico II University, Napoli, Italy
| | - Siraj Misbah
- Clinical Immunology, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Gesmar R Segundo
- Allergy and Immunology Division, Pediatrics Department, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Anna Shcherbina
- Dmitry Rogachev National Research and Clinical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mary Slatter
- Translational and Clinical Research Institute, Newcastle University, and Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Robert Sokolic
- Hematologic Malignancies Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Children's Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Polina Stepensky
- Bone Marrow Transplantation Department, Hadassah-Hebrew, University Medical Center, Jerusalem, Israel
| | - Joris M van Montfrans
- Department of Pediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Samppa Ryhänen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Pediatric Research Center, Helsinki, Finland
| | | | - John B Ziegler
- School of Women's & Children's Health, University of New South Wales, Sydney, Australia
| | - Xiaodong Zhao
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Aiuti
- Pediatric Immunohematology and Stem Cell Program, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Hans D Ochs
- University of Washington School of Medicine, Seattle, WA
| | - Michael H Albert
- Pediatric Hematology/Oncology, Dr von Hauner University Children's Hospital, Munich, Germany
| |
Collapse
|
8
|
Strauss T, Körholz J, Kuehn HS, Gil Silva AA, Taube F, Trautmann-Grill K, Stittrich A, Pietzsch L, Wiedemuth R, Wahn V, von Bernuth H, Rosenzweig SD, Fasshauer M, Krüger R, Schuetz C. IKAROS-how many feathers have you lost: mild and severe phenotypes in IKZF1 deficiency. Front Pediatr 2024; 12:1345730. [PMID: 38813543 PMCID: PMC11135284 DOI: 10.3389/fped.2024.1345730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/15/2024] [Indexed: 05/31/2024] Open
Abstract
Heterozygous germline variants in human IKZF1 encoding for IKAROS define an inborn error of immunity with immunodeficiency, immune dysregulation and risk of malignancy with a broad phenotypic spectrum. Growing evidence of underlying pathophysiological genotype-phenotype correlations helps to improve our understanding of IKAROS-associated diseases. We describe 6 patients from 4 kindreds with two novel IKZF1 variants leading to haploinsufficiency from 3 centers in Germany. We also provide an overview of first symptoms to a final diagnosis including data from the literature.
Collapse
Affiliation(s)
- Timmy Strauss
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, University Center for Chronic Immunodeficiencies (UCID), Technische Universität Dresden, Dresden, Germany
| | - Julia Körholz
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, University Center for Chronic Immunodeficiencies (UCID), Technische Universität Dresden, Dresden, Germany
| | - Hye Sun Kuehn
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD, United States
| | - Agustin A. Gil Silva
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD, United States
| | - Franziska Taube
- Faculty of Medicine and University Hospital Carl Gustav Carus, University Center for Chronic Immunodeficiencies (UCID), Technische Universität Dresden, Dresden, Germany
- Department of Hematology and Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Karolin Trautmann-Grill
- Department of Hematology and Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anna Stittrich
- Department of Human Genetics, Labor Berlin Charité-Vivantes GmbH, Berlin, Germany
| | - Leonora Pietzsch
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, University Center for Chronic Immunodeficiencies (UCID), Technische Universität Dresden, Dresden, Germany
| | - Ralf Wiedemuth
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, University Center for Chronic Immunodeficiencies (UCID), Technische Universität Dresden, Dresden, Germany
| | - Volker Wahn
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Sergio D. Rosenzweig
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD, United States
| | - Maria Fasshauer
- ImmunoDeficiencyCenter Leipzig (IDCL), Hospital St. Georg GGmbH Leipzig, Academic Teaching Hospital of the University of Leipzig, Leipzig, Germany
| | - Renate Krüger
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Catharina Schuetz
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, University Center for Chronic Immunodeficiencies (UCID), Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Körholz J, Chen LS, Strauss T, Schuetz C, Dalpke AH. One gene to rule them all - clinical perspectives of a potent suppressor of cytokine signaling - SOCS1. Front Immunol 2024; 15:1385190. [PMID: 38711523 PMCID: PMC11070515 DOI: 10.3389/fimmu.2024.1385190] [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: 02/12/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024] Open
Abstract
The discovery of Suppressor of Cytokine Signaling 1 (SOCS1) in 1997 marked a significant milestone in understanding the regulation of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling pathways. Subsequent research deciphered its cellular functions, and recent insights into SOCS1 deficiencies in humans underscored its critical role in immune regulation. In humans, SOCS-haploinsufficiency (SOCS1-HI) presents a diverse clinical spectrum, encompassing autoimmune diseases, infection susceptibility, and cancer. Variability in disease manifestation, even within families sharing the same genetic variant, raises questions about clinical penetrance and the need for individualized treatments. Current therapeutic strategies include JAK inhibition, with promising results in controlling inflammation in SOCS1-HI patients. Hematopoietic stem cell transplantation and gene therapy emerge as promising avenues for curative treatments. The evolving landscape of SOCS1 research, emphasizes the need for a nuanced understanding of genetic variants and their functional consequences.
Collapse
Affiliation(s)
- Julia Körholz
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Chronic Immunodeficiencies (UCID), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lan-Sun Chen
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, University Heidelberg, Heidelberg, Germany
- University Hospital Heidelberg, Heidelberg, Germany
| | - Timmy Strauss
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Chronic Immunodeficiencies (UCID), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Catharina Schuetz
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Chronic Immunodeficiencies (UCID), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Center for Rare Diseases, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alexander H. Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, University Heidelberg, Heidelberg, Germany
- University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
10
|
Bucciol G, Delafontaine S, Meyts I, Poli C. Inborn errors of immunity: A field without frontiers. Immunol Rev 2024; 322:15-27. [PMID: 38062988 DOI: 10.1111/imr.13297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
The study of primary immunodeficiencies or inborn errors of immunity continues to drive our knowledge of the function of the human immune system. From the outset, the study of inborn errors has focused on unraveling genetic etiologies and molecular mechanisms. Aided by the continuous growth in genetic diagnostics, the field has moved from the study of an infection dominated phenotype to embrace and unravel diverse manifestations of autoinflammation, autoimmunity, malignancy, and severe allergy in all medical disciplines. It has now moved from the study of ultrarare presentations to producing meaningful impact in conditions as diverse as inflammatory bowel disease, neurological conditions, and hematology. Beyond offering immunogenetic diagnosis, the study of underlying inborn errors of immunity in these conditions points to targeted treatment which can be lifesaving.
Collapse
Affiliation(s)
- Giorgia Bucciol
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Inborn Errors of Immunity, KU Leuven, Leuven, Belgium
| | - Selket Delafontaine
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Inborn Errors of Immunity, KU Leuven, Leuven, Belgium
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Inborn Errors of Immunity, KU Leuven, Leuven, Belgium
| | - Cecilia Poli
- Facultad de Medicina Universidad del Desarrollo-Clínica Alemana, Santiago, Chile
- Unidad de Inmunología y Reumatología, Hospital Roberto del Río, Santiago, Chile
| |
Collapse
|
11
|
Segura-Tudela A, López-Nevado M, Nieto-López C, García-Jiménez S, Díaz-Madroñero MJ, Delgado Á, Cabrera-Marante O, Pleguezuelo D, Morales P, Paz-Artal E, Gil-Niño J, Marco FM, Serrano C, González-Granado LI, Quesada-Espinosa JF, Allende LM. Enrichment of Immune Dysregulation Disorders in Adult Patients with Human Inborn Errors of Immunity. J Clin Immunol 2024; 44:61. [PMID: 38363452 PMCID: PMC10873437 DOI: 10.1007/s10875-024-01664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/26/2024] [Indexed: 02/17/2024]
Abstract
Human inborn errors of immunity (IEI) comprise a group of diseases resulting from molecular variants that compromise innate and adaptive immunity. Clinical features of IEI patients are dominated by susceptibility to a spectrum of infectious diseases, as well as autoimmune, autoinflammatory, allergic, and malignant phenotypes that usually appear in childhood, which is when the diagnosis is typically made. However, some IEI patients are identified in adulthood due to symptomatic delay of the disease or other reasons that prevent the request for a molecular study. The application of next-generation sequencing (NGS) as a diagnostic technique has given rise to an ever-increasing identification of IEI-monogenic causes, thus improving the diagnostic yield and facilitating the possibility of personalized treatment. This work was a retrospective study of 173 adults with IEI suspicion that were sequenced between 2005 and 2023. Sanger, targeted gene-panel, and whole exome sequencing were used for molecular diagnosis. Disease-causing variants were identified in 44 of 173 (25.43%) patients. The clinical phenotype of these 44 patients was mostly related to infection susceptibility (63.64%). An enrichment of immune dysregulation diseases was found when cohorts with molecular diagnosis were compared to those without. Immune dysregulation disorders, group 4 from the International Union of Immunological Societies Expert Committee (IUIS), were the most prevalent among these adult patients. Immune dysregulation as a new item in the Jeffrey Model Foundation warning signs for adults significantly increases the sensitivity for the identification of patients with an IEI-producing molecular defect.
Collapse
Affiliation(s)
- Alejandro Segura-Tudela
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
| | - Marta López-Nevado
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
| | - Celia Nieto-López
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
| | - Sandra García-Jiménez
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
| | - María J Díaz-Madroñero
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
| | - Ángeles Delgado
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
| | - Oscar Cabrera-Marante
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
| | - Daniel Pleguezuelo
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
| | - Pablo Morales
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
| | - Estela Paz-Artal
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
- School of Medicine, Complutense University of Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Gil-Niño
- Department of Internal Medicine, University Hospital, 12 de Octubre, Madrid, Spain
| | - Francisco M Marco
- Unit of Immunology, University Hospital General Dr Balmis, Alicante, Spain
| | - Cristina Serrano
- Department of Immunology, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Luis I González-Granado
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
- School of Medicine, Complutense University of Madrid, Madrid, Spain
- Unit of Immunodeficiencies, Department of Pediatrics, University Hospital, 12 de Octubre, Madrid, Spain
| | - Juan F Quesada-Espinosa
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain
- Department of Genetics, University Hospital, 12 de Octubre, Madrid, Spain
| | - Luis M Allende
- Department of Immunology, University Hospital, 12 de Octubre, Avda. de Andalucía S/N, 28041, Madrid, Spain.
- Research Institute Hospital, 12 Octubre (imas12), Madrid, Spain.
- School of Medicine, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
12
|
Anantharachagan A, Loh SY, Burns SO, Laurence A, Tadros S, Tholouli E, Lwin Y, Martinez-Calle N, Vaitla P, Morris EC. Allogeneic hematopoietic stem cell transplantation outcome in oldest known surviving patients with Wiskott-Aldrich syndrome. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100191. [PMID: 38187865 PMCID: PMC10770606 DOI: 10.1016/j.jacig.2023.100191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 01/09/2024]
Abstract
Regardless of their age, adult patients with Wiskott-Aldrich syndrome should be considered for hematopoietic stem cell transplantation if clinically indicated.
Collapse
Affiliation(s)
- Ariharan Anantharachagan
- Department of Allergy and Clinical Immunology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
- Department of Immunology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
| | - Sook Yin Loh
- Department of Allergy and Clinical Immunology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, United Kingdom
| | - Siobhan O. Burns
- University College London Institute of Immunity and Transplantation, London, United Kingdom
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Arian Laurence
- University College London Institute of Immunity and Transplantation, London, United Kingdom
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
- Department of Clinical Haematology, University College London NHS Foundation Trust, London, United Kingdom
| | - Susan Tadros
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Eleni Tholouli
- Manchester University NHS Foundation Trust, Department of Haematology, Manchester, United Kingdom
| | - Yadanar Lwin
- Department of Haematology, Nottingham, United Kingdom
| | | | - P. Vaitla
- Department of Immunology Nottingham University Hospitals, NHS Trust, Nottingham, United Kingdom
| | - Emma C. Morris
- University College London Institute of Immunity and Transplantation, London, United Kingdom
- Department of Immunology, The Royal Free London NHS Foundation Trust, London, United Kingdom
- Department of Clinical Haematology, University College London NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
13
|
Wang JJF, Dhir A, Hildebrand KJ, Turvey SE, Schellenberg R, Chen LYC, Pourshahnazari P, Biggs CM. Inborn errors of immunity in adulthood. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2024; 20:6. [PMID: 38233962 DOI: 10.1186/s13223-023-00862-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/26/2023] [Indexed: 01/19/2024]
Abstract
Inborn errors of immunity (IEIs) are a group of conditions whereby parts of the immune system are missing or dysfunctional. Once thought to primarily be a pediatric disorder, it is now estimated that more than 50% of worldwide incident IEI cases are accounted for by adults. Delayed diagnosis, late symptom onset, and IEI phenocopies can all lead to adult-onset recognition of IEIs. Lack of awareness regarding the diversity of IEI manifestations in adults contributes to diagnostic and treatment delays. Prompt referral to immunology is critical so that patients can receive a precise molecular diagnosis and targeted therapy when available. This article serves as a primer on IEIs in adulthood, highlighting the pathophysiology, epidemiology and clinical features. We present clinical vignettes of three key IEIs to assist clinicians in building illness scripts on their presentations. We provide a framework for the laboratory evaluation of IEIs and their initial treatment, with the aim of improving recognition and management of these conditions.
Collapse
Affiliation(s)
- Joanne J F Wang
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Arün Dhir
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Kyla J Hildebrand
- Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | | | - Luke Y C Chen
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | | | - Catherine M Biggs
- Department of Medicine, University of British Columbia, Vancouver, Canada.
- Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
14
|
Cattoni A, Nicolosi ML, Capitoli G, Gadda A, Molinari S, Louka S, Buonsante A, Orlandi S, Salierno G, Bellani I, Vendemini F, Ottaviano G, Gaiero A, Fichera G, Biondi A, Balduzzi A. Pubertal attainment and Leydig cell function following pediatric hematopoietic stem cell transplantation: a three-decade longitudinal assessment. Front Endocrinol (Lausanne) 2023; 14:1292683. [PMID: 38152128 PMCID: PMC10751351 DOI: 10.3389/fendo.2023.1292683] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/01/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction Impaired testosterone secretion is a frequent sequela following hematopoietic stem cell transplantation (HSCT) in pediatrics, but long-term longitudinal trendlines of clinical and biochemical findings are still scanty. Methods Monocentric, retrospective analysis. Male patients transplanted <18 years between 1992 and 2021, surviving ≥2 years after HSCT and showing, upon enrollment, clinical and biochemical signs consistent with pubertal onset and progression were included. Clinical and biochemical data collected every 6-12 months were recorded. Results Of 130 patients enrolled, 56% were prepubertal, while 44% were peri-/postpubertal upon HSCT. Overall, 44% showed spontaneous progression into puberty and normal gonadal profile, while the remaining experienced pubertal arrest (1%), isolated increase of FSH (19%), compensated (23%) or overt (13%) hypergonadotropic hypogonadism. Post-pubertal testicular volume (TV) was statistically smaller among patients still pre-pubertal upon HSCT (p 0.049), whereas no differences were recorded in adult testosterone levels. LH and testosterone levels showed a specular trend between 20 and 30 years, as a progressive decrease in sexual steroids was associated with a compensatory increase of the luteinizing hormone. A variable degree of gonadal dysfunction was reported in 85%, 51%, 32% and 0% of patients following total body irradiation- (TBI), busulfan-, cyclophosphamide- and treosulfan-based regimens, respectively. TBI and busulfan cohorts were associated with the lowest probability of gonadal event-free course (p<0.0001), while it achieved 100% following treosulfan. A statistically greater gonadotoxicity was detected after busulfan than treosulfan (p 0.024). Chemo-only regimens were associated with statistically larger TV (p <0.001), higher testosterone (p 0.008) and lower gonadotropin levels (p <0.001) than TBI. Accordingly, the latter was associated with a 2-fold increase in the risk of gonadal failure compared to busulfan (OR 2.34, CI 1.08-8.40), whereas being pre-pubertal upon HSCT was associated with a reduced risk (OR 0.15, CI 0.08-0.30). Conclusions a) patients pre-pubertal upon HSCT showed a reduced risk of testicular endocrine dysfunction, despite smaller adult TV; b) patients showed downwards trend in testosterone levels after full pubertal attainment, despite a compensatory increase in LH; c) treosulfan was associated to a statistically lower occurrence of hypogonadism than busulfan, with a trend towards larger TV, higher testosterone levels and lower gonadotropins.
Collapse
Affiliation(s)
- Alessandro Cattoni
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Maria Laura Nicolosi
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Capitoli
- Bicocca Bioinformatics, Biostatistics and Bioimaging B4 Centre, University of Milano-Bicocca, Monza, Italy
| | - Alberto Gadda
- Bicocca Bioinformatics, Biostatistics and Bioimaging B4 Centre, University of Milano-Bicocca, Monza, Italy
| | - Silvia Molinari
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Sotiris Louka
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Andrea Buonsante
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Simona Orlandi
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Gianluca Salierno
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Iacopo Bellani
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Francesca Vendemini
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giorgio Ottaviano
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alberto Gaiero
- Department of Pediatrics and Neonatology, IRCCS Gaslini Savona e Pietra Ligure, Savona, Italy
| | | | - Andrea Biondi
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Adriana Balduzzi
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
15
|
Jiang D, Rosenlind K, Baxter S, Gernsheimer T, Gulsuner S, Allenspach EJ, Keel SB. Evaluating the prevalence of inborn errors of immunity in adults with chronic immune thrombocytopenia or Evans syndrome. Blood Adv 2023; 7:7202-7208. [PMID: 37792884 PMCID: PMC10702780 DOI: 10.1182/bloodadvances.2023011042] [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: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 10/06/2023] Open
Abstract
Inborn errors of immunity (IEIs) are monogenic disorders that predispose patients to immune dysregulation, autoimmunity, and infection. Autoimmune cytopenias, such as immune thrombocytopenia (ITP) and Evans syndrome (a combination of ITP and autoimmune hemolytic anemia), are increasingly recognized phenotypes of IEI. Although recent findings suggest that IEIs may commonly underlie pediatric ITP and Evans syndrome, its prevalence in adult patients with these disorders remains undefined. This study sought to estimate the prevalence of underlying IEIs among adults with persistent or chronic ITP or Evans syndrome using a next-generation sequencing panel encompassing >370 genes implicated in IEIs. Forty-four subjects were enrolled from an outpatient adult hematology clinic at a tertiary referral center in the United States, with a median age of 49 years (range, 20-83). Fourteen subjects (31.8%) had secondary ITP, including 8 (18.2%) with Evans syndrome. No cases of IEI were identified despite a high representation of subjects with a personal history of autoimmunity (45.5%) and early onset of disease (median age at diagnosis of 40 years [range, 2-77]), including 20.5% who were initially diagnosed as children. Eight subjects (18.2%) were found to be carriers of pathogenic IEI variants, which, in their heterozygous state, are not disease-causing. One case of TUBB1-related congenital thrombocytopenia was identified. Although systematic screening for IEI has been proposed for pediatric patients with Evans syndrome, findings from this real-world study suggest that inclusion of genetic testing for IEI in the routine work-up of adults with ITP and Evans syndrome has a low diagnostic yield.
Collapse
MESH Headings
- Humans
- Adult
- Child
- Young Adult
- Middle Aged
- Aged
- Aged, 80 and over
- Child, Preschool
- Adolescent
- Anemia, Hemolytic, Autoimmune/epidemiology
- Anemia, Hemolytic, Autoimmune/genetics
- Anemia, Hemolytic, Autoimmune/complications
- Purpura, Thrombocytopenic, Idiopathic/epidemiology
- Purpura, Thrombocytopenic, Idiopathic/genetics
- Purpura, Thrombocytopenic, Idiopathic/complications
- Autoimmunity
- Prevalence
- Thrombocytopenia/epidemiology
- Thrombocytopenia/genetics
- Thrombocytopenia/complications
Collapse
Affiliation(s)
- Debbie Jiang
- Division of Hematology, University of Washington, Seattle, WA
- Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology, Massachusetts General Hospital, Boston, MA
| | | | - Sarah Baxter
- Division of Rheumatology, Seattle Children’s Hospital, Seattle, WA
| | - Terry Gernsheimer
- Division of Hematology, University of Washington, Seattle, WA
- Fred Hutchinson Cancer Center, Seattle, WA
| | | | | | - Siobán B. Keel
- Division of Hematology, University of Washington, Seattle, WA
- Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
16
|
Arnold DE, Pai SY. Progress in the field of hematopoietic stem cell-based therapies for inborn errors of immunity. Curr Opin Pediatr 2023; 35:663-670. [PMID: 37732933 PMCID: PMC10872717 DOI: 10.1097/mop.0000000000001292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
PURPOSE OF REVIEW Hematopoietic stem cell-based therapies, including allogeneic hematopoietic cell transplantation (HCT) and autologous gene therapy (GT), have been used as curative therapy for many inborn errors of immunity (IEI). As the number of genetically defined IEI and the use of HCT and GT increase, valuable data on outcomes and approaches for specific disorders are available. We review recent progress in HCT and GT for IEI in this article. RECENT FINDINGS Novel approaches to prevention of allogeneic complications and experience in adolescents and young adults have expanded the use of HCT. Universal newborn screening for severe combined immunodeficiency (SCID) has led to improved outcome after HCT. Analysis of outcomes of HCT and GT for SCID, Wiskott-Aldrich syndrome (WAS) and chronic granulomatous disease (CGD) reveal risk factors for survival, the impact of specific conditioning regimens, and vector- or disease-specific impacts on efficacy and safety. Preclinical studies of GT and gene editing show potential for translation to the clinic. SUMMARY Emerging data on outcome after HCT for specific IEI support early evaluation and treatment, before development of co-morbidities. Data in large cooperative retrospective databases continues to yield valuable insights clinicians can use in patient selection and choice of therapy.
Collapse
Affiliation(s)
- Danielle E. Arnold
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Sung-Yun Pai
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Mehta P, Tsilifis C, Lum SH, Slatter MA, Hambleton S, Owens S, Williams E, Flood T, Gennery AR, Nademi Z. Outcome of Second Allogeneic HSCT for Patients with Inborn Errors of Immunity: Retrospective Study of 20 Years' Experience. J Clin Immunol 2023; 43:1812-1826. [PMID: 37452206 DOI: 10.1007/s10875-023-01549-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
A significant complication of HSCT is graft failure, although few studies focus on this problem in patients with inborn errors of immunity (IE). We explored outcome of second HSCT for IEI by a retrospective, single-centre study between 2002 and 2022. Four hundred ninety-three patients underwent allogeneic HSCT for severe combined immunodeficiency (SCID; n = 113, 22.9%) or non-SCID IEI (n = 380, 77.1%). Thirty patients (6.0%) required second HSCT. Unconditioned infusion or no serotherapy at first HSCT was more common in patients who required second transplant. Median interval between first and second HSCT was 0.97 years (range: 0.19-8.60 years); a different donor was selected for second HSCT in 24/30 (80.0%) patients. Conditioning regimens for second HSCT were predominately treosulfan-based (with thiotepa: n = 18, 60.0%; without, n = 6, 20.0%). Patients received grafts from peripheral blood stem cell (n = 25, 83.3%) or bone marrow (n = 5, 16.7%) with median stem cell dose 9.5 × 106 CD34 + cells/kilogram (range: 1.4-32.3). Median follow-up was 1.92 years (0.22-16.0). Overall survival was 80.8% and event-free survival was 64.7%. Four patients died, two of early-transplant related complications, and two of late sepsis post-second HSCT. Three patients required third HSCT; all are alive with 100% donor chimerism. Cumulative incidence of acute graft-versus-host disease was 28.4%, (all grade I-II). Viral reactivation was seen in 13/30 (43.3%) patients, including HHV6 (n = 6), CMV (n = 4), and adenovirus (n = 2). At latest follow-up, 25/26 surviving patients have donor chimerism ≥ 90% and 16/25 (64.0%) have discontinued immunoglobulin replacement. Second HSCT offers IEI patients with graft failure curative treatment with good overall survival and immunological recovery.
Collapse
Affiliation(s)
- Priti Mehta
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
| | - Christo Tsilifis
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Su Han Lum
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Mary A Slatter
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Sophie Hambleton
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Stephen Owens
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
| | - Eleri Williams
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
| | - Terry Flood
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
| | - Andrew R Gennery
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Zohreh Nademi
- Children's Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| |
Collapse
|
18
|
Izadi N, Sullivan KE. Difficult Cases in Primary Immunodeficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:3559-3560.e9. [PMID: 37945212 DOI: 10.1016/j.jaip.2023.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 11/12/2023]
Affiliation(s)
- Neema Izadi
- Division of Clinical Immunology and Allergy, Children's Hospital Los Angeles, Keck School of Medicine of USC, Los Angeles, Calif.
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Perelman School of Medicine of UPenn, Philadelphia, Pa
| |
Collapse
|
19
|
Tsilifis C, Torppa T, Williams EJ, Albert MH, Hauck F, Soncini E, Kang E, Malech H, Schuetz C, von Bernuth H, Slatter MA, Gennery AR. Allogeneic HSCT for Symptomatic Female X-linked Chronic Granulomatous Disease Carriers. J Clin Immunol 2023; 43:1964-1973. [PMID: 37620741 PMCID: PMC10661721 DOI: 10.1007/s10875-023-01570-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
X-linked chronic granulomatous disease (XL-CGD) is an inherited disorder of superoxide production, causing failure to generate the oxidative burst in phagocytes. It is characterized by invasive bacterial and fungal infections, inflammation, and chronic autoimmune disease. While XL-CGD carriers were previously assumed to be healthy, a range of clinical manifestations with significant morbidity have recently been described in a subgroup of carriers with impaired neutrophil oxidative burst due to skewed lyonization. Allogeneic hematopoietic stem cell transplantation (HSCT) is the standard curative treatment for CGD but has rarely been reported in individual symptomatic carriers to date. We undertook a retrospective international survey of outcome of HSCT for symptomatic XL-CGD carriers. Seven symptomatic female XL-CGD carriers aged 1-56 years underwent HSCT in four centers, indicated for severe and recurrent infection, colitis, and autoimmunity. Two patients died from transplant-related complications, following donor engraftment and restoration of oxidative burst. All surviving patients demonstrated resolution of their neutrophil oxidative burst defect with concordant reduction in infection and inflammatory symptoms and freedom from further immunosuppressive therapy. In conclusion, allogeneic HSCT may cure the phagocyte defect in symptomatic XL-CGD carriers and improve their recurrent and disabling infective and inflammatory symptoms but risks transplant-related complications.
Collapse
Affiliation(s)
- Christo Tsilifis
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Ward 3, Newcastle Upon Tyne, NE1 4LP, UK.
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.
| | - Tuulia Torppa
- School of Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Eleri J Williams
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Ward 3, Newcastle Upon Tyne, NE1 4LP, UK
| | - Michael H Albert
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, LMU, Munich, Germany
| | - Fabian Hauck
- Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, LMU, Munich, Germany
| | - Elena Soncini
- Paediatric Haematopoietic Stem Cell Transplant Unit, Children's Hospital ASST Spedali Civili, Brescia, Italy
| | - Elizabeth Kang
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Harry Malech
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Catharina Schuetz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Mary A Slatter
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Ward 3, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Andrew R Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Ward 3, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
20
|
Giardino S, Pierri F, Faraci M. How to optimize outcome of patients undergoing HLA-matched related haematopoietic stem cell transplantation in acquired and inherited bone marrow failure syndromes. Br J Haematol 2023; 203:158-160. [PMID: 37583352 DOI: 10.1111/bjh.19047] [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: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
Up-front allogeneic haematopoietic stem cell transplantation (allo-HSCT) after a reduced intensity conditioning regimen is the standard treatment in children with acquired severe aplastic anaemia (aSAA) and inherited bone marrow failure syndromes (iBMFs) in the presence of a healthy matched related donor (MRD). The paper by Alsultan et al. report the safety and efficacy of MRD HSCT conditioned with low-dose cyclophosphamide, fludarabine and thymoglobulin in both aSAA and non-Fanconi iBMFs, strengthening the concept of the pivotal role of immunosuppressive approach in allo-HSCT for specific subgroups of non-malignant diseases requiring a reduced risk of toxicities, offering the opportunity to discuss the essential points for achieving patients' long-term survival after MRD HSCT in BMF. Commentary on: Alsultan et al. Human leucocyte antigen-matched related haematopoietic stem cell transplantation using low-dose cyclophosphamide, fludarabine and thymoglobulin in children with severe aplastic anaemia. Br J Haematol 2023;203:255-263.
Collapse
Affiliation(s)
- Stefano Giardino
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology and Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Filomena Pierri
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology and Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Maura Faraci
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology and Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
21
|
Ersoy GZ, Çipe F, Fışgın T, Aksoy BA, Öner ÖB, Hashemi N, Aydoğdu S, Erdem M, Dikme G, Murat K, Bozkurt C. The impact of Treosulfan-based conditioning for inborn errors of immunity: Is dose monitoring crucial? Clin Transplant 2023; 37:e15083. [PMID: 37534623 DOI: 10.1111/ctr.15083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION In children with inborn errors of immunity (IEI) who will receive a hematopoietic stem cell transplant (HSCT) treosulfan-based conditioning is currently preferred. The aim of this study was to investigate early and late outcomes in pediatric IEI patients receiving pre-HSCT treosulfan and to examine the effect of treosulfan dose monitoring on outcomes. METHODS Seventy-three pediatric patients receiving this management between 2015 and 2022 were included. RESULTS Overall survival rate was 80%, and event-free survival was 67.8%. A larger treosulfan dose AUC after first application increased the rate of early toxicity (p = .034) and slowed lymphocyte engraftment (r = .290; p = .030). Underlying disease, treosulfan AUC, donor type, stem cell type, number of immunosuppressive agents, the dose of anti-thymocyte globulin, and post-transplantation cyclophosphamide did not to increase risk of acute graft-versus-host disease. The risk of mixed chimerism (MC) in patients with autoimmune lymphoproliferative syndrome and leukocyte adhesion deficiency were higher than those with severe combined immunodeficiency (p = .021 and p = .014, respectively). The risk of MC was lower in those receiving peripheral blood stem cells (SC) compared with bone marrow derived SC (OR = .204, p = .022). CONCLUSION The AUC of the treosulfan dose was not associated with poorer late outcomes. Treosulfan is an agent that can be used safely in the IEI patient group, level measurement appears essential to identify early toxicities. Prospective studies with more extended follow-up periods are needed.
Collapse
Affiliation(s)
- Gizem Zengin Ersoy
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Funda Çipe
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Allergy-Immunology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Tunç Fışgın
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Basak Adakli Aksoy
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Özlem Başoğlu Öner
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Nazlı Hashemi
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Selime Aydoğdu
- Umraniye Research & Training Hospital Pediatric Hematology & Oncology Department, Medical Sciences University, İstanbul, Turkey
| | - Melek Erdem
- İstinye University Pediatric Hematology Oncology, Gaziosmanpaşa Medical Park Hospital, İstanbul, Turkey
| | - Gürcan Dikme
- Aydin University Pediatric Hematology Oncology, Florya Medical Park Hospital, İstanbul, Turkey
| | | | - Ceyhun Bozkurt
- Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstinye University Pediatric Hematology Oncology, İstanbul, Turkey
| |
Collapse
|
22
|
Grunebaum E, Booth C, Cuvelier GDE, Loves R, Aiuti A, Kohn DB. Updated Management Guidelines for Adenosine Deaminase Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1665-1675. [PMID: 36736952 DOI: 10.1016/j.jaip.2023.01.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/23/2022] [Accepted: 01/07/2023] [Indexed: 02/04/2023]
Abstract
Inherited defects in the adenosine deaminase (ADA) gene typically cause severe combined immunodeficiency. In addition to infections, ADA-deficient patients can present with neurodevelopmental, behavioral, hearing, skeletal, lung, heart, skin, kidney, urogenital, and liver abnormalities. Some patients also suffer from autoimmunity and malignancies. In recent years, there have been remarkable advances in the management of ADA deficiency. Most ADA-deficient patients can be identified by newborn screening for severe combined immunodeficiency, which facilitates early diagnosis and treatment of asymptomatic infants. Most patients benefit from enzyme replacement therapy (ERT). Allogeneic hematopoietic cell transplantation from an HLA-matched sibling donor or HLA-matched family member donor with no conditioning is currently the preferable treatment. When matched sibling donor or matched family member donor is not available, autologous ADA gene therapy with nonmyeloablative conditioning and ERT withdrawal, which is reported in recent studies to result in 100% overall survival and 90% to 95% engraftment, should be pursued. If gene therapy is not immediately available, ERT can be continued for a few years, although its excessive cost might be prohibitive. The recent improved outcome of hematopoietic cell transplantation using HLA-mismatched family-related donors or HLA-matched unrelated donors, after reduced-intensity conditioning, suggests that such procedures might also be considered rather than continuing ERT for prolonged periods. Long-term follow-up will further assist in determining the optimal treatment approach for ADA-deficient patients.
Collapse
Affiliation(s)
- Eyal Grunebaum
- Division of Immunology and Allergy, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Claire Booth
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital, London, United Kingdom
| | - Geoffrey D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robyn Loves
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, and the Università Vita-Salute San Raffaele, Milan, Italy
| | - Donald B Kohn
- Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, Calif
| |
Collapse
|
23
|
Laberko A, Mukhinа A, Machneva E, Pashchenko O, Bykova T, Vahonina L, Bronin G, Skvortsova Y, Skorobogatova E, Kondratenko I, Fechina L, Shcherbina A, Zubarovskaya L, Balashov D, Rumiantsev A. Allogeneic Hematopoietic Stem Cell Transplantation Activity in Inborn Errors of Immunity in Russian Federation. J Clin Immunol 2023:10.1007/s10875-023-01476-w. [PMID: 37009957 PMCID: PMC10068234 DOI: 10.1007/s10875-023-01476-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/20/2023] [Indexed: 04/04/2023]
Abstract
PURPOSE Allogeneic hematopoietic stem cell transplantation (HSCT) is an established therapy for many inborn errors of immunity (IEI). The indications for HSCT have expanded over the last decade. The study aimed to collect and analyze the data on HSCT activity in IEI in Russia. METHODS The data were collected from the Russian Primary Immunodeficiency Registry and complemented with information from five Russian pediatric transplant centers. Patients diagnosed with IEI by the age of 18 years and who received allogeneic HSCT by the end of 2020 were included. RESULTS From 1997 to 2020, 454 patients with IEI received 514 allogeneic HSCT. The median number of HSCTs per year has risen from 3 in 1997-2009 to 60 in 2015-2020. The most common groups of IEI were immunodeficiency affecting cellular and humoral immunity (26%), combined immunodeficiency with associated/syndromic features (28%), phagocyte defects (21%), and diseases of immune dysregulation (17%). The distribution of IEI diagnosis has changed: before 2012, the majority (65%) had severe combined immunodeficiency (SCID) and hemophagocytic lymphohistiocytosis (HLH), and after 2012, only 24% had SCID and HLH. Of 513 HSCTs, 48.5% were performed from matched-unrelated, 36.5% from mismatched-related (MMRD), and 15% from matched-related donors. In 349 transplants T-cell depletion was used: 325 TCRαβ/CD19+ depletion, 39 post-transplant cyclophosphamide, and 27 other. The proportion of MMRD has risen over the recent years. CONCLUSION The practice of HSCT in IEI has been changing in Russia. Expanding indications to HSCT and SCID newborn screening implementation may necessitate additional transplant beds for IEI in Russia.
Collapse
Affiliation(s)
- Alexandra Laberko
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Anna Mukhinа
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Russian National Association of Experts in Primary Immunodeficiency Registry, Moscow, Russia
| | - Elena Machneva
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Olga Pashchenko
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Tatiana Bykova
- RM Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Pavlov University, St. Petersburg, Russia
| | - Larisa Vahonina
- Sverdlovsk Regional Children's Hospital №1, Institute of Medical Cell Technologies, Yekaterinburg, Russia
| | | | - Yulia Skvortsova
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Elena Skorobogatova
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Irina Kondratenko
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Larisa Fechina
- Sverdlovsk Regional Children's Hospital №1, Institute of Medical Cell Technologies, Yekaterinburg, Russia
| | - Anna Shcherbina
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Ludmila Zubarovskaya
- RM Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Pavlov University, St. Petersburg, Russia
| | - Dmitry Balashov
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexander Rumiantsev
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
24
|
Fevang B. Treatment of inflammatory complications in common variable immunodeficiency (CVID): current concepts and future perspectives. Expert Rev Clin Immunol 2023; 19:627-638. [PMID: 36996348 DOI: 10.1080/1744666x.2023.2198208] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
INTRODUCTION Patients with Common variable immunodeficiency (CVID) have a high frequency of inflammatory complications like autoimmune cytopenias, interstitial lung disease and enteropathy. These patients have poor prognosis and effective, timely and safe treatment of inflammatory complications in CVID are essential, but guidelines and consensus on therapy are often lacking. AREAS COVERED This review will focus on current medical treatment of inflammatory complications in CVID and point out some future perspectives based on literature indexed in PubMed. There are a number of good observational studies and case reports on treatment of specific complications but randomized controlled trials are scarce. EXPERT OPINION In clinical practice, the most urgent issues that need to be addressed are the preferred treatment of GLILD, enteropathy and liver disease. Treating the underlying immune dysregulation and immune exhaustion in CVID is an alternative approach that potentially could alleviate these and other organ-specific inflammatory complications. Therapies of potential interest and wider use in CVID include mTOR-inhibitors like sirolimus, JAK-inhibitors like tofacitinib, the monoclonal IL-12/23 antibody ustekinumab, the anti-BAFF antibody belimumab and abatacept. For all inflammatory complications, there is a need for prospective therapeutic trials, preferably randomized controlled trials, and multi-center collaborations with larger cohorts of patients will be essential.
Collapse
Affiliation(s)
- Børre Fevang
- Centre for Rare Disorders, Oslo University Hospital, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Research Institute for Internal Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
25
|
Schuetz C, Gerke J, Ege M, Walter J, Kusters M, Worth A, Kanakry JA, Dimitrova D, Wolska-Kuśnierz B, Chen K, Unal E, Karakukcu M, Pashchenko O, Leiding J, Kawai T, Amrolia PJ, Berghuis D, Buechner J, Buchbinder D, Cowan MJ, Gennery AR, Güngör T, Heimall J, Miano M, Meyts I, Morris EC, Rivière J, Sharapova SO, Shaw PJ, Slatter M, Honig M, Veys P, Fischer A, Cavazzana M, Moshous D, Schulz A, Albert MH, Puck JM, Lankester AC, Notarangelo LD, Neven B. Hypomorphic RAG deficiency: impact of disease burden on survival and thymic recovery argues for early diagnosis and HSCT. Blood 2023; 141:713-724. [PMID: 36279417 PMCID: PMC10082356 DOI: 10.1182/blood.2022017667] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/19/2022] [Accepted: 10/04/2022] [Indexed: 11/20/2022] Open
Abstract
Patients with hypomorphic mutations in the RAG1 or RAG2 gene present with either Omenn syndrome or atypical combined immunodeficiency with a wide phenotypic range. Hematopoietic stem cell transplantation (HSCT) is potentially curative, but data are scarce. We report on a worldwide cohort of 60 patients with hypomorphic RAG variants who underwent HSCT, 78% of whom experienced infections (29% active at HSCT), 72% had autoimmunity, and 18% had granulomas pretransplant. These complications are frequently associated with organ damage. Eight individuals (13%) were diagnosed by newborn screening or family history. HSCT was performed at a median of 3.4 years (range 0.3-42.9 years) from matched unrelated donors, matched sibling or matched family donors, or mismatched donors in 48%, 22%, and 30% of the patients, respectively. Grafts were T-cell depleted in 15 cases (25%). Overall survival at 1 and 4 years was 77.5% and 67.5% (median follow-up of 39 months). Infection was the main cause of death. In univariable analysis, active infection, organ damage pre-HSCT, T-cell depletion of the graft, and transplant from a mismatched family donor were predictive of worse outcome, whereas organ damage and T-cell depletion remained significant in multivariable analysis (hazard ratio [HR] = 6.01, HR = 8.46, respectively). All patients diagnosed by newborn screening or family history survived. Cumulative incidences of acute and chronic graft-versus-host disease were 35% and 22%, respectively. Cumulative incidences of new-onset autoimmunity was 15%. Immune reconstitution, particularly recovery of naïve CD4+ T cells, was faster and more robust in patients transplanted before 3.5 years of age, and without organ damage. These findings support the indication for early transplantation.
Collapse
Affiliation(s)
- C. Schuetz
- Department of Paediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - J. Gerke
- Department of Paediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - M. Ege
- Dr. von Hauner Children’s Hospital at Ludwig-Maximilians-Universität, München, Germany
- Helmholtz Zentrum München, Neuherberg, Germany
| | - J. Walter
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - M. Kusters
- Department of Immunology and Gene therapy, Great Ormond Street Hospital, NHS Foundation trust, London, United Kingdom
| | - A. Worth
- Department of Immunology and Gene therapy, Great Ormond Street Hospital, NHS Foundation trust, London, United Kingdom
| | - J. A. Kanakry
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - D. Dimitrova
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - B. Wolska-Kuśnierz
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
| | - K. Chen
- Division of Allergy and Immunology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT
| | - E. Unal
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Erciyes University, Kayseri, Turkey
| | - M. Karakukcu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Erciyes University, Kayseri, Turkey
| | - O. Pashchenko
- Department of Immunology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - J. Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Orlando Health Arnold Pamer Hospital for Children, Orlando, FL
| | - T. Kawai
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - P. J. Amrolia
- Bone Marrow Transplant Unit, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - D. Berghuis
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - J. Buechner
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
| | - D. Buchbinder
- Division of Hematology, Children's Hospital of Orange County, Orange, CA
| | - M. J. Cowan
- Division of Allergy, Immunology, and Blood and Marrow Transplant, Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - A. R. Gennery
- Translational and Clinical Research Institute, Newcastle University, Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children’s Hospital, Newcastle upon Tyne, United Kingdom
| | - T. Güngör
- Department of Hematology/Oncology/Immunology, Gene-therapy, and Stem Cell Transplantation, University Children’s Hospital Zurich–Eleonore Foundation & Children’s Research Center, Zürich, Switzerland
| | - J. Heimall
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - M. Miano
- IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - I. Meyts
- Department of Pediatrics, Department of Microbiology and Immunology, University Hospitals Leuven, Leuven, Belgium
| | - E. C. Morris
- UCL Institute of Immunity & Transplantation, University College London Hospitals NHS Foundation Trust, Royal Free London Hospital NHS Foundation Trust, London, United Kingdom
| | - J. Rivière
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d'Hebron Research Institute, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - S. O. Sharapova
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - P. J. Shaw
- Blood Transplant and Cell Therapies, Children’s Hospital at Westmead, Sydney, Australia
| | - M. Slatter
- Paediatric Immunology & HSCT, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - M. Honig
- Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| | - P. Veys
- Bone Marrow Transplant Unit, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - A. Fischer
- Paediatric Immunology, Department of Immunology, Haematology and Rheumatology, Necker-Enfants Malades, Paris, France
- Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
- Collège de France, Paris, France
| | - M. Cavazzana
- Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
- Département de Biothérapie, Hôpital Universitaire Necker-Enfants Malades, Groupe Hospitalier Paris Centre, Assistance Publique–Hopitaux de Paris, Paris, France
- Centre d’Investigation Clinique Biothérapie, Groupe hospitalier Universitaire paris centre, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France
| | - D. Moshous
- Paediatric Immunology, Department of Immunology, Haematology and Rheumatology, Necker-Enfants Malades, Paris, France
- Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - A. Schulz
- Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| | - M. H. Albert
- Pediatric SCT Program, Dr. von Hauner University Children’s Hospital, Ludwig-Maximilians Universität, München, Germany
| | - J. M. Puck
- Division of Allergy, Immunology, and Blood and Marrow Transplant, Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - A. C. Lankester
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - L. D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - B. Neven
- Paediatric Immunology, Department of Immunology, Haematology and Rheumatology, Necker-Enfants Malades, Paris, France
| | - Inborn Errors Working Party (IEWP) of the European Society for Immunodeficiencies (ESID) and European Society for Blood and Marrow Transplantation (EBMT) and the Primary Immune Deficiency Treatment Consortium (PIDTC)
- Department of Paediatrics, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Dr. von Hauner Children’s Hospital at Ludwig-Maximilians-Universität, München, Germany
- Helmholtz Zentrum München, Neuherberg, Germany
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
- Department of Immunology and Gene therapy, Great Ormond Street Hospital, NHS Foundation trust, London, United Kingdom
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
- Division of Allergy and Immunology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Erciyes University, Kayseri, Turkey
- Department of Immunology, Pirogov Russian National Research Medical University, Moscow, Russia
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Orlando Health Arnold Pamer Hospital for Children, Orlando, FL
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Bone Marrow Transplant Unit, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pediatric Hematology and Oncology, Oslo University Hospital, Oslo, Norway
- Division of Hematology, Children's Hospital of Orange County, Orange, CA
- Division of Allergy, Immunology, and Blood and Marrow Transplant, Department of Pediatrics, University of California San Francisco, San Francisco, CA
- Translational and Clinical Research Institute, Newcastle University, Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children’s Hospital, Newcastle upon Tyne, United Kingdom
- Department of Hematology/Oncology/Immunology, Gene-therapy, and Stem Cell Transplantation, University Children’s Hospital Zurich–Eleonore Foundation & Children’s Research Center, Zürich, Switzerland
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Pediatrics, Department of Microbiology and Immunology, University Hospitals Leuven, Leuven, Belgium
- UCL Institute of Immunity & Transplantation, University College London Hospitals NHS Foundation Trust, Royal Free London Hospital NHS Foundation Trust, London, United Kingdom
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d'Hebron Research Institute, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
- Blood Transplant and Cell Therapies, Children’s Hospital at Westmead, Sydney, Australia
- Paediatric Immunology & HSCT, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
- Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
- Bone Marrow Transplant Unit, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
- Paediatric Immunology, Department of Immunology, Haematology and Rheumatology, Necker-Enfants Malades, Paris, France
- Institut Imagine, Paris Descartes-Sorbonne Paris Cité University, Paris, France
- Collège de France, Paris, France
- Département de Biothérapie, Hôpital Universitaire Necker-Enfants Malades, Groupe Hospitalier Paris Centre, Assistance Publique–Hopitaux de Paris, Paris, France
- Centre d’Investigation Clinique Biothérapie, Groupe hospitalier Universitaire paris centre, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France
- Pediatric SCT Program, Dr. von Hauner University Children’s Hospital, Ludwig-Maximilians Universität, München, Germany
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
26
|
Allogeneic stem cell transplantation compared to conservative management in adults with inborn errors of immunity. Blood 2023; 141:60-71. [PMID: 36167031 DOI: 10.1182/blood.2022015482] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 01/10/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) is curative for severe inborn errors of immunity (IEIs), with recent data suggesting alloSCT in adulthood is safe and effective in selected patients. However, questions remain regarding the indications for and optimal timing of transplant. We retrospectively compared outcomes of transplanted vs matched nontransplanted adults with severe IEIs. Seventy-nine patients (aged ≥ 15 years) underwent alloSCT between 2008 and 2018 for IEIs such as chronic granulomatous disease (n = 20) and various combined immune deficiencies (n = 59). A cohort of nontransplanted patients from the French Centre de Référence Déficits Immunitaires Héréditaires registry was identified blindly for case-control analysis, with ≤3 matched controls per index patient, without replacement. The nontransplanted patients were matched for birth decade, age at last review greater than index patient age at alloSCT, chronic granulomatous disease or combined immune deficiencies, and autoimmune/lymphoproliferative complications. A total of 281 patients were included (79 transplanted, 202 nontransplanted). Median age at transplant was 21 years. Transplant indications were mainly lymphoproliferative disease (n = 23) or colitis (n = 15). Median follow-up was 4.8 years (interquartile range, 2.5-7.2). One-year transplant-related mortality rate was 13%. Estimated disease-free survival at 5 years was higher in transplanted patients (58% vs 33%; P = .007). Nontransplanted patients had an ongoing risk of severe events, with an increased mean cumulative number of recurrent events compared with transplanted patients. Sensitivity analyses removing patients with common variable immune deficiency and their matched transplanted patients confirm these results. AlloSCT prevents progressive morbidity associated with IEIs in adults, which may outweigh the negative impact of transplant-related mortality.
Collapse
|