1
|
Eissa H, Cowan MJ, Heimall J. Late effects following hematopoietic cell transplantation for severe combined immunodeficiency: critical factors and therapeutic options. Expert Rev Clin Immunol 2025; 21:73-82. [PMID: 39307944 DOI: 10.1080/1744666x.2024.2402948] [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: 05/26/2024] [Accepted: 09/06/2024] [Indexed: 01/02/2025]
Abstract
INTRODUCTION Severe combined immunodeficiency (SCID) is an inborn error of immunity that is fatal without hematopoietic cell transplantation (HCT) or gene therapy (GT). Survival outcomes have improved, largely due to implementation of SCID newborn screening. A better understanding of the long-term outcomes and late effects to address critical aspects of monitoring immune and general health life-long is needed. AREAS COVERED In a comprehensive review of PubMed indexed articles with publication dates 2008-2024 we describe the current knowledge of chronic and late effects (CLE) of HCT survivors for SCID as well as the role of GT and advances for specific SCID genotypes. We review factors affecting the development of CLE including disease related factors (genotype, trigger for diagnosis and presence of infection prior to HCT), transplant related factors (type of donor, conditioning regimen, immune reconstitution and graft versus host disease (GVHD) and describe causes and factors associated with higher risk for late mortality in this unique population. We further describe monitoring and potential therapeutic strategies for management of common CLE in this patient population. EXPERT OPINION Ongoing research efforts are needed to better describe CLE in survivors, to develop prospective clinical trials aimed at mitigating these CLE, and developing genotype-based approaches for management and follow-up of these patients.
Collapse
Affiliation(s)
- Hesham Eissa
- Division of Pediatric Hematology-Oncology-BMT, University of Colorado, Aurora, CO, USA
| | - Morton J Cowan
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, CA, USA
- Department of Pediatrics, UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Jennifer Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
2
|
Bhatt NS, Harris AC, Gorfinkel L, Ibanez K, Tkaczyk ER, Mitchell SA, Albuquerque S, Schechter T, Pavletic S, Duncan CN, Rotz SJ, Williams K, Carpenter PA, Cuvelier GDE. Pediatric Transplant and Cellular Therapy Consortium RESILIENT Conference on Pediatric Chronic Graft-Versus-Host Disease Survivorship After Hematopoietic Cell Transplantation: Part I. Phases of Chronic GVHD, Supportive Care, and Systemic Therapy Discontinuation. Transplant Cell Ther 2024:S2666-6367(24)00812-1. [PMID: 39701289 DOI: 10.1016/j.jtct.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Current literature lacks details on the impact of pediatric chronic graft-versus-host disease (cGVHD) on long-term survivorship after allogeneic hematopoietic cell transplantation (HCT). Nonetheless, cGVHD remains a leading cause of post-transplant morbidity and mortality in children and adolescents, which is particularly relevant given the longer life-expectancy after HCT (measured in decades) compared to older adults. To address this knowledge gap, leaders of the Pediatric Transplant and Cellular Therapy Consortium convened a multidisciplinary taskforce of experts in pediatric cGVHD and HCT late effects known as RESILIENT after Chronic GVHD (Research and Education towards Solutions for Late effects to Innovate, Excel, and Nurture after cGVHD). Our goals were to define: (1) the current state of understanding about how cGVHD impacts long-term survivorship in children transplanted <18 yr of age; (2) practical aspects of care to help clinicians managing long-term pediatric cGVHD survivors; and (3) develop a research framework for the next decade to further our knowledge. Four working groups were formed, each tasked with addressing a unique theme: (1) cGVHD natural history (phases of cGVHD) and its impact on clinicians' ability to taper and durably discontinue systemic therapy; (2) organ dysfunction and immune reconstitution in relation to survivorship; (3) how cGVHD and its treatment impact growth, metabolism, and development in children; and (4) psychosocial health and patient reported outcomes. The 4 groups met before the 2024 BMT Tandem Meeting in San Antonio, Texas, and then convened a larger in-person RESILIENT conference held on February 20, 2024, at the Tandem meeting to put forth recommendations from their respective working groups and garner feedback. These recommendations are now presented in a series of 4 manuscripts. This current manuscript focuses on the first theme and discusses the phases of cGVHD, challenges in differentiating clinically active from quiescent cGVHD in clinical practice, and the resultant difficulties in determining when and if to taper systemic therapy. To overcome these challenges, we propose revised categorization of long-term cGVHD outcomes and practical recommendations for clinicians and researchers around the long-term follow-up for these patients, including determining when and if to taper systemic therapy, along with the integration of non-immunosuppressive supportive care interventions.
Collapse
Affiliation(s)
- Neel S Bhatt
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, Washington
| | - Andrew C Harris
- Pediatric Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center; New York, New York
| | - Lev Gorfinkel
- Department of Pediatric Hematology-Oncology, Boston Children's Hospital, Dana Farber Cancer Institute; Boston, Massachusetts
| | - Katarzyna Ibanez
- Department of Neurology, Rehabilitation Service, Memorial Sloan Kettering Cancer Center; New York, New York
| | - Eric R Tkaczyk
- Department of Dermatology, Vanderbilt University Medical Center and Department of Veteran Affairs; Nashville, Tennessee
| | - Sandra A Mitchell
- Outcomes Research Branch, Division of Cancer Control and Population Sciences, National Cancer Institute; Rockville, Maryland
| | - Stacey Albuquerque
- Boston Children's Hospital Cancer and Blood Disorders Center, Dana Farber Cancer Institute; Boston, Massachusetts
| | - Tal Schechter
- Division of Pediatric Hematology, Oncology, BMT, and Cellular Therapy, The Hospital for Sick Children, University of Toronto; Toronto, Ontario, Canada
| | - Steven Pavletic
- Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, Maryland
| | - Christine N Duncan
- Department of Pediatric Hematology-Oncology, Boston Children's Hospital, Dana Farber Cancer Institute; Boston, Massachusetts
| | - Seth J Rotz
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Cleveland Clinic; Cleveland, Ohio
| | - Kirsten Williams
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University; Atlanta, Georgia
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center; Seattle, Washington
| | - Geoffrey D E Cuvelier
- Department of Pediatric Oncology and Transplantation, Alberta Children's Hospital, University of Calgary; Calgary, Alberta, Canada.
| |
Collapse
|
3
|
DiGiacomo D, Barmettler S. Secondary hypogammaglobulinemia: diagnosis and management of a pediatric condition of clinical importance. Curr Opin Pediatr 2024; 36:659-667. [PMID: 39254658 PMCID: PMC11560679 DOI: 10.1097/mop.0000000000001396] [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: 09/11/2024]
Abstract
PURPOSE OF REVIEW Secondary hypogammaglobulinemia, or low serum immunoglobulins, is associated with a variety of medications or medical conditions and may be symptomatic and lead to increased infectious risk. There is limited data regarding the study of acquired, or secondary, hypogammaglobulinemia (SHG) in pediatrics. The data to date has suffered from methodologic issues including retrospective study design, lack of baseline immunoglobulin measurements, and limited longitudinal follow-up. RECENT FINDINGS There is emerging research on the impact of B-cell depleting therapies, specifically rituximab and chimeric antigen T-cells, along with other autoimmune and malignant disease states, in the development of SHG in pediatric patients. This review will also summarize other relevant pediatric conditions related to SHG. SUMMARY The clinical relevance of SHG in pediatrics is increasingly appreciated. Improved understanding of the specific etiologies, risk factors, and natural history of SHG have informed screening and management recommendations.
Collapse
Affiliation(s)
- Daniel DiGiacomo
- Department of Pediatrics, K. Hovnanian Children’s Hospital, Jersey Shore University Medical Center, Neptune, New Jersey, USA
- Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - Sara Barmettler
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
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
|
5
|
Bense JE, Guilonard N, Zwaginga F, Stiggelbout AM, Louwerens M, Mekelenkamp H, Lankester AC, Pieterse AH, de Pagter APJ. The value of using patient-reported outcomes for health screening during long-term follow-up after paediatric stem cell transplantation for nonmalignant diseases. Health Expect 2024; 27:e13902. [PMID: 38102818 PMCID: PMC10768862 DOI: 10.1111/hex.13902] [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: 06/26/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 12/17/2023] Open
Abstract
INTRODUCTION The assessment of using patient-reported outcomes (PROs) within comprehensive care follow-up programmes, specifically focused on health screening, remains largely unexplored. PROs were implemented in our late effects and comprehensive care programme after paediatric hematopoietic stem cell transplantation (HSCT) for nonmalignant diseases. The programme focuses solely on screening of physical and mental health and on discussing PROs during the consultation. METHODS The primary method of this study was semistructured interviews to explore the perspective of both patients and healthcare providers' (HCP) on the use of PROs, which were thematically analyzed. Additionally, an explorative quantitative approach with patient-reported experience measures (PREMS) was used, with a pretest-posttest design, to assess whether the use of PROs was accompanied by more patient-centred care. RESULTS From the patient-interviews (N = 15) four themes were extracted: use of PROs (1) help to discuss topics; (2) make the patients feel understood; (3) create a moment of self-reflection; and (4) make consultations more efficient. Pre- and postimplementation analysis of PREMs (N = 40) did not show significant differences in terms of patient-centeredness. CONCLUSION Our results demonstrate the added value of integrating PROs for health screening purposes within the long-term follow-up programme after paediatric HSCT, as perceived by both patient and HCP. With the active use of PROs, patients are stimulated to consciously assess their health status. PATIENT CONTRIBUTION This study included patients as participants. Caregivers were approached if patients were below a certain age. Additionally, preliminary results were shared with all patients (including nonparticipants) during a patient conference day.
Collapse
Affiliation(s)
- Joëll E. Bense
- Department of Pediatrics, Willem‐Alexander Children's Hospital, Division of Stem Cell TransplantationLeiden University Medical CenterLeidenThe Netherlands
| | - Nicole Guilonard
- Department of Pediatrics, Willem‐Alexander Children's Hospital, Division of Stem Cell TransplantationLeiden University Medical CenterLeidenThe Netherlands
| | - Femke Zwaginga
- Department of Pediatrics, Willem‐Alexander Children's Hospital, Division of Stem Cell TransplantationLeiden University Medical CenterLeidenThe Netherlands
| | - Anne M. Stiggelbout
- Department of Biomedical Data Sciences, Medical Decision MakingLeiden University Medical CenterLeidenThe Netherlands
- Erasmus School of Health Policy and ManagementErasmus University RotterdamRotterdamThe Netherlands
| | - Marloes Louwerens
- Department of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Hilda Mekelenkamp
- Department of Pediatrics, Willem‐Alexander Children's Hospital, Division of Stem Cell TransplantationLeiden University Medical CenterLeidenThe Netherlands
| | - Arjan C. Lankester
- Department of Pediatrics, Willem‐Alexander Children's Hospital, Division of Stem Cell TransplantationLeiden University Medical CenterLeidenThe Netherlands
| | - Arwen H. Pieterse
- Department of Biomedical Data Sciences, Medical Decision MakingLeiden University Medical CenterLeidenThe Netherlands
| | - Anne P. J. de Pagter
- Department of Pediatrics, Willem‐Alexander Children's Hospital, Division of Stem Cell TransplantationLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
6
|
Eissa H, Thakar MS, Shah AJ, Logan BR, Griffith LM, Dong H, Parrott RE, O'Reilly RJ, Dara J, Kapoor N, Forbes Satter L, Chandra S, Kapadia M, Chandrakasan S, Knutsen A, Jyonouchi SC, Molinari L, Rayes A, Ebens CL, Teira P, Dávila Saldaña BJ, Burroughs LM, Chaudhury S, Chellapandian D, Gillio AP, Goldman F, Malech HL, DeSantes K, Cuvelier GDE, Rozmus J, Quinones R, Yu LC, Broglie L, Aquino V, Shereck E, Moore TB, Vander Lugt MT, Mousallem TI, Oved JH, Dorsey M, Abdel-Azim H, Martinez C, Bleesing JH, Prockop S, Kohn DB, Bednarski JJ, Leiding J, Marsh RA, Torgerson T, Notarangelo LD, Pai SY, Pulsipher MA, Puck JM, Dvorak CC, Haddad E, Buckley RH, Cowan MJ, Heimall J. Posttransplantation late complications increase over time for patients with SCID: A Primary Immune Deficiency Treatment Consortium (PIDTC) landmark study. J Allergy Clin Immunol 2024; 153:287-296. [PMID: 37793572 PMCID: PMC11294800 DOI: 10.1016/j.jaci.2023.09.027] [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: 03/30/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND The Primary Immune Deficiency Treatment Consortium (PIDTC) enrolled children in the United States and Canada onto a retrospective multicenter natural history study of hematopoietic cell transplantation (HCT). OBJECTIVE We investigated outcomes of HCT for severe combined immunodeficiency (SCID). METHODS We evaluated the chronic and late effects (CLE) after HCT for SCID in 399 patients transplanted from 1982 to 2012 at 32 PIDTC centers. Eligibility criteria included survival to at least 2 years after HCT without need for subsequent cellular therapy. CLE were defined as either conditions present at any time before 2 years from HCT that remained unresolved (chronic), or new conditions that developed beyond 2 years after HCT (late). RESULTS The cumulative incidence of CLE was 25% in those alive at 2 years, increasing to 41% at 15 years after HCT. CLE were most prevalent in the neurologic (9%), neurodevelopmental (8%), and dental (8%) categories. Chemotherapy-based conditioning was associated with decreased-height z score at 2 to 5 years after HCT (P < .001), and with endocrine (P < .001) and dental (P = .05) CLE. CD4 count of ≤500 cells/μL and/or continued need for immunoglobulin replacement therapy >2 years after transplantation were associated with lower-height z scores. Continued survival from 2 to 15 years after HCT was 90%. The presence of any CLE was associated with increased risk of late death (hazard ratio, 7.21; 95% confidence interval, 2.71-19.18; P < .001). CONCLUSION Late morbidity after HCT for SCID was substantial, with an adverse impact on overall survival. This study provides evidence for development of survivorship guidelines based on disease characteristics and treatment exposure for patients after HCT for SCID.
Collapse
Affiliation(s)
- Hesham Eissa
- Division of Pediatric Hematology-Oncology-BMT, University of Colorado, Aurora, Wash.
| | - Monica S Thakar
- Fred Hutchinson Cancer Center, Seattle, Wash; Department of Pediatrics, University of Washington, Seattle, Wash
| | - Ami J Shah
- Pediatrics [Hematology/Oncology/Stem Cell Transplantation and Regenerative Medicine], Stanford University/Lucille Packard Children's Hospital, Palo Alto, Calif
| | - Brent R Logan
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wis
| | - Linda M Griffith
- Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Huaying Dong
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wis
| | | | - Richard J O'Reilly
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jasmeen Dara
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Neena Kapoor
- Division of Hematology, Oncology and Blood and Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, Calif
| | - Lisa Forbes Satter
- Immunology, Allergy, and Rheumatology, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Malika Kapadia
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Mass
| | | | - Alan Knutsen
- St Louis University, Cardinal Glennon Children's Hospital, St Louis, Mo
| | - Soma C Jyonouchi
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| | | | - Ahmad Rayes
- Division of Hematology, Oncology, Transplantation, and Immunology, Primary Children's Hospital, Huntsman Cancer Institute, Spense Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah
| | - Christen L Ebens
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapy, University of Minnesota Masonic Children's Hospital, Minneapolis, Minn
| | - Pierre Teira
- Paediatric Haematology Oncology, Ste-Justine Hospital, Montreal, Canada
| | | | - Lauri M Burroughs
- Fred Hutchinson Cancer Center, Seattle, Wash; Department of Pediatrics, University of Washington, Seattle, Wash
| | - Sonali Chaudhury
- Hematology, Oncology, Neuro-oncology & Stem Cell Transplantation Division, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Deepak Chellapandian
- Center for Cell and Gene Therapy for Non-malignant Conditions, Johns Hopkins All Children's Hospital, St Petersburg, Fla
| | - Alfred P Gillio
- Children's Cancer Institute, Hackensack University Medical Center, Hackensack, NJ
| | - Fredrick Goldman
- Division of Pediatric Hematology and Oncology and Bone Marrow Transplant, University of Alabama at Birmingham, Birmingham, Ala
| | | | - Kenneth DeSantes
- Division of Pediatric Hematology-Oncology & Bone Marrow Transplant, University of Wisconsin, American Family Children's Hospital, Madison, Wis
| | - Geoff D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, Winnipeg, Canada
| | - Jacob Rozmus
- Children's & Women's Health Centre of British Columbia, Vancouver, Canada
| | - Ralph Quinones
- Division of Pediatric Hematology-Oncology-BMT, University of Colorado, Aurora, Wash
| | - Lolie C Yu
- Division of Heme-Onc/HSCT, Children's Hospital/LSUHSC, New Orleans, La
| | - Larisa Broglie
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Medical College of Wisconsin, Milwaukee, Wis
| | - Victor Aquino
- Division of Pediatric Hematology and Oncology, The University of Texas Southwestern Medical Center, Dallas, Tex
| | - Evan Shereck
- Division of Pediatric Hematology/Oncology, Oregon Health and Science University, Portland, Ore
| | - Theodore B Moore
- Department of Pediatric Hematology-Oncology, Mattel Children's Hospital, University of California, Los Angeles, Calif
| | - Mark T Vander Lugt
- Blood and Marrow Transplant Program, University of Michigan, Ann Arbor, Mich
| | | | - Joeseph H Oved
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Morna Dorsey
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Hisham Abdel-Azim
- Division of Hematology, Oncology and Blood and Marrow Transplant, Children's Hospital Los Angeles, Los Angeles, Calif; Loma Linda University School of Medicine, Cancer Center, Children Hospital and Medical Center, Loma Linda, Calif
| | - Caridad Martinez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Tex
| | - Jacob H Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Susan Prockop
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Mass
| | | | - Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Jennifer Leiding
- Orlando Health Arnold Palmer Hospital for Children, Orlando, Fla
| | - Rebecca A Marsh
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | | | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Md
| | - Sung-Yun Pai
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Md
| | - Michael A Pulsipher
- Division of Hematology, Oncology, Transplantation, and Immunology, Primary Children's Hospital, Huntsman Cancer Institute, Spense Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah
| | - Jennifer M Puck
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Christopher C Dvorak
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Elie Haddad
- Department of Pediatrics and the Department of Microbiology, Immunology, and Infectious Diseases, University of Montreal, CHU Sainte-Justine, Montreal, Canada
| | | | - Morton J Cowan
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Jennifer Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| |
Collapse
|
7
|
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
|
8
|
Patel NC, Torgerson T, Thakar MS, Younger MEM, Sriaroon P, Pozos TC, Buckley RH, Morris D, Vilkama D, Heimall J. Safety and Efficacy of Hizentra® Following Pediatric Hematopoietic Cell Transplant for Treatment of Primary Immunodeficiencies. J Clin Immunol 2023; 43:1557-1565. [PMID: 37266769 PMCID: PMC10499723 DOI: 10.1007/s10875-023-01482-y] [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: 10/29/2022] [Accepted: 04/01/2023] [Indexed: 06/03/2023]
Abstract
Primary immunodeficiency disease (PIDD) comprises a group of disorders of immune function. Some of the most severe PIDD can be treated with hematopoietic cell transplant (HCT). Hizentra® is a 20% liquid IgG product approved for subcutaneous administration in adults and children greater than 2 years of age with PIDD-associated antibody deficiency. Limited information is available on the use of Hizentra® in children following HCT for PIDD. A multicenter retrospective chart review demonstrated 37 infants and children (median age 70.1 [range 12.0 to 176.4] months) with PIDD treated by HCT who received Hizentra® infusions over a median duration of 31 (range 4-96) months post-transplant. The most common indication for HCT was IL2RG SCID (n = 16). Thirty-two patients switched from IVIG to SCIG administration, due to one or more of the following reasons: patient/caregiver (n = 17) or physician (n = 12) preference, discontinuation of central venous catheter (n = 16), desire for home infusion (n = 12), improved IgG serum levels following lower levels on IVIG (n = 10), and loss of venous access (n = 8). Serious bacterial infections occurred at a rate of 0.041 per patient-year while on therapy. Weight percentile increased by a mean of 16% during the observation period, with females demonstrating the largest gains. Mild local reactions were observed in 24%; 76% had no local reactions. One serious adverse event (death from sepsis) was reported. Hizentra® was discontinued in 15 (41%) patients, most commonly due to recovery of B cell function (n = 11). These data demonstrate that Hizentra® is a safe and effective option in children who have received HCT for PIDD.
Collapse
Affiliation(s)
- Niraj C Patel
- Department of Pediatrics, Division of Allergy and Immunology, Duke University, Durham, NC, USA.
- Atrium Health, Charlotte, NC, USA.
| | | | - Monica S Thakar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - M Elizabeth M Younger
- Division of Allergy, Immunology and Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Panida Sriaroon
- Division of Allergy and Immunology, University of South Florida, Tampa, FL, USA
| | - Tamara C Pozos
- Department of Clinical Immunology, Children's Minnesota, Minneapolis, MN, USA
| | - Rebecca H Buckley
- Department of Pediatrics, Division of Allergy and Immunology, Duke University, Durham, NC, USA
| | | | - Diana Vilkama
- Department of Clinical Immunology, Children's Minnesota, Minneapolis, MN, USA
| | - Jennifer Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
9
|
Elgarten C, Wohlschlaeger A, Levy E, Tadley K, Wang L, Atkinson M, Roberson H, Olson T, Bunin N, Heimall J, Fisher B, Grupp S, Freedman J. Quality Improvement Initiative to Improve Time and Adherence to Revaccination after Hematopoietic Cell Transplantation: Implementation of a Revaccination Clinic within the Transplantation Program. Transplant Cell Ther 2023; 29:635.e1-635.e8. [PMID: 37517611 PMCID: PMC10592250 DOI: 10.1016/j.jtct.2023.07.020] [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: 06/15/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
Revaccination after hematopoietic cell transplantation (HCT) is critical to prevent morbidity and mortality from vaccine-preventable illnesses. The global aim of our quality improvement initiative was to enhance timely, correct, and effective revaccination after pediatric HCT. The SMART aim of our project was to decrease median unvaccinated time by 4 months by decreasing the time to vaccine eligibility, time from eligibility to vaccine initiation, and time to completion of the vaccine series. A multidisciplinary group performed a cross-sectional quantitative and qualitative evaluation of revaccination practices at our institution. We identified factors associated with delayed, incorrect, or incomplete revaccination. Several plan-do-study-act interventions were implemented to address these drivers, including revising immune readiness criteria, increasing auditing of primary care administered immunizations, and, importantly, establishing a dedicated revaccination clinic within the HCT clinic at our center. The time to vaccine eligibility decreased from 12.6 months to 10 months (a 20% decrease), and the time to complete the vaccine series decreased from 19.3 months to 15.7 months (a 19% decrease). With a quality improvement initiative, we addressed the many causes of delayed or incomplete revaccination post-HCT and through a team-based approach successfully decreased the time to vaccine start and time to vaccine completion at our center.
Collapse
Affiliation(s)
- Caitlin Elgarten
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Anne Wohlschlaeger
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia
| | - Ellen Levy
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia
| | - Katharine Tadley
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia
| | - Lei Wang
- Data Science and Biostatistics Unit, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Megan Atkinson
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia
| | - Houston Roberson
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia
| | - Timothy Olson
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nancy Bunin
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer Heimall
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Allergy/Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Brian Fisher
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Stephan Grupp
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason Freedman
- Cellular Therapy and Transplantation Section, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Petit A, Neven B, Min V, Mahlaoui N, Moshous D, Castelle M, Allouche M, Stérin A, Visentin S, Saultier P, Boucekine M, Shawket AM, Picard C, Auquier P, Michel G, Fischer A, Barlogis V. Impact of Graft Function on Health Status and Quality of Life in Very Long-Term Survivors Who Received an HSCT for Inborn Errors of Immunity, a Prospective Study of the CEREDIH. Transplant Cell Ther 2023; 29:582.e1-582.e6. [PMID: 37321401 DOI: 10.1016/j.jtct.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/17/2023]
Abstract
The overall survival rate after hematopoietic stem cell transplantation (HSCT) for inborn errors of immunity (IEI) has improved considerably, and its indications have broadened. As a consequence, addressing the issue of long-term health-related quality of life (HRQoL) has become crucial. Our study focuses on the health and HRQoL of post-HSCT survivors. We conducted a multicenter prospective follow-up study enrolling IEI patients who underwent transplantation in childhood before 2009. Self-reported data from the French Childhood Immune Deficiency Long-term Cohort and the 36-item Short Form questionnaires were compiled. One hundred twelve survivors were included with a median duration period from HSCT of 15 years (range 5-37), of whom 55 underwent transplantation for a combined immunodeficiency. We show that in patients evaluated at least 5 years after HSCT, 55% are still affected by a poor or very poor health status. Poor and very poor health status correlated with an abnormal graft function, defined as host or mixed chimerism, abnormal CD3+ count, or diagnosis of chronic graft-versus-host disease (poor health: odds ratio [OR] = 2.6, 95% confidence interval [CI], 1.1-5.9, P = .028; very poor health: OR = 3.6, 95% CI, 1.1-13, P = .049). Poor health was directly linked to a poorer HRQoL. Significant improvements in graft procedures have translated into better survival rates, but we show here that about half of the transplanted patients remain affected by an altered health status with a correlation to both abnormal graft function and impaired HRQoL. Additional studies are needed to confirm the impact of those improvements on long-term health status and HRQoL.
Collapse
Affiliation(s)
- Audrey Petit
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France.
| | - Bénédicte Neven
- Department of Pediatric Hematology-Immunology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; Imagine Institute, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Paris, France; CEREDIH, French National Reference Centre for Primary ImmunoDeficiencies, Paris, France
| | - Victoria Min
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France
| | - Nizar Mahlaoui
- Department of Pediatric Hematology-Immunology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; CEREDIH, French National Reference Centre for Primary ImmunoDeficiencies, Paris, France
| | - Despina Moshous
- Department of Pediatric Hematology-Immunology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; CEREDIH, French National Reference Centre for Primary ImmunoDeficiencies, Paris, France; Imagine Institute, Laboratory of Genome Dynamics in the Immune System, Paris, France
| | - Martin Castelle
- Department of Pediatric Hematology-Immunology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; CEREDIH, French National Reference Centre for Primary ImmunoDeficiencies, Paris, France
| | - Maya Allouche
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France
| | - Arthur Stérin
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France
| | - Sandrine Visentin
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France
| | - Paul Saultier
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France
| | - Mohamed Boucekine
- CEReSS Research Unit EA 3279 and Department of Public Health, Aix Marseille University, School of medicine, La Timone Medical Campus, Marseille, France; Aix Marseille University, Marseille France
| | | | - Capucine Picard
- Department of Pediatric Hematology-Immunology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; CEREDIH, French National Reference Centre for Primary ImmunoDeficiencies, Paris, France; Study Center for Primary Immunodeficiencies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; Imagine Institute, Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Paris, France
| | - Pascal Auquier
- CEReSS Research Unit EA 3279 and Department of Public Health, Aix Marseille University, School of medicine, La Timone Medical Campus, Marseille, France; Aix Marseille University, Marseille France
| | - Gérard Michel
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France; CEReSS Research Unit EA 3279 and Department of Public Health, Aix Marseille University, School of medicine, La Timone Medical Campus, Marseille, France; Aix Marseille University, Marseille France
| | - Alain Fischer
- Department of Pediatric Hematology-Immunology and Rheumatology, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; CEREDIH, French National Reference Centre for Primary ImmunoDeficiencies, Paris, France; Collège de France, Paris, France
| | - Vincent Barlogis
- Department of Pediatric Hematology, Immunology and Oncology, APHM, Hôpital de la Timone Enfants, Marseille, France; CEReSS Research Unit EA 3279 and Department of Public Health, Aix Marseille University, School of medicine, La Timone Medical Campus, Marseille, France; Aix Marseille University, Marseille France
| |
Collapse
|
11
|
Justiz-Vaillant AA, Gopaul D, Akpaka PE, Soodeen S, Arozarena Fundora R. Severe Combined Immunodeficiency-Classification, Microbiology Association and Treatment. Microorganisms 2023; 11:1589. [PMID: 37375091 DOI: 10.3390/microorganisms11061589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Severe combined immunodeficiency (SCID) is a primary inherited immunodeficiency disease that presents before the age of three months and can be fatal. It is usually due to opportunistic infections caused by bacteria, viruses, fungi, and protozoa resulting in a decrease in number and impairment in the function of T and B cells. Autosomal, X-linked, and sporadic forms exist. Evidence of recurrent opportunistic infections and lymphopenia very early in life should prompt immunological investigation and suspicion of this rare disorder. Adequate stem cell transplantation is the treatment of choice. This review aimed to provide a comprehensive approach to the microorganisms associated with severe combined immunodeficiency (SCID) and its management. We describe SCID as a syndrome and summarize the different microorganisms that affect children and how they can be investigated and treated.
Collapse
Affiliation(s)
- Angel A Justiz-Vaillant
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Darren Gopaul
- Department of Internal Medicine, Port of Spain General Hospital, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Patrick Eberechi Akpaka
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, Trinidad and Tobago
| | - Sachin Soodeen
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Rodolfo Arozarena Fundora
- Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Champs Fleurs, Trinidad and Tobago
- Department of Clinical and Surgical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
12
|
Slatter MA, Gennery AR. Advances in the treatment of severe combined immunodeficiency. Clin Immunol 2022; 242:109084. [DOI: 10.1016/j.clim.2022.109084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
|
13
|
Late endocrine effects after hematopoietic stem cell transplantation in children with nonmalignant diseases. Bone Marrow Transplant 2022; 57:1564-1572. [PMID: 35840745 DOI: 10.1038/s41409-022-01755-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 11/08/2022]
Abstract
The number of children undergoing hematopoietic stem cell transplantation (HSCT) for nonmalignant diseases has increased in recent years. Endocrine complications are common after HSCT for malignant diseases, while little is known about long-term prevalence and risk factors in children transplanted for nonmalignant diseases. We retrospectively evaluated gonadal function, near adult height and thyroid function in 197 survivors of pediatric HSCT for hemoglobinopathies (n = 66), inborn errors of immunity/metabolism (n = 74) and bone marrow failure disorders (n = 57); median follow-up was 6.2 years (range 3.0-10.5). Gonadal dysfunction occurred in 55% of (post)pubertal females, was still present at last assessment in 43% and was more common after busulfan- than treosulfan-based conditioning (HR 10.6, CI 2.2-52.7; adjusted for HSCT indication). Gonadal dysfunction occurred in 39% of (post)pubertal males, was still present at last assessment in 32% and was less common in those who were prepubertal compared to (post)pubertal at HSCT (HR 0.11; CI 0.05-0.21). Near adult height was more than 2 SDS below mean parental height in 21% of males and 8% of females. Hypothyroidism occurred in 16% of patients; 4% received thyroxin treatment. In conclusion, endocrine complications, especially gonadal dysfunction, are common after pediatric HSCT for nonmalignant conditions. In females, treosulfan seems less gonadotoxic than busulfan. Careful long-term endocrine follow-up is indicated.
Collapse
|
14
|
Nicholson B, Goodman R, Day J, Worth A, Carpenter B, Sandford K, Morris EC, Burns SO, Ridout D, Titman P, Campbell M. Quality of Life and Social and Psychological Outcomes in Adulthood Following Allogeneic HSCT in Childhood for Inborn Errors of Immunity. J Clin Immunol 2022; 42:1451-1460. [PMID: 35723794 DOI: 10.1007/s10875-022-01286-6] [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: 11/16/2021] [Accepted: 05/01/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Hematopoietic stem cell transplant (HSCT) is well established as a corrective treatment for many inborn errors of immunity (IEIs) presenting in childhood. Due to improved techniques, more transplants are undertaken and patients are living longer. However, long-term complications can significantly affect future health and quality of life. Previous research has focused on short-term medical outcomes and little is known about health or psychosocial outcomes in adulthood. OBJECTIVE This project aimed to ascertain the long-term social and psychological outcomes for adults who underwent HSCT for IEI during childhood. METHODS Adult patients, who had all undergone HSCT for IEI during childhood at two specialist immunology services at least 5 years previously, were invited to participate in the study. Questionnaires and practical tasks assessed their current functioning and circumstances. Information was also gathered from medical notes. Data was compared with population norms and a control group of participant-nominated siblings or friends. RESULTS Eighty-three patients and 46 matched controls participated in the study. Patients reported significantly better physical health-related quality of life than the general population norm, but significantly worse than matched controls. Patient's self-reported physical health status and the perceived impact of their physical health on everyday life were worse than matched controls and patients reported higher levels of anxiety and lower mood than the general population. For those where their IEI diagnosis was not associated with a learning disability, cognitive function was generally within the normal range. CONCLUSIONS Patients who have had a HSCT in childhood report mixed psychosocial outcomes in adulthood. More research is needed to establish screening protocols and targeted interventions to maximize holistic outcomes. CLINICAL IMPLICATIONS Screening for holistic needs and common mental health difficulties should be part of routine follow-up. Information should be provided to patients and families in order to support decision-making regarding progression to transplant and the early identification of any difficulties.
Collapse
Affiliation(s)
- Bethany Nicholson
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Rupert Goodman
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - James Day
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, UK.,UCL Institute of Immunity & Transplantation, London, UK
| | - Austen Worth
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ben Carpenter
- University College London Hospitals NHS Foundation Trust, London, UK
| | | | - Emma C Morris
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, UK.,UCL Institute of Immunity & Transplantation, London, UK.,University College London Hospitals NHS Foundation Trust, London, UK
| | - Siobhan O Burns
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, UK.,UCL Institute of Immunity & Transplantation, London, UK
| | - Deborah Ridout
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Penny Titman
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Mari Campbell
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, UK. .,UCL Institute of Immunity & Transplantation, London, UK.
| |
Collapse
|
15
|
Chitty-Lopez M, Duff C, Vaughn G, Trotter J, Monforte H, Lindsay D, Haddad E, Keller MD, Oshrine BR, Leiding JW. Case Report: Unmanipulated Matched Sibling Donor Hematopoietic Cell Transplantation In TBX1 Congenital Athymia: A Lifesaving Therapeutic Approach When Facing a Systemic Viral Infection. Front Immunol 2022; 12:721917. [PMID: 35095830 PMCID: PMC8794793 DOI: 10.3389/fimmu.2021.721917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Congenital athymia can present with severe T cell lymphopenia (TCL) in the newborn period, which can be detected by decreased T cell receptor excision circles (TRECs) on newborn screening (NBS). The most common thymic stromal defect causing selective TCL is 22q11.2 deletion syndrome (22q11.2DS). T-box transcription factor 1 (TBX1), present on chromosome 22, is responsible for thymic epithelial development. Single variants in TBX1 causing haploinsufficiency cause a clinical syndrome that mimics 22q11.2DS. Definitive therapy for congenital athymia is allogeneic thymic transplantation. However, universal availability of such therapy is limited. We present a patient with early diagnosis of congenital athymia due to TBX1 haploinsufficiency. While evaluating for thymic transplantation, she developed Omenn Syndrome (OS) and life-threatening adenoviremia. Despite treatment with anti-virals and cytotoxic T lymphocytes (CTLs), life threatening adenoviremia persisted. Given the imminent need for rapid establishment of T cell immunity and viral clearance, the patient underwent an unmanipulated matched sibling donor (MSD) hematopoietic cell transplant (HCT), ultimately achieving post-thymic donor-derived engraftment, viral clearance, and immune reconstitution. This case illustrates that because of the slower immune recovery that occurs following thymus transplantation and the restricted availability of thymus transplantation globally, clinicians may consider CTL therapy and HCT to treat congenital athymia patients with severe infections.
Collapse
Affiliation(s)
- Maria Chitty-Lopez
- Division of Pediatric Allergy and Immunology, University of South Florida, Tampa, FL, United States
| | - Carla Duff
- Division of Pediatric Allergy and Immunology, University of South Florida, Tampa, FL, United States
| | - Gretchen Vaughn
- Center for Cell and Gene Therapy for Non-Malignant Conditions, Cancer and Blood Disorders Institute at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, United States
| | - Jessica Trotter
- Division of Pediatric Allergy and Immunology, University of South Florida, Tampa, FL, United States
| | - Hector Monforte
- Department of Pathology, Johns Hopkins All Children’s Hospital, St. Petersburg, FL, United States
- Division of Allergy and Immunology, Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | - David Lindsay
- Division of Allergy and Immunology, Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
- Division of Immuno-Allergy and Rheumatology, The Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Elie Haddad
- Division of Immuno-Allergy and Rheumatology, The Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Division of Allergy and Immunology, Children’s National Hospital, Washington, DC, United States
| | - Michael D. Keller
- Division of Allergy and Immunology, Children’s National Hospital, Washington, DC, United States
| | - Benjamin R. Oshrine
- Center for Cell and Gene Therapy for Non-Malignant Conditions, Cancer and Blood Disorders Institute at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, United States
| | - Jennifer W. Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, United States
- Infectious Diseases and Immunology Division. Arnold Palmer Hospital for Children, Orlando, FL, United States
| |
Collapse
|
16
|
Atay D, Akcay A, Akinci B, Yenigurbuz FD, Ovali E, Ozturk G. Co-transplantation of mesenchymal stromal cell and haploidentical hematopoietic stem cell with TCR αβ depletion in children with primary immunodeficiency syndromes. Pediatr Transplant 2021; 25:e14120. [PMID: 34409718 DOI: 10.1111/petr.14120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Haploidentical HSCT is a good option for children with PIDs lacking an HLA-matched donor. Co-transplantation of MSCs during haploidentical HSCT in patients with PIDs may enhance engraftment, decrease the risk of GVHD, and ensure stable donor chimerism. METHODS Twenty-seven pediatric patients (median age, 1.4 years; range, .3-10.9) with PIDs undergoing thirty haploidentical HSCT with TCR αβ depletion and co-transplantation of MSCs were enrolled to study. Most patients (73.3%) received myeloablative conditioning consisting of treosulfan or busulfan, fludarabine, and thiotepa. The median duration of follow-up was 14.3 months (range, 1-69 months). RESULTS Acute GVHD occurred in 7 patients (grade I-II n = 5, grade III-IV n = 2). Chronic GVHD was observed in only one patient. Twenty-one patients (70.2%) had 100% donor chimerism in all cell lines including T-cell and B-cell lineages. Primary graft failure was observed in 7 patients (25.9%). The cumulative incidences of TRM were 20% at day 100, and 26.7% at one year and five years. Probabilities of OS were 80% at day 100, and 71.9% at 1 year and 5 years. Infants transplanted younger than 6 months of age had the highest 5-year survival rate (85.7%). CONCLUSION We conclude that use of TCR αβ depleted haploidentical transplantation with MSCs may ensure a rapid engraftment rate, low incidence of significant acute and chronic GVHD, and acceptable post-transplantation morbidity, especially in patients diagnosed with SCID and may be considered in children with PIDs. In younger patients (≤6 months), survival is comparable between HLA-matched graft and CD3+ TCRαβ depleted HLA-mismatched graft recipients.
Collapse
Affiliation(s)
- Didem Atay
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University, Istanbul, Turkey
| | - Arzu Akcay
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University, Istanbul, Turkey
| | - Burcu Akinci
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University, Istanbul, Turkey
| | - Fatma Demir Yenigurbuz
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University, Istanbul, Turkey
| | - Ercument Ovali
- Acibadem Labcell Cellular Therapy Laboratory, Istanbul, Turkey
| | - Gulyuz Ozturk
- Department of Pediatric Hematology/Oncology & Bone Marrow Transplantation Unit, Acıbadem University, Istanbul, Turkey
| |
Collapse
|
17
|
Baloh CH, Borkar SA, Chang KF, Yao J, Hershfield MS, Parikh SH, Kohn DB, Goodenow MM, Sleasman JW, Yin L. Normal IgH Repertoire Diversity in an Infant with ADA Deficiency After Gene Therapy. J Clin Immunol 2021; 41:1597-1606. [PMID: 34184208 PMCID: PMC9906566 DOI: 10.1007/s10875-021-01034-2] [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: 11/14/2020] [Accepted: 04/05/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Adenosine deaminase (ADA) deficiency causes severe combined immunodeficiency (SCID) through an accumulation of toxic metabolites within lymphocytes. Recently, ADA deficiency has been successfully treated using lentiviral-transduced autologous CD34+ cells carrying the ADA gene. T and B cell function appears to be fully restored, but in many patients' B cell numbers remain low, and assessments of the immunoglobulin heavy (IgHV) repertoire following gene therapy are lacking. METHODS We performed deep sequencing of IgHV repertoire in peripheral blood lymphocytes from a child following lentivirus-based gene therapy for ADA deficiency and compared to the IgHV repertoire in healthy infants and adults. RESULTS After gene therapy, Ig diversity increased over time as evidenced by V, D, and J gene usage, N-additions, CDR3 length, extent of somatic hypermutation, and Ig class switching. There was the emergence of predominant IgHM, IgHG, and IgHA CDR3 lengths after gene therapy indicating successful oligoclonal expansion in response to antigens. This provides proof of concept for the feasibility and utility of molecular monitoring in following B cell reconstitution following gene therapy for ADA deficiency. CONCLUSION Based on deep sequencing, gene therapy resulted in an IgHV repertoire with molecular diversity similar to healthy infants.
Collapse
Affiliation(s)
- Carolyn H Baloh
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Samiksha A Borkar
- Molecular HIV Host Interaction Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Kai-Fen Chang
- Molecular HIV Host Interaction Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Jiqiang Yao
- Department of Biostatistics and bioinformatics, Moffitt Cancer Center, Tampa, FL
| | - Michael S Hershfield
- Division of Rheumatology and Immunology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Suhag H Parikh
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Donald B Kohn
- Division of Hematology & Oncology, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA.,Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA
| | - Maureen M Goodenow
- Molecular HIV Host Interaction Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - John W Sleasman
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA.
| | - Li Yin
- Molecular HIV Host Interaction Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| |
Collapse
|
18
|
Smith H, Scalchunes C, Cowan MJ, Puck J, Heimall J. Expectations and experience: Parent and patient perspectives regarding treatment for Severe Combined Immunodeficiency (SCID). Clin Immunol 2021; 229:108778. [PMID: 34144198 PMCID: PMC8559521 DOI: 10.1016/j.clim.2021.108778] [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: 02/23/2021] [Revised: 05/13/2021] [Accepted: 06/06/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Infants with SCID are treated with hematopoietic cell transplantation (HCT) or gene therapy (GT). Caregiver perceptions of pre-treatment counseling and understanding of durability of HCT/GT are poorly understood. METHODS A survey was designed and distributed to families of patients with SCID. Topics in the questionnaire included SCID genotype and treatment, family recollections of pre-treatment counseling and present clinical status. RESULTS 151 surveys were analyzed. 132 were treated with HCT, 19 with GT. From counseling received, 37% expected HCT/GT would lead to "cure"; 43% expected HCT/GT would last a lifetime. Of 136 living patients, 59% reported overall good health but 65% reported some persistent health challenges. CONCLUSIONS For some, interpretation of the word "cure" varied, leading to misunderstanding regarding need for continued medical evaluations and additional therapies. Clear communication regarding the importance of lifelong follow-up, no matter the treatment outcome, will help to optimize good health and quality of life.
Collapse
Affiliation(s)
| | | | - Morton J Cowan
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplantation; Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Puck
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplantation; Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, USA.
| |
Collapse
|
19
|
Lugthart G, Jordans CCE, de Pagter APJ, Bresters D, Jol-van der Zijde CM, Bense JE, van Rooij-Kouwenhoven RWG, Sukhai RN, Louwerens M, Dorresteijn EM, Lankester AC. Chronic kidney disease ten years after pediatric allogeneic hematopoietic stem cell transplantation. Kidney Int 2021; 100:906-914. [PMID: 34102218 DOI: 10.1016/j.kint.2021.05.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/15/2021] [Accepted: 05/06/2021] [Indexed: 12/19/2022]
Abstract
Chronic kidney disease (CKD) is an important sequela of hematopoietic stem cell transplantation (HSCT), but data regarding CKD after pediatric HSCT are limited. In this single center cohort study, we evaluated the estimated glomerular filtration rate (eGFR) dynamics, proteinuria and hypertension in the first decade after HSCT and assessed risk factors for CKD in 216 pediatric HSCT survivors, transplanted 2002-2012. The eGFR decreased from a median of 148 to 116 ml/min/1.73 m2 between pre-HSCT to ten years post-HSCT. CKD (KDIGO stages G2 or A2 or more; eGFR under 90 ml/min/1.73m2 and/or albuminuria) occurred in 17% of patients. In multivariate analysis, severe prolonged stage 2 or more acute kidney injury (AKI), with an eGFR under 60ml/min/1.73m2 and duration of 28 days or more, was the main risk factor for CKD (hazard ratio 9.5, 95% confidence interval 3.4-27). Stage 2 or more AKI with an eGFR of 60ml/min/1.73m2 or more and KDIGO stage 2 or more AKI with eGFR under 60ml/min/1.73m2 but recovery within 28 days were not associated with CKD. Furthermore, hematological malignancy as HSCT indication was an independent risk factor for CKD. One third of patients had both CKD criteria, one third had isolated eGFR reduction and one third only had albuminuria. Hypertension occurred in 27% of patients with CKD compared to 4.4% of patients without. Tubular proteinuria was present in 7% of a subgroup of 71 patients with available β2-microglobulinuria. Thus, a significant proportion of pediatric HSCT recipients developed CKD within ten years. Our data stress the importance of structural long-term monitoring of eGFR, urine and blood pressure after HSCT to identify patients with incipient CKD who can benefit from nephroprotective interventions.
Collapse
Affiliation(s)
- Gertjan Lugthart
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands; Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Carlijn C E Jordans
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne P J de Pagter
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Dorine Bresters
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands; Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Cornelia M Jol-van der Zijde
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Joell E Bense
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Roos W G van Rooij-Kouwenhoven
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Ram N Sukhai
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Marloes Louwerens
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Eiske M Dorresteijn
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands; Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Arjan C Lankester
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
20
|
Guilcher GMT, Rivard L, Huang JT, Wright NAM, Anderson L, Eissa H, Pelletier W, Ramachandran S, Schechter T, Shah AJ, Wong K, Chow EJ. Immune function in childhood cancer survivors: a Children's Oncology Group review. THE LANCET. CHILD & ADOLESCENT HEALTH 2021; 5:284-294. [PMID: 33600774 PMCID: PMC8725381 DOI: 10.1016/s2352-4642(20)30312-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 11/20/2022]
Abstract
Childhood cancer and its treatment often impact the haematopoietic and lymphatic systems, with immunological consequences. Immunological assessments are not routinely included in surveillance guidelines for most survivors of childhood cancer, although a robust body of literature describes immunological outcomes, testing recommendations, and revaccination guidelines after allogeneic haematopoietic cell transplantation. Survivorship care providers might not fully consider the impaired recovery of a child's immune system after cancer treatment if the child has not undergone haematopoietic cell transplantation. We did a scoping review to collate the existing literature describing immune function after childhood cancer therapy, including both standard-dose chemotherapy and high-dose chemotherapy with haematopoietic cell rescue. This Review aims to summarise: the principles of immunology and testing of immune function; the body of literature describing immunological outcomes after childhood cancer therapy, with an emphasis on the risk of infection, when is testing indicated, and preventive strategies; and knowledge gaps and opportunities for future research.
Collapse
Affiliation(s)
- Gregory M T Guilcher
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Linda Rivard
- Pediatric Hematology and Oncology, Advocate Children's Hospital, Oak Lawn, IL, USA
| | - Jennifer T Huang
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Nicola A M Wright
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Hesham Eissa
- Department of Pediatrics, University of Colorado, Aurora, CO, USA; Center for Cancer and Blood Disorders, Children's Hospital of Colorado, Aurora, CO, USA
| | - Wendy Pelletier
- Section of Pediatric Oncology and BMT, Alberta Children's Hospital, Calgary, AB, Canada
| | - Shanti Ramachandran
- School of Paediatrics and Child Health, University of Western Australia, Nedland, WA, Australia; Department of Oncology, Haematology, Blood and Marrow Transplantation, Child and Adolescent Health Services, Perth Children's Hospital, Nedland, WA, Australia
| | - Tal Schechter
- Division of Hematology and Oncology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Ami J Shah
- Department of Pediatrics, Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Palo Alto, CA, USA
| | - Ken Wong
- Department of Radiology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA; Children's Hospital Los Angeles, Cancer and Blood Disease Institute, Los Angeles, CA, USA
| | - Eric J Chow
- Fred Hutchinson Cancer Research Center, Clinical Research and Public Health Sciences Divisions, Seattle, WA, USA
| |
Collapse
|
21
|
Béziat V, Meyts I. The "Editors" Take to RAG: Promise of CRISPR/Cas9/rAAV6-Based Gene Therapy for RAG2 Deficiency. J Clin Immunol 2021; 41:849-851. [PMID: 33740170 DOI: 10.1007/s10875-021-01024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France. .,Imagine Institute, Paris University, Paris, France. .,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA.
| | - Isabelle Meyts
- Department of Microbiology, Immunology and Transplantation, Laboratory for Inborn Errors of Immunity, Department of Pediatrics, University Hospitals Leuven and KU Leuven, Leuven, Belgium.
| |
Collapse
|
22
|
Blanco E, Izotova N, Booth C, Thrasher AJ. Immune Reconstitution After Gene Therapy Approaches in Patients With X-Linked Severe Combined Immunodeficiency Disease. Front Immunol 2020; 11:608653. [PMID: 33329605 PMCID: PMC7729079 DOI: 10.3389/fimmu.2020.608653] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
X-linked severe immunodeficiency disease (SCID-X1) is an inherited, rare, and life-threating disease. The genetic origin is a defect in the interleukin 2 receptor γ chain (IL2RG) gene and patients are classically characterized by absence of T and NK cells, as well as presence of partially-functional B cells. Without any treatment the disease is usually lethal during the first year of life. The treatment of choice for these patients is hematopoietic stem cell transplantation, with an excellent survival rate (>90%) if an HLA-matched sibling donor is available. However, when alternative donors are used, the success and survival rates are often lower. Gene therapy has been developed as an alternative treatment initially using γ-retroviral vectors to correct the defective γ chain in the absence of pre-conditioning treatment. The results were highly promising in SCID-X1 infants, showing long-term T-cell recovery and clinical benefit, although NK and B cell recovery was less robust. However, some infants developed T-cell acute lymphoblastic leukemia after the gene therapy, due to vector-mediated insertional mutagenesis. Consequently, considerable efforts have been made to develop safer vectors. The most recent clinical trials using lentiviral vectors together with a low-dose pre-conditioning regimen have demonstrated excellent sustained T cell recovery, but also B and NK cells, in both children and adults. This review provides an overview about the different gene therapy approaches used over the last 20 years to treat SCID-X1 patients, particularly focusing on lymphoid immune reconstitution, as well as the developments that have improved the process and outcomes.
Collapse
Affiliation(s)
- Elena Blanco
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Natalia Izotova
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Claire Booth
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Paediatric Immunology, Great Ormond Street Hospital NHS Trust, London, United Kingdom
| | - Adrian James Thrasher
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Paediatric Immunology, Great Ormond Street Hospital NHS Trust, London, United Kingdom
| |
Collapse
|
23
|
Chitty-Lopez M, Westermann-Clark E, Dawson I, Ujhazi B, Csomos K, Dobbs K, Le K, Yamazaki Y, Sadighi Akha AA, Chellapandian D, Oshrine B, Notarangelo LD, Sunkersett G, Leiding JW, Walter JE. Asymptomatic Infant With Atypical SCID and Novel Hypomorphic RAG Variant Identified by Newborn Screening: A Diagnostic and Treatment Dilemma. Front Immunol 2020; 11:1954. [PMID: 33117328 PMCID: PMC7552884 DOI: 10.3389/fimmu.2020.01954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
The T-cell receptor excision circle (TREC) assay detects T-cell lymphopenia (TCL) in newborns and is especially important to identify severe combined immunodeficiency (SCID). A spectrum of SCID variants and non-SCID conditions that present with TCL are being discovered with increasing frequency by newborn screening (NBS). Recombination-activating gene (RAG) deficiency is one the most common causes of classical and atypical SCID and other conditions with immune dysregulation. We present the case of an asymptomatic male with undetectable TRECs on NBS at 1 week of age. The asymptomatic newborn was found to have severe TCL, but normal B cell quantities and lymphocyte proliferation upon mitogen stimulation. Next generation sequencing revealed compound heterozygous hypomorphic RAG variants, one of which was novel. The moderately decreased recombinase activity of the RAG variants (16 and 40%) resulted in abnormal T and B-cell receptor repertoires, decreased fraction of CD3+ TCRVα7.2+ T cells and an immune phenotype consistent with the RAG hypomorphic variants. The patient underwent successful treatment with hematopoietic stem cell transplantation (HSCT) at 5 months of age. This case illustrates how after identification of a novel RAG variant, in vitro studies are important to confirm the pathogenicity of the variant. This confirmation allows the clinician to expedite definitive treatment with HSCT in an asymptomatic phase, mitigating the risk of serious infectious and non-infectious complications.
Collapse
Affiliation(s)
- Maria Chitty-Lopez
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Emma Westermann-Clark
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Irina Dawson
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Boglarka Ujhazi
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Krisztian Csomos
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Khuong Le
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Yasuhiro Yamazaki
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Amir A Sadighi Akha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Deepak Chellapandian
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Ben Oshrine
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Gauri Sunkersett
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Jennifer W Leiding
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Jolan E Walter
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States.,Division of Pediatric Allergy and Immunology, Massachusetts General Hospital for Children, Boston, MA, United States
| |
Collapse
|
24
|
Kahn JM, Brazauskas R, Tecca HR, Bo-Subait S, Buchbinder D, Battiwala M, Flowers MED, Savani BN, Phelan R, Broglie L, Abraham AA, Keating AK, Daly A, Wirk B, George B, Alter BP, Ustun C, Freytes CO, Beitinjaneh AM, Duncan C, Copelan E, Hildebrandt GC, Murthy HS, Lazarus HM, Auletta JJ, Myers KC, Williams KM, Page KM, Vrooman LM, Norkin M, Byrne M, Diaz MA, Kamani N, Bhatt NS, Rezvani A, Farhadfar N, Mehta PA, Hematti P, Shaw PJ, Kamble RT, Schears R, Olsson RF, Hayashi RJ, Gale RP, Mayo SJ, Chhabra S, Rotz SJ, Badawy SM, Ganguly S, Pavletic S, Nishihori T, Prestidge T, Agrawal V, Hogan WJ, Inamoto Y, Shaw BE, Satwani P. Subsequent neoplasms and late mortality in children undergoing allogeneic transplantation for nonmalignant diseases. Blood Adv 2020; 4:2084-2094. [PMID: 32396620 PMCID: PMC7218429 DOI: 10.1182/bloodadvances.2019000839] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 02/11/2020] [Indexed: 02/07/2023] Open
Abstract
We examined the risk of subsequent neoplasms (SNs) and late mortality in children and adolescents undergoing allogeneic hematopoietic cell transplantation (HCT) for nonmalignant diseases (NMDs). We included 6028 patients (median age, 6 years; interquartile range, 1-11; range, <1 to 20) from the Center for International Blood and Marrow Transplant Research (1995-2012) registry. Standardized mortality ratios (SMRs) in 2-year survivors and standardized incidence ratios (SIRs) were calculated to compare mortality and SN rates with expected rates in the general population. Median follow-up of survivors was 7.8 years. Diagnoses included severe aplastic anemia (SAA; 24%), Fanconi anemia (FA; 10%), other marrow failure (6%), hemoglobinopathy (15%), immunodeficiency (23%), and metabolic/leukodystrophy syndrome (22%). Ten-year survival was 93% (95% confidence interval [95% CI], 92% to 94%; SMR, 4.2; 95% CI, 3.7-4.8). Seventy-one patients developed SNs (1.2%). Incidence was highest in FA (5.5%), SAA (1.1%), and other marrow failure syndromes (1.7%); for other NMDs, incidence was <1%. Hematologic (27%), oropharyngeal (25%), and skin cancers (13%) were most common. Leukemia risk was highest in the first 5 years posttransplantation; oropharyngeal, skin, liver, and thyroid tumors primarily occurred after 5 years. Despite a low number of SNs, patients had an 11-fold increased SN risk (SIR, 11; 95% CI, 8.9-13.9) compared with the general population. We report excellent long-term survival and low SN incidence in an international cohort of children undergoing HCT for NMDs. The risk of SN development was highest in patients with FA and marrow failure syndromes, highlighting the need for long-term posttransplantation surveillance in this population.
Collapse
Affiliation(s)
- Justine M Kahn
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, Columbia University, New York, NY
| | - Ruta Brazauskas
- Center for International Blood and Marrow Transplant Research, Department of Medicine, and
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | - Heather R Tecca
- Center for International Blood and Marrow Transplant Research, Department of Medicine, and
| | - Stephanie Bo-Subait
- Center for International Blood and Marrow Transplant Research, Department of Medicine, and
| | - David Buchbinder
- Division of Pediatric Hematology, Children's Hospital of Orange County, Orange, CA
| | - Minoo Battiwala
- Hematology Branch, Sarah Cannon Bone and Marrow Transplant Program, Nashville, TN
| | - Mary E D Flowers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Rachel Phelan
- Center for International Blood and Marrow Transplant Research, Department of Medicine, and
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Larisa Broglie
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, Columbia University, New York, NY
| | - Allistair A Abraham
- Division of Blood and Marrow Transplantation, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC
| | - Amy K Keating
- Children's Hospital Colorado and University of Colorado, Aurora, CO
| | - Andrew Daly
- Tom Baker Cancer Center, Calgary, AB, Canada
| | - Baldeep Wirk
- Division of Bone Marrow Transplant, Seattle Cancer Alliance, Seattle, WA
| | - Biju George
- Department of Hematology, Christian Medical College, Vellore, India
| | - Blanche P Alter
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Celalettin Ustun
- Division of Hematology/Oncology/Cell Therapy, Rush University, Chicago, IL
| | | | - Amer M Beitinjaneh
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL
| | - Christine Duncan
- Department of Pediatric Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA
| | - Edward Copelan
- Levine Cancer Institute, Atrium Health, Carolinas HealthCare System, Charlotte, NC
| | | | - Hemant S Murthy
- Division of Hematology/Oncology, College of Medicine, University of Florida, Gainesville, FL
| | - Hillard M Lazarus
- Department of Medicine, University Hospitals Case Medical Center and Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Jeffery J Auletta
- Blood and Marrow Transplant Program and Host Defense Program, Division of Hematology/Oncology/Bone Marrow Transplant and Infectious Diseases, Nationwide Children's Hospital, Columbus, OH
| | - Kasiani C Myers
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kirsten M Williams
- Children's Research Institute, Children's National Health Systems, Washington, DC
| | - Kristin M Page
- Division of Pediatric Blood and Marrow Transplantation, Duke University Medical Center, Durham, NC
| | - Lynda M Vrooman
- Department of Pediatric Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA
| | - Maxim Norkin
- Division of Hematology/Oncology, College of Medicine, University of Florida, Gainesville, FL
| | - Michael Byrne
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Miguel Angel Diaz
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Naynesh Kamani
- Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC
| | - Neel S Bhatt
- St Jude Children's Research Hospital, Memphis, TN
| | | | - Nosha Farhadfar
- Division of Hematology/Oncology, College of Medicine, University of Florida, Gainesville, FL
| | - Parinda A Mehta
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Peiman Hematti
- Division of Hematology/Oncology/Bone Marrow Transplantation, Department of Medicine, University of Wisconsin-Madison, Madison, WI
| | - Peter J Shaw
- The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Rammurti T Kamble
- Division of Hematology and Oncology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Raquel Schears
- Division of Hematology/Bone Marrow Transplant, Mayo Clinic, Rochester, MN
| | - Richard F Olsson
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Clinical Research Sormland, Uppsala University, Uppsala, Sweden
| | - Robert J Hayashi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO
| | - Robert Peter Gale
- Hematology Research Center, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Samantha J Mayo
- Lawrence S. Bloomberg Faculty of Nursing, University of Toronto, Toronto, ON, Canada
| | - Saurabh Chhabra
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Seth J Rotz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sherif M Badawy
- Division of Hematology, Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Siddhartha Ganguly
- Division of Hematological Malignancy and Cellular Therapeutics, University of Kansas Health System, Kansas City, KS
| | - Steven Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Tim Prestidge
- Blood and Cancer Center, Starship Children's Hospital, Auckland, New Zealand
| | - Vaibhav Agrawal
- Simon Cancer Center, Indiana University, Indianapolis, IN; and
| | - William J Hogan
- Division of Hematology and Oncology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
- Division of Hematology/Bone Marrow Transplant, Mayo Clinic, Rochester, MN
| | - Yoshihiro Inamoto
- Division of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Bronwen E Shaw
- Center for International Blood and Marrow Transplant Research, Department of Medicine, and
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, Columbia University, New York, NY
| |
Collapse
|
25
|
Kohn DB, Hershfield MS, Puck JM, Aiuti A, Blincoe A, Gaspar HB, Notarangelo LD, Grunebaum E. Consensus approach for the management of severe combined immune deficiency caused by adenosine deaminase deficiency. J Allergy Clin Immunol 2019; 143:852-863. [PMID: 30194989 PMCID: PMC6688493 DOI: 10.1016/j.jaci.2018.08.024] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/07/2018] [Accepted: 08/28/2018] [Indexed: 12/29/2022]
Abstract
Inherited defects in adenosine deaminase (ADA) cause a subtype of severe combined immunodeficiency (SCID) known as severe combined immune deficiency caused by adenosine deaminase defects (ADA-SCID). Most affected infants can receive a diagnosis while still asymptomatic by using an SCID newborn screening test, allowing early initiation of therapy. We review the evidence currently available and propose a consensus management strategy. In addition to treatment of the immune deficiency seen in patients with ADA-SCID, patients should be followed for specific noninfectious respiratory, neurological, and biochemical complications associated with ADA deficiency. All patients should initially receive enzyme replacement therapy (ERT), followed by definitive treatment with either of 2 equal first-line options. If an HLA-matched sibling donor or HLA-matched family donor is available, allogeneic hematopoietic stem cell transplantation (HSCT) should be pursued. The excellent safety and efficacy observed in more than 100 patients with ADA-SCID who received gammaretrovirus- or lentivirus-mediated autologous hematopoietic stem cell gene therapy (HSC-GT) since 2000 now positions HSC-GT as an equal alternative. If HLA-matched sibling donor/HLA-matched family donor HSCT or HSC-GT are not available or have failed, ERT can be continued or reinstituted, and HSCT with alternative donors should be considered. The outcomes of novel HSCT, ERT, and HSC-GT strategies should be evaluated prospectively in "real-life" conditions to further inform these management guidelines.
Collapse
Affiliation(s)
- Donald B Kohn
- Department of Microbiology, Immunology and Molecular Genetics, and the Division of Hematology & Oncology, Department of Pediatrics, David Geffen School of Medicine University of California, Los Angeles, Calif
| | - Michael S Hershfield
- Department of Medicine and Biochemistry, Duke University Medical Center, Durham, NC
| | - Jennifer M Puck
- Department of Pediatrics, Division of Allergy, Immunology, and Bone Marrow Transplantation, University of California San Francisco, San Francisco, Calif
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, and Università Vita Salute San Raffaele, Milan, Italy
| | - Annaliesse Blincoe
- Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - H Bobby Gaspar
- Infection, Immunity, Inflammation, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Eyal Grunebaum
- Division of Immunology and Allergy, and the Department of Pediatrics, Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
Ardura MI. Overview of Infections Complicating Pediatric Hematopoietic Cell Transplantation. Infect Dis Clin North Am 2019; 32:237-252. [PMID: 29406976 DOI: 10.1016/j.idc.2017.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hematopoietic cell transplantations (HCT) are increasingly being performed in children for the treatment of malignant and nonmalignant diseases. Infections remain an important cause of morbidity and mortality after HCT, where the type and timing of infection is influenced by host, transplant, and pathogen-related factors. Herein, an overview of the epidemiology of infections is presented and organized by timing before and after HCT, understanding that infection may occur at any time point until there is successful immune reconstitution.
Collapse
Affiliation(s)
- Monica I Ardura
- Pediatric Infectious Diseases, Host Defense Program, The Ohio State University, Nationwide Children's Hospital, 700 Children's Drive, C5C-J5428, Columbus, OH 43205, USA.
| |
Collapse
|
27
|
Gennery AR, Albert MH, Slatter MA, Lankester A. Hematopoietic Stem Cell Transplantation for Primary Immunodeficiencies. Front Pediatr 2019; 7:445. [PMID: 31737589 PMCID: PMC6831554 DOI: 10.3389/fped.2019.00445] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/14/2019] [Indexed: 01/01/2023] Open
Abstract
The field of primary immunodeficiencies has pioneered many of the advances in haematopoietic stem cell transplantation and cellular therapies over the last 50 years. The first patients to demonstrate sustained benefit and prolonged cure from the primary genetic defect following allogeneic haematopoietic stem cell transplantation were patients with primary immunodeficiencies. Although primary immunodeficiency patients began the modern era of haematopoietic stem cell transplantation, the history is nevertheless short-in answer to the question "what is the long term outcome of patients transplanted for primary immunodeficiencies?" we often have to say that we do not know. We believe that most patients who undergo haematopoietic stem cell transplantation for primary immunodeficiencies should live a normal lifespan with a fully corrected immune system. We are now beginning to understanding long term outcomes, the relationship to the underlying genetic defect, age, and pre-morbid condition of the patient at time of transplantation, stem cell source and donor, and effect of pre-transplant cytoreductive chemotherapy conditioning. The long term consequences of post-transplant complications such as graft vs. host disease, veno-occlusive disease, or immune dysregulation are also being recognized. Additionally, some genetic defects have a systemic distribution, and we are learning the natural history of these defects once the immunodeficiency has been removed.
Collapse
Affiliation(s)
- Andrew R Gennery
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Pediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Michael H Albert
- Pediatric SCT Program, Dr. von Hauner University Children's Hospital, Ludwig-Maximilians Universität, Munich, Germany
| | - Mary A Slatter
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Pediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Arjan Lankester
- Department of Pediatrics, Stem Cell Transplantation Program, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
28
|
Shamriz O, Chandrakasan S. Update on Advances in Hematopoietic Cell Transplantation for Primary Immunodeficiency Disorders. Immunol Allergy Clin North Am 2018; 39:113-128. [PMID: 30466768 DOI: 10.1016/j.iac.2018.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) in patients with primary immunodeficiency disorders (PIDDs) is being increasingly used as a curative option. Understanding the critical components, such as disease's nature and activity and pre-HSCT and post-HSCT patient care is key to a successful outcome. HSCT should be tailored to the underlying PIDD, as different PIDDs, such as severe combined immune deficiency, Treg dysfunction, and phagocytic disorders, have different transplant approaches. Therefore, successful HSCT in patients with PIDDs requires teamwork between immunologists and transplant physicians. In this article, the authors elaborate on various aspects of PIDD-HSCT and highlight recent advances.
Collapse
Affiliation(s)
- Oded Shamriz
- Division of Bone Marrow Transplant, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, 2015 Uppergate Drive, ECC Room 418, Atlanta, GA 30030, USA; Pediatric Division, Hadassah-Hebrew University Medical Center, Ein-Kerem, POB 12000, Jerusalem, Israel 91120
| | - Shanmuganathan Chandrakasan
- Division of Bone Marrow Transplant, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, 2015 Uppergate Drive, ECC Room 418, Atlanta, GA 30030, USA.
| |
Collapse
|
29
|
Haddad E, Logan BR, Griffith LM, Buckley RH, Parrott RE, Prockop SE, Small TN, Chaisson J, Dvorak CC, Murnane M, Kapoor N, Abdel-Azim H, Hanson IC, Martinez C, Bleesing JJH, Chandra S, Smith AR, Cavanaugh ME, Jyonouchi S, Sullivan KE, Burroughs L, Skoda-Smith S, Haight AE, Tumlin AG, Quigg TC, Taylor C, Dávila Saldaña BJ, Keller MD, Seroogy CM, Desantes KB, Petrovic A, Leiding JW, Shyr DC, Decaluwe H, Teira P, Gillio AP, Knutsen AP, Moore TB, Kletzel M, Craddock JA, Aquino V, Davis JH, Yu LC, Cuvelier GDE, Bednarski JJ, Goldman FD, Kang EM, Shereck E, Porteus MH, Connelly JA, Fleisher TA, Malech HL, Shearer WT, Szabolcs P, Thakar MS, Vander Lugt MT, Heimall J, Yin Z, Pulsipher MA, Pai SY, Kohn DB, Puck JM, Cowan MJ, O'Reilly RJ, Notarangelo LD. SCID genotype and 6-month posttransplant CD4 count predict survival and immune recovery. Blood 2018; 132:1737-1749. [PMID: 30154114 PMCID: PMC6202916 DOI: 10.1182/blood-2018-03-840702] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/20/2018] [Indexed: 12/26/2022] Open
Abstract
The Primary Immune Deficiency Treatment Consortium (PIDTC) performed a retrospective analysis of 662 patients with severe combined immunodeficiency (SCID) who received a hematopoietic cell transplantation (HCT) as first-line treatment between 1982 and 2012 in 33 North American institutions. Overall survival was higher after HCT from matched-sibling donors (MSDs). Among recipients of non-MSD HCT, multivariate analysis showed that the SCID genotype strongly influenced survival and immune reconstitution. Overall survival was similar for patients with RAG, IL2RG, or JAK3 defects and was significantly better compared with patients with ADA or DCLRE1C mutations. Patients with RAG or DCLRE1C mutations had poorer immune reconstitution than other genotypes. Although survival did not correlate with the type of conditioning regimen, recipients of reduced-intensity or myeloablative conditioning had a lower incidence of treatment failure and better T- and B-cell reconstitution, but a higher risk for graft-versus-host disease, compared with those receiving no conditioning or immunosuppression only. Infection-free status and younger age at HCT were associated with improved survival. Typical SCID, leaky SCID, and Omenn syndrome had similar outcomes. Landmark analysis identified CD4+ and CD4+CD45RA+ cell counts at 6 and 12 months post-HCT as biomarkers predictive of overall survival and long-term T-cell reconstitution. Our data emphasize the need for patient-tailored treatment strategies depending upon the underlying SCID genotype. The prognostic significance of CD4+ cell counts as early as 6 months after HCT emphasizes the importance of close follow-up of immune reconstitution to identify patients who may need additional intervention to prevent poor long-term outcome.
Collapse
Affiliation(s)
- Elie Haddad
- Pediatric Immunology and Rheumatology Division, CHU Sainte-Justine, University of Montreal, Montreal, QC, Canada
| | - Brent R Logan
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI
| | - Linda M Griffith
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | | | - Susan E Prockop
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Trudy N Small
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jessica Chaisson
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christopher C Dvorak
- Pediatric Allergy, Immunology, and Blood and Marrow Transplant Division, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Megan Murnane
- Pediatric Allergy, Immunology, and Blood and Marrow Transplant Division, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Neena Kapoor
- Blood and Marrow Transplant Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Hisham Abdel-Azim
- Blood and Marrow Transplant Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | - Caridad Martinez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX
| | - Jack J H Bleesing
- Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sharat Chandra
- Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Angela R Smith
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN
| | | | - Soma Jyonouchi
- Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Kathleen E Sullivan
- Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Lauri Burroughs
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Seattle Children's Hospital, Seattle, WA
| | | | - Ann E Haight
- Aflac Cancer and Blood Disorders Center, Emory/Children's Healthcare of Atlanta, Atlanta, GA
| | - Audrey G Tumlin
- Aflac Cancer and Blood Disorders Center, Emory/Children's Healthcare of Atlanta, Atlanta, GA
| | - Troy C Quigg
- Texas Transplant Institute, Methodist Children's Hospital, San Antonio, TX
| | - Candace Taylor
- Texas Transplant Institute, Methodist Children's Hospital, San Antonio, TX
| | - Blachy J Dávila Saldaña
- Division of Blood and Marrow Transplantation, Children's National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Michael D Keller
- Division of Blood and Marrow Transplantation, Children's National Health System, George Washington University School of Medicine and Health Sciences, Washington, DC
| | | | - Kenneth B Desantes
- American Family Children's Hospital, University of Wisconsin, Madison, WI
| | - Aleksandra Petrovic
- Blood and Marrow Transplant, John Hopkins All Children's Hospital, St. Petersburg, FL
| | - Jennifer W Leiding
- Blood and Marrow Transplant, John Hopkins All Children's Hospital, St. Petersburg, FL
- Division of Allergy and Immunology, Department of Pediatrics, University of South Florida, St. Petersburg, FL
| | - David C Shyr
- Department of Pediatrics, Primary Children's Hospital, University of Utah, Salt Lake City, UT
| | - Hélène Decaluwe
- Pediatric Immunology and Rheumatology Division, CHU Sainte-Justine, University of Montreal, Montreal, QC, Canada
| | - Pierre Teira
- Pediatric Immunology and Rheumatology Division, CHU Sainte-Justine, University of Montreal, Montreal, QC, Canada
| | - Alfred P Gillio
- Institute for Pediatric Cancer and Blood Disorders, Hackensack University Medical Center, Hackensack, NJ
| | - Alan P Knutsen
- Pediatric Allergy and Immunology, Saint Louis University, Cardinal Glennon Children's Medical Center, St. Louis, MO
| | - Theodore B Moore
- Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Morris Kletzel
- Division of Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - John A Craddock
- Children's Hospital of Colorado, University of Colorado School of Medicine, Aurora, CO
| | - Victor Aquino
- Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jeffrey H Davis
- Pediatrics, British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Lolie C Yu
- Division of Hematology/Oncology and Hematopoietic Stem Cell Transplantation, The Center for Cancer and Blood Disorders, Children's Hospital/Louisiana State University Medical Center, New Orleans, LA
| | - Geoffrey D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | | | - Frederick D Goldman
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL
| | - Elizabeth M Kang
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Evan Shereck
- Division of Pediatric Hematology/Oncology, Oregon Health and Science University, Portland, OR
| | - Matthew H Porteus
- Pediatric Stem Cell Transplantation, Stanford University, Stanford, CA
| | | | - Thomas A Fleisher
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Harry L Malech
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | | | - Monica S Thakar
- Pediatric Blood and Marrow Transplant Program, Division of Hematology, Oncology, and Blood Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Mark T Vander Lugt
- Pediatric Hematology/Oncology, University of Michigan, Ann Arbor, MI; and
| | - Jennifer Heimall
- Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Ziyan Yin
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI
| | - Michael A Pulsipher
- Blood and Marrow Transplant Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Sung-Yun Pai
- Hematology-Oncology, Boston Children's Hospital, Boston, MA
| | - Donald B Kohn
- Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | - Jennifer M Puck
- Pediatric Allergy, Immunology, and Blood and Marrow Transplant Division, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Morton J Cowan
- Pediatric Allergy, Immunology, and Blood and Marrow Transplant Division, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Richard J O'Reilly
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
30
|
Braunlin E, Steinberger J, DeFor T, Orchard P, Kelly AS. Metabolic Syndrome and Cardiovascular Risk Factors after Hematopoietic Cell Transplantation in Severe Mucopolysaccharidosis Type I (Hurler Syndrome). Biol Blood Marrow Transplant 2018; 24:1289-1293. [DOI: 10.1016/j.bbmt.2018.01.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 01/20/2018] [Indexed: 01/08/2023]
|
31
|
Zinter MS, Holubkov R, Steurer MA, Dvorak CC, Duncan CN, Sapru A, Tamburro RF, McQuillen PS, Pollack MM. Pediatric Hematopoietic Cell Transplant Patients Who Survive Critical Illness Frequently Have Significant but Recoverable Decline in Functional Status. Biol Blood Marrow Transplant 2017; 24:330-336. [PMID: 29128553 DOI: 10.1016/j.bbmt.2017.10.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/31/2017] [Indexed: 12/28/2022]
Abstract
The number of pediatric hematopoietic cell transplant (HCT) patients who survive pediatric intensive care unit (PICU) admission is increasing, yet little is known about their functional morbidity after PICU discharge. We hypothesized that relative to control subjects, pediatric HCT patients who survive PICU admission would have greater rates of new functional morbidity at the time of PICU discharge and only some of these patients would return to their functional baseline by the end of the hospitalization. We performed a retrospective cohort study with secondary data analysis of the Trichotomous Outcomes in Pediatric Critical Care dataset. The pediatric HCT cohort was identified by querying International Classification of Diseases, 9th edition, diagnostic codes. A control group consisted of previously healthy patients matched 4:1 on age, sex, and illness severity, as estimated by the Pediatric Risk of Mortality (PRISM) score. We benchmarked our findings by comparing with a previously healthy group of children with lower respiratory tract infections. Functional impairment was measured by the Functional Status Scale, wherein new morbidity was defined as an increase of ≥3 points relative to the prehospital baseline. Relative to matched control subjects, HCT patients had similar admission PRISM scores (P = .516) but greater PICU mortality (12.9% [11/85] versus 6.2% [21/340], P = .035). However, among those who survived to PICU discharge, HCT patients had similar rates of new morbidity at PICU discharge (14.9% [11/74] versus 17.2% [55/319], P = .622) and similar rates of resolution of new morbidity by hospital discharge (54.5% [6/11] versus 60.0% [33/55], P = .737). Relative to the comparison group with lower respiratory tract infections, HCT patients had both greater admission PRISM scores (P < .001) and greater PICU mortality (12.9% [11/85] versus 1.6% [5/308], P < .001). However, among those who survived to PICU discharge, HCT patients again displayed similar rates of new morbidity at PICU discharge (14.9% [11/74] versus 22.1% [67/303], P = .168) as well as resolution of new morbidity by hospital discharge (54.5% [6/11] versus 71.6% [48/67], P = .299). For pediatric HCT patients PICU survival with new functional morbidity is as prevalent an outcome as PICU mortality. Although pediatric HCT patients have greater PICU mortality than age-, sex-, and PRISM-matched control subjects, they have similar rates of new functional morbidity at PICU discharge and similar resolution of new functional morbidity at hospital discharge. Future interventions focused on improving functional status in pediatric HCT survivors of critical illness are warranted.
Collapse
Affiliation(s)
- Matt S Zinter
- Department of Pediatrics, Division of Critical Care, Benioff Children's Hospital, University of California-San Francisco School of Medicine, San Francisco, California.
| | - Richard Holubkov
- Department of Pediatrics, Division of Critical Care, University of Utah School of Medicine, Salt Lake City, Utah
| | - Martina A Steurer
- Department of Pediatrics, Division of Critical Care, Benioff Children's Hospital, University of California-San Francisco School of Medicine, San Francisco, California
| | - Christopher C Dvorak
- Department of Pediatrics, Division of Allergy, Immunology, and Blood & Marrow Transplantation, Benioff Children's Hospital, University of California-San Francisco School of Medicine, San Francisco, California
| | - Christine N Duncan
- Department of Pediatrics, Division of Stem Cell Transplantation, Boston Children's Hospital, Harvard University School of Medicine, Boston, Massachusetts
| | - Anil Sapru
- Department of Pediatrics, Division of Critical Care, Mattel Children's Hospital, University of California-Los Angeles School of Medicine, San Francisco, California
| | | | - Patrick S McQuillen
- Department of Pediatrics, Division of Critical Care, Benioff Children's Hospital, University of California-San Francisco School of Medicine, San Francisco, California
| | - Murray M Pollack
- Department of Pediatrics, Division of Critical Care, Children's National Medical Center, George Washington University School of Medicine, Washington, DC
| |
Collapse
|
32
|
Heimall J, Cowan MJ. Long term outcomes of severe combined immunodeficiency: therapy implications. Expert Rev Clin Immunol 2017; 13:1029-1040. [PMID: 28918671 PMCID: PMC6019104 DOI: 10.1080/1744666x.2017.1381558] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/15/2017] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Newborn screening has led to a better understanding of the prevalence of Severe Combined Immunodeficiency (SCID) overall and in terms of specific genotypes. Survival has improved following hematopoietic stem cell transplantation (HCT) with the best outcomes seen following use of a matched sibling donor. However, questions remain regarding the optimal alternative donor source, appropriate use of conditioning and the impact of these decisions on immune reconstitution and other late morbidities. Areas covered: The currently available literature reporting late effects after HCT for SCID and use of alternative therapies including enzyme replacement, alternative donors and gene therapy are reviewed. A literature search was performed on Pubmed and ClinicalTrials.gov using key words 'Severe Combined Immunodeficiency', 'SCID', 'hematopoietic stem cell transplant', 'conditioning', 'gene therapy', 'SCID newborn screening', 'TREC' and 'late effects'. Expert commentary: Newborn screening has dramatically changed the clinical presentation of newborn SCID. While the majority of patients with SCID survive HCT, data regarding late effects in these patients is limited and additional studies focused on genotype specific late effects are needed. Prospective studies aimed at minimizing the use of alkylating agents and reducing late effects beyond survival are needed. Gene therapy is being developed and will likely become a more commonly used treatment that will require separate consideration of survival and late effects.
Collapse
Affiliation(s)
- Jennifer Heimall
- Allergy/Immunology Attending Physician, Perelman School of Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Morton J. Cowan
- Allergy Immunology and Blood and Marrow Transplant Division, University of California San Francisco, Benioff Children’s Hospital, San Francisco, CA, USA
| |
Collapse
|