1
|
Goebel GA, Cunha LAO, Minafra FG, Pinto JA. Newborn Screening Followed By Early Treatment is Essential to Improve Survival in SCID. J Clin Immunol 2025; 45:94. [PMID: 40374985 PMCID: PMC12081553 DOI: 10.1007/s10875-025-01887-x] [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: 07/19/2024] [Accepted: 05/01/2025] [Indexed: 05/18/2025]
Abstract
Severe combined immunodeficiency (SCID) is a heterogeneous genetic disease characterized by severe T-cell lymphopenia with a profound impairment of T- and B-cells' function and, in some types, also NK cells. Hematopoietic cell transplantation (HCT) is the only curative treatment currently available in Brazil. Late diagnosis and treatment are the main factors affecting the survival of these children. This study aims to describe the demographic, phenotypic, genotypic, and clinical characteristics of twenty SCID patients (including typical SCID, leaky-SCID, and Omenn Syndrome) followed at a Brazilian referral center and correlate these data with their clinical outcome. The children were analyzed into two groups: patients diagnosed early by newborn screening (NBS) or family history, n = 7, and patients with late diagnosis, by clinical presentation, n = 13. The 2-year overall survival (OS) of the late group was 29.2%, in contrast to the 2-year OS of the early diagnosis group of 71.4% (p = 0.053). However, despite early diagnosis in the first group, timely access to HCT was delayed, with a median of 11 months. This research reveals that survival depends not only on timely diagnosis but also on early definitive treatment. To improve SCID survival rates, developing countries need public policies that allow rapid access to curative treatment for these patients.
Collapse
Affiliation(s)
- Gabriela Assunção Goebel
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena, 110, Belo Horizonte, Minas Gerais, 30.130-100, Brazil.
| | - Luciana Araújo Oliveira Cunha
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena, 110, Belo Horizonte, Minas Gerais, 30.130-100, Brazil
| | - Fernanda Gontijo Minafra
- Department of Pediatrics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jorge Andrade Pinto
- Department of Pediatrics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
2
|
Botaro MH, Maria E Silva J, Jamra SRA, Geraldino SZ, Roxo-Junior P. BCG vaccination in children with severe combined immunodeficiency in a tertiary center: evaluation of complications and risks. J Pediatr (Rio J) 2025; 101:224-230. [PMID: 39681319 PMCID: PMC11889692 DOI: 10.1016/j.jped.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVE To describe the complications and risks associated with BCG (Bacillus Calmette-Guérin) vaccination in patients diagnosed with SCID (Severe Combined Immunodeficiency). METHODS This is a descriptive case series study. Medical charts were retrospectively reviewed for demographics, clinical manifestation, laboratory findings at diagnosis, outcome, and diagnosis of BCG vaccine-associated complications. RESULTS Eleven patients diagnosed with SCID were enrolled. Ten were male. Seven (64 %) were considered probable SCID, while four (36 %) were considered definite SCID (genetically confirmed). The median age at the onset of symptoms was one month; the median age at SCID diagnosis was four months. Respiratory symptoms were the most frequent. Eight patients were vaccinated within seven days of life. Seven (87 %) of these patients experienced BCG vaccine-associated complications (86 % disseminated reactions; 14 % localized reactions). BCG vaccine-associated complications were the first clinical manifestation in 75 % of the vaccinated patients. Less than half of the patients (36 %) underwent hematopoietic stem cell transplantation. The overall death rate was elevated (73 %); the death rate related to BCG vaccination was 25 %. CONCLUSIONS Patients with SCID can present a high rate of BCG vaccine-associated complications, which negatively impact the clinical outcome and mortality. Pediatricians must be aware that BCG vaccine-associated complications can be the first presentation and a warning sign of SCID. Implementing newborn screening for SCID in Brazil may represent a worthy opportunity to impact the health outcomes of affected infants significantly.
Collapse
Affiliation(s)
- Matheus Henrique Botaro
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Pediatria, Divisão de Imunologia e Alergia Pediátrica, Ribeirão Preto, SP, Brazil
| | - Jorgete Maria E Silva
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Pediatria, Divisão de Imunologia e Alergia Pediátrica, Ribeirão Preto, SP, Brazil
| | - Soraya Regina Abu Jamra
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Pediatria, Divisão de Imunologia e Alergia Pediátrica, Ribeirão Preto, SP, Brazil
| | - Stephanie Zago Geraldino
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Pediatria, Divisão de Imunologia e Alergia Pediátrica, Ribeirão Preto, SP, Brazil
| | - Persio Roxo-Junior
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Pediatria, Divisão de Imunologia e Alergia Pediátrica, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
3
|
Nguyen Ngoc Quynh L, Nguyen Thanh B, Luong Thi L, Nguyen Thi Dieu T, Dang Anh D, P. Lee P, Cao Viet T, Tran Minh D. Haploidentical stem cell transplantation with posttransplant cyclophosphamide in children with Wiskott-Aldrich syndrome: a case report. Front Immunol 2025; 16:1495666. [PMID: 39967667 PMCID: PMC11832547 DOI: 10.3389/fimmu.2025.1495666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is a condition characterized by a low platelet count, eczema, and a weakened immune system. Hematopoietic stem cell transplantation (HSCT) is the only curative treatment option. Haploidentical HSCT with posttransplant cyclophosphamide (PTCy) is an emerging approach for children with noncancerous conditions. This case describes a WAS patient who was early diagnosed and successfully treated with haploidentical HSCT. A 3-month-old boy presented with widespread eczema, a low platelet count, and severe infections in infancy. The diagnosis of WAS was quickly confirmed by genetic test. He received immunoglobulin replacement therapy and antimicrobial prophylaxis and underwent HSCT at 4 years 3 months of age. After failed unrelated cord blood HSCT, second rescue haploidentical HSCT had been performed using the patient's mother as the donor, with stem cells collected from peripheral blood. The conditioning regimen included anti-thymocyte globulin, melphalan, and fludarabine. The stem cell dose was 2.63 × 106 CD34+ cells/kg. GVHD prevention included PTCy, mycophenolat mofetil, and tacrolimus. The patient had no significant complications after the transplant. Neutrophil and platelet engraftment occurred promptly. At 32 months post-HSCT, the patient had complete hematological and immune reconstitution, with full donor chimerism and no GVHD. In conclusion, the PTCy approach to haploidentical HSCT was a safe and effective treatment for this WAS patient.
Collapse
Affiliation(s)
| | - Binh Nguyen Thanh
- Pathophysiology and Immunology Department, Hanoi Medical University, Hanoi, Vietnam
| | - Lien Luong Thi
- Pediatric Department, Hanoi Medical University Hospital, Hanoi, Vietnam
| | | | - Duong Dang Anh
- Surgical Intensive Care Unit, Vietnam National Children’s Hospital, Hanoi, Vietnam
| | - Pamela P. Lee
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tung Cao Viet
- Children Heart Center, National Children’s Hospital, Hanoi, Vietnam
| | - Dien Tran Minh
- Surgical Intensive Care Unit, Vietnam National Children’s Hospital, Hanoi, Vietnam
| |
Collapse
|
4
|
Anchoo C, Lev A, Simon AJ, Levy S, Mandola A, Frizinsky S, Somekh I, NaserEddin A, Adwan RSA, Toren A, Golan H, Bielorai B, Hutt D, Adam E, Somech R. Outcome of hematopoietic stem cell transplantation for severe combined immunodeficiency and impact of newborn screening on overall survival: A single referral center study. J Allergy Clin Immunol 2025:S0091-6749(25)00118-6. [PMID: 39900265 DOI: 10.1016/j.jaci.2025.01.029] [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: 11/20/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Hematopoietic stem cell transplantation (HSCT) is a curative treatment for infants with severe combined immunodeficiency (SCID). Different factors determine HSCT success and overall survival (OS). Specifically, prompt diagnosis of SCID, preferably through newborn screening (NBS), is critical. OBJECTIVE We explored variable factors including the impact of NBS that are associated with HSCT outcomes and OS in a multiethnic SCID cohort. METHODS One hundred patients with SCID diagnosed and treated with HSCT at Sheba Medical Center in Israel between 1996 and 2024 were studied. RESULTS We distinguished 3 groups: Palestinians (62%), Israeli Jews (16%), and Israeli non-Jews (22%). The OS rate was 68%, increasing to 84% when excluding mortality in the first month after transplantation. Better outcomes were significantly associated with the use of bone marrow (BM) as a stem cell source (P = .003), the availability of matched related donors (P = .045), and the use of conditioning (P = .0006). As a result of delayed diagnosis, Palestinian patients had more infections, more events of early post-HSCT death, and inferior OS rates compared to other patients. SCID cases identified by NBS demonstrated superior OS (93%) compared to cases identified by clinical presentation (P = .04). Improvement in OS was most significant after the implementation of the NBS program for SCID in Israel (P = .03). CONCLUSION Our study delineates and reinforces specific factors that influence OS after undergoing HSCT for SCID. Importantly, it raises the value of early diagnosis and treatment of affected infants, highlighting the benefit of NBS for SCID in determining the clinical outcome.
Collapse
Affiliation(s)
- Chen Anchoo
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Atar Lev
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Amos J Simon
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Hemato-Immunology Unit, Hematology Lab, Sheba Medical Center, Tel Hashomer, Israel
| | - Shiran Levy
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Amarilla Mandola
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shirly Frizinsky
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ido Somekh
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adeeb NaserEddin
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem (Israel); H. Clinic Specialty Hospital, Ramallah, Palestine
| | - Rabee S A Adwan
- Infectious Diseases Unit, Al-Makassed Hospital, and the Al-Quds University School of Medicine, Jerusalem (Palestine)
| | - Amos Toren
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology, Oncology, and Bone Marrow Transplant, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Hana Golan
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology, Oncology, and Bone Marrow Transplant, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Bella Bielorai
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology, Oncology, and Bone Marrow Transplant, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Daphna Hutt
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Division of Pediatric Hematology, Oncology, and Bone Marrow Transplant, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Etai Adam
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology, Oncology, and Bone Marrow Transplant, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Raz Somech
- Pediatric Department A, Immunology Service and Lab, Jeffrey Modell Foundation Center, Tel Hashomer, Israel; Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
5
|
Karaaslan BG, Demirkale ZH, Turan I, Aydemir S, Meric Z, Taskin Z, Kilinc OC, Burtecene N, Topcu B, Yucel E, Aydogmus C, Cokugras H, Kiykim A. Evaluation of T-cell repertoire by flow cytometric analysis in primary immunodeficiencies with DNA repair defects. Scand J Immunol 2025; 101:e70003. [PMID: 39967281 PMCID: PMC11836546 DOI: 10.1111/sji.70003] [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: 07/05/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 02/20/2025]
Abstract
The group of patients with DNA-repair-defects increases susceptibility to infections due to impaired repertoire diversity. In this context, we aimed to investigate the TCRvβ-repertoire by flow cytometric analysis and its correlation with clinical entities in a group of IEI patients with DNA repair defects. Peripheral lymphocyte subset and TCRvβ-repertoire analyses were performed by flow cytometric analysis. The aim was to explore the changing TCR-Vβ-repertoire that can predict some clinical entities by investigating the repertoire using flow-cytometric-analysis-based TCR-Vβ and its interaction with clinical entities in a group of IEI patients with DNA repair defects. TCR-repertoire of the patients with DNA-repair-defects and healthy controls was analysed with flow-cytometer. The potential of flow-cytometric analysis of the TCR repertoire as a practical and easily accessible clinical prediction method was investigated. Thirty-nine-IEI patients with DNA-repair-defects and 15 age-matched healthy-controls were included in this study. Peripheral lymphocyte subset and TCR-Vβ repertoire analyses were performed by flow cytometry. Compared to the control group, 9 out of 24 clones (37.5%) exhibited a statistically significant reduction, while only 3 clones showed a statistically significant increase (p < 0.05). Preferential use of vβ-genes was associated with some clinical entities. Lower TCR-vβ-9 and TCR-vβ23, higher TCR-vβ7.2 were found in the patients with pneumonia (n = 13) (p = 0.018, p = 0.044 p = 0.032). AT patients with pneumonia had lower TCR-vβ-9 clone than patients without pneumonia (p = 0.008). Skewed proliferation of most TCR-vβ clones was seen DNA-repair-defects, especially AT. In addition, this study showed that preferential use of TCR-vβ genes could be predictive for some clinical entities.
Collapse
Affiliation(s)
- Betul Gemici Karaaslan
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Zeynep Hizli Demirkale
- Istanbul Medical Faculty, Department of Pediatric Immunology and AllergyIstanbul UniversityIstanbulTürkiye
| | - Isilay Turan
- Department of Pediatric Immunology and AllergyBasaksehir Cam and Sakura City HospitalIstanbulTürkiye
| | - Sezin Aydemir
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Zeynep Meric
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Zuleyha Taskin
- Cerrahpasa School of MedicineIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Ozgur Can Kilinc
- Cerrahpasa School of MedicineIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Nihan Burtecene
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Birol Topcu
- Department of BiostatisticsTekirdag Namik Kemal UniversityTekirdagTürkiye
| | - Esra Yucel
- Istanbul Medical Faculty, Department of Pediatric Immunology and AllergyIstanbul UniversityIstanbulTürkiye
| | - Cigdem Aydogmus
- Department of Pediatric Immunology and AllergyBasaksehir Cam and Sakura City HospitalIstanbulTürkiye
| | - Haluk Cokugras
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Ayca Kiykim
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| |
Collapse
|
6
|
De Felipe B, Delgado-Pecellin C, Lopez-Lobato M, Olbrich P, Blanco-Lobo P, Marquez-Fernandez J, Salamanca C, Mendoza B, Castro-Serrano R, Duque C, Moreno-Prieto M, Madruga-Garrido M, Lucena JM, Fernandez RM, Ruiz-Camacho M, Varona A, Neth O. Neonatal Screening for Spinal Muscular Atrophy and Severe T- and B-Cell Lymphopenias in Andalusia: A Prospective Study. Int J Neonatal Screen 2025; 11:11. [PMID: 39982345 PMCID: PMC11843956 DOI: 10.3390/ijns11010011] [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: 10/15/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/22/2025] Open
Abstract
Spinal muscular atrophy (SMA) and severe T- and/or B-cell lymphopenias (STBCL) in the form of severe combined immunodeficiencies (SCID) or X-linked agammaglobulinemia (XLA) are rare but potentially fatal pathologies. In January 2021, we initiated the first pilot study in Spain to evaluate the efficacy of a very early detection technique for SMA and SCID. RT-PCR was performed on prospectively collected dried blood spots (DBSs) from newborns in Western Andalusia (Spain). Internal and external controls (SCID, XLA and SMA) were included. The determination of SMA was relative (positive/negative) and that of TRECs and KRECs was quantitative (copies/punch). A total of 14.035 prospective samples were analysed. All controls were correctly identified while no cases of SMA or SCID/XLA were prospectively identified. DBS analysis of infants with suspected SMA or STBCL that presented to our centre showed pathological values in two cases each for SMA and SCID and one for XLA, all of them being subsequently confirmed genetically. In this prospective pilot study, no infants with SMA or STBCL were detected; however, the technique applied here was shown to be reliable and fast, further supporting the benefits and need to include SMA and SCID in national newborn screening (NBS) programs, as it will allow early supportive and curative therapy.
Collapse
Affiliation(s)
- Beatriz De Felipe
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
| | - Carmen Delgado-Pecellin
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
- Clinical Biochemistry Department, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | | | - Peter Olbrich
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
- Department of Pharmacology, Pediatrics and Radiology, University of Seville, 41008 Seville, Spain
| | - Pilar Blanco-Lobo
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
- Department of Pharmacology, Pediatrics and Radiology, University of Seville, 41008 Seville, Spain
| | | | - Carmen Salamanca
- Neonatology Unit, Hospital Universitario Virgen de Macarena, 41008 Seville, Spain
| | - Beatriz Mendoza
- Neonatology Service, Juan Ramón Jiménez Hospital, 21005 Huelva, Spain (M.R.-C.)
| | - Rocio Castro-Serrano
- Clinical Biochemistry Department, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | - Cristina Duque
- Neonatology Unit, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | - Mariana Moreno-Prieto
- Hospital Viamed Santa Angela de la Cruz, Sevilla and Neurolinkia, 41018 Seville, Spain (M.M.-G.)
| | - Marcos Madruga-Garrido
- Hospital Viamed Santa Angela de la Cruz, Sevilla and Neurolinkia, 41018 Seville, Spain (M.M.-G.)
| | - Jose M. Lucena
- Unidad de Inmunología, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | - Raquel M. Fernandez
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Maria Ruiz-Camacho
- Neonatology Service, Juan Ramón Jiménez Hospital, 21005 Huelva, Spain (M.R.-C.)
| | - Alberto Varona
- Paediatrics Service of Riotinto Hospital, 21660 Huelva, Spain
| | - Olaf Neth
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
| |
Collapse
|
7
|
Arman Bilir Ö, Karaatmaca B, Ok Bozkaya İ, Kanbur ŞM, Kaçar D, Metin A, Özbek NY. Haematopoietic stem cell transplantation in children with inborn errors of immunity: A single centre experience. Scand J Immunol 2025; 101:e13431. [PMID: 39781591 DOI: 10.1111/sji.13431] [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: 08/27/2024] [Revised: 11/18/2024] [Accepted: 12/08/2024] [Indexed: 01/12/2025]
Abstract
This study retrospectively analyzed the outcomes of 61 pediatric patients with inborn errors of immunity (IEI) who underwent hematopoietic stem cell transplantation (HSCT) between 2011 and 2023. Patients were categorized into primary immunodeficiency disorders (PIDD), primary immune dysregulation disorders (PIRD), and congenital defects of phagocyte number or function (CDP). Median ages at diagnosis and HSCT were 9 and 30 months, respectively. With a median follow-up of 51 months, the overall survival (OS) was 70%, with a 100-day post-transplant OS of 80%. Transplant-related mortality (TRM) was 29%, with rates of 42%, 22.5%, and 27% for PIRD, PIDD, and CDP, respectively. This study highlights the importance of early diagnosis and HSCT in improving survival for IEI patients, while also emphasizing the need for continuous improvements in transplant protocols to minimize TRM and enhance quality of life.
Collapse
Affiliation(s)
- Özlem Arman Bilir
- Department of Pediatric Hematology Oncology & Bone Marrow Transplantation Unit, University of Health Sciences Ankara Bilkent City Hospital, Ankara, Turkey
| | - Betül Karaatmaca
- Department of Pediatric Allergy and Immunology, University of Health Sciences Ankara Bilkent City Hospital, Ankara, Turkey
| | - İkbal Ok Bozkaya
- Department of Pediatric Hematology Oncology & Bone Marrow Transplantation Unit, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Şerife Mehtap Kanbur
- Department of Pediatric Hematology Oncology & Bone Marrow Transplantation Unit, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Dilek Kaçar
- Department of Pediatric Hematology Oncology & Bone Marrow Transplantation Unit, University of Health Sciences Ankara Bilkent City Hospital, Ankara, Turkey
| | - Ayşe Metin
- Department of Pediatric Allergy and Immunology, University of Health Sciences Ankara Bilkent City Hospital, Ankara, Turkey
| | - Namık Yaşar Özbek
- Department of Pediatric Hematology Oncology & Bone Marrow Transplantation Unit, University of Health Sciences Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
8
|
Medina D, Castro JO, Castro DE, Beltrán E, Manzi E, Franco AA, Olaya M. Haploidentical hematopoietic stem cell transplantation using post-transplant cyclophosphamide in patients with inborn errors of immunity: Experience in a reference center in Colombia. BIOMEDICA : REVISTA DEL INSTITUTO NACIONAL DE SALUD 2024; 44:118-130. [PMID: 39836841 PMCID: PMC12014220 DOI: 10.7705/biomedica.7560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/26/2024] [Indexed: 01/23/2025]
Abstract
INTRODUCTION Inborn errors of immunity is a diverse group of rare diseases caused by over 400 genetic mutations affecting the immune system and increasing infection susceptibility, autoimmunity, and malignancy. Hematopoietic stem cell transplantation offers a curative option for some inborn errors of immunity, with haploidentical donors providing a viable alternative when identical donors are unavailable. OBJECTIVE To determine survival, usefulness of weekly chimerism monitoring, immune reconstitution, and complications in patients with inborn errors of immunity who underwent haploidentical hematopoietic stem cell transplantation at a reference center in Colombia. MATERIALS AND METHODS We conducted a retrospective and observational study of a case series of pediatric patients who underwent haploidentical hematopoietic stem cell transplantation with post-transplant cyclophosphamide and follow-up with weekly chimerism. Survival analysis was performed using the Kaplan-Meier method. RESULTS Sixteen patients with haploidentical familial donor transplantation were included. The most frequent diagnosis was severe combined immunodeficiency (n=5). Eleven out of seventeen patients received a non-myeloablative conditioning regimen. Twelve out of sixteen patients developed acute graft-versus-host disease. Out of these, 3 corresponded to grades III-IV. Post-transplant infections affected 14 of the subjects, predominating bacterial agents. Median T-cell chimerism was greater than 80% during the follow-up. Reconstitution of B and T lymphocytes was achieved in more than 80%. Overall survival at five years was 81%. Survival at 100 days was 94%. CONCLUSION Haploidentical hematopoietic stem cell transplantation using post-transplant cyclophosphamide is a viable alternative for inborn errors of immunity when an identical donor is unavailable. Serial chimerism monitoring is useful for graft follow-up.
Collapse
Affiliation(s)
- Diego Medina
- Unidad de Trasplante de Progenitores Hematopoyéticos, Servicio de Hemato-Oncología Pediátrica, Departamento Materno Infantil, Fundación Valle del Lili, Cali, ColombiaFundación Valle del LiliUnidad de Trasplante de Progenitores HematopoyéticosServicio de Hemato-Oncología Pediátrica, Departamento Materno InfantilFundación Valle del LiliCaliColombia
- Facultad de Ciencias de la Salud, Universidad ICESI, Cali, ColombiaUniversidad ICESIUniversidad ICESICaliColombia
| | - Jhonier Orlando Castro
- Facultad de Ciencias de la Salud, Universidad ICESI, Cali, ColombiaUniversidad ICESIUniversidad ICESICaliColombia
| | - David Esteban Castro
- Facultad de Ciencias de la Salud, Universidad ICESI, Cali, ColombiaUniversidad ICESIUniversidad ICESICaliColombia
| | - Estefanía Beltrán
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, Cali, ColombiaFundación Valle del LiliFundación Valle del LiliCaliColombia
| | - Eliana Manzi
- Facultad de Ciencias de la Salud, Universidad ICESI, Cali, ColombiaUniversidad ICESIUniversidad ICESICaliColombia
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, Cali, ColombiaFundación Valle del LiliFundación Valle del LiliCaliColombia
| | - Alexis Antonio Franco
- Unidad de Trasplante de Progenitores Hematopoyéticos, Servicio de Hemato-Oncología Pediátrica, Departamento Materno Infantil, Fundación Valle del Lili, Cali, ColombiaFundación Valle del LiliUnidad de Trasplante de Progenitores HematopoyéticosServicio de Hemato-Oncología Pediátrica, Departamento Materno InfantilFundación Valle del LiliCaliColombia
- Facultad de Ciencias de la Salud, Universidad ICESI, Cali, ColombiaUniversidad ICESIUniversidad ICESICaliColombia
| | - Manuela Olaya
- Facultad de Ciencias de la Salud, Universidad ICESI, Cali, ColombiaUniversidad ICESIUniversidad ICESICaliColombia
- Servicio de Alergología e Inmunología Pediátrica, Departamento Materno-Infantil, Fundación Valle del Lili, Cali, ColombiaFundación Valle del LiliFundación Valle del LiliCaliColombia
| |
Collapse
|
9
|
Lum SH, Albert MH, Gilbert P, Sirait T, Algeri M, Muratori R, Fournier B, Laberko A, Karakukcu M, Unal E, Ayas M, Yadav SP, Fisgin T, Elfeky R, Fernandes J, Faraci M, Cole T, Schulz A, Meisel R, Zecca M, Ifversen M, Biffi A, Diana JS, Vallée T, Giardino S, Ersoy GZ, Moshous D, Gennery AR, Balashov D, Bonfim C, Locatelli F, Lankester A, Neven B, Slatter M. Outcomes of HLA-mismatched HSCT with TCRαβ/CD19 depletion or post-HSCT cyclophosphamide for inborn errors of immunity. Blood 2024; 144:565-580. [PMID: 38669631 DOI: 10.1182/blood.2024024038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/27/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
ABSTRACT HLA-mismatched transplants with either in vitro depletion of CD3+ T-cell receptor (TCR)αβ/CD19 (TCRαβ) cells or in vivo T-cell depletion using posttransplant cyclophosphamide (PTCY) have been increasingly used for patients with inborn errors of immunity (IEIs). We performed a retrospective multicenter study via the EBMT registry on 306 children with IEIs undergoing their first transplant between 2010 and 2019 from an HLA-mismatched donor using TCRαβ (n = 167) or PTCY (n = 139). The median age for hematopoietic stem cell transplantation (HSCT) was 1.2 years (range, 0.03-19.6 years). The 3-year overall survival (OS) was 78% (95% confidence interval (CI), 71-84) after TCRαβ and 66% (57-74) after PTCY (P = .013). Pre-HSCT morbidity score (hazard ratio [HR], 2.27; 1.07-4.80, P = .032) and non-busulfan/treosulfan conditioning (HR, 3.12; 1.98-4.92, P < .001) were the only independent predictors of unfavorable OS. The 3-year event-free survival (EFS) was 58% (50%-66%) after TCRαβ and 57% (48%-66%) after PTCY (P = .804). The cumulative incidence of severe acute graft-versus-host disease (GvHD) was higher after PTCY (15%, 9%-21%) than TCRαβ (6%, 2%-9%, P = .007), with no difference in chronic GvHD (PTCY, 11%, 6%-17%; TCRαβ, 7%, 3%-11%, P = .173). The 3-year GvHD-free EFS was 53% (44%-61%) after TCRαβ and 41% (32%-50%) after PTCY (P = .080). PTCY had significantly higher rates of veno-occlusive disease (14.4% vs TCRαβ 4.9%, P = .009), acute kidney injury (12.7% vs 4.6%, P = .032), and pulmonary complications (38.2% vs 24.1%, P = .017). Adenoviremia (18.3% vs PTCY 8.0%, P = .015), primary graft failure (10% vs 5%, P = .048), and second HSCT (17.4% vs 7.9%, P = .023) were significantly higher in TCRαβ. In conclusion, this study demonstrates that both approaches are suitable options in patients with IEIs, although they are characterized by different advantages and outcomes.
Collapse
Affiliation(s)
- Su Han Lum
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Michael H Albert
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Mattia Algeri
- Department of Paediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Magna Graecia University, Catanzaro, Italy
| | - Rafaella Muratori
- Pediatric Hematology and Transplantation Unit, Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil
| | - Benjamin Fournier
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Alexandra Laberko
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Musa Karakukcu
- Erciyes University, KANKA Pediatric Hematology/Oncology and BMT Hospital, Kayseri, Turkey
| | - Elrem Unal
- Hasan KALYONCU University and Medicalpoint Hospital, Gaziantep, Turkey
| | - Mouhab Ayas
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Tunc Fisgin
- Pediatric Hematology/Oncology and BMT Unit, Altinbas University Faculty of Medicine Medical Park Bahcelievler Hospital, Istanbul, Turkey
| | - Reem Elfeky
- Department of Paediatric Immunology, Great Ormand Street Children's Hospital, London, United Kingdom
| | - Juliana Fernandes
- Stem Cell Transplantation Unit, ITACI-Instituto da Criança-Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
- Hematology and Stem Cell Transplantation Unit, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Hematology and Stem Cell Transplantation Unit, Hospital 9 de Julho, São Paulo, Brazil
| | | | - Theresa Cole
- Department of Allergy and Immunology, Royal Children's Hospital, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Marco Zecca
- Paediatric Haematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marianne Ifversen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Alessandra Biffi
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Women and Child Health Department, University of Padua and Padua University Hospital, Padua, Italy
| | - Jean-Sebastien Diana
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Tanja Vallée
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Gizem Zengin Ersoy
- Pediatric Hematology/Oncology and BMT Unit, Altinbas University Faculty of Medicine Medical Park Bahcelievler Hospital, Istanbul, Turkey
| | - Despina Moshous
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Andrew R Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Dmitry Balashov
- Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Carmem Bonfim
- Instituto de Pesquisa Pele Pequeno Príncipe/Faculdades Pequeno Príncipe, Pediatric Blood and Marrow Transplantation Service Hospital Pequeno Príncipe, Curitiba, Brazil
| | - Franco Locatelli
- Department of Paediatric Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Arjan Lankester
- Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Bénédicte Neven
- Pediatric Immunology, Hematology and Rheumatology Department, Necker-Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mary Slatter
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Paediatric Stem Cell Transplantation Unit, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
10
|
Marszołek A, Leśniak M, Sekunda A, Siwek A, Skiba Z, Lejman M, Zawitkowska J. Haploidentical HSCT in the Treatment of Pediatric Hematological Disorders. Int J Mol Sci 2024; 25:6380. [PMID: 38928087 PMCID: PMC11204214 DOI: 10.3390/ijms25126380] [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: 04/26/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation has become a treatment option for otherwise non-curative conditions, both malignant and benign, affecting children and adults. Nevertheless, the latest research has been focusing extensively on transplantation from related and unrelated haploidentical donors, suitable for patients requiring emergent hematopoietic stem cell transplantation (HSCT) in the absence of an HLA-matched donor. Haploidentical HSCT (haplo-HSCT) can be an effective treatment for non-malignant pediatric disorders, such as primary immunodeficiencies or hemoglobinopathies, by enabling a much quicker selection of the appropriate donor for virtually all patients, low incidence of graft-versus-host disease (GVHD), and transplant-related mortality (TRM). Moreover, the outcomes of haplo-HSCT among children with hematological malignancies have improved radically. The most demanding tasks for clinicians are minimizing T-cell-mediated alloreactivity as well as early GVHD prevention. As a result, several T-cell depletion approaches, such as ex vivo T-cell depletion (TCD), and T-cell replete approaches, such as a combination of anti-thymocyte globulin (ATG), post-transplantation cyclophosphamide (PTCy), cyclosporine/tacrolimus, mycophenolate mofetil, or methotrexate, have been taken up. As more research is needed to establish the most beneficial form of therapy, haplo-HSCT is currently considered an alternative donor strategy for pediatric and adult patients with complications like viral and bacterial infections, invasive fungal disease, and GVHD.
Collapse
Affiliation(s)
- Anna Marszołek
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.M.); (M.L.); (A.S.); (A.S.); (Z.S.)
| | - Maria Leśniak
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.M.); (M.L.); (A.S.); (A.S.); (Z.S.)
| | - Anna Sekunda
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.M.); (M.L.); (A.S.); (A.S.); (Z.S.)
| | - Aleksander Siwek
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.M.); (M.L.); (A.S.); (A.S.); (Z.S.)
| | - Zuzanna Skiba
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.M.); (M.L.); (A.S.); (A.S.); (Z.S.)
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
11
|
Chong-Neto HJ, Radwan N, Condino-Neto A, Rosário Filho NA, Ortega-Martell JA, El-Sayed ZA. Newborn screening for inborn errors of immunity: The status worldwide. World Allergy Organ J 2024; 17:100920. [PMID: 38974948 PMCID: PMC11225001 DOI: 10.1016/j.waojou.2024.100920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 07/09/2024] Open
Abstract
Background Newborn screening (NBS) for the early detection of inborn errors of immunity (IEI) has been implemented in a few countries. The objective of this study was to verify the situation and define obstacles to the implementation of NBS worldwide. Methods A questionnaire was developed by the Inborn Errors of Immunity Committee of the World Allergy Organization (WAO) with 17 questions regarding NBS for IEI in the physician's workplace, NBS test type, problems hindering NBS implementation, reimbursement for IEI therapy, presence of a national IEI registry, referral centers, molecular diagnosis, hematopoietic stem cell transplantation centers, gene therapy, and immunoglobulin replacement therapy. The survey was sent by email once a week to doctors and others associated with WAO and the main immunology societies worldwide as a Google Form™ to be completed during September and October 2021. Results Two hundred twenty-nine questionnaires were completed, of which 216 (94.3%) were completed by physicians. One hundred seventy-six (76.8%) physicians were both allergists and immunologists. The agreement between allergists/immunologists and non-allergists/non-immunologists for the question "Is there NBS for IEI in the country you work in?" was good (κ = 0,64: 95% CI 0.55-0.69). Ninety-eight (42.8%) participants were from Latin America, 35 (15.3%) from North America, 29 (12.6%) from Europe, 18 (7.9%) from Africa, 44 (19.2%) from Asia, and 5 (2.2%) from Oceania. More than half the participants (n = 124, 54.2%) regularly treated patients with IEI, followed by occasional treatment (n = 77, 33.6%), or never (n = 28, 12.2%). Of the respondents, 14.8% reported that their countries performed NBS for IEI, whereas 42.2% reported their countries did not. T-cell receptor excision circles was the most widely used technique in some countries, with 75 (59.9%) for the diagnosis of NBS for IEI, followed by combined use with kappa deleting-recombination excision circles. Only 13 participants (10.3%) underwent neonatal exon screening in their respective countries. Financial and technical issues were among the major obstacles to the implementation of NBS for IEI. Conclusions This pilot study showed that few countries have implemented NBS for IEI, despite the presence of immunology referral centers and the availability of hematopoietic stem cell transplantation and intravenous immunoglobulin replacement therapy. The findings highlight the difficulties, mainly financial and technical, hindering wide application of NBS. Sharing experiences, technologies, and resources at the international level can help overcome these difficulties.
Collapse
Affiliation(s)
- Herberto José Chong-Neto
- Division of Allergy and Immunology, Complexo Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil
| | - Nesrine Radwan
- Pediatric Allergy, Immunology and Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | | | - Nelson Augusto Rosário Filho
- Division of Allergy and Immunology, Complexo Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil
| | | | - Zeinab A. El-Sayed
- Pediatric Allergy, Immunology and Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
Alzyoud R, Alsuweiti M, Maaitah H, Aladaileh B, Noubani M, Nsour H. Inborn Errors of Immunity in Jordan: First Report from a Tertiary Referral Center. J Clin Immunol 2024; 44:101. [PMID: 38630413 DOI: 10.1007/s10875-024-01709-6] [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: 12/02/2023] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Inborn errors of immunity (IEI) are a heterogeneous group of diseases with variable clinical phenotypes. This study was conducted to describe the epidemiology, clinical presentations, treatment, and outcome of IEI in Jordanian children. METHODS A retrospective data analysis was conducted for children under 15 years diagnosed with IEI from the pediatric Allergy, Immunology, and Rheumatology Division-based registry at Queen Rania Children's Hospital, Amman, Jordan, between 2010 and 2022. RESULTS A total of 467 patients, 263 (56.3%) males and 204 (43.7%) females, were diagnosed with IEI. The mean age at symptom onset was 18 months (1 week to 144 months), a positive family history of IEI was reported in 43.5%, and the consanguinity rate was 47.9%. The most common IEI category was immunodeficiencies affecting cellular and humoral immunity at 33.2%, followed by predominantly antibody deficiencies at 16.9%. The overall median diagnostic delay (range) was 6 (0-135) months; patients with a positive family history of IEI had a statistically significant shorter diagnostic delay. Pulmonary and gastrointestinal clinical features were the most common at 55.2% and 45.6%, respectively. The overall mortality was 33.2%; the highest rate was reported in severe combined immunodeficiency at 56.2%. CONCLUSIONS The high minimal estimated IEI prevalence at 16.2/100,000 Jordanian children compared to the regional and worldwide data, with the diversities in clinical presentation and distribution of IEI categories in our cohort point to unique features of IEI in Jordanian children, call for national registry establishment, regional and international collaborative networks.
Collapse
Affiliation(s)
- Raed Alzyoud
- Pediatric Allergy, Immunology, and Rheumatology Division, Queen Rania Children's Hospital, Jordanian Royal Medical Service, Amman, Jordan.
| | - Motasem Alsuweiti
- Pediatric Allergy, Immunology, and Rheumatology Division, Queen Rania Children's Hospital, Jordanian Royal Medical Service, Amman, Jordan
| | - Heba Maaitah
- Pediatric Allergy, Immunology, and Rheumatology Division, Queen Rania Children's Hospital, Jordanian Royal Medical Service, Amman, Jordan
| | - Boshra Aladaileh
- Pediatric Allergy, Immunology, and Rheumatology Division, Queen Rania Children's Hospital, Jordanian Royal Medical Service, Amman, Jordan
| | - Mohammed Noubani
- Pediatric Allergy, Immunology, and Rheumatology Division, Queen Rania Children's Hospital, Jordanian Royal Medical Service, Amman, Jordan
| | - Hamazh Nsour
- Pediatric Allergy, Immunology, and Rheumatology Division, Queen Rania Children's Hospital, Jordanian Royal Medical Service, Amman, Jordan
| |
Collapse
|
13
|
Tsilifis C, Spegarova JS, Good R, Griffin H, Engelhardt KR, Graham S, Hughes S, Arkwright PD, Hambleton S, Gennery AR. Omenn Syndrome in Two Infants with Different Hypomorphic Variants in Janus Kinase 3. J Clin Immunol 2024; 44:98. [PMID: 38598033 PMCID: PMC11006754 DOI: 10.1007/s10875-024-01699-5] [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: 02/11/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024]
Abstract
Biallelic null or hypomorphic variants in JAK3 cause SCID and less frequently Omenn syndrome. We investigated homozygous hypomorphic JAK3 mutations in two patients, and expression and function of a novel JAK3R431P variant in Omenn syndrome. Immunophenotyping of PBMC from the patient with the novel JAK3R431P variant was undertaken, by flow cytometry and Phosflow after stimulation with IL-2, IL-7, and IL-15. JAK3 expression was investigated by Western blotting. We report two patients with homozygous hypomorphic JAK3 variants and clinical features of Omenn syndrome. One patient had a previously described JAK3R775H variant, and the second had a novel JAK3R431P variant. One patient with a novel JAK3R431P variant had normal expression of JAK3 in immortalised EBV-LCL cells but reduced phosphorylation of STAT5 after stimulation with IL-2, IL-7, and IL-15 consistent with impaired kinase activity. These results suggest the JAK3R431P variant to be hypomorphic. Both patients are alive and well after allogeneic haematopoietic stem cell transplantation. They have full donor chimerism, restitution of thymopoiesis and development of appropriate antibody responses following vaccination. We expand the phenotype of hypomorphic JAK3 deficiency and demonstrate the importance of functional testing of novel variants in disease-causing genes.
Collapse
Affiliation(s)
- Christo Tsilifis
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Victoria Wing, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Ross Good
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Helen Griffin
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Karin R Engelhardt
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Graham
- Faculty of Medical Sciences, Medical School, Newcastle University, Newcastle Upon Tyne, UK
| | - Stephen Hughes
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Sophie Hambleton
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Victoria Wing, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Andrew R Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital, Victoria Wing, Royal Victoria Infirmary, Newcastle Upon Tyne, NE1 4LP, UK.
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
14
|
Goebel GA, de Assis CS, Cunha LAO, Minafra FG, Pinto JA. Survival After Hematopoietic Stem Cell Transplantation in Severe Combined Immunodeficiency (SCID): A Worldwide Review of the Prognostic Variables. Clin Rev Allergy Immunol 2024; 66:192-209. [PMID: 38689103 DOI: 10.1007/s12016-024-08993-5] [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] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
This study aims to perform an extensive review of the literature that evaluates various factors that affect the survival rates of patients with severe combined immunodeficiency (SCID) after hematopoietic stem cell transplantation (HSCT) in developed and developing countries. An extensive search of the literature was made in four different databases (PubMed, Embase, Scopus, and Web of Science). The search was carried out in December 2022 and updated in July 2023, and the terms such as "hematopoietic stem cell transplantation," "bone marrow transplant," "mortality," "opportunistic infections," and "survival" associated with "severe combined immunodeficiency" were sought based on the MeSH terms. The language of the articles was "English," and only articles published from 2000 onwards were selected. Twenty-three articles fulfilled the inclusion criteria for review and data extraction. The data collected corroborates that early HSCT, but above all, HSCT in patients without active infections, is related to better overall survival. The universal implementation of newborn screening for SCID will be a fundamental pillar for enabling most transplants to be carried out in this "ideal scenario" at an early age and free from infection. HSCT with an HLA-identical sibling donor is also associated with better survival rates, but this is the least common scenario. For this reason, transplantation with matched unrelated donors (MUD) and mismatched related donors (mMRD/Haploidentical) appear as alternatives. The results obtained with MUD are improving and show survival rates similar to those of MSD, as well as they do not require manipulation of the graft with expensive technologies. However, they still have high rates of complications after HSCT. Transplants with mMRD/Haplo are performed just in a few large centers because of the high costs of the technology to perform CD3/CD19 depletion and TCRαβ/CD19 depletion or CD34 + selection techniques in vitro. The new possibility of in vivo T cell depletion using post-transplant cyclophosphamide could also be a viable alternative for performing mMRD transplants in centers that do not have this technology, especially in developing countries.
Collapse
Affiliation(s)
- Gabriela Assunção Goebel
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena, 110, Belo Horizonte, Minas Gerais, Brazil.
| | - Cíntia Silva de Assis
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena, 110, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana Araújo Oliveira Cunha
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena, 110, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Gontijo Minafra
- Department of Pediatrics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jorge Andrade Pinto
- Department of Pediatrics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
15
|
Keller MD, Schattgen SA, Chandrakasan S, Allen EK, Jensen-Wachspress MA, Lazarski CA, Qayed M, Lang H, Hanley PJ, Tanna J, Pai SY, Parikh S, Berger SI, Gottschalk S, Pulsipher MA, Thomas PG, Bollard CM. Secondary bone marrow graft loss after third-party virus-specific T cell infusion: Case report of a rare complication. Nat Commun 2024; 15:2749. [PMID: 38553461 PMCID: PMC10980733 DOI: 10.1038/s41467-024-47056-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/20/2023] [Indexed: 04/02/2024] Open
Abstract
Virus-specific T cells (VST) from partially-HLA matched donors have been effective for treatment of refractory viral infections in immunocompromised patients in prior studies with a good safety profile, but rare adverse events have been described. Here we describe a unique and severe adverse event of VST therapy in an infant with severe combined immunodeficiency, who receives, as part of a clinical trial (NCT03475212), third party VSTs for treating cytomegalovirus viremia following bone marrow transplantation. At one-month post-VST infusion, rejection of graft and reversal of chimerism is observed, as is an expansion of T cells exclusively from the VST donor. Single-cell gene expression and T cell receptor profiling demonstrate a narrow repertoire of predominantly activated CD4+ T cells in the recipient at the time of rejection, with the repertoire overlapping more with that of peripheral blood from VST donor than the infused VST product. This case thus demonstrates a rare but serious side effect of VST therapy.
Collapse
Affiliation(s)
- Michael D Keller
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Division of Allergy and Immunology, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University, Washington, DC, USA
| | - Stefan A Schattgen
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - E Kaitlynn Allen
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Christopher A Lazarski
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Hospital of Atlanta, Atlanta, GA, USA
| | - Haili Lang
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Jay Tanna
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Sung-Yun Pai
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Suhag Parikh
- Aflac Cancer and Blood Disorders Center, Children's Hospital of Atlanta, Atlanta, GA, USA
| | - Seth I Berger
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Stephen Gottschalk
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael A Pulsipher
- Division of Pediatric Hematology/Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.
- GW Cancer Center, George Washington University, Washington, DC, USA.
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA.
| |
Collapse
|
16
|
Arnold DE, Pai SY. Progress in the field of hematopoietic stem cell-based therapies for inborn errors of immunity. Curr Opin Pediatr 2023; 35:663-670. [PMID: 37732933 PMCID: PMC10872717 DOI: 10.1097/mop.0000000000001292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
PURPOSE OF REVIEW Hematopoietic stem cell-based therapies, including allogeneic hematopoietic cell transplantation (HCT) and autologous gene therapy (GT), have been used as curative therapy for many inborn errors of immunity (IEI). As the number of genetically defined IEI and the use of HCT and GT increase, valuable data on outcomes and approaches for specific disorders are available. We review recent progress in HCT and GT for IEI in this article. RECENT FINDINGS Novel approaches to prevention of allogeneic complications and experience in adolescents and young adults have expanded the use of HCT. Universal newborn screening for severe combined immunodeficiency (SCID) has led to improved outcome after HCT. Analysis of outcomes of HCT and GT for SCID, Wiskott-Aldrich syndrome (WAS) and chronic granulomatous disease (CGD) reveal risk factors for survival, the impact of specific conditioning regimens, and vector- or disease-specific impacts on efficacy and safety. Preclinical studies of GT and gene editing show potential for translation to the clinic. SUMMARY Emerging data on outcome after HCT for specific IEI support early evaluation and treatment, before development of co-morbidities. Data in large cooperative retrospective databases continues to yield valuable insights clinicians can use in patient selection and choice of therapy.
Collapse
Affiliation(s)
- Danielle E. Arnold
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Sung-Yun Pai
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
17
|
Ruttens D, Philippet P, Bucciol G, Meyts I. Haploidentical Stem Cell Transplantation with Post-transplantation Cyclophosphamide in High-Risk Chronic Granulomatous Disease Patient with Invasive Mucormycosis. J Clin Immunol 2023; 43:1758-1765. [PMID: 37578614 DOI: 10.1007/s10875-023-01567-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Affiliation(s)
- D Ruttens
- Department of Pediatrics, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - P Philippet
- Division of Pediatric Immuno-hematology and Oncology, CHC MontLégia, Liège, Belgium
| | - G Bucciol
- Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Childhood Immunology, Department of Pediatrics, University Hospitals Leuven, ERN-RITA Core Member, Herestraat 49, 3000, Leuven, Belgium
| | - I Meyts
- Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
- Childhood Immunology, Department of Pediatrics, University Hospitals Leuven, ERN-RITA Core Member, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
18
|
Allen D, Knop O, Itkowitz B, Kalter N, Rosenberg M, Iancu O, Beider K, Lee YN, Nagler A, Somech R, Hendel A. CRISPR-Cas9 engineering of the RAG2 locus via complete coding sequence replacement for therapeutic applications. Nat Commun 2023; 14:6771. [PMID: 37891182 PMCID: PMC10611791 DOI: 10.1038/s41467-023-42036-5] [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: 02/08/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
RAG2-SCID is a primary immunodeficiency caused by mutations in Recombination-activating gene 2 (RAG2), a gene intimately involved in the process of lymphocyte maturation and function. ex-vivo manipulation of a patient's own hematopoietic stem and progenitor cells (HSPCs) using CRISPR-Cas9/rAAV6 gene editing could provide a therapeutic alternative to the only current treatment, allogeneic hematopoietic stem cell transplantation (HSCT). Here we show an innovative RAG2 correction strategy that replaces the entire endogenous coding sequence (CDS) for the purpose of preserving the critical endogenous spatiotemporal gene regulation and locus architecture. Expression of the corrective transgene leads to successful development into CD3+TCRαβ+ and CD3+TCRγδ+ T cells and promotes the establishment of highly diverse TRB and TRG repertoires in an in-vitro T-cell differentiation platform. Thus, our proof-of-concept study holds promise for safer gene therapy techniques of tightly regulated genes.
Collapse
Affiliation(s)
- Daniel Allen
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Orli Knop
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Bryan Itkowitz
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Nechama Kalter
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Michael Rosenberg
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Ortal Iancu
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Katia Beider
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, 5266202, Israel
| | - Yu Nee Lee
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, 5266202, Israel
| | - Arnon Nagler
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, 5266202, Israel
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, 5266202, Israel
| | - Ayal Hendel
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel.
| |
Collapse
|
19
|
Wadbudhe AM, Meshram RJ, Tidke SC. Severe Combined Immunodeficiency (SCID) and Its New Treatment Modalities. Cureus 2023; 15:e47759. [PMID: 38022338 PMCID: PMC10676291 DOI: 10.7759/cureus.47759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Severe combined immunodeficiency (SCID) is a rare condition with very high mortality. SCID is mainly caused by the multiple mutations of genes affecting the entire immune cells. Children with this disease are born with an impaired immune system. The child appears healthy but the consequences of the impaired immune system lead to various secondary infections such as meningeal infections and respiratory infections further leading to consolidation, diarrhea, inflammation of skin and other systemic diseases. Severe combined immunodeficiency is also known as "bubble boy disease" or "living in the bubble" syndrome, as in early days for treatment the physicians decided to completely isolate them until they got the perfect match for the bone marrow transplantation. It is one of the pediatric emergencies and is to be treated as soon as possible. SCID involves multiple genes which leads to makes diagnosis of the disease cumbersome. In early years many infants were diagnosed almost after half a year and in severe conditions which led to the decrease in the survival rate of the children. But now due to advanced newborn screening modalities and other monitoring systems it can be diagnosed as early as within three months of age. The various treatment modalities include hematopoietic stem cell transplantation, gene therapy, enzyme replacement therapy and chemotherapy. This narrative review article describes about the severe combined immunodeficiency and its newer treatment modalities.
Collapse
Affiliation(s)
- Akshad M Wadbudhe
- Department of Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Revat J Meshram
- Department of Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Shivangi C Tidke
- Department of Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
20
|
Ersoy GZ, Çipe F, Fışgın T, Aksoy BA, Öner ÖB, Hashemi N, Aydoğdu S, Erdem M, Dikme G, Murat K, Bozkurt C. The impact of Treosulfan-based conditioning for inborn errors of immunity: Is dose monitoring crucial? Clin Transplant 2023; 37:e15083. [PMID: 37534623 DOI: 10.1111/ctr.15083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION In children with inborn errors of immunity (IEI) who will receive a hematopoietic stem cell transplant (HSCT) treosulfan-based conditioning is currently preferred. The aim of this study was to investigate early and late outcomes in pediatric IEI patients receiving pre-HSCT treosulfan and to examine the effect of treosulfan dose monitoring on outcomes. METHODS Seventy-three pediatric patients receiving this management between 2015 and 2022 were included. RESULTS Overall survival rate was 80%, and event-free survival was 67.8%. A larger treosulfan dose AUC after first application increased the rate of early toxicity (p = .034) and slowed lymphocyte engraftment (r = .290; p = .030). Underlying disease, treosulfan AUC, donor type, stem cell type, number of immunosuppressive agents, the dose of anti-thymocyte globulin, and post-transplantation cyclophosphamide did not to increase risk of acute graft-versus-host disease. The risk of mixed chimerism (MC) in patients with autoimmune lymphoproliferative syndrome and leukocyte adhesion deficiency were higher than those with severe combined immunodeficiency (p = .021 and p = .014, respectively). The risk of MC was lower in those receiving peripheral blood stem cells (SC) compared with bone marrow derived SC (OR = .204, p = .022). CONCLUSION The AUC of the treosulfan dose was not associated with poorer late outcomes. Treosulfan is an agent that can be used safely in the IEI patient group, level measurement appears essential to identify early toxicities. Prospective studies with more extended follow-up periods are needed.
Collapse
Affiliation(s)
- Gizem Zengin Ersoy
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Funda Çipe
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Allergy-Immunology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Tunç Fışgın
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Basak Adakli Aksoy
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Özlem Başoğlu Öner
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Nazlı Hashemi
- Altınbaş University Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Selime Aydoğdu
- Umraniye Research & Training Hospital Pediatric Hematology & Oncology Department, Medical Sciences University, İstanbul, Turkey
| | - Melek Erdem
- İstinye University Pediatric Hematology Oncology, Gaziosmanpaşa Medical Park Hospital, İstanbul, Turkey
| | - Gürcan Dikme
- Aydin University Pediatric Hematology Oncology, Florya Medical Park Hospital, İstanbul, Turkey
| | | | - Ceyhun Bozkurt
- Medical Park Bahçelievler Hospital Pediatric Hematology Oncology & Pediatric Bone Marrow Transplantation Unit, İstinye University Pediatric Hematology Oncology, İstanbul, Turkey
| |
Collapse
|
21
|
Peddi NC, Vuppalapati S, Sreenivasulu H, Muppalla SK, Reddy Pulliahgaru A. Guardians of Immunity: Advances in Primary Immunodeficiency Disorders and Management. Cureus 2023; 15:e44865. [PMID: 37809154 PMCID: PMC10560124 DOI: 10.7759/cureus.44865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/10/2023] Open
Abstract
Primary immunodeficiency disorders (PIDs) are a heterogeneous group of genetic conditions profoundly impacting immune function. The investigation spans various PID categories, offering insights into their distinct pathogenic mechanisms and clinical manifestations. Within the adaptive immune system, B-cell, T-cell, and combined immunodeficiencies are dissected, emphasizing their critical roles in orchestrating effective immune responses. In the realm of the innate immune system, focus is directed toward phagocytes and complement deficiencies, underscoring the pivotal roles of these components in initial defense against infections. Furthermore, the review delves into disorders of immune dysregulation, encompassing hemophagocytic lymphohistiocytosis (HLH), autoimmune lymphoproliferative syndrome (ALPS), immune dysregulation, polyendocrinopathy, enteropathy, and X-linked(IPEX), and autoimmunity polyendocrinopathy candidiasis-ectodermal dystrophy(APECED), elucidating the intricate interplay between immune tolerance and autoimmunity prevention. Diagnostic strategies for PIDs are explored, highlighting advancements in genetic and molecular techniques that enable precise identification of underlying genetic mutations and alterations in immune function. We have also outlined treatment modalities for PIDs, which often entail a multidisciplinary approach involving immunoglobulin replacement, antimicrobial prophylaxis, and, in select cases, hematopoietic stem cell transplantation. Emerging therapies, including gene therapy, hold promise for targeted interventions. In essence, this review encapsulates the complexity of PIDs, emphasizing the critical importance of early diagnosis and tailored therapeutic interventions. As research advances, a clearer understanding of these disorders emerges, fostering optimism for enhanced patient care and management in the future.
Collapse
Affiliation(s)
| | - Sravya Vuppalapati
- General Physician, People's Education Society (PES) Institute of Medical Sciences and Research, Kuppam, IND
| | - Himabindu Sreenivasulu
- General Physician, People's Education Society (PES) Institute of Medical Sciences and Research, Kuppam, IND
| | - Sudheer Kumar Muppalla
- Pediatrics, People's Education Society (PES) Institute of Medical Sciences and Research, kuppam, IND
| | - Apeksha Reddy Pulliahgaru
- Pediatrics, People's Education Society (PES) Institute of Medical Sciences and Research, Kuppam, IND
| |
Collapse
|
22
|
Staudacher O, Klein J, Thee S, Ullrich J, Wahn V, Unterwalder N, Kölsch U, Lankes E, Stittrich A, Dedieu C, Dinges S, Völler M, Schuetz C, Schulte J, Boztug K, Meisel C, Kuehl JS, Krüger R, Blankenstein O, von Bernuth H. Screening Newborns for Low T Cell Receptor Excision Circles (TRECs) Fails to Detect Immunodeficiency, Centromeric Instability, and Facial Anomalies Syndrome. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2872-2883. [PMID: 37302792 DOI: 10.1016/j.jaip.2023.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Assessment of T-cell receptor excision circles (TRECs) in dried blood spots of newborns allows the detection of severe combined immunodeficiency (SCID) (T cells <300/μL at birth) with a presumed sensitivity of 100%. TREC screening also identifies patients with selected combined immunodeficiency (CID) (T cells >300/μL, yet <1500/μL at birth). Nevertheless, relevant CIDs that would benefit from early recognition and curative treatment pass undetected. OBJECTIVE We hypothesized that TREC screening at birth cannot identify CIDs that develop with age. METHODS We analyzed the number of TRECs in dried blood spots in archived Guthrie cards of 22 children who had been born in the Berlin-Brandenburg area between January 2006 and November 2018 and who had undergone hematopoietic stem-cell transplantation (HSCT) for inborn errors of immunity. RESULTS All patients with SCID would have been identified by TREC screening, but only 4 of 6 with CID. One of these patients had immunodeficiency, centromeric instability, and facial anomalies syndrome type 2 (ICF2). Two of 3 patients with ICF whom we have been following up at our institution had TREC numbers above the cutoff value suggestive of SCID at birth. Yet all patients with ICF had a severe clinical course that would have justified earlier HSCT. CONCLUSIONS In ICF, naïve T cells may be present at birth, yet they decline with age. Therefore, TREC screening cannot identify these patients. Early recognition is nevertheless crucial, as patients with ICF benefit from HSCT early in life.
Collapse
Affiliation(s)
- Olga Staudacher
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Jeanette Klein
- Newborn Screening Laboratory, Charité Universitätsmedizin, Berlin, Germany
| | - Stephanie Thee
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan Ullrich
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Volker Wahn
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nadine Unterwalder
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Uwe Kölsch
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Erwin Lankes
- Newborn Screening Laboratory, Charité Universitätsmedizin, Berlin, Germany; Department of Pediatric Endocrinology, Charité-Uninrsitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anna Stittrich
- Department of Human Genetics, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Cinzia Dedieu
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sarah Dinges
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mirjam Völler
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Catharina Schuetz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Johannes Schulte
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; St. Anna Children's Cancer Research Institute, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Christian Meisel
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany; Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörn-Sven Kuehl
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Pediatric Oncology, Hematology and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Renate Krüger
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany; Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.
| |
Collapse
|
23
|
Paris K, Wall LA. The Treatment of Primary Immune Deficiencies: Lessons Learned and Future Opportunities. Clin Rev Allergy Immunol 2023; 65:19-30. [PMID: 35776401 PMCID: PMC9247903 DOI: 10.1007/s12016-022-08950-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/30/2022]
Abstract
Primary immunodeficiency is a group of disorders associated with susceptibility to infectious agents and the development of various comorbidities. Many primary immunodeficiencies are complicated by immune dysregulation, autoinflammation, or autoimmunity which impacts multiple organ systems. Major advances in the treatment of these disorders have occurred over the last half-century, and deeper molecular understanding of many disorders combined with clinically available genetic testing is allowing for use of precision therapy for several primary immunodeficiencies. Patients with antibody deficiencies who rely on immunoglobulin replacement therapy now have many treatment options with products that are much safer and better tolerated compared to the past. Newborn screening for severe combined immunodeficiency, now implemented throughout the USA and in many countries worldwide, has lowered the age at which many patients are diagnosed with these diseases. Early diagnosis of severe combined immunodeficiency allows infants to proceed to definitive therapy such as stem cell transplantation or gene therapy prior to facing potentially life-threatening infections. While stem cell transplantation continues to carry significant risks, knowledge gained over recent decades is allowing for improved survival with less toxicity and less graft versus host disease.
Collapse
Affiliation(s)
- Kenneth Paris
- Department of Pediatrics, Division of Allergy and Immunology, Louisiana State University Health Sciences Center New Orleans and Children’s Hospital New Orleans, New Orleans, LA USA
| | - Luke A. Wall
- Department of Pediatrics, Division of Allergy and Immunology, Louisiana State University Health Sciences Center New Orleans and Children’s Hospital New Orleans, New Orleans, LA USA
| |
Collapse
|
24
|
Thakar MS, Logan BR, Puck JM, Dunn EA, Buckley RH, Cowan MJ, O'Reilly RJ, Kapoor N, Satter LF, Pai SY, Heimall J, Chandra S, Ebens CL, Chellapandian D, Williams O, Burroughs LM, Saldana BD, Rayes A, Madden LM, Chandrakasan S, Bednarski JJ, DeSantes KB, Cuvelier GDE, Teira P, Gillio AP, Eissa H, Knutsen AP, Goldman FD, Aquino VM, Shereck EB, Moore TB, Caywood EH, Lugt MTV, Rozmus J, Broglie L, Yu LC, Shah AJ, Andolina JR, Liu X, Parrott RE, Dara J, Prockop S, Martinez CA, Kapadia M, Jyonouchi SC, Sullivan KE, Bleesing JJ, Chaudhury S, Petrovic A, Keller MD, Quigg TC, Parikh S, Shenoy S, Seroogy C, Rubin T, Decaluwe H, Routes JM, Torgerson TR, Leiding JW, Pulsipher MA, Kohn DB, Griffith LM, Haddad E, Dvorak CC, Notarangelo LD. Measuring the effect of newborn screening on survival after haematopoietic cell transplantation for severe combined immunodeficiency: a 36-year longitudinal study from the Primary Immune Deficiency Treatment Consortium. Lancet 2023; 402:129-140. [PMID: 37352885 PMCID: PMC10386791 DOI: 10.1016/s0140-6736(23)00731-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/25/2023] [Accepted: 04/03/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND Severe combined immunodeficiency (SCID) is fatal unless durable adaptive immunity is established, most commonly through allogeneic haematopoietic cell transplantation (HCT). The Primary Immune Deficiency Treatment Consortium (PIDTC) explored factors affecting the survival of individuals with SCID over almost four decades, focusing on the effects of population-based newborn screening for SCID that was initiated in 2008 and expanded during 2010-18. METHODS We analysed transplantation-related data from children with SCID treated at 34 PIDTC sites in the USA and Canada, using the calendar time intervals 1982-89, 1990-99, 2000-09, and 2010-18. Categorical variables were compared by χ2 test and continuous outcomes by the Kruskal-Wallis test. Overall survival was estimated by the Kaplan-Meier method. A multivariable analysis using Cox proportional hazards regression models examined risk factors for HCT outcomes, including the variables of time interval of HCT, infection status and age at HCT, trigger for diagnosis, SCID type and genotype, race and ethnicity of the patient, non-HLA-matched sibling donor type, graft type, GVHD prophylaxis, and conditioning intensity. FINDINGS For 902 children with confirmed SCID, 5-year overall survival remained unchanged at 72%-73% for 28 years until 2010-18, when it increased to 87% (95% CI 82·1-90·6; n=268; p=0·0005). For children identified as having SCID by newborn screening since 2010, 5-year overall survival was 92·5% (95% CI 85·8-96·1), better than that of children identified by clinical illness or family history in the same interval (79·9% [69·5-87·0] and 85·4% [71·8-92·8], respectively [p=0·043]). Multivariable analysis demonstrated that the factors of active infection (hazard ratio [HR] 2·41, 95% CI 1·56-3·72; p<0·0001), age 3·5 months or older at HCT (2·12, 1·38-3·24; p=0·001), Black or African-American race (2·33, 1·56-3·46; p<0·0001), and certain SCID genotypes to be associated with lower overall survival during all time intervals. Moreover, after adjusting for several factors in this multivariable analysis, HCT after 2010 no longer conveyed a survival advantage over earlier time intervals studied (HR 0·73, 95% CI 0·43-1·26; p=0·097). This indicated that younger age and freedom from infections at HCT, both directly driven by newborn screening, were the main drivers for recent improvement in overall survival. INTERPRETATION Population-based newborn screening has facilitated the identification of infants with SCID early in life, in turn leading to prompt HCT while avoiding infections. Public health programmes worldwide can benefit from this definitive demonstration of the value of newborn screening for SCID. FUNDING National Institute of Allergy and Infectious Diseases, Office of Rare Diseases Research, and National Center for Advancing Translational Sciences.
Collapse
Affiliation(s)
- Monica S Thakar
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA.
| | - Brent R Logan
- Division of Biostatistics, Medical College of Wisconsin, WI, USA; Center for International Blood and Marrow Transplant Research, Milwaukee, WI, USA
| | - Jennifer M Puck
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplantation, University of California San Francisco, CA, USA; UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Elizabeth A Dunn
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplantation, University of California San Francisco, CA, USA
| | - Rebecca H Buckley
- Department of Allergy and Immunology, Department of Pediatrics and Immunology, Duke University Medical Center, Durham, NC, USA
| | - Morton J Cowan
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplantation, University of California San Francisco, CA, USA; UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Richard J O'Reilly
- Stem Cell Transplantation and Cellular Therapy, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neena Kapoor
- Transplant and Cell Therapy Program and Laboratory, Department of Pediatrics, Keck School of Medicine, University of Southern California, CA, USA; Hematology, Oncology and TCT, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Lisa Forbes Satter
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Immunology Allergy and Retrovirology, Center for Human Immunobiology, Texas Children's Hospital Infusion Center, Houston, TX, USA
| | - Sung-Yun Pai
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute (NCI)/NIH, Bethesda, MD, USA
| | - Jennifer Heimall
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, PA, USA; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sharat Chandra
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christen L Ebens
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, USA
| | - Deepak Chellapandian
- Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Cell and Gene Therapy for Nonmalignant Conditions, Johns Hopkins All Children's Hospital, St Petersburg, FL, USA
| | - Olatundun Williams
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA; Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, New York-Presbyterian Morgan Stanley Children's Hospital, New York, NY, USA
| | - Lauri M Burroughs
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Blachy Davila Saldana
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington DC, USA; Division of Blood and Marrow Transplantation and Center for Cancer and Immunology Research, Children's National Hospital, Washington DC, USA
| | - Ahmad Rayes
- Pediatric Immunology and Blood and Marrow Transplant Program, University of Utah, Salt Lake City, UT, USA; Intermountain Primary Children's Hospital, Salt Lake City, UT, USA
| | - Lisa M Madden
- Pediatric Bone Marrow Transplant Program, Texas Transplant Institute, San Antonio, TX, USA
| | - Shanmuganathan Chandrakasan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | | | - Geoffrey D E Cuvelier
- University of Manitoba, Winnipeg, MB, Canada; Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, Winnipeg, MB, Canada
| | - Pierre Teira
- Department of Pediatrics and Department of Microbiology, Immunology and Infectious Diseases, University of Montreal, Montreal, QC, Canada; Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Alfred P Gillio
- Pediatric Stem Cell and Cellular Therapy Division, Joseph M Sanzari Children's Hospital at HMH Hackensack University Medical Center, Hackensack, NJ, USA
| | - Hesham Eissa
- Department of Pediatrics, University of Colorado, Aurora, CO, USA; Bone Marrow Transplant and Cellular Therapeutics, Children's Hospital of Colorado, Aurora, CO, USA
| | - Alan P Knutsen
- Pediatric Allergy and Immunology, St Louis University, St Louis, MO, USA; Jeffrey Modell Diagnostic & Research Center for Primary Immunodeficiencies, Cardinal Glennon Children's Hospital, St Louis, MO, USA
| | - Frederick D Goldman
- Division of Hematology/Oncology/BMT, Department of Pediatrics, University of Alabama, Birmingham, AL, USA
| | - Victor M Aquino
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Evan B Shereck
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Theodore B Moore
- Pediatric Blood and Marrow Transplant Program, Division of Pediatric Hematology/Oncology in the Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, USA
| | - Emi H Caywood
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Nemours Children's Health, Delaware, Wilmington, DE, USA
| | | | - Jacob Rozmus
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada; British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Larisa Broglie
- Center for International Blood and Marrow Transplant Research, Milwaukee, WI, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lolie C Yu
- Louisiana State University Health New Orleans School of Medicine, New Orleans, LA, USA; Children's Hospital of New Orleans, New Orleans, LA, USA
| | - Ami J Shah
- Division of Hematology/Oncology/Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Palo Alto, CA, USA
| | - Jeffrey R Andolina
- Department of Pediatrics, Golisano Children's Hospital, University of Rochester Medical Center, Rochester, NY, USA
| | - Xuerong Liu
- Division of Biostatistics, Medical College of Wisconsin, WI, USA
| | - Roberta E Parrott
- Department of Allergy and Immunology, Department of Pediatrics and Immunology, Duke University Medical Center, Durham, NC, USA
| | - Jasmeen Dara
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplantation, University of California San Francisco, CA, USA; UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Susan Prockop
- Department of Pediatrics, Harvard University Medical School, Boston, MA, USA; Dana Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Caridad A Martinez
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Immunology Allergy and Retrovirology, Center for Human Immunobiology, Texas Children's Hospital Infusion Center, Houston, TX, USA
| | - Malika Kapadia
- Department of Pediatrics, Harvard University Medical School, Boston, MA, USA; Dana Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Soma C Jyonouchi
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, PA, USA; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen E Sullivan
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, PA, USA; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jack J Bleesing
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sonali Chaudhury
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Aleksandra Petrovic
- Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Michael D Keller
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington DC, USA; Division of Blood and Marrow Transplantation and Center for Cancer and Immunology Research, Children's National Hospital, Washington DC, USA; Intermountain Primary Children's Hospital, Salt Lake City, UT, USA
| | - Troy C Quigg
- Pediatrics, Michigan State University College of Human Medicine, Grand Rapids, MI, USA; Pediatric Blood and Marrow Transplant and Cellular Therapy Program, Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Suhag Parikh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Shalini Shenoy
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Christine Seroogy
- Division of Allergy, Immunology, and Rheumatology, University of Wisconsin, Madison, WI, USA
| | - Tamar Rubin
- Division of Pediatric Allergy and Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Hélène Decaluwe
- Department of Pediatrics and Department of Microbiology, Immunology and Infectious Diseases, University of Montreal, Montreal, QC, Canada; Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - John M Routes
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Troy R Torgerson
- Experimental Immunology, Allen Institute for Immunology, Seattle, WA, USA
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Michael A Pulsipher
- Pediatric Immunology and Blood and Marrow Transplant Program, University of Utah, Salt Lake City, UT, USA; Intermountain Primary Children's Hospital, Salt Lake City, UT, USA
| | - Donald B Kohn
- Pediatric Blood and Marrow Transplant Program, Division of Pediatric Hematology/Oncology in the Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, USA
| | - Linda M Griffith
- Division of Allergy, Immunology and Transplantation, (NIAID)/NIH, Bethesda, MD, USA
| | - Elie Haddad
- Department of Pediatrics and Department of Microbiology, Immunology and Infectious Diseases, University of Montreal, Montreal, QC, Canada; Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - Christopher C Dvorak
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplantation, University of California San Francisco, CA, USA; UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID)/NIH, Bethesda, MD, USA
| |
Collapse
|
25
|
Slatter MA, Maschan MA, Gennery AR. T-lymphocyte depleted transplants for inborn errors of immunity. Expert Rev Clin Immunol 2023; 19:1315-1324. [PMID: 37554030 DOI: 10.1080/1744666x.2023.2245146] [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: 04/13/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023]
Abstract
INTRODUCTION Hematopoietic stem cell transplantation is a curative treatment for many inborn errors of immunity (IEI). Incremental improvements and advances in care have led to high rates of >85% survival and cure in many of these diseases. Improvements in HLA-classification and matching have led to increased survival using HLA-matched donors, but survival using T-lymphocyte-depleted mismatched grafts remained significantly worse until fairly recently. Advances in T-lymphocyte depletion methods and graft engineering, although not specific to IEI, have been widely adopted and instrumental in changing the landscape of donor selection, such that a donor should now be possible for every patient. AREAS COVERED A literature review focusing on T-lymphocyte depletion methodologies and treatment results was performed. The importance of early T-lymphocyte immunoreconstitution to protect against viral infection is reviewed. Two main platforms now dominate the field - immune-magnetic selection of specific cell types and post-transplant chemotherapeutic targeting of rapidly proliferating allo-reactive T-lymphocytes - the emerging literature on these reports, focusing on IEI, is explored, as well as the impact of serotherapy on early immunoreconstitution. EXPERT OPINION Pharmacokinetic monitoring of serotherapy agents, and use of co-stimulatory molecule blockade are likely to become more widespread. Post-transplant cyclophosphamide or TCR depletion strategies are likely to become the dominant methods of transplantation for nonmalignant diseases.
Collapse
Affiliation(s)
- M A Slatter
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, UK
| | - M A Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Department of Hematology, Oncology and Radiation Therapy, Pirogov Russian National Research Medical University, Moscow, Russia
| | - A R Gennery
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Paediatric Stem Cell Transplant Unit, Great North Children's Hospital, Newcastle Upon Tyne, UK
| |
Collapse
|
26
|
Riller Q, Fourgeaud J, Bruneau J, De Ravin SS, Smith G, Fusaro M, Meriem S, Magerus A, Luka M, Abdessalem G, Lhermitte L, Jamet A, Six E, Magnani A, Castelle M, Lévy R, Lecuit MM, Fournier B, Winter S, Semeraro M, Pinto G, Abid H, Mahlaoui N, Cheikh N, Florkin B, Frange P, Jeziorski E, Suarez F, Sarrot-Reynauld F, Nouar D, Debray D, Lacaille F, Picard C, Pérot P, Regnault B, Da Rocha N, de Cevins C, Delage L, Pérot BP, Vinit A, Carbone F, Brunaud C, Marchais M, Stolzenberg MC, Asnafi V, Molina T, Rieux-Laucat F, Notarangelo LD, Pittaluga S, Jais JP, Moshous D, Blanche S, Malech H, Eloit M, Cavazzana M, Fischer A, Ménager MM, Neven B. Late-onset enteric virus infection associated with hepatitis (EVAH) in transplanted SCID patients. J Allergy Clin Immunol 2023; 151:1634-1645. [PMID: 36638922 PMCID: PMC10336473 DOI: 10.1016/j.jaci.2022.12.822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Allogenic hematopoietic stem cell transplantation (HSCT) and gene therapy (GT) are potentially curative treatments for severe combined immunodeficiency (SCID). Late-onset posttreatment manifestations (such as persistent hepatitis) are not uncommon. OBJECTIVE We sought to characterize the prevalence and pathophysiology of persistent hepatitis in transplanted SCID patients (SCIDH+) and to evaluate risk factors and treatments. METHODS We used various techniques (including pathology assessments, metagenomics, single-cell transcriptomics, and cytometry by time of flight) to perform an in-depth study of different tissues from patients in the SCIDH+ group and corresponding asymptomatic similarly transplanted SCID patients without hepatitis (SCIDH-). RESULTS Eleven patients developed persistent hepatitis (median of 6 years after HSCT or GT). This condition was associated with the chronic detection of enteric viruses (human Aichi virus, norovirus, and sapovirus) in liver and/or stools, which were not found in stools from the SCIDH- group (n = 12). Multiomics analysis identified an expansion of effector memory CD8+ T cells with high type I and II interferon signatures. Hepatitis was associated with absence of myeloablation during conditioning, split chimerism, and defective B-cell function, representing 25% of the 44 patients with SCID having these characteristics. Partially myeloablative retransplantation or GT of patients with this condition (which we have named as "enteric virus infection associated with hepatitis") led to the reconstitution of T- and B-cell immunity and remission of hepatitis in 5 patients, concomitantly with viral clearance. CONCLUSIONS Enteric virus infection associated with hepatitis is related to chronic enteric viral infection and immune dysregulation and is an important risk for transplanted SCID patients with defective B-cell function.
Collapse
Affiliation(s)
- Quentin Riller
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Jacques Fourgeaud
- University of Paris Cité, Paris, France; Microbiology Department, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, Paris, France; Prise en Charge des Anomalies Congénitales et leur Traitement, Unit 7328, Imagine Institute, University of Paris Cité, Paris, France
| | - Julie Bruneau
- University of Paris Cité, Paris, France; Pathology Department, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Imagine Institute, INSERM UMR 1163, Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Suk See De Ravin
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Grace Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Mathieu Fusaro
- Study Center for Primary Immunodeficiencies, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Samy Meriem
- Laboratory of Biostatistics, University of Paris Cité, Paris, France
| | - Aude Magerus
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Marine Luka
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Ghaith Abdessalem
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Ludovic Lhermitte
- University of Paris Cité, Paris, France; Laboratory of Onco-Haematology, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; the Institut Necker-Enfants Malades (INEM), INSERM UMR 1151, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anne Jamet
- University of Paris Cité, Paris, France; Microbiology Department, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; the Institut Necker-Enfants Malades (INEM), INSERM UMR 1151, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Emmanuelle Six
- University of Paris Cité, Paris, France; Laboratory of Human Lympho-Hematopoiesis, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Alessandra Magnani
- Department of Biotherapy, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Martin Castelle
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Romain Lévy
- University of Paris Cité, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Mathilde M Lecuit
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Benjamin Fournier
- University of Paris Cité, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sarah Winter
- University of Paris Cité, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Michaela Semeraro
- University of Paris Cité, Paris, France; Clinical Investigation Center, Clinical Research Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Graziella Pinto
- Pediatric Endocrinology, Gynecology, Diabetology, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Hanène Abid
- University of Paris Cité, Paris, France; Microbiology Department, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nizar Mahlaoui
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nathalie Cheikh
- Pediatric Hematology Oncology Unit, University Hospital of Besançon, Besançon, France
| | - Benoit Florkin
- Immuno-Hémato-Rhumatologie Pédiatrique, Service de Pédiatrie, CHR Citadelle, Liege, Belgium
| | - Pierre Frange
- University of Paris Cité, Paris, France; Microbiology Department, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Eric Jeziorski
- Department of Pediatrics, Infectious Diseases, and Immunology, University of Montpellier, CHU Montpellier, Montpellier, France
| | - Felipe Suarez
- University of Paris Cité, Paris, France; Imagine Institute, INSERM UMR 1163, Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Hematology Department, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Dalila Nouar
- Service d'Immunologie Clinique et d'Allergologie, Centre Hospitalier Régional Universitaire, Tours, France
| | - Dominique Debray
- Pediatric Liver Unit, National Reference Center for Rare Diseases, Biliary Atresia and Genetic Cholestasis, Inflammatory Biliary Diseases and Autoimmune Hepatitis, ERN Rare Liver, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Florence Lacaille
- Gastroenterology-Hepatology-Nutrition Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Capucine Picard
- Study Center for Primary Immunodeficiencies, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Philippe Pérot
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, Paris, France; OIE Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Paris, France
| | - Béatrice Regnault
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, Paris, France; OIE Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Paris, France
| | - Nicolas Da Rocha
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, Paris, France; OIE Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Paris, France
| | - Camille de Cevins
- University of Paris Cité, Paris, France; Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Imagine Institute, INSERM UMR 1163, Paris, France; Artificial Intelligence & Deep Analytics (AIDA) Group, Data & Data Science (DDS), Sanofi R&D, Chilly-Mazarin, France
| | - Laure Delage
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Brieuc P Pérot
- University of Paris Cité, Paris, France; Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Angélique Vinit
- Sorbonne Université, UMS037, PASS, Plateforme de Cytométrie de la Pitié-Salpêtrière CyPS, Paris, France
| | - Francesco Carbone
- University of Paris Cité, Paris, France; Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France; Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Camille Brunaud
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Manon Marchais
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Marie-Claude Stolzenberg
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Vahid Asnafi
- University of Paris Cité, Paris, France; Laboratory of Onco-Haematology, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; the Institut Necker-Enfants Malades (INEM), INSERM UMR 1151, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Thierry Molina
- University of Paris Cité, Paris, France; Pathology Department, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Frédéric Rieux-Laucat
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | - Jean Philippe Jais
- University of Paris Cité, Paris, France; Laboratory of Biostatistics, University of Paris Cité, Paris, France
| | - Despina Moshous
- University of Paris Cité, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue contre le Cancer, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Stephane Blanche
- University of Paris Cité, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Harry Malech
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marc Eloit
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, Paris, France; OIE Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Paris, France; Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Marina Cavazzana
- University of Paris Cité, Paris, France; Laboratory of Onco-Haematology, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory of Human Lympho-Hematopoiesis, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Alain Fischer
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Collège de France, Paris, France
| | - Mickaël M Ménager
- University of Paris Cité, Paris, France; Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France; Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Bénédicte Neven
- University of Paris Cité, Paris, France; Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, INSERM UMR 1163, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| |
Collapse
|
27
|
Wang P, Liu C, Wei Z, Jiang W, Sun H, Wang Y, Hou J, Sun J, Huang Y, Wang H, Wang Y, He X, Wang X, Qian X, Zhai X. Nomogram for Predicting Early Mortality after Umbilical Cord Blood Transplantation in Children with Inborn Errors of Immunity. J Clin Immunol 2023:10.1007/s10875-023-01505-8. [PMID: 37155023 DOI: 10.1007/s10875-023-01505-8] [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: 03/24/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE Pediatric patients with inborn errors of immunity (IEI) undergoing umbilical cord blood transplantation (UCBT) are at risk of early mortality. Our aim was to develop and validate a prediction model for early mortality after UCBT in pediatric IEI patients based on pretransplant factors. METHODS Data from 230 pediatric IEI patients who received their first UCBT between 2014 and 2021 at a single center were analyzed retrospectively. Data from 2014-2019 and 2020-2021 were used as training and validation sets, respectively. The primary outcome of interest was early mortality. Machine learning algorithms were used to identify risk factors associated with early mortality and to build predictive models. The model with the best performance was visualized using a nomogram. Discriminative ability was measured using the area under the curve (AUC) and decision curve analysis. RESULTS Fifty days was determined as the cutoff for distinguishing early mortality in pediatric IEI patients undergoing UCBT. Of the 230 patients, 43 (18.7%) suffered early mortality. Multivariate logistic regression with pretransplant albumin, CD4 (absolute count), elevated C-reactive protein, and medical history of sepsis showed good discriminant AUC values of 0.7385 (95% CI, 0.5824-0.8945) and 0.827 (95% CI, 0.7409-0.9132) in predicting early mortality in the validation and training sets, respectively. The sensitivity and specificity were 0.5385 and 0.8154 for validation and 0.7667 and 0.7705 for training, respectively. The final model yielded net benefits across a reasonable range of risk thresholds. CONCLUSION The developed nomogram can predict early mortality in pediatric IEI patients undergoing UCBT.
Collapse
Affiliation(s)
- Ping Wang
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Chao Liu
- Yidu Cloud Technology Inc, Beijing, 100083, China
- Nanjing YiGenCloud Institute, Nanjing, 211899, China
| | - Zhongling Wei
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Wenjin Jiang
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Hua Sun
- Department of Gastroenterology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yuhuan Wang
- Department of Gastroenterology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Jia Hou
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Jinqiao Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Ying Huang
- Department of Gastroenterology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Hongsheng Wang
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Yao Wang
- Yidu Cloud Technology Inc, Beijing, 100083, China
| | - Xinjun He
- Yidu Cloud Technology Inc, Beijing, 100083, China
- Nanjing YiGenCloud Institute, Nanjing, 211899, China
| | - Xiaochuan Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Xiaowen Qian
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China
| | - Xiaowen Zhai
- Department of Hematology/Oncology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 201102, China.
| |
Collapse
|
28
|
Slatter M, Lum SH. Personalized hematopoietic stem cell transplantation for inborn errors of immunity. Front Immunol 2023; 14:1162605. [PMID: 37090739 PMCID: PMC10113466 DOI: 10.3389/fimmu.2023.1162605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Patients with inborn errors of immunity (IEI) have been transplanted for more than 50 years. Many long-term survivors have ongoing medical issues showing the need for further improvements in how hematopoietic stem cell transplantation (HSCT) is performed if patients in the future are to have a normal quality of life. Precise genetic diagnosis enables early treatment before recurrent infection, autoimmunity and organ impairment occur. Newborn screening for severe combined immunodeficiency (SCID) is established in many countries. For newly described disorders the decision to transplant is not straight-forward. Specific biologic therapies are effective for some diseases and can be used as a bridge to HSCT to improve outcome. Developments in reduced toxicity conditioning and methods of T-cell depletion for mismatched donors have made transplant an option for all eligible patients. Further refinements in conditioning plus precise graft composition and additional cellular therapy are emerging as techniques to personalize the approach to HSCT for each patient.
Collapse
Affiliation(s)
- Mary Slatter
- Paediatric Immunology and HSCT, Newcastle University, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Great North Children’s Hospital, Newcastle upon Tyne, United Kingdom
| | - Su Han Lum
- Paediatric Immunology and HSCT, Newcastle University, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Great North Children’s Hospital, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
29
|
Laberko A, Mukhinа A, Machneva E, Pashchenko O, Bykova T, Vahonina L, Bronin G, Skvortsova Y, Skorobogatova E, Kondratenko I, Fechina L, Shcherbina A, Zubarovskaya L, Balashov D, Rumiantsev A. Allogeneic Hematopoietic Stem Cell Transplantation Activity in Inborn Errors of Immunity in Russian Federation. J Clin Immunol 2023:10.1007/s10875-023-01476-w. [PMID: 37009957 PMCID: PMC10068234 DOI: 10.1007/s10875-023-01476-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/20/2023] [Indexed: 04/04/2023]
Abstract
PURPOSE Allogeneic hematopoietic stem cell transplantation (HSCT) is an established therapy for many inborn errors of immunity (IEI). The indications for HSCT have expanded over the last decade. The study aimed to collect and analyze the data on HSCT activity in IEI in Russia. METHODS The data were collected from the Russian Primary Immunodeficiency Registry and complemented with information from five Russian pediatric transplant centers. Patients diagnosed with IEI by the age of 18 years and who received allogeneic HSCT by the end of 2020 were included. RESULTS From 1997 to 2020, 454 patients with IEI received 514 allogeneic HSCT. The median number of HSCTs per year has risen from 3 in 1997-2009 to 60 in 2015-2020. The most common groups of IEI were immunodeficiency affecting cellular and humoral immunity (26%), combined immunodeficiency with associated/syndromic features (28%), phagocyte defects (21%), and diseases of immune dysregulation (17%). The distribution of IEI diagnosis has changed: before 2012, the majority (65%) had severe combined immunodeficiency (SCID) and hemophagocytic lymphohistiocytosis (HLH), and after 2012, only 24% had SCID and HLH. Of 513 HSCTs, 48.5% were performed from matched-unrelated, 36.5% from mismatched-related (MMRD), and 15% from matched-related donors. In 349 transplants T-cell depletion was used: 325 TCRαβ/CD19+ depletion, 39 post-transplant cyclophosphamide, and 27 other. The proportion of MMRD has risen over the recent years. CONCLUSION The practice of HSCT in IEI has been changing in Russia. Expanding indications to HSCT and SCID newborn screening implementation may necessitate additional transplant beds for IEI in Russia.
Collapse
Affiliation(s)
- Alexandra Laberko
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Anna Mukhinа
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Russian National Association of Experts in Primary Immunodeficiency Registry, Moscow, Russia
| | - Elena Machneva
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Olga Pashchenko
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Tatiana Bykova
- RM Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Pavlov University, St. Petersburg, Russia
| | - Larisa Vahonina
- Sverdlovsk Regional Children's Hospital №1, Institute of Medical Cell Technologies, Yekaterinburg, Russia
| | | | - Yulia Skvortsova
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Elena Skorobogatova
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Irina Kondratenko
- Russian Children's Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - Larisa Fechina
- Sverdlovsk Regional Children's Hospital №1, Institute of Medical Cell Technologies, Yekaterinburg, Russia
| | - Anna Shcherbina
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Ludmila Zubarovskaya
- RM Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Pavlov University, St. Petersburg, Russia
| | - Dmitry Balashov
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexander Rumiantsev
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
30
|
Iancu O, Allen D, Knop O, Zehavi Y, Breier D, Arbiv A, Lev A, Lee YN, Beider K, Nagler A, Somech R, Hendel A. Multiplex HDR for disease and correction modeling of SCID by CRISPR genome editing in human HSPCs. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:105-121. [PMID: 36618262 PMCID: PMC9813580 DOI: 10.1016/j.omtn.2022.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Severe combined immunodeficiency (SCID) is a group of disorders caused by mutations in genes involved in the process of lymphocyte maturation and function. CRISPR-Cas9 gene editing of the patient's own hematopoietic stem and progenitor cells (HSPCs) ex vivo could provide a therapeutic alternative to allogeneic hematopoietic stem cell transplantation, the current gold standard for treatment of SCID. To eliminate the need for scarce patient samples, we engineered genotypes in healthy donor (HD)-derived CD34+ HSPCs using CRISPR-Cas9/rAAV6 gene-editing, to model both SCID and the therapeutic outcomes of gene-editing therapies for SCID via multiplexed homology-directed repair (HDR). First, we developed a SCID disease model via biallelic knockout of genes critical to the development of lymphocytes; and second, we established a knockin/knockout strategy to develop a proof-of-concept single-allelic gene correction. Based on these results, we performed gene correction of RAG2-SCID patient-derived CD34+ HSPCs that successfully developed into CD3+ T cells with diverse TCR repertoires in an in vitro T cell differentiation platform. In summary, we present a strategy to determine the optimal configuration for CRISPR-Cas9 gene correction of SCID using HD-derived CD34+ HSPCs, and the feasibility of translating this gene correction approach in patient-derived CD34+ HSPCs.
Collapse
Affiliation(s)
- Ortal Iancu
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Daniel Allen
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Orli Knop
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Yonathan Zehavi
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Dor Breier
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Adaya Arbiv
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Atar Lev
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
| | - Yu Nee Lee
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Katia Beider
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
| | - Arnon Nagler
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- The Division of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
| | - Raz Somech
- Pediatric Department A and the Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-HaShomer, Ramat Gan 5266202, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ayal Hendel
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| |
Collapse
|
31
|
Castiello MC, Ferrari S, Villa A. Correcting inborn errors of immunity: From viral mediated gene addition to gene editing. Semin Immunol 2023; 66:101731. [PMID: 36863140 PMCID: PMC10109147 DOI: 10.1016/j.smim.2023.101731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/25/2023] [Accepted: 02/14/2023] [Indexed: 03/04/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation is an effective treatment to cure inborn errors of immunity. Remarkable progress has been achieved thanks to the development and optimization of effective combination of advanced conditioning regimens and use of immunoablative/suppressive agents preventing rejection as well as graft versus host disease. Despite these tremendous advances, autologous hematopoietic stem/progenitor cell therapy based on ex vivo gene addition exploiting integrating γ-retro- or lenti-viral vectors, has demonstrated to be an innovative and safe therapeutic strategy providing proof of correction without the complications of the allogeneic approach. The recent advent of targeted gene editing able to precisely correct genomic variants in an intended locus of the genome, by introducing deletions, insertions, nucleotide substitutions or introducing a corrective cassette, is emerging in the clinical setting, further extending the therapeutic armamentarium and offering a cure to inherited immune defects not approachable by conventional gene addition. In this review, we will analyze the current state-of-the art of conventional gene therapy and innovative protocols of genome editing in various primary immunodeficiencies, describing preclinical models and clinical data obtained from different trials, highlighting potential advantages and limits of gene correction.
Collapse
Affiliation(s)
- Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy; Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy.
| |
Collapse
|
32
|
Sobrino S, Magnani A, Semeraro M, Martignetti L, Cortal A, Denis A, Couzin C, Picard C, Bustamante J, Magrin E, Joseph L, Roudaut C, Gabrion A, Soheili T, Cordier C, Lortholary O, Lefrere F, Rieux-Laucat F, Casanova JL, Bodard S, Boddaert N, Thrasher AJ, Touzot F, Taque S, Suarez F, Marcais A, Guilloux A, Lagresle-Peyrou C, Galy A, Rausell A, Blanche S, Cavazzana M, Six E. Severe hematopoietic stem cell inflammation compromises chronic granulomatous disease gene therapy. Cell Rep Med 2023; 4:100919. [PMID: 36706754 PMCID: PMC9975109 DOI: 10.1016/j.xcrm.2023.100919] [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: 07/06/2022] [Revised: 10/20/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023]
Abstract
X-linked chronic granulomatous disease (CGD) is associated with defective phagocytosis, life-threatening infections, and inflammatory complications. We performed a clinical trial of lentivirus-based gene therapy in four patients (NCT02757911). Two patients show stable engraftment and clinical benefits, whereas the other two have progressively lost gene-corrected cells. Single-cell transcriptomic analysis reveals a significantly lower frequency of hematopoietic stem cells (HSCs) in CGD patients, especially in the two patients with defective engraftment. These two present a profound change in HSC status, a high interferon score, and elevated myeloid progenitor frequency. We use elastic-net logistic regression to identify a set of 51 interferon genes and transcription factors that predict the failure of HSC engraftment. In one patient, an aberrant HSC state with elevated CEBPβ expression drives HSC exhaustion, as demonstrated by low repopulation in a xenotransplantation model. Targeted treatments to protect HSCs, coupled to targeted gene expression screening, might improve clinical outcomes in CGD.
Collapse
Affiliation(s)
- Steicy Sobrino
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Alessandra Magnani
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Michaela Semeraro
- Clinical Investigation Center CIC 1419, Necker-Enfants Malades Hospital, GH Paris Centre, Université Paris Cité, AP-HP, Paris, France
| | - Loredana Martignetti
- Clinical Bioinformatics Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Akira Cortal
- Clinical Bioinformatics Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Adeline Denis
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Chloé Couzin
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Capucine Picard
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades Hospital, AP-HP, Université Paris Cité, Paris, France; Lymphocyte Activation and Susceptibility to EBV Infection Laboratory, INSERM UMR 1163, Imagine Institute, Paris, France; Centre de Références des Déficits Immunitaires Héréditaires (CEREDIH), Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Jacinta Bustamante
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades Hospital, AP-HP, Université Paris Cité, Paris, France; Human Genetics of Infectious Diseases Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Elisa Magrin
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Laure Joseph
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Cécile Roudaut
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Aurélie Gabrion
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Tayebeh Soheili
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Corinne Cordier
- Plateforme de Cytométrie en Flux, Structure Fédérative de Recherche Necker, INSERM US24-CNRS UAR3633, Paris, France
| | - Olivier Lortholary
- Necker-Pasteur Center for Infectious Diseases and Tropical Medicine, Necker-Enfants Malades Hospital, AP-HP, Université Paris Cité, Imagine Institute, Paris, France
| | - François Lefrere
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Department of Adult Hematology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Frédéric Rieux-Laucat
- Immunogenetics of Pediatric Autoimmune Diseases Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Jean-Laurent Casanova
- Human Genetics of Infectious Diseases Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Sylvain Bodard
- Department of Adult Radiology, Necker Enfants-Malades Hospital, AP-HP, Université Paris Cité, Paris, France; Laboratoire d'Imagerie Biomédicale, LIB, Sorbonne Université, CNRS, INSERM, Paris, France
| | - Nathalie Boddaert
- Département de Radiologie Pédiatrique, INSERM UMR 1163 and UMR 1299, Imagine Institute, AP-HP, Necker-Enfants Malades Hospital, Paris, France
| | - Adrian J Thrasher
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fabien Touzot
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Sophie Taque
- CHU de Rennes, Département de Pédiatrie, Rennes, France
| | - Felipe Suarez
- Necker-Pasteur Center for Infectious Diseases and Tropical Medicine, Necker-Enfants Malades Hospital, AP-HP, Université Paris Cité, Imagine Institute, Paris, France; Imagine Institute, Université Paris Cité, Paris, France
| | - Ambroise Marcais
- Necker-Pasteur Center for Infectious Diseases and Tropical Medicine, Necker-Enfants Malades Hospital, AP-HP, Université Paris Cité, Imagine Institute, Paris, France
| | - Agathe Guilloux
- Mathematics and Modelization Laboratory, CNRS, Université Paris-Saclay, Université d'Evry, Evry, France
| | - Chantal Lagresle-Peyrou
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Anne Galy
- Genethon, Evry-Courcouronnes, France; Université Paris-Saclay, University Evry, Inserm, Genethon (UMR_S951), Evry-Courcouronnes, France
| | - Antonio Rausell
- Clinical Bioinformatics Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France; Service de Médecine Génomique des Maladies Rares, AP-HP, Necker-Enfants Malades Hospital, Paris, France
| | - Stephane Blanche
- Department of Pediatric Immunology, Hematology, and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Marina Cavazzana
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France; Imagine Institute, Université Paris Cité, Paris, France.
| | - Emmanuelle Six
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| |
Collapse
|
33
|
Dell’Orso G, Bagnasco F, Giardino S, Pierri F, Ferrando G, Di Martino D, Micalizzi C, Guardo D, Volpi S, Sabatini F, Miano M, Gattorno M, Dufour C, Faraci M. Hematopoietic stem cell transplantation for inborn errors of immunity: 30-year single-center experience. Front Immunol 2023; 14:1103080. [PMID: 36825011 PMCID: PMC9941625 DOI: 10.3389/fimmu.2023.1103080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents an effective treatment for a variety of inborn errors of immunity (IEI). We report the experience of children affected by IEI who received allo-HSCT over a period of 32 years at IRCCS Istituto Giannina Gaslini, Genoa, Italy. HSCTs were performed in 67 children with IEI. Kaplan-Meier estimates of overall survival (OS) rate at 5 years in the whole group of patients was 83.4% after a median follow-up of 4 years. Median age at transplant was 2.5 years. Eight allo-HSCTs were complicated by either primary or secondary graft failure (GF), the overall incidence of this complication being 10.9%. Incidence of grade 3-4 acute GvHD (aGvHD) was 18.7%, significantly lower in the haploidentical transplant cohort (p = 0.005). Year of transplant (≤2006 vs. >2006) was the main factor influencing the outcome. In fact, a significant improvement in 5-year OS was demonstrated (92.5% >2006 vs. 65% ≤2006, p = 0.049). Frequency of severe aGvHD was significantly reduced in recent years (≤2006 61.5%, vs. >2006 20%, p = 0.027). A significant progress has been the introduction of the TCR αβ/CD19-depleted haploidentical platform, which was associated with the absence of severe aGvHD. However, it was associated with 23.5% incidence of GF. All but one patient experiencing GF in the this specific cohort were successfully retransplanted. In summary, allo-HSCT is confirmed to be an effective treatment for children with IEI, even in the absence of an HLA-matched donor.
Collapse
Affiliation(s)
- Gianluca Dell’Orso
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology-Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Francesca Bagnasco
- Scientific Directorate, Epidemiology and Biostatistics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Stefano Giardino
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology-Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Filomena Pierri
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology-Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giulia Ferrando
- Infectious Diseases Unit and COVID-Hospital, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | | | - Daniela Guardo
- Hematology Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Stefano Volpi
- Center for Autoinflammatory Diseases and Immunodeficiencies, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Federica Sabatini
- Stem Cells and Cell Therapies Laboratory, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Maurizio Miano
- Hematology Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Gattorno
- Center for Autoinflammatory Diseases and Immunodeficiencies, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Carlo Dufour
- Hematology Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Maura Faraci
- Hematopoietic Stem Cell Transplantation Unit, Department of Hematology-Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
34
|
Klein OR, Bonfim C, Abraham A, Ruggeri A, Purtill D, Cohen S, Wynn R, Russell A, Sharma A, Ciccocioppo R, Prockop S, Boelens JJ, Bertaina A. Transplant for non-malignant disorders: an International Society for Cell & Gene Therapy Stem Cell Engineering Committee report on the role of alternative donors, stem cell sources and graft engineering. Cytotherapy 2023; 25:463-471. [PMID: 36710227 DOI: 10.1016/j.jcyt.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 01/30/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) is curative for many non-malignant disorders. As HSCT and supportive care technologies improve, this life-saving treatment may be offered to more and more patients. With the development of new preparative regimens, expanded alternative donor availability, and graft manipulation techniques, there are many options when choosing the best regimen for patients. Herein the authors review transplant considerations, transplant goals, conditioning regimens, donor choice, and graft manipulation strategies for patients with non-malignant disorders undergoing HSCT.
Collapse
Affiliation(s)
- Orly R Klein
- Division of Hematology, Oncology and Stem Cell Transplant and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA.
| | - Carmem Bonfim
- Pediatric Blood and Marrow Transplantation Division and Pele Pequeno Principe Research Institute, Hospital Pequeno Principe, Curitiba, Brazil
| | - Allistair Abraham
- Center for Cancer and Immunology Research, Cell Enhancement and Technologies for Immunotherapy, Children's National Hospital, Washington, DC, USA
| | - Annalisa Ruggeri
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Duncan Purtill
- Department of Hematology, Fiona Stanley Hospital, Perth, Australia
| | - Sandra Cohen
- Université de Montréal and Maisonneuve Rosemont Hospital, Montréal, Canada
| | - Robert Wynn
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Athena Russell
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy
| | - Susan Prockop
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Alice Bertaina
- Division of Hematology, Oncology and Stem Cell Transplant and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
35
|
Reilly L, Emonts M. Recurrent or unusual infections in children - when to worry about inborn errors of immunity. Ther Adv Infect Dis 2023; 10:20499361231162978. [PMID: 37089444 PMCID: PMC10116010 DOI: 10.1177/20499361231162978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/23/2023] [Indexed: 04/25/2023] Open
Abstract
Recurrent infections are a common presenting feature in paediatrics and, while most times considered part of normal growing up, they are also a classical hallmark of inborn errors of immunity (IEI). We aimed to outline the value of currently used signs for IEI and the influence of the changing epidemiology of infectious diseases due to implementation of new vaccines and the effect of the COVID-19 pandemic on the assessment of children with recurrent infections. Warning signs for IEI have been developed, but the supporting evidence for their effectiveness is limited, and immune dysregulation is more commonly recognised as a feature for IEI, making reliable identification of children who should be screened for IEI on clinical grounds difficult. In addition, the epidemiology of infectious diseases is changing due to restrictions related to Covid-19 as well as immunisations, which may change the threshold to screen children for IEI. Treatments for IEI are evolving and are often more effective and less complicated when started early. Screening for IEI can be initiated by the non-immunologist and should be considered early to ensure optimal treatment outcomes.
Collapse
Affiliation(s)
- Liam Reilly
- Paediatric Immunology, Infectious Diseases & Allergy, Great North Children’s Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | |
Collapse
|
36
|
Impact of Genetic Diagnosis on the Outcome of Hematopoietic Stem Cell Transplant in Primary Immunodeficiency Disorders. J Clin Immunol 2023; 43:636-646. [PMID: 36495401 PMCID: PMC9958161 DOI: 10.1007/s10875-022-01403-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 10/31/2022] [Indexed: 12/14/2022]
Abstract
To evaluate the relationship between knowledge of genetic diagnosis before HSCT and outcome, we reviewed all HSCTs for primary immune deficiencies (PID) performed at UCSF from 2007 through 2018. SCID, a distinct entity identified since 2010 in California by newborn screening and treated early, was considered separately. The underlying genetic condition was known at the time of HSCT in 85% of cases. Graft failure was less frequent in patients with a genetic diagnosis (19% with a genetic diagnosis versus 47% without, p = 0.020). Furthermore, event-free survival and overall survival (OS) at 5 years were better for those with a genetic diagnosis (78% with versus 44% without, p = 0.006; and 93% versus 60% without, p = 0.0002, respectively). OS at 5 years was superior for known-genotype patients with both SCID (p = 0.010) and non-SCID PID (p = 0.010). There was no difference in OS between HSCT done in 2007-2010 compared to more recently (p = 0.19). These data suggest that outcomes of HSCT for PID with known genotype may reflect specific experience and literature, or that a substantial proportion of patients with PID of undetermined genotype may have had underlying conditions for which HSCT may carry greater risk. The higher rate of graft failure in PID with unknown genotype may be in part explained by insufficient conditioning, which in turn could be dictated by compromised organ function in patients undergoing HSCT late in the course. Widespread availability of PID gene sequencing as standard care can provide genetic diagnoses for most patients with PID prior to HSCT, permitting optimization of transplant approach.
Collapse
|
37
|
Farshbafnadi M, Razi S, Rezaei N. Transplantation. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
38
|
Labrosse R, Boufaied I, Bourdin B, Gona S, Randolph HE, Logan BR, Bourbonnais S, Berthe C, Chan W, Buckley RH, Parrott RE, Cuvelier GDE, Kapoor N, Chandra S, Dávila Saldaña BJ, Eissa H, Goldman FD, Heimall J, O'Reilly R, Chaudhury S, Kolb EA, Shenoy S, Griffith LM, Pulsipher M, Kohn DB, Notarangelo LD, Pai SY, Cowan MJ, Dvorak CC, Haddad É, Puck JM, Barreiro LB, Decaluwe H. Aberrant T-cell exhaustion in severe combined immunodeficiency survivors with poor T-cell reconstitution after transplantation. J Allergy Clin Immunol 2023; 151:260-271. [PMID: 35987350 PMCID: PMC9924130 DOI: 10.1016/j.jaci.2022.08.004] [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: 04/11/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Severe combined immunodeficiency (SCID) comprises rare inherited disorders of immunity that require definitive treatment through hematopoietic cell transplantation (HCT) or gene therapy for survival. Despite successes of allogeneic HCT, many SCID patients experience incomplete immune reconstitution, persistent T-cell lymphopenia, and poor long-term outcomes. OBJECTIVE We hypothesized that CD4+ T-cell lymphopenia could be associated with a state of T-cell exhaustion in previously transplanted SCID patients. METHODS We analyzed markers of exhaustion in blood samples from 61 SCID patients at a median of 10.4 years after HCT. RESULTS Compared to post-HCT SCID patients with normal CD4+ T-cell counts, those with poor T-cell reconstitution showed lower frequency of naive CD45RA+/CCR7+ T cells, recent thymic emigrants, and TCR excision circles. They also had a restricted TCR repertoire, increased expression of inhibitory receptors (PD-1, 2B4, CD160, BTLA, CTLA-4), and increased activation markers (HLA-DR, perforin) on their total and naive CD8+ T cells, suggesting T-cell exhaustion and aberrant activation, respectively. The exhaustion score of CD8+ T cells was inversely correlated with CD4+ T-cell count, recent thymic emigrants, TCR excision circles, and TCR diversity. Exhaustion scores were higher among recipients of unconditioned HCT, especially when further in time from HCT. Patients with fewer CD4+ T cells showed a transcriptional signature of exhaustion. CONCLUSIONS Recipients of unconditioned HCT for SCID may develop late post-HCT T-cell exhaustion as a result of diminished production of T-lineage cells. Elevated expression of inhibitory receptors on their T cells may be a biomarker of poor long-term T-cell reconstitution.
Collapse
Affiliation(s)
- Roxane Labrosse
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Ines Boufaied
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Benoîte Bourdin
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Saideep Gona
- Genetics, Genomics, and Systems Biology, Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Ill
| | - Haley E Randolph
- Genetics, Genomics, and Systems Biology, Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Ill
| | - Brent R Logan
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wis
| | - Sara Bourbonnais
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Chloé Berthe
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada
| | - Wendy Chan
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, and UCSF Benioff Children's Hospital, San Francisco, Calif
| | | | | | - Geoffrey D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - 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, Calif
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Blachy J Dávila Saldaña
- Division of Blood and Marrow Transplantation, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Hesham Eissa
- Children's Hospital of Colorado, University of Colorado School of Medicine, Aurora, Colo
| | - Fred D Goldman
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, Ala
| | - Jennifer Heimall
- Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Richard O'Reilly
- Department of Pediatrics, Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sonali Chaudhury
- Division of Hematology, Oncology, and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Edward A Kolb
- Nemours Children's Health, Center for Cancer and Blood Disorders, Wilmington, Del
| | - Shalini Shenoy
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Linda M Griffith
- Division of Allergy, Immunology, and Transplantation, National Institutes of Health, Bethesda, Md
| | - Michael 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, Calif
| | - Donald B Kohn
- Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Calif
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Health, Bethesda, Md
| | - Sung-Yun Pai
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Morton J Cowan
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, 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, and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Élie Haddad
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Jennifer M Puck
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Luis B Barreiro
- Genetics, Genomics, and Systems Biology, Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Ill
| | - Hélène Decaluwe
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada; Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Montreal, Quebec, Canada.
| |
Collapse
|
39
|
Margarit-Soler A, Deyà-Martínez À, Canizales JT, Vlagea A, García-García A, Marsal J, Del Castillo MT, Planas S, Simó S, Esteve-Sole A, Grande MSL, Badell I, Tarrats MR, Fernández-Avilés F, Alsina L. Case report: Challenges in immune reconstitution following hematopoietic stem cell transplantation for CTLA-4 insufficiency-like primary immune regulatory disorders. Front Immunol 2022; 13:1070068. [PMID: 36636328 PMCID: PMC9831655 DOI: 10.3389/fimmu.2022.1070068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/28/2022] [Indexed: 12/28/2022] Open
Abstract
Cytotoxic T-lymphocyte antigen-4 (CTLA-4) haploinsufficiency is a T-cell hyperactivation disorder that can manifest with both immunodeficiency and immune dysregulation. Approximately one-third of patients may present mild symptoms and remain stable under supportive care. The remaining patients may develop severe multiorgan autoimmunity requiring lifelong immunosuppressive treatment. Hematopoietic stem cell transplantation (HSCT) is potentially curable for patients with treatment-resistant immune dysregulation. Nevertheless, little experience is reported regarding the management of complications post-HSCT. We present case 1 (CTLA-4 haploinsufficiency) and case 2 (CTLA-4 insufficiency-like phenotype) manifesting with severe autoimmunity including cytopenia and involvement of the central nervous system (CNS), lung, and gut and variable impairment of humoral responses. Both patients underwent HSCT for which the main complications were persistent mixed chimerism, infections, and immune-mediated complications [graft-versus-host disease (GVHD) and nodular lung disease]. Detailed management and outcomes of therapeutic interventions post-HSCT are discussed. Concretely, post-HSCT abatacept and human leukocyte antigen (HLA)-matched sibling donor lymphocyte infusions may be used to increase T-cell donor chimerism with the aim of correcting the immune phenotype of CTLA-4 haploinsufficiency.
Collapse
Affiliation(s)
- Adriana Margarit-Soler
- Bone Marrow Transplant Unit, Oncology Service, Hospital Sant Joan de Déu, Barcelona, Spain,*Correspondence: Adriana Margarit-Soler, ; Laia Alsina,
| | - Àngela Deyà-Martínez
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain,Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain,Clinical Immunology Program Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | - Juan Torres Canizales
- Clinical Immunology Unit, Department of Immunology, Biomedical Diagnostic Center, Hospital Clínic of Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alexandru Vlagea
- Clinical Immunology Unit, Department of Immunology, Biomedical Diagnostic Center, Hospital Clínic of Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana García-García
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain,Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain,Clinical Immunology Program Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | - Júlia Marsal
- Bone Marrow Transplant Unit, Oncology Service, Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - Sílvia Planas
- Department of Pathology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Sílvia Simó
- Infectious Diseases Unit, Department of Pediatrics, Hospital Sant Joan de Déu, Barcelona, Spain,Center for Biomedical Network Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Ana Esteve-Sole
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain,Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain,Clinical Immunology Program Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain
| | - María Suárez-Lledó Grande
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain,Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Isabel Badell
- Bone Marrow Transplant Unit, Oncology Service, Hospital Sant Joan de Déu, Barcelona, Spain,Pediatric Haematology and Stem Cell Transplantation Unit, Pediatric Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Montserrat Rovira Tarrats
- Bone Marrow Transplant Unit, Oncology Service, Hospital Sant Joan de Déu, Barcelona, Spain,Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Francesc Fernández-Avilés
- Hematopoietic Transplantation Unit, Hematology Department, Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain,Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain,Clinical Immunology Program Hospital Sant Joan de Déu-Hospital Clínic Barcelona, Barcelona, Spain,Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain,*Correspondence: Adriana Margarit-Soler, ; Laia Alsina,
| |
Collapse
|
40
|
Khan S, Siddiqui K, ElSolh H, AlJefri A, AlAhmari A, Ghemlas I, AlSaedi H, AlEnazi A, AlSeraihi A, Ayas M. Outcomes of blood and marrow transplantation in children less than 2-years of age: 23 years of experience at a single center. Int J Pediatr Adolesc Med 2022; 9:190-195. [PMID: 36937328 PMCID: PMC10019952 DOI: 10.1016/j.ijpam.2022.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/28/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVES Allogeneic hematopoietic cell transplantation (Allo-HCT) is a curative option for children with various malignant and non-malignant diseases. Most reports studied all age groups amongst children. Herein we analyzed our data in children transplanted at or less than 2-years of age. PATIENTS AND METHODS We reviewed medical charts of 618 patients who underwent 666 transplantation at our center between 1993 and 2015. There were 340 boys and 278 girls. Median age was 0.7 years (range 0.04-2). Stem cell source was bone marrow (BM) in 492 (73.9%), unrelated umbilical cord blood (UCB) in 161 (24.2%) followed by peripheral blood stem cell (PBSC) in 13 (2%) patients. Matched siblings were the most common donors (n = 356, 53.5%), followed by unrelated (n = 161, 24.2%) with haploidentical family member donors in 29 (4.4%) transplants. Disease groups were categorized as benign hematology (Thalassemia, Fanconi, Aplastic anemia etc.), benign neoplasm (Langerhans cell histiocytosis, Hemophagocytic Lymphohistiocytosis etc.), non-neoplasms (metabolic disorders, immunodeficiency disorders etc.) and Leukemia/lymphomas (myeloid and lymphoid malignancies etc.). RESULTS Cumulative incidence of acute GvHD (I-IV) was 31.5% (n = 210) and grade III-IV GvHD was 8.7% (n = 58). At median follow-up of 115.1 months, the cumulative probability of overall survival (OS) at 5 years was 70.0% ± 1.9%. Our mortality rate was 31.2% (n = 193). The five-year OS was significantly better in patients transplanted for benign hematological disorders (P = .001). Patients transplanted using BM/PBSC as source of stem cells fared significantly better compared to those in which CB was used (P<.001). Post-transplant graft failure remains the leading cause requiring further transplants in this age group. In conclusion, the cumulative probability of OS at 5 years was about 70.0% for all with an OS of 61% in our haploidentical recipients. CONCLUSION Analyzing our institutional data over time has enabled us to develop tentative strategies to minimize transplant related toxicities in very young children who are candidates for allo-HCT.
Collapse
Affiliation(s)
- Saadiya Khan
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Khawar Siddiqui
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Hasan ElSolh
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Abdullah AlJefri
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Ali AlAhmari
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Ibrahim Ghemlas
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Hawazen AlSaedi
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Awatif AlEnazi
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Amal AlSeraihi
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Mouhab Ayas
- Department of Pediatric Hematology/Oncology, MBC 53, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| |
Collapse
|
41
|
Ozturk E, Catak MC, Kiykim A, Baser D, Bilgic Eltan S, Yalcin K, Kasap N, Nain E, Bulutoglu A, Akgun G, Can Y, Sefer AP, Babayeva R, Caki-Kilic S, Tezcan Karasu G, Yesilipek A, Ozen A, Karakoc-Aydiner E, Baris S. Clinical and Laboratory Factors Affecting the Prognosis of Severe Combined Immunodeficiency. J Clin Immunol 2022; 42:1036-1050. [PMID: 35451701 DOI: 10.1007/s10875-022-01262-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Severe combined immunodeficiency (SCID) is one of the most severe forms of inborn errors of immunity characterized by absence or loss of function in T cells. The long-term outcomes of all forms of SCID have been evaluated in a limited number of studies. We aimed to evaluate the pre- and post-transplant manifestations of SCID patients and determine the factors affecting the survival of patients. METHODS We included 54 SCID patients (classical SCID, Omenn syndrome, atypical SCID (AS)) in this study. We evaluated the clinical presentation, infections, and outcome of hematopoietic stem cell transplantation (HSCT). Lymphocyte subsets and T-cell receptor (TCR) repertoire were analyzed by flow cytometry. RESULTS The median age at diagnosis was 5 (range: 3-24) months and follow-up time was 25 (range: 5-61) months. Symptom onset and diagnostic ages were significantly higher in AS compared to others (p = 0.001; p < 0.001). The most common SCID phenotype was T-B-NK + , and mutations in recombination-activating genes (RAG1/2) were the prominent genetic defect among patients. The overall survival (OS) rate was 83.3% after HSCT, higher than in non-transplanted patients (p = 0.001). Peripheral blood stem cell sources and genotypes other than RAG had a significant favorable impact on CD4+ T cells immune reconstitution after transplantation (p = 0.044, p = 0.035; respectively). Gender matching transplantations from human leukocyte antigen (HLA)-identical and non-identical donors and using peripheral blood stem cell source yielded higher B-cell reconstitution (p = 0.002, p = 0.028; respectively). Furthermore, receiving a conditioning regimen provided better B-cell reconstitution and chimerism (p = 0.003, p = 0.001). Post-transplant TCR diversity was sufficient in the patients and showed an equal distribution pattern as healthy controls. The OS rate was lower in patients who underwent transplant with active infection or received stem cells from mismatched donors (p = 0.030, p = 0.015; respectively). CONCLUSION This study identifies diagnostic and therapeutic approaches predictive of favorable outcomes for patients with SCID.
Collapse
Affiliation(s)
- Elif Ozturk
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Mehmet Cihangir Catak
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Ayca Kiykim
- Faculty of Medicine, Pediatric Allergy and Immunology, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Dilek Baser
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Sevgi Bilgic Eltan
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Koray Yalcin
- Pediatric Bone Marrow Transplantation Unit, Medical Park Goztepe Hospital, Istanbul, Turkey
| | - Nurhan Kasap
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Ercan Nain
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Alper Bulutoglu
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Gamze Akgun
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Yasemin Can
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Asena Pinar Sefer
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Royala Babayeva
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Suar Caki-Kilic
- Division of Pediatric Hematology, Umraniye Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Gulsun Tezcan Karasu
- Pediatric Bone Marrow Transplantation Unit, Medical Park Goztepe Hospital, Istanbul, Turkey
| | - Akif Yesilipek
- Pediatric Bone Marrow Transplantation Unit, Medical Park Goztepe Hospital, Istanbul, Turkey
| | - Ahmet Ozen
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey.,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
| | - Safa Baris
- Faculty of Medicine, Division of Pediatric Allergy and Immunology, Marmara University, Fevzi Çakmak Mah. No: 41, Pendik, Istanbul, Turkey. .,Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey.
| |
Collapse
|
42
|
Conditioning regimens for inborn errors of immunity: current perspectives and future strategies. Int J Hematol 2022; 116:7-15. [PMID: 35675025 DOI: 10.1007/s12185-022-03389-7] [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: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
Abstract
Inborn errors of immunity (IEI) are caused by germline genetic mutations, resulting in defects of innate or acquired immunity. Hematopoietic cell transplantation (HCT) is indicated for curative therapy especially in patients with IEI who develop fatal opportunistic infections or severe manifestations of immune dysregulation. The first successful HCT for severe combined immunodeficiency (SCID) was reported in 1968. Since then, the indications for HCT have expanded from SCID to various non-SCID IEI. In general, HCT for IEI differs from that for other hematological malignancies in that the goal is not to eradicate certain immune cells but to achieve immune reconstitution. European Society for Blood and Marrow Transplantation/European Society for Immunodeficiencies guidelines recommend reduced-intensity conditioning to avoid treatment-related toxicity, and the optimal conditioning regimen should be considered for each IEI. We review conditioning regimens for some representative IEI disorders in Japanese and worldwide cohort studies, and future strategies for treating IEI.
Collapse
|
43
|
Barreiros LA, Sousa JL, Geier C, Leiss-Piller A, Kanegae MPP, França TT, Boisson B, Lima AM, Costa-Carvalho BT, Aranda CS, de Moraes-Pinto MI, Segundo GRS, Ferreira JFS, Tavares FS, Guimarães FATDM, Toledo EC, da Matta Ain AC, Moreira IF, Soldatelli G, Grumach AS, de Barros Dorna M, Weber CW, Di Gesu RSW, Dantas VM, Fernandes FR, Torgerson TR, Ochs HD, Bustamante J, Walter JE, Condino-Neto A. SCID and Other Inborn Errors of Immunity with Low TRECs - the Brazilian Experience. J Clin Immunol 2022; 42:1171-1192. [PMID: 35503492 DOI: 10.1007/s10875-022-01275-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/17/2022] [Indexed: 11/26/2022]
Abstract
Severe combined immunodeficiency, SCID, is a pediatric emergency that represents the most critical group of inborn errors of immunity (IEI). Affected infants present with early onset life-threatening infections due to absent or non-functional T cells. Without early diagnosis and curative treatment, most die in early infancy. As most affected infants appear healthy at birth, newborn screening (NBS) is essential to identify and treat patients before the onset of symptoms. Here, we report 47 Brazilian patients investigated between 2009 and 2020 for SCID due to either a positive family history and/or clinical impression and low TRECs. Based on clinical presentation, laboratory finding, and genetic information, 24 patients were diagnosed as typical SCID, 14 as leaky SCID, and 6 as Omenn syndrome; 2 patients had non-SCID IEI, and 1 remained undefined. Disease onset median age was 2 months, but at the time of diagnosis and treatment, median ages were 6.5 and 11.5 months, respectively, revealing considerable delay which affected negatively treatment success. While overall survival was 51.1%, only 66.7% (30/45) lived long enough to undergo hematopoietic stem-cell transplantation, which was successful in 70% of cases. Forty-three of 47 (91.5%) patients underwent genetic testing, with a 65.1% success rate. Even though our patients did not come from the NBS programs, the diagnosis of SCID improved in Brazil during the pilot programs, likely due to improved medical education. However, we estimate that at least 80% of SCID cases are still missed. NBS-SCID started to be universally implemented in the city of São Paulo in May 2021, and it is our hope that other cities will follow, leading to early diagnosis and higher survival of SCID patients in Brazil.
Collapse
Affiliation(s)
- Lucila Akune Barreiros
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Jusley Lira Sousa
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | | | | | - Marilia Pylles Patto Kanegae
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Tábata Takahashi França
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | | | | | | | | | | | | | | | | | | | - Ana Carolina da Matta Ain
- Departamento de Pediatria E Imunologia, Hospital Universitário de Taubaté, Universidade de Taubaté, Taubate, SP, Brazil
| | | | - Gustavo Soldatelli
- Hospital das Clínicas, Universidade Federal de Santa Caratina, Florianopolis, SC, Brazil
| | | | - Mayra de Barros Dorna
- Instituto da Criança, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil
| | | | | | - Vera Maria Dantas
- Departamento de Pediatria, Universidade Federal Do Rio Grande Do Norte, Natal, RN, Brazil
| | | | | | - Hans Dietrich Ochs
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, Seattle, USA
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Jolan Eszter Walter
- University of South Florida at Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA
- Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Antonio Condino-Neto
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil.
- Immunogenic Laboratories Inc, Sao Paulo, SP, Brazil.
| |
Collapse
|
44
|
Pastorczak A, Attarbaschi A, Bomken S, Borkhardt A, van der Werff ten Bosch J, Elitzur S, Gennery AR, Hlavackova E, Kerekes A, Křenová Z, Mlynarski W, Szczepanski T, Wassenberg T, Loeffen J. Consensus Recommendations for the Clinical Management of Hematological Malignancies in Patients with DNA Double Stranded Break Disorders. Cancers (Basel) 2022; 14:2000. [PMID: 35454905 PMCID: PMC9029535 DOI: 10.3390/cancers14082000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/03/2022] Open
Abstract
Patients with double stranded DNA repair disorders (DNARDs) (Ataxia Telangiectasia (AT) and Nijmegen Breakage syndrome (NBS)) are at a very high risk for developing hematological malignancies in the first two decades of life. The most common neoplasms are T-cell lymphoblastic malignancies (T-cell ALL and T-cell LBL) and diffuse large B cell lymphoma (DLBCL). Treatment of these patients is challenging due to severe complications of the repair disorder itself (e.g., congenital defects, progressive movement disorders, immunological disturbances and progressive lung disease) and excessive toxicity resulting from chemotherapeutic treatment. Frequent complications during treatment for malignancies are deterioration of pre-existing lung disease, neurological complications, severe mucositis, life threating infections and feeding difficulties leading to significant malnutrition. These complications make modifications to commonly used treatment protocols necessary in almost all patients. Considering the rarity of DNARDs it is difficult for individual physicians to obtain sufficient experience in treating these vulnerable patients. Therefore, a team of experts assembled all available knowledge and translated this information into best available evidence-based treatment recommendations.
Collapse
Affiliation(s)
- Agata Pastorczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, 91-738 Lodz, Poland;
| | - Andishe Attarbaschi
- Department of Pediatrics, Pediatric Hematology and Oncology, St. Anna Children’s Hospital, Medical University of Vienna, 1090 Vienna, Austria;
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Simon Bomken
- Great North Children’s Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK; (S.B.); (A.R.G.)
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children’s Hospital, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany;
| | - Jutte van der Werff ten Bosch
- Department of Pediatric Hematology, Oncology and Immunology, University Hospital Brussels, 1090 Jette Brussels, Belgium;
| | - Sarah Elitzur
- Pediatric Hematology-Oncology, Schneider Children’s Medical Center, Petach Tikvah 4920235, Israel;
| | - Andrew R. Gennery
- Great North Children’s Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK; (S.B.); (A.R.G.)
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Eva Hlavackova
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, 662 63 Brno, Czech Republic; (E.H.); (Z.K.)
- Department of Clinical Immunology and Allergology, St. Anne’s University Hospital in Brno, Faculty of Medicine, Masaryk University, 662 63 Brno, Czech Republic;
| | - Arpád Kerekes
- Department of Clinical Immunology and Allergology, St. Anne’s University Hospital in Brno, Faculty of Medicine, Masaryk University, 662 63 Brno, Czech Republic;
| | - Zdenka Křenová
- Department of Pediatric Oncology, University Hospital and Faculty of Medicine, Masaryk University, 662 63 Brno, Czech Republic; (E.H.); (Z.K.)
| | - Wojciech Mlynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, 91-738 Lodz, Poland;
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Medical University of Silesia (SUM), 41-800 Zabrze, Poland;
| | - Tessa Wassenberg
- Department of Neurology and Child Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Jan Loeffen
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
| |
Collapse
|
45
|
Giardino S, Bagnasco F, Falco M, Miano M, Pierri F, Risso M, Terranova P, Martino DD, Massaccesi E, Ricci M, Chianucci B, Dell'Orso G, Sabatini F, Podestà M, Lanino E, Faraci M. HAPLOIDENTICAL STEM CELL TRANSPLANTATION AFTER TCR αβ +AND CD19 + CELLS DEPLETION IN CHILDREN WITH CONGENITAL NON-MALIGNANT DISEASE. Transplant Cell Ther 2022; 28:394.e1-394.e9. [PMID: 35405368 DOI: 10.1016/j.jtct.2022.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND . Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) represents a valuable alternative for children with non-malignant disease and ex-vivo negative selection of TCR αβ+-cells is an emerging graft manipulation option that carries several potential advantages in terms of reduced risk of Graft versus Host Disease (GvHD) and improved immune reconstitution. METHODS . We reported all consecutive patients with a diagnosis of non-malignant disease who received a TCR-αβ+ and CD19+depleted haplo-HSCT at "IRCCS Istituto Giannina Gaslini" from 2013 to 2019; the conditioning regimen was myeloablative or non-myeloablative, depending on underlying disease; all patients received anti-thymocyte globulin and rituximab. No post-transplant GvHD prophylaxis was given in presence of a TCR-αβ+ cell-dose in the graft lower than the threshold of 1 × 105/kg of the recipient's weight. RESULTS . Among 20 HSCTs, engraftment occurred in 17 (85%) after a median of 14 and 12 days from graft infusion for neutrophils and platelets respectively. Primary graft failure was diagnosed in 3 (15%) patients, two (10%) experienced secondary rejection; all of these underwent a second HSCT. The cumulative incidence of a-GvHD and c-GvHD was 15% (2 grade 1, 1 grade 4) at 90 days and 5% (1 grade 1) at 7 months, respectively. Cytomegalovirus reactivation requiring pre-emptive treatment was observed in 9 patients (45%). One patient developed a JC virus-related progressive multifocal leukoencephalopathy, successfully managed with donor-derived virus-specific T-cell infusions. A complete immunological recovery was reached in most patients within 6 months. After a median follow-up of 4 years, 18 patients are alive, with a cumulative survival probability of 90%. CONCLUSION . Haplo-HSCT after ex-vivo TCR-αβ+/CD19+ negative selection may be considered a good option for children with non-malignant diseases since it ensures a high engraftment rate with an acceptable risk of graft failure, very low incidence of significant GvHD, and good immune reconstitution with low frequency of severe virus-related disease. However, the control of viral infection/reactivation should be kept high in order to promptly provide pre-emptive treatments and approaches of antiviral adoptive immunotherapy.
Collapse
Affiliation(s)
- Stefano Giardino
- Hematopoietic stem cell transplantation Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy.
| | - Francesca Bagnasco
- Epidemiology and Biostatistics Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Michela Falco
- Laboratory of Clinical and Experimental Immunology, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Maurizio Miano
- Hematology Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Filomena Pierri
- Hematopoietic stem cell transplantation Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Risso
- Immunohematology and Transfusional Department, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Paola Terranova
- Laboratory of Hematology, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | | | | | - Margherita Ricci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Liguria, Italy
| | - Benedetta Chianucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Liguria, Italy
| | - Gianluca Dell'Orso
- Hematopoietic stem cell transplantation Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Federica Sabatini
- Stem Cells and Cell Therapies Laboratory, IRCSS IstitutoGianninaGaslini, Genoa, Italy
| | - Marina Podestà
- Stem Cells and Cell Therapies Laboratory, IRCSS IstitutoGianninaGaslini, Genoa, Italy
| | - Edoardo Lanino
- Hematopoietic stem cell transplantation Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Maura Faraci
- Hematopoietic stem cell transplantation Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
46
|
Hardin O, Lokhnygina Y, Buckley RH. Long-Term Clinical Outcomes of Severe Combined Immunodeficiency Patients Given Nonablative Marrow Transplants. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1077-1083. [PMID: 34942385 DOI: 10.1016/j.jaip.2021.11.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/08/2021] [Accepted: 11/26/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Severe combined immunodeficiency (SCID) is a syndrome characterized by profound T-cell deficiency that is universally fatal in infancy unless immune reconstitution is achieved by hematopoietic stem cell transplantation, gene therapy, or enzyme replacement. Published long-term clinical follow-up is limited for transplanted patients with SCID. OBJECTIVE To characterize the long-term outcomes of patients with SCID treated at a single center. METHODS We examined the clinical outcomes of 177 successive SCID infants given allogeneic bone marrow over 38 years without pretransplant chemotherapy or post-transplant graft-versus-host disease prophylaxis. A total of 90% received T-cell-depleted haploidentical parental marrow. Clinical status was assessed by questionnaires delivered by mail or electronically. Molecular type of SCID, demographics, and type, date and age of transplant were obtained from a database. RESULTS Eighty-eight questionnaires were completed for survivors ranging in age from 2 to 38 years. Survival remained higher in those transplanted before 3.5 months of age. Half of the cohort remained on immunoglobulin replacement. Health conditions reported included rashes, anxiety, depression, warts, and mouth ulcers. Most reported that these were transient, self-resolving issues. Attention-deficit/hyperactivity disorder, warts, and learning disabilities were reportedly more prevalent than in the general population. Most reported having no active concerns about their health. We found substantial scholastic achievement, with half of adult patients reporting college attendance. Most patients had a healthy body mass index. CONCLUSIONS Overall, our findings are consistent with those in the last update in 2009 in this population. Age at transplant remains a key variable in survival.
Collapse
Affiliation(s)
- Olga Hardin
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Yuliya Lokhnygina
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - Rebecca H Buckley
- Department of Pediatrics, Duke University Medical Center, Durham, NC.
| |
Collapse
|
47
|
Delmonte OM, Castagnoli R, Yu J, Dvorak CC, Cowan MJ, Dávila Saldaña BJ, De Ravin SS, Mamcarz E, Chang CK, Daley SR, Griffith LM, Notarangelo LD, Puck JM. Poor T-cell receptor β repertoire diversity early posttransplant for severe combined immunodeficiency predicts failure of immune reconstitution. J Allergy Clin Immunol 2022; 149:1113-1119. [PMID: 34384841 PMCID: PMC9132846 DOI: 10.1016/j.jaci.2021.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Development of a diverse T-cell receptor β (TRB) repertoire is associated with immune recovery following hematopoietic cell transplantation (HCT) for severe combined immunodeficiency (SCID). High-throughput sequencing of the TRB repertoire allows evaluation of clonotype dynamics during immune reconstitution. OBJECTIVES We investigated whether longitudinal analysis of the TRB repertoire would accurately describe T-cell receptor diversity and illustrate the quality of T-cell reconstitution following HCT or gene therapy for SCID. METHODS We used high-throughput sequencing to study composition and diversity of the TRB repertoire in 27 infants with SCID at 3, 6, and 12 months and yearly posttreatment(s). Total RNA from peripheral blood was used as template to amplify TRB rearrangements. RESULTS TRB sequence analysis showed poor diversity at 3 months, followed by significant improvement by 6 months after cellular therapies. Kinetics of development of TRB diversity were similar in patients with a range of underlying gene defects. However, in patients with RAG and DCLRE1C defects, HCT with no conditioning or immune suppression only resulted in lower diversity than did HCT with conditioning. HCT from a matched donor correlated with higher diversity than did HCT from a mismatched donor. Naive CD4+ T-cell count at 6 months post-HCT correlated with higher TRB diversity. A Shannon index of diversity of 5.2 or lower 3 months after HCT predicted a need for a second intervention. CONCLUSIONS TRB repertoire after hematopoietic cell therapies for SCID provides a quantitative and qualitative measure of diversity of T-cell reconstitution and permits early identification of patients who may require a second intervention.
Collapse
Affiliation(s)
- Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Md
| | - Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Md
| | - Jason Yu
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, Calif; Smith Cardiovascular Research Institute, San Francisco, Calif; 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, San Francisco, Calif; UCSF Benioff Children's Hospital, San Francisco, Calif
| | - 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, Calif; Smith Cardiovascular Research Institute, San Francisco, Calif; UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Blachy J Dávila Saldaña
- Division of Blood and Marrow Transplantation, Children's National Medical Center, Washington, DC
| | - Suk See De Ravin
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Md
| | - Ewelina Mamcarz
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tenn
| | - Catherine K Chang
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, Calif; UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Stephen R Daley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Linda M Griffith
- Division of Allergy Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Md.
| | - Jennifer M Puck
- Division of Allergy, Immunology and Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, Calif; Smith Cardiovascular Research Institute, San Francisco, Calif; UCSF Benioff Children's Hospital, San Francisco, Calif.
| |
Collapse
|
48
|
Ochoa-Fernández B, Galán-Gómez V, Mestre C, González-Vicent M, Pascual A, Alonso L, Regueiro A, Plaza M, Pérez Hurtado JM, Benito A, Fuster JL, Bueno D, Mozo Y, Vicario JL, Balas A, Sisinni L, Díaz de Heredia C, Pérez-Martínez A. Haploidentical hematopoietic stem cell transplantation in pediatric and adolescent patients: A study of the Spanish hematopoietic stem cell transplantation group (GETH). Med Clin (Barc) 2022; 159:411-419. [PMID: 35241283 DOI: 10.1016/j.medcli.2022.01.009] [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: 10/03/2021] [Revised: 12/28/2021] [Accepted: 01/01/2022] [Indexed: 10/31/2022]
Abstract
INTRODUCTION The main advantages of haploidentical hematopoietic stem cell transplantation (haplo-HSCT) are the immediate availability of donors, the possibility of developing cell therapy approaches with different novel transplant platforms, and the procedure's cost savings. METHODOLOGY We retrospectively analyzed the pediatric haplo-HSCT activity of the Spanish hematopoietic stem-cell transplantation group (GETH) between 1999 and 2016, aiming to study clinical characteristics and outcomes by describing patient groups with non-malignant disease (NMD) or malignant disease (MD) and the impact of 2 different periods (1999-2009 and 2010-2016) on long-term outcomes. RESULTS Twelve centers performed 232 haplo-HSCTs in 227 children, representing 10% of all pediatric allogeneic HSCT activity in Spain from 1999 to 2016, with a notable increase since 2013. Most haplo-HSCTs (86.7%) were performed in patients with MD; 95% received peripheral blood stem cells from donors, and 78.9% received ex vivo T-cell depleted grafts. Non-manipulated grafts using post-transplantation cyclophosphamide have been incorporated since 2012. We observed a higher percentage of graft failure in NMD versus MD (32% vs. 15.6%; p=0.029). Relapse and transplant-related mortality were the procedure's main limitations in MD and NMD, respectively. Five-year overall survival was 48.5% (SE 3.9), with no statistically significant difference when comparing the MD and NMD cohorts. Patients who received previously a HSCT the overall survival was significantly decreased. We observed no survival improvement over time. CONCLUSIONS Although haplo-HSCT is an increasingly employed treatment option, our patients' results need improvement. We need to develop reference centers, especially for NMD whose rarity makes it difficult to gain experience.
Collapse
Affiliation(s)
| | | | - Carmen Mestre
- La Paz Health Research Institute, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | | | | | | | | | - Mercedes Plaza
- Virgen de la Arrixaca University Clinical Hospital; Biomedical Research Institute of Murcia (IMIB), Spain
| | | | | | - José Luis Fuster
- Virgen de la Arrixaca University Clinical Hospital; Biomedical Research Institute of Murcia (IMIB), Spain
| | | | | | | | - Antonio Balas
- Transfusion Center of the Community of Madrid, Spain
| | | | | | - Antonio Pérez-Martínez
- La Paz University Hospital, Madrid, Spain; La Paz Health Research Institute, IdiPAZ, La Paz University Hospital, Madrid, Spain; Faculty of Medicine, Autonomous University of Madrid, Spain.
| |
Collapse
|
49
|
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: 3] [Impact Index Per Article: 1.0] [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
|
50
|
Hosahalli Vasanna S, Pereda MA, Dalal J. Clinical Features, Cancer Biology, Transplant Approach and Other Integrated Management Strategies for Wiskott-Aldrich Syndrome. J Multidiscip Healthc 2022; 14:3497-3512. [PMID: 34992377 PMCID: PMC8711845 DOI: 10.2147/jmdh.s295386] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/24/2021] [Indexed: 11/23/2022] Open
Abstract
Wiskott–Aldrich syndrome (WAS) is a rare X-linked recessive inborn error of immunity (IEI) first described in 1937. Classic WAS is characterized by the triad of thrombocytopenia with small platelets, recurrent infections due to combined immunodeficiency, and eczema. Hematopoietic stem cell transplantation (HSCT) was the only curative option available for five decades, with excellent outcomes reported for matched sibling donors (MSD) and matched unrelated donors (MUD). More recently, alternative donor transplants such as umbilical cord blood (UCB) and haploidentical transplant have emerged as viable options due to improvements in better graft selection, cell dosing, and effective allograft manipulation measures. Gene therapy is another potential curative option with promising results, yet currently is offered only as part of a clinical trial.
Collapse
Affiliation(s)
- Smitha Hosahalli Vasanna
- Department of Pediatrics, Division of Pediatric Hematology Oncology, Rainbow Babies and Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - Maria A Pereda
- Department of Pediatrics, Division of Pediatric Hematology Oncology, Rainbow Babies and Children's Hospital, University Hospitals, Cleveland, OH, USA
| | - Jignesh Dalal
- Department of Pediatrics, Division of Pediatric Hematology Oncology, Rainbow Babies and Children's Hospital, University Hospitals, Cleveland, OH, USA
| |
Collapse
|