1
|
Bayegi SN, Hamidieh AA, Behfar M, Saghazadeh A, Bozorgmehr M, Karamlou Y, Shekarabi M, Tajik N, Delbandi AA, Zavareh FT, Delavari S, Rezaei N. T helper 17 and regulatory T-cell profile and graft-versus-host disease after allogeneic hematopoietic stem cell transplantation in pediatric patients with beta-thalassemia. Transpl Immunol 2023; 77:101803. [PMID: 36842567 DOI: 10.1016/j.trim.2023.101803] [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: 11/30/2022] [Revised: 01/29/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an effective treatment option for hereditary hemoglobin disorders, such as beta-thalassemia; However, this procedure is not without constraints, mainly engendering complications such as acute graft-versus-host disease (aGvHD), chronic GvHD (cGvHD), and susceptibility to infections. The clinical outcomes of allo-HSCT are highly dependant on the quality and quantity of T-cell subsets reconstitution. Following the allo-HSCT of six pediatric patients afflicted with beta-thalassemia, their mononuclear cells were isolated, and then cultured with a combination of phorbol myristate acetate (PMA)/ionomycin and Brefeldin A. The content of CD4 T-cell subsets, including T helper 17 (Th17) cells and regulatory T cells (Tregs), were determined by specific conjugated-monoclonal antibodies three and six months post-HSCT. An increased frequency of total CD4 T-cells, Tregs and Th17 cells was observed at day 90 and 180 after allo-HSCT, albeit the numbers were still lower than that of our healthy controls. In patients who developed cGvHD, a lower Th17/Treg ratio was observed, owing it to a decreased proportion of Th17 cells. In conclusion, creating balance between Th17 and Treg subsets may prevent acute and chronic GvHD in patients after allo-HSCT.
Collapse
Affiliation(s)
- Shideh Namazi Bayegi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amene Saghazadeh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmood Bozorgmehr
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Yalda Karamlou
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shekarabi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nader Tajik
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Tofighi Zavareh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Delavari
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
2
|
Sperl D, Lang P, Benesch M, Bainschab A, Urban C, Wilfing R, Feuchtinger T, Döring M, Seitz C, Strenger V, Lackner H, Seidel MG, Perwein T, Handgretinger R, Sipurzynski S, Rosskopf K, Schwinger W. Immunological recovery following HLA-matched CD3+ TCR αß+/CD19+ depleted hematopoietic stem cell transplantation in children. Pediatr Transplant 2022; 26:e14285. [PMID: 35441401 DOI: 10.1111/petr.14285] [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: 07/30/2021] [Revised: 02/08/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative option for children with both malignant and nonmalignant diseases. T-cell depletion techniques may result in reduced transplant-related mortality compared with unmanipulated grafts due to a lower incidence of GvHD. METHODS Immune recovery and outcome were analyzed in a cohort of 23 patients with malignant and nonmalignant diseases who received CD3+TCRαβ+ T- and B-cell-depleted allografts from matched donors after reduced-intensity or myeloablative conditioning. The median number of CD34+, CD3+TCRαβ+, and CD19+B-cells infused was 12.7 × 106 /kg, 16.8 × 103 /kg, and 96 × 103 /kg bodyweight. RESULTS With a median follow-up of 36 (range 1-73) months, overall survival and disease-free survival at 3 years were 65.2% and 60.8%. Eight patients died, six due to the underlying disease and two of extended visceral cGvHD. Immune reconstitution, disease-free, and overall survivals were similar compared with a historical cohort of 23 patients transplanted with matched unmanipulated bone marrow. A significant lower rate of higher grade (III-IV) aGvHD was observed in the manipulated HSCT group (8.7% vs. 26%; p = 0.001), whereas the incidence of cGvHD was equal. CONCLUSIONS Our data suggest that this graft manipulation strategy could be a safe and effective alternative to conventional HSCT techniques in matched donors.
Collapse
Affiliation(s)
- Daniela Sperl
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Peter Lang
- Children's University Hospital University of Tuebingen, Tuebingen, Germany
| | - Martin Benesch
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Antonia Bainschab
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Christian Urban
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Roland Wilfing
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Tobias Feuchtinger
- Department of Pediatric Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Dr. von Hauner University Children's Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Michaela Döring
- Children's University Hospital University of Tuebingen, Tuebingen, Germany
| | - Christian Seitz
- Children's University Hospital University of Tuebingen, Tuebingen, Germany
| | - Volker Strenger
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Herwig Lackner
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Markus G Seidel
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | - Thomas Perwein
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | | | - Sabine Sipurzynski
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Konrad Rosskopf
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Wolfgang Schwinger
- Division of Pediatric Hematology/Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
3
|
Jarduli-Maciel LR, de Azevedo JTC, Clave E, Costa TCDM, Arruda LCM, Fournier I, Palma PVB, Lima KC, Elias JB, Stracieri ABP, Pieroni F, Cunha R, Darrigo-Júnior LG, Grecco CES, Covas DT, Silva-Pinto AC, De Santis GC, Simões BP, Oliveira MC, Toubert A, Malmegrim KCR. Allogeneic haematopoietic stem cell transplantation resets T- and B-cell compartments in sickle cell disease patients. Clin Transl Immunology 2022; 11:e1389. [PMID: 35474905 PMCID: PMC9035210 DOI: 10.1002/cti2.1389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/12/2022] Open
Abstract
Objectives Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is the only currently available curative treatment for sickle cell disease (SCD). Here, we comprehensively evaluated the reconstitution of T- and B-cell compartments in 29 SCD patients treated with allo-HSCT and how it correlated with the development of acute graft-versus-host disease (aGvHD). Methods T-cell neogenesis was assessed by quantification of signal-joint and β-chain TCR excision circles. B-cell neogenesis was evaluated by quantification of signal-joint and coding-joint K-chain recombination excision circles. T- and B-cell peripheral subset numbers were assessed by flow cytometry. Results Before allo-HSCT (baseline), T-cell neogenesis was normal in SCD patients compared with age-, gender- and ethnicity-matched healthy controls. Following allo-HSCT, T-cell neogenesis declined but was fully restored to healthy control levels at one year post-transplantation. Peripheral T-cell subset counts were fully restored only at 24 months post-transplantation. Occurrence of acute graft-versus-host disease (aGvHD) transiently affected T- and B-cell neogenesis and overall reconstitution of T- and B-cell peripheral subsets. B-cell neogenesis was significantly higher in SCD patients at baseline than in healthy controls, remaining high throughout the follow-up after allo-HSCT. Notably, after transplantation SCD patients showed increased frequencies of IL-10-producing B-regulatory cells and IgM+ memory B-cell subsets compared with baseline levels and with healthy controls. Conclusion Our findings revealed that the T- and B-cell compartments were normally reconstituted in SCD patients after allo-HSCT. In addition, the increase of IL-10-producing B-regulatory cells may contribute to improve immune regulation and homeostasis after transplantation.
Collapse
Affiliation(s)
- Luciana Ribeiro Jarduli-Maciel
- Graduate Program in Biosciences Applied to Pharmacy School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | - Júlia Teixeira Cottas de Azevedo
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Graduate Program in Basic and Applied Immunology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | | | - Thalita Cristina de Mello Costa
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | | | - Isabelle Fournier
- Laboratoire d'Immunologie et d'Histocompatibilité Hôpital Saint-Louis AP-HP Paris France
| | - Patrícia Vianna Bonini Palma
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | - Keli Cristina Lima
- Graduate Program in Biosciences Applied to Pharmacy School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | | | | | - Fabiano Pieroni
- Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Renato Cunha
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | | | | | - Dimas Tadeu Covas
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Ana Cristina Silva-Pinto
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Gil Cunha De Santis
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Belinda Pinto Simões
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Antoine Toubert
- Université de Paris INSERM UMR 1160 IRSL Paris France.,Laboratoire d'Immunologie et d'Histocompatibilité Hôpital Saint-Louis AP-HP Paris France
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Department of Clinical Analysis, Toxicology and Food Sciences School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| |
Collapse
|
4
|
Vellaichamy Swaminathan V, Ravichandran N, Ramanan KM, Meena SK, Varla H, Ramakrishnan B, Jayakumar I, Uppuluri R, Raj R. Augmented immunosuppression and PTCY-based haploidentical hematopoietic stem cell transplantation for thalassemia major. Pediatr Transplant 2021; 25:e13893. [PMID: 33111490 DOI: 10.1111/petr.13893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 11/25/2022]
Abstract
Alternate donor HSCT for thalassemia major from a matched unrelated donor or haploidentical family donor is a feasible therapeutic option in children with no matched family donor. Aggressive pretransplant immunosuppression, reduced toxicity conditioning, and PTCY result in excellent thalassemia-free survival. We describe here our experience in this cohort. We performed a retrospective analysis of the data on children who underwent a haploidentical HSCT for thalassemia major with PTCY at our center from August 2017 to August 2019. All children received pretransplant immune suppression for 6 weeks with fludarabine and dexamethasone, hypertransfusion and chelation with intravenous desferrioxamine. Conditioning included thiotepa, fludarabine, rabbit ATG, and cyclophosphamide, and GvHD prophylaxis included PTCY with tacrolimus. Twenty children were included and nineteen children engrafted. Acute hypertension occurred in five children, bacterial infection in eight children and viral respiratory infection in three children. Three children suffered from graft rejection. Reactivation of viruses namely CMV, adenovirus, and BK virus was seen in 60% of children. Grades 1-2 acute GvHD of the skin in four children (20%) and limited chronic GvHD of the skin in four children (20%). Immune cytopenia was documented in three children (15%). Haploidentical HSCT offers a therapeutic option for children with thalassemia major with no suitably matched family or unrelated donors. Our reduced toxicity regimen with PTCY offers a DFS of 75% and OS of 95% with low transplant-related mortality of 5%.
Collapse
Affiliation(s)
| | - Nikila Ravichandran
- Department of Paediatric Hematology, Oncology and Blood and Marrow Transplantation, Apollo Cancer Institutes, Chennai, India
| | - Kesavan Melarcode Ramanan
- Department of Paediatric Hematology, Oncology and Blood and Marrow Transplantation, Apollo Cancer Institutes, Chennai, India
| | - Satish Kumar Meena
- Department of Paediatric Hematology, Oncology and Blood and Marrow Transplantation, Apollo Cancer Institutes, Chennai, India
| | - Harika Varla
- Department of Paediatric Hematology, Oncology and Blood and Marrow Transplantation, Apollo Cancer Institutes, Chennai, India
| | | | - Indra Jayakumar
- Department of Pediatric Intensive Care, Apollo Cancer Institutes, Chennai, India
| | - Ramya Uppuluri
- Department of Paediatric Hematology, Oncology and Blood and Marrow Transplantation, Apollo Cancer Institutes, Chennai, India
| | - Revathi Raj
- Department of Paediatric Hematology, Oncology and Blood and Marrow Transplantation, Apollo Cancer Institutes, Chennai, India
| |
Collapse
|
5
|
Lertthammakiat S, Sitthirat P, Anurathapan U, Songdej D, Pakakasama S, Chuansumrit A, Putawornsub N, Sirasittikarn S, Wantanawijarn S, Kadegasem P, Hongeng S, Sirachainan N. No differences in hemostatic and endothelial activations between haploidentical and matched-donor hematopoietic stem cell transplantation in thalassemia disease. Thromb J 2020; 18:21. [PMID: 33327955 PMCID: PMC7739460 DOI: 10.1186/s12959-020-00232-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/07/2020] [Indexed: 11/25/2022] Open
Abstract
Hemostatic changes and endothelial activations have been recognized in β-thalassemic patients after matched-donor hematopoietic stem cell transplantation (HSCT) but there are limited studies for haploidentical HSCT. This report demonstrates that the levels of hemostatic and endothelial markers, including thrombin antithrombin complex, prothrombin fragment, D-dimer, von Willebrand factor antigen and thrombomodulin levels, were not significantly different between haploidentical and matched-donor HSCT patients.
Collapse
Affiliation(s)
- Surapong Lertthammakiat
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand.,Chakri Naruebodindr Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Peerasit Sitthirat
- Department of Biology, Mahidol Wittayanusorn School, Nakhon Pathom, Thailand
| | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand
| | - Duantida Songdej
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand
| | - Samart Pakakasama
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand
| | - Ampaiwan Chuansumrit
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand
| | | | | | | | - Praguywan Kadegasem
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand
| | - Nongnuch Sirachainan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi District, Bangkok, Thailand.
| |
Collapse
|
6
|
Hematopoietic stem cell transplantation for adult sickle cell disease in the era of universal donor availibility. Bone Marrow Transplant 2018; 53:1390-1400. [DOI: 10.1038/s41409-018-0193-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/13/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
|
7
|
Hematopoietic stem cell transplantation for homozygous β-thalassemia and β-thalassemia/hemoglobin E patients from haploidentical donors. Bone Marrow Transplant 2016; 51:813-8. [PMID: 26878659 DOI: 10.1038/bmt.2016.7] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 01/26/2023]
Abstract
Thalassemia-free survival after allogeneic stem cell transplantation (SCT) is about 80-90% with either matched-related or -unrelated donors. We explored the use of a mismatched-related ('haplo- ') donor. All patients received two courses of pretransplant immunosuppressive therapy (PTIS) with fludarabine (Flu) and dexamethasone (Dxm). After two courses of PTIS, a conditioning regimen of rabbit antithymocyte globulin, Flu and IV busulfan (Bu) was given followed by T-cell-replete peripheral blood progenitor cells. GvHD prophylaxis consisted of cyclophosphamide (Cy) on days SCT +3 and +4 (post-Cy), and on day SCT +5 tacrolimus or sirolimus was started together with a short course of mycophenolate mofetil. Thirty-one patients underwent haplo-SCT. Their median age was 10 years (range, 2-20 years). Twenty-nine patients engrafted with 100% donor chimerism. Two patients suffered primary graft failure. Median time to neutrophil engraftment was 14 days (range, 11-18 days). Five patients developed mild to moderate, reversible veno-occlusive disease, while nine patients developed acute GvHD grade II. Only five patients developed limited-chronic GvHD. Projected overall and event-free survival rates at 2 years are 95% and 94%, respectively. The median follow up time is 12 months (range, 7-33 months).
Collapse
|
8
|
Andreani M, Gianolini ME, Testi M, Battarra M, Tiziana G, Morrone A, Sodani P, Lucarelli G, Roncarolo MG, Gregori S. Mixed chimerism evolution is associated with T regulatory type 1 (Tr1) cells in a β-thalassemic patient after haploidentical haematopoietic stem cell transplantation. CHIMERISM 2015; 5:75-9. [PMID: 26650878 DOI: 10.1080/19381956.2015.1103423] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In a cohort of β-Thalassemia (β-Thal) transplanted with haploidentical-HSCT we identified one transplanted patient characterized by persistent mixed chimerism (PMC) for several months after HSCT. In this unique β-Thal patient we assessed the donor engraftment overtime after transplantation, the potential loss of the non-shared HLA haplotype, and the presence of CD49b(+)LAG-3(+) T regulatory type 1 (Tr1) cells, previously demonstrated to be associated with PMC after HLA-related HSCT for β-Thal. The majority of the patient's erythrocytes were of donor origin, whereas T cells were initially mostly derived from the recipient, no HLA loss, but an increased frequency of circulating Tr1 cells were observed. For the first time, we showed that when the proportion of residual donor cells decreases, the frequency of CD49b(+)LAG-3(+) Tr1 cells declines, reaching the levels present in healthy subjects. These findings confirm previous results obtained in transplant related settings for β-Thal, and supported the central role of Tr1 cells in promoting and maintaining PMC after allo-HSCT.
Collapse
Affiliation(s)
- Marco Andreani
- a Laboratory of Immunogenetics and Transplant Biology; IME Foundation at Polyclinic of Tor Vergata ; Rome , Italy
| | - Monica Emma Gianolini
- b San Raffaele Telethon Institute for Gene Therapy (TIGET); Division of Regenerative Medicine; Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute ; Milan , Italy
| | - Manuela Testi
- a Laboratory of Immunogenetics and Transplant Biology; IME Foundation at Polyclinic of Tor Vergata ; Rome , Italy
| | - MariaRosa Battarra
- a Laboratory of Immunogenetics and Transplant Biology; IME Foundation at Polyclinic of Tor Vergata ; Rome , Italy
| | - Galluccio Tiziana
- a Laboratory of Immunogenetics and Transplant Biology; IME Foundation at Polyclinic of Tor Vergata ; Rome , Italy
| | - Aldo Morrone
- a Laboratory of Immunogenetics and Transplant Biology; IME Foundation at Polyclinic of Tor Vergata ; Rome , Italy
| | - Pietro Sodani
- c International Center for Transplantation in Thalassemia and Sickle Cell Anemia-Mediterranean Institute of Hematology; Policlinic of the University of Rome Tor Vergata ; Rome , Italy
| | - Guido Lucarelli
- c International Center for Transplantation in Thalassemia and Sickle Cell Anemia-Mediterranean Institute of Hematology; Policlinic of the University of Rome Tor Vergata ; Rome , Italy
| | - Maria-Grazia Roncarolo
- b San Raffaele Telethon Institute for Gene Therapy (TIGET); Division of Regenerative Medicine; Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute ; Milan , Italy.,d Department of Pediatric Stem Cell Transplantation and Regenerative Medicine ; Stanford School of Medicine ; Stanford , CA USA
| | - Silvia Gregori
- b San Raffaele Telethon Institute for Gene Therapy (TIGET); Division of Regenerative Medicine; Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute ; Milan , Italy
| |
Collapse
|
9
|
Peripheral red blood cell split chimerism as a consequence of intramedullary selective apoptosis of recipient red blood cells in a case of sickle cell disease. Mediterr J Hematol Infect Dis 2014; 6:e2014066. [PMID: 25408852 PMCID: PMC4235471 DOI: 10.4084/mjhid.2014.066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/23/2014] [Indexed: 11/25/2022] Open
Abstract
Allogeneic cellular gene therapy through hematopoietic stem cell transplantation is the only radical cure for congenital hemoglobinopathies like thalassemia and sickle cell anemia. Persistent mixed hematopoietic chimerism (PMC) has been described in thalassemia and sickle cell anemia. Here, we describe the clinical course of a 6-year-old girl who had received bone marrow transplant for sickle cell anemia. After the transplant, the patient showed 36% donor hematopoietic stem cells in the bone marrow, whereas in the peripheral blood there was evidence of 80% circulating donor red blood cells (RBC). The analysis of apoptosis at the Bone Marrow level suggests that Fas might contribute to the cell death of host erythroid precursors. The increase in NK cells and the regulatory T cell population observed in this patient suggests that these cells might contribute to the condition of mixed chimerism.
Collapse
|
10
|
Isgrò A, Sodani P, Marziali M, Gaziev J, Fraboni D, Paciaroni K, Gallucci C, De Angelis G, Alfieri C, Ribersani M, Armiento D, Roveda A, Andreani M, Testi M, Lucarelli G. Reduction of intramedullary apoptosis after stem cell transplantation in black african variant of pediatric sickle cell anemia. Mediterr J Hematol Infect Dis 2014; 6:e2014054. [PMID: 25045462 PMCID: PMC4103501 DOI: 10.4084/mjhid.2014.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/04/2014] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND AND PURPOSE Allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative treatment for sickle cell anemia (SCA). We report our experience with transplantation in children with the Black African variant of SCA and the effects of transplant on erythroid compartment in bone marrow (BM). PATIENTS AND METHODS Twenty-seven consecutive patients who underwent BM transplantation from HLA-identical donors following a myeloablative conditioning regimen were included. Using both CD71 and FSC parameters, we obtained three erythroid populations: EryA-C. Ery A (CD71(high) FSC(high)) are basophilic; Ery B (CD71(high) FSC(low)) are late basophilic and polychromatic; and Ery C (CD71(low) FSC(low)) are orthochromatic erythroblasts and reticulocytes. To analyze the effect of transplantation on intramedullary apoptosis, we studied Fas (CD95+) and caspase-3 expression in erythroblast subpopulations. RESULTS All patients experienced sustained engraftment, and all surviving patients remained free of SCA-related events after transplantation. The erythroid population showed expansion in the BM at baseline. After transplant, levels decreased, especially of Ery C, in parallel to reduced Fas expression and an initial caspase 3 increase in erythroid population, similar to reported later steps of "normal" erythroid maturation. CONCLUSIONS The results suggest a good chance of cure for children with SCA, with an excellent survival rate. We also observed "normalization" of erythroid populations in parallel with a decreased intramedullary apoptosis rate, suggesting normal erythroid maturation in ex-SCA patients after HSCT.
Collapse
Affiliation(s)
- Antonella Isgrò
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Pietro Sodani
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Marco Marziali
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Javid Gaziev
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Daniela Fraboni
- Laboratory of Oncohematology, Department of Laboratory Medicine, Policlinic of the University of Roma Tor Vergata, Rome, Italy
| | - Katia Paciaroni
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Cristiano Gallucci
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Gioia De Angelis
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Cecilia Alfieri
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Michela Ribersani
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Daniele Armiento
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Andrea Roveda
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Marco Andreani
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Manuela Testi
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| | - Guido Lucarelli
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinic of the University of Roma Tor Vergata., Rome, Italy
| |
Collapse
|
11
|
Ding L, Zhu H, Yang Y, Wang ZD, Zheng XL, Yan HM, Dong L, Zhang HH, Han DM, Xue M, Liu J, Zhu L, Guo ZK, Wang HX. Functional mesenchymal stem cells remain present in bone marrow microenvironment of patients with leukemia post-allogeneic hematopoietic stem cell transplant. Leuk Lymphoma 2014; 55:1635-44. [PMID: 24180332 DOI: 10.3109/10428194.2013.858815] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mesenchymal stem cells (MSCs) and their progenies are important supporting cells in the bone marrow (BM) microenvironment. However, the function and kinetics of MSCs post-hematopoietic stem cell transplant (HSCT) remain unknown. In the present study, MSCs were cultured from a total of 76 BM samples from 15 patients receiving HSCT. Colony-forming unit fibroblasts in BM before pre-conditioning and 1, 3, 6 and 9 months post-HSCT were cultured and counted to quantify MSCs. Hematopoiesis-supporting activity of MSCs was observed with long-term culture of hematopoietic progenitors. An inhibitory effect of MSCs on in vitro lymphocyte proliferation was also observed. Results showed that post-HSCT MSCs supported in vitro hematopoiesis and inhibited lymphocyte growth. Moreover, the quantity of MSCs was reduced at an early stage and restored to baseline level 9 months post-transplant. The results indicate that functional MSCs remain present in the BM microenvironment, and these findings shed light on the understanding of BM microenvironment reconstitution post-HSCT.
Collapse
Affiliation(s)
- Li Ding
- Department of Hematology, General Hospital of the Air Force , PLA, Beijing , China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Immune reconstitution after haploidentical hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2013; 20:440-9. [PMID: 24315844 DOI: 10.1016/j.bbmt.2013.11.028] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/29/2013] [Indexed: 12/12/2022]
Abstract
Haploidentical hematopoietic stem cell transplantation (HSCT) offers the benefits of rapid and nearly universal donor availability and has been accepted worldwide as an alternative treatment for patients with hematologic malignancies who do not have a completely HLA-matched sibling or who require urgent transplantation. Unfortunately, serious infections and leukemia relapse resulting from slow immune reconstitution remain the 2 most frequent causes of mortality in patients undergoing haploidentical HSCT, particularly in those receiving extensively T cell-depleted megadose CD34(+) allografts. This review summarizes advances in immune recovery after haploidentical HSCT, focusing on the immune subsets likely to have the greatest impact on clinical outcomes. The progress made in accelerating immune reconstitution using different strategies after haploidentical HSCT is also discussed. It is our belief that a predictive immune subset-guided strategy to improve immune recovery might represent a future clinical direction.
Collapse
|
13
|
Dvorak CC, Gilman AL, Horn B, Oon CY, Dunn EA, Baxter-Lowe LA, Cowan MJ. Haploidentical related-donor hematopoietic cell transplantation in children using megadoses of CliniMACs-selected CD34(+) cells and a fixed CD3(+) dose. Bone Marrow Transplant 2012. [PMID: 23178543 DOI: 10.1038/bmt.2012.186] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We conducted a prospective phase II trial utilizing the CliniMACs system to perform CD34(+)-cell selection of PBSCs from haploidentical donors to evaluate engraftment and hematoimmunological reconstitution. In total, 21 children with hematological malignancies or nonmalignant conditions underwent conditioning with 1200 cGy TBI, thiotepa, fludarabine and Thymoglobulin. Patients received megadoses of CD34(+) cells (median: 22 × 10(6)/kg) with a fixed dose of 3 × 10(4)/kg CD3(+) cells/kg, and engraftment occurred in 90% with prompt recovery of neutrophils and platelets. Grade II acute GVHD (aGVHD) was seen in 32% (95% confidence interval (CI), 15-54%) of evaluable patients, there was no grade III-IV aGVHD, and chronic extensive GVHD was seen in 35% (95% CI, 17-59%) of patients. The estimated 2-year EFS was 62% (95% CI, 48-83%) with a median survivor follow-up of 49 months (range: 18-119 months). Patients with nonmalignant diseases had an estimated 2-year EFS of 100% (95% CI, 56-100%) and patients with malignancies in remission had an estimated 2-year EFS of 56% (95% CI, 22-89%). Megadose CD34(+) cells with a fixed CD3(+) cell dose from haploidentical related donors resulted in good outcomes for pediatric patients with nonmalignant diseases and those with malignant diseases transplanted in remission.
Collapse
Affiliation(s)
- C C Dvorak
- Department of Pediatrics, Division of Allergy, Immunology, and Blood and Marrow Transplant, UCSF Benioff Children's Hospital, University of California San Francisco, San Francisco, CA 94143-1278, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Sodani P, Isgrò A, Gaziev J, Paciaroni K, Marziali M, Simone MD, Roveda A, De Angelis G, Gallucci C, Torelli F, Isacchi G, Zinno F, Landi F, Adorno G, Lanti A, Testi M, Andreani M, Lucarelli G. T cell-depleted hla-haploidentical stem cell transplantation in thalassemia young patients. Pediatr Rep 2011; 3 Suppl 2:e13. [PMID: 22053275 PMCID: PMC3206538 DOI: 10.4081/pr.2011.s2.e13] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 06/04/2011] [Indexed: 11/23/2022] Open
Abstract
The cure for thalassemia involves correcting the genetic defect in a hematopoietic stem cell that results in reduced or absent β-globin synthesis and an excess of α-globin dimers. [...]
Collapse
Affiliation(s)
- Pietro Sodani
- International Center for Transplantation in Thalassemia and Sickle Cell Anemia, Mediterranean Institute of Hematology, Policlinico Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Cao A, Moi P, Galanello R. Recent advances in β-thalassemias. Pediatr Rep 2011; 3:e17. [PMID: 21772954 PMCID: PMC3133499 DOI: 10.4081/pr.2011.e17] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/15/2011] [Accepted: 04/15/2011] [Indexed: 01/25/2023] Open
Abstract
β-thalassemias are heterogeneous hereditary anemias characterized by a reduced output of β-globin chains. The disease is most frequent in the temperate regions of the world, where it represents an important health problem. In the last decades, several programs, aimed at controlling the birth rate of thalassemia newborns by screening and prenatal diagnosis of populations with high risk of β-thalassemia, have been successful accomplished. Bone marrow transplantation has offered a definitive cure for the fraction of patients with available donors. In the same time, steady improvements were made in the traditional clinical management of β-thalassemia patients. The introduction of the oral iron chelators deferiprone that preferentially chelates hearth iron and the development of novel NMR diagnostic methods has led to reduced morbility, increased survival and improved quality of life. More recently, major advances have being made in the discovery of critical modifier genes, such as Myb and especially BCL11A (B cell lymphoma 11A), a master regulator of HbF (fetal hemoglobin) and hemoglobin switching. Polimorphysms of BCL11A, Myb and γ-globin genes account for most of the variability in the clinical phenotypes in β-thalassemia and sickle cell anemia patients. Finally, the year 2010 has brought in the first successful experiment of gene therapy in a β-thalassemia patient, opening up the perspective of a generalized cure for all β- thalassemia patients.
Collapse
Affiliation(s)
- Antonio Cao
- Biomedical and Biotechnology Department, University of Cagliari, Cagliari, Italy
| | | | | |
Collapse
|