1
|
Kleinschmidt K, Penkivech G, Troeger A, Foell J, Hanafee-Alali T, Leszczak S, Jakob M, Kramer S, Kietz S, Hoffmann P, Behrendt-Böhm C, Kaess C, Brosig A, Offner R, Wolff D, Corbacioglu S. αß T-cell depleted haploidentical stem cell transplantation for pediatric and young adult patients with transfusion-dependent thalassemia. Bone Marrow Transplant 2025; 60:682-689. [PMID: 40102568 PMCID: PMC12061755 DOI: 10.1038/s41409-025-02546-w] [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: 11/02/2024] [Revised: 01/08/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Life expectancy of patients with severe transfusion-dependent beta-thalassemia (TDT) remains below that of the general population. Allogenic hematopoietic stem cell transplantation (HSCT) is the standard curative treatment. Due to the paucity of matched donor (MD) availability, haploidentical HSCT (haplo-HSCT) is a reasonable alternative. Twenty patients with TDT (median age 10 years; range 2-23) received either a matched sibling donor (MSD; n = 7) or a haplo-HSCT (n = 13) in a single center (Regensburg, Germany) between 2016 and 2022, including two patients referred for a haplo-HSCT as rescue failing prior MD- and haplo-HSCT, respectively. The conditioning regimen consisted of anti-thymocyte globulin (ATG; Grafalon®), treosulfan, thiotepa, and fludarabine (FTT). Immunosuppression consisted of a calcineurin inhibitor and mycophenolate mofetil (MMF). At a median follow-up of 37 months (range 6-90), overall survival (OS) was 100% with a disease-free survival (DFS) of 100% in MSD and 92% in haplo-HSCT, respectively. Two patients in haplo-HSCT experienced graft failure, one achieving DFS after a second haplo-HSCT. No acute graft-versus-host disease (aGvHD) ≥ °III or severe chronic GvHD (cGvHD) were observed. No sinusoidal obstruction syndrome (SOS) was observed in this high-risk population. Treosulfan-based T-cell depleted haplo-HSCT can achieve comparable OS and DFS even in young adult TDT patients with no SOS/VOD.
Collapse
Affiliation(s)
- Katharina Kleinschmidt
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Gina Penkivech
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Anja Troeger
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Juergen Foell
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Tarek Hanafee-Alali
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Stefanie Leszczak
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Marcus Jakob
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Sonja Kramer
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Silke Kietz
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Petra Hoffmann
- Department of Internal Medicine III, University of Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Claudia Behrendt-Böhm
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Carina Kaess
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Andreas Brosig
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Robert Offner
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Daniel Wolff
- Department of Internal Medicine III, University of Regensburg, Regensburg, Germany
| | - Selim Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
2
|
Xiao H, Huang Q, Lai Y, Liu R. Haploidentical Hematopoietic Stem Cell Transplantation in Pediatric Transfusion-Dependent Thalassemia: A Systematic Review and Meta-Analysis. Transplant Cell Ther 2025; 31:101.e1-101.e12. [PMID: 39647520 DOI: 10.1016/j.jtct.2024.12.001] [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: 07/23/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) presents a promising therapeutic option for pediatric transfusion-dependent thalassemia, particularly in the scarcity of matched donors. Despite its potential, the comprehensive evaluation of this method through large-scale prospective studies remains lacking. This study aims to systematically summarize the efficacy and safety of haplo-HSCT in thalassemia, thereby providing further evidence-based insights for clinical practice. A comprehensive literature search was conducted across PubMed, Embase, and Web of Science databases through June 2024 to ensure a robust analysis of the available evidence. Data extraction was independently performed by 2 reviewers. The analysis utilized the inverse variance method with a 95% confidence interval (95% CI) to calculate the pooled proportion. To assess the heterogeneity among the studies, Cochran's Q test and Higgins' I-squared statistical methods were utilized. A random-effects model was employed to accommodate the variability between study results. Furthermore, subgroup analyses were explored differences in outcomes based on conditioning regimens and graft versus host disease (GVHD) prophylaxis. Conditioning regimens were categorized into reduced-intensity conditioning and myeloablative conditioning regimens. GVHD prophylaxis was classified into post-transplantation cyclophosphamide and non-post-transplantation cyclophosphamide. In this meta-analysis, we reviewed data from 10 studies encompassing 356 patients with thalassemia who underwent haplo-HSCT. Out of these, 328 patients survived until the follow-up date, resulting in a pooled overall survival rate of 92.4% (95% CI, 86.9-96.7; I² = 54.32%). The thalassemia-free survival was 84.5% (95% CI, 75.3-91.9; I² = 77.64%), and the graft failure rate was 8.1% (95% CI, 2.5-16.4; I² = 81.78%). The transplantation-related mortality stood at 7.4% (95% CI, 3.6-12.5; I² = 55.74%), with infections noted as the primary cause of death. The pooled proportion of acute graft versus host disease (aGVHD), grade 2-4 aGVHD, and grade 3-4 aGVHD were 29.6% (95% CI, 16.7-42.5, I² = 92.48%), 22.3% (95% CI, 10.1-42.1, I² = 80.06%), and 9.1% (95% CI, 2.8-17.7, I² = 67.92%), respectively. Subgroup analyses revealed no significant differences in these outcomes when comparing myeloablative conditioning to reduced-intensity conditioning, or post-transplantation cyclophosphamide to non-post-transplantation cyclophosphamide prophylaxis. However, variations in sample size, patient's age and geographic region among the studies suggest these factors as potential sources of heterogeneity. Haploidentical hematopoietic stem cell transplantation utilizes donors who are partially HLA-matched, typically family members, making it a viable option for transfusion-dependent thalassemia when fully matched donors are not available.
Collapse
Affiliation(s)
- Hongwen Xiao
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qiulin Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yongrong Lai
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; NHC Key Laboratory of Thalassemia Medicine, Nanning, Guangxi, China; Guangxi Key laboratory of Thalassemia Research, Nanning, Guangxi, China
| | - Rongrong Liu
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; NHC Key Laboratory of Thalassemia Medicine, Nanning, Guangxi, China; Guangxi Key laboratory of Thalassemia Research, Nanning, Guangxi, China.
| |
Collapse
|
3
|
Origa R, Issa L. Beta Thalassemia in Children: Established Approaches, Old Issues, New Non-Curative Therapies, and Perspectives on Healing. J Clin Med 2024; 13:6966. [PMID: 39598110 PMCID: PMC11594693 DOI: 10.3390/jcm13226966] [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: 10/12/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Despite a decrease in prevalence and incidence rates, beta thalassemia continues to represent a significant public health challenge worldwide. In high-resource settings, children with thalassemia have an open prognosis, with a high chance of reaching adulthood and old age with a good quality of life. This is achievable if transfusion therapy is properly managed, effectively mitigating ineffective erythropoiesis and its associated complications while also minimizing excessive iron accumulation. Adequate iron chelation is essential to maintain reactive forms of iron within the normal range throughout life, thus preventing organ damage caused by hemosiderosis, which inevitably results from a regular transfusion regimen. New therapies, both curative, such as gene therapy, and non-curative, such as modulators of erythropoiesis, are becoming available for patients with transfusion-dependent beta thalassemia. Two curative approaches based on gene therapy have been investigated in both adults and children with thalassemia. The first approach uses a lentivirus to correct the genetic defect, delivering a functional gene copy to the patient's cells. The second approach employs CRISPR/Cas9 gene editing to directly modify the defective gene at the molecular level. No non-curative therapies have received approval for pediatric use. Among adults, the only available drug is luspatercept, which is currently undergoing clinical trials in pediatric populations. However, in many countries around the world, the new therapeutic options remain a mirage, and even transfusion therapy itself is not guaranteed for most patients, while the choice of iron chelation therapy depends on drug availability and affordability.
Collapse
Affiliation(s)
- Raffaella Origa
- Department of Medical Sciences and Public Health, University of Cagliari, Ospedale Pediatrico Microcitemico A. Cao, ASL Cagliari, 09121 Cagliari, Italy
| | - Layal Issa
- Karma Association for Diseased Children and Adolescents, Furn El Chebbak, Beirut VG9G+3GV, Lebanon;
| |
Collapse
|
4
|
Youssry I, Ayad N. Sickle cell disease: combination new therapies vs. CRISPR-Cas9 potential and challenges - review article. Ann Hematol 2024; 103:2613-2619. [PMID: 37867187 DOI: 10.1007/s00277-023-05510-0] [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: 06/16/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023]
Abstract
In 2022, sickle cell disease (SCD) continues to affect the lives of millions of people, being one of the most frequently inherited blood disorders worldwide. Recently, several new therapies have been FDA approved for the treatment of SCD. The complexity of the pathophysiology of sickling has given opportunity to the evolution of several modalities of therapies. Nonetheless, the potential for complementary targeting of HbS polymerization, vasocclusion, and other inflammatory pathways remains controversial. None of these drugs can be considered a single curative line of treatment. With the advancement of CRISPR/Cas9 technology, autologous transplant of gene-edited hematopoietic stem cells could possibly provide a cure for most patients with SCD. The advantage of this approach over the conventional stem cell transplantation is that it decreases the need for immuno-suppressive drugs and the risk of graft-versus-host disease. In addition, recent technological advances can reduce the off-target effects, but long-term monitoring is needed to ensure the reliability of these methods in the clinical setting. This review explores the efficacy and safety of combination therapies and contrasting this alternative with the challenges that exist with sickle cell gene therapy using CRISPR.
Collapse
Affiliation(s)
- Ilham Youssry
- Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nardeen Ayad
- Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
5
|
Jaiswal SR, Agarwal M, Bhagawati G, Das BC, Baligar P, Garg M, Biswas S, Chakrabarti S. Long-Term Follow-Up of Abatacept, Post-Transplantation Cyclophosphamide, and Sirolimus-Based Haploidentical Transplantation in Younger Patients with Nonmalignant Diseases. Transplant Cell Ther 2024; 30:605.e1-605.e13. [PMID: 38490295 DOI: 10.1016/j.jtct.2024.03.010] [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: 02/02/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024]
Abstract
Haploidentical (haplo) hematopoietic cell transplantation (HCT) for nonmalignant disease (NMD) carries inherent challenges of both alloreactivity and graft failure. Building on promising results from pilot studies in which abatacept was combined with post-transplantation cyclophosphamide (PTCy) and sirolimus (AbaCyS) in younger NMD patients undergoing haplo-HCT, we present the long-term outcomes of this protocol. On the back of uniform disease-specific conditioning regimens containing antithymocyte globulin 4.5 mg/kg from day -9 to day -7, GVHD prophylaxis with AbaCyS consisted of abatacept administered on days 0, +5, +20, +35, and monthly until 180 days with PTCy and sirolimus. The patients were followed up with longitudinal assessment of immune reconstitution, growth, and reproductive development and quality of life (QoL) analyses. Among 40 patients (aplastic anemia, n = 24; hemoglobinopathies, n = 14; and primary immunodeficiencies, n = 2) with a median age of 10 years (range, 2 to 25 years), 95% achieved sustained engraftment. Post-transplantation hemophagocytic syndrome was detected in 3 patients, leading to graft failure in 2 cases. The incidence of acute graft-versus-host disease (GVHD) was 2.6%, and that of chronic GVHD (cGVHD) was 14.3%. Cytomegalovirus, adenovirus, and Epstein-Barr virus infections were observed in 45%, 5%, and 0% respectively. Rates of nonrelapse mortality, overall survival, event-free survival, and GVHD-free, event-free survival were 5%, 95%, 90%, and 82%, respectively, at a median follow-up of 4.6 years. Absence of cGVHD correlated with younger patient age and early sustained recovery of regulatory T cells and mature natural killer cells, which in turn was associated with improved QoL and lack of late infections. The AbaCyS protocol was associated with excellent long-term survival, with attenuation of both early and late alloreactivity in >80% of younger patients undergoing haplo-HCT for NMD. This study sheds light on predispositions to cGVHD and its impact on QoL, warranting further optimization of this approach.
Collapse
Affiliation(s)
- Sarita Rani Jaiswal
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India; Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, Kolkata, India; Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India.
| | - Mahak Agarwal
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India; Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, Kolkata, India
| | - Gitali Bhagawati
- Department of Microbiology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Bhudev Chandra Das
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Suparno Chakrabarti
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India; Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, Kolkata, India; Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
6
|
Pai VB, Tansmore J, Song E. Pharmacokinetic analysis driven letermovir dosing in pediatric hematopoietic cell transplantation patients with resistant CMV disease. Pediatr Transplant 2024; 28:e14580. [PMID: 37466088 DOI: 10.1111/petr.14580] [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: 05/14/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Reactivation of cytomegalovirus (CMV) in CMV-seropositive patients after haploidentical T-cell receptor αβ+ /CD19+ depleted hematopoietic cell transplant (HCT) is common. Due to delayed CMV-specific immune reconstitution, patients may require prolonged antiviral therapy, including secondary prophylaxis (SP). We present our clinical experience with the off-label use of letermovir for SP in a severely immunocompromised 2-year-old toddler with refractory pre-B-cell ALL and bilateral retinitis caused by resistant CMV (A594V UL97 mutation) following a haploidentical TCRαβ+ /CD19+ depleted HCT. METHODS The patient underwent measurement of two separate sets of letermovir serum concentrations, drawn at pre-dose, 1 and 4 h (and 8 h during the second therapeutic drug monitoring) post-dose. Pharmacokinetic parameters, including AUC0-24 were calculated, and dose adjustment was performed based on the drug level. RESULTS While receiving oral letermovir 240 mg once daily without cyclosporine, the observed AUC0-24 was high (75 815 ng h/mL) with a Cmin of 209 ng/mL. The dose was reduced by 25% to 180 mg once daily. Despite the dose reduction, both AUC0-24 and Cmin values further increased to 119 095 ng h/L and 959 ng/mL, respectively. The patient continued oral letermovir 180 mg once daily for about 3 months, with adequate viral suppression (CMV viral load in plasma <150 IU/mL) and no recurrent CMV end-organ disease or adverse events. CONCLUSIONS Given limited options for anti-CMV therapy in young children with resistant CMV, letermovir could be considered as an alternative antiviral for SP. Further studies are warranted to evaluate the pharmacokinetics of letermovir in pediatric allogeneic HCT recipients.
Collapse
Affiliation(s)
- Vinita B Pai
- Nationwide Children's Hospital, Columbus, Ohio, USA
- College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | | | - Eunkyung Song
- Nationwide Children's Hospital, Columbus, Ohio, USA
- College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Di Ianni M, Liberatore C, Santoro N, Ranalli P, Guardalupi F, Corradi G, Villanova I, Di Francesco B, Lattanzio S, Passeri C, Lanuti P, Accorsi P. Cellular Strategies for Separating GvHD from GvL in Haploidentical Transplantation. Cells 2024; 13:134. [PMID: 38247827 PMCID: PMC10814899 DOI: 10.3390/cells13020134] [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] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
GvHD still remains, despite the continuous improvement of transplantation platforms, a fearful complication of transplantation from allogeneic donors. Being able to separate GvHD from GvL represents the greatest challenge in the allogeneic transplant setting. This may be possible through continuous improvement of cell therapy techniques. In this review, current cell therapies are taken into consideration, which are based on the use of TCR alpha/beta depletion, CD45RA depletion, T regulatory cell enrichment, NK-cell-based immunotherapies, and suicide gene therapies in order to prevent GvHD and maximally amplify the GvL effect in the setting of haploidentical transplantation.
Collapse
Affiliation(s)
- Mauro Di Ianni
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carmine Liberatore
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
| | - Nicole Santoro
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
| | - Paola Ranalli
- Hematology Unit, Pescara Hospital, 65124 Pescara, Italy; (C.L.); (N.S.); (P.R.)
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Francesco Guardalupi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giulia Corradi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ida Villanova
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| | - Barbara Di Francesco
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| | - Stefano Lattanzio
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Cecilia Passeri
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, 66100 Chieti, Italy; (F.G.); (G.C.); (S.L.); (P.L.)
- Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, 66100 Chieti, Italy
| | - Patrizia Accorsi
- Blood Bank Unit, Pescara Hospital, 65124 Pescara, Italy; (I.V.); (B.D.F.); (C.P.); (P.A.)
| |
Collapse
|
8
|
Greenmyer JR, Ariagno S, Ali A, Pence L, O'Shea M, Greenmyer LA, Khan S, Kuhn A, Martin C, Ferdjallah A, Kohorst M. Autoimmune cytopenias following pediatric hematopoietic cell transplant. Bone Marrow Transplant 2024; 59:117-120. [PMID: 37794111 DOI: 10.1038/s41409-023-02116-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Affiliation(s)
- Jacob R Greenmyer
- Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Sydney Ariagno
- Hospice and Palliative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Asma Ali
- Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lindy Pence
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mary O'Shea
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Shakila Khan
- Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Alexis Kuhn
- Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
- Pediatric Pharmacy, Mayo Clinic, Rochester, MN, USA
| | - Catherine Martin
- Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
- Pediatric Pharmacy, Mayo Clinic, Rochester, MN, USA
| | - Asmaa Ferdjallah
- Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | - Mira Kohorst
- Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Kassim AA. The range of haploidentical transplant protocols in sickle cell disease: all haplos are not created equally. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:532-541. [PMID: 38066894 PMCID: PMC10727056 DOI: 10.1182/hematology.2023000486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The ideal curative therapy for sickle cell disease (SCD) must be applicable across all ages and include individuals with strokes and preexisting heart, lung, and kidney disease. Myeloablative, matched sibling donor hematopoietic stem cell transplant (HCT) for children with SCD has shown excellent outcomes over the past 3 decades but has been restricted due to the limited availability of a human leukocyte antigen-matched sibling donor (10%-15%) and increased treatment-related death in adults with myeloablative conditioning. To overcome these 2 significant barriers to curative therapy in SCD, related haploidentical HCT has become an active area of research. The use of related haploidentical donors (first- and second-degree relatives) increases the donor pool to at least 90% of those eligible across the life span. Importantly, most adults, even with strokes or significant comorbidities, can tolerate the nonmyeloablative conditioning regimen without treatment-related death. Since 2013, at least 3 related haploidentical HCT strategies have emerged as potential curative therapies for SCD: (1) a nonmyeloablative, T-cell replete, bone marrow transplant with thiotepa and posttransplant cyclophosphamide with a goal of complete donor chimerism; (2) a nonmyeloablative, in vivo T-cell depletion, using peripheral blood stem cells (PBSCs) with a goal of stable mixed donor-recipient chimerism; and (3) a myeloablative, ex vivo T-cell depletion using PBSCs and advanced-technology graft manipulation, with a goal of complete donor chimerism. We review the similarities, differences, outcomes, and gaps in knowledge with these 3 haploidentical HCT approaches for SCD.
Collapse
Affiliation(s)
- Adetola A. Kassim
- Department of Hematology/Oncology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
10
|
Cuvelier GDE, Paulson K, Bow EJ. Updates in hematopoietic cell transplant and cellular therapies that enhance the risk for opportunistic infections. Transpl Infect Dis 2023; 25 Suppl 1:e14101. [PMID: 37461887 DOI: 10.1111/tid.14101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Infectious disease physicians may be asked to evaluate and manage a variety of infections in immunocompromised hosts undergoing hematopoietic cell transplant (HCT) and cellular therapies. Over the last decade, several advances in cellular therapy have occurred, with implications for the types of infectious complications that may be seen. AIMS The purpose of this review is to update the infectious disease physician on newer advances in HCT and cellular therapy, including haploidentical transplant, expanding indications for transplant in older individuals and children, and chimeric antigen receptor T-cells. We will review how these advances might influence infectious disease complications following HCT. We will also provide a perspective that infectious disease physicians can use to evaluate the degree of immune suppression in an individual patient to help determine the type of infections that may be encountered.
Collapse
Affiliation(s)
- Geoffrey D E Cuvelier
- Department of Paediatrics and Child Health, Section of Paediatric Haematology/Oncology-BMT, Max Rady College of Medicine, the University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Paediatric Haematology/Oncology-BMT, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Blood and Marrow Transplant Programme, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | - Kristjan Paulson
- Manitoba Blood and Marrow Transplant Programme, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Haematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, the University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Oncology and Haematology, CancerCare Manitoba, Winnipeg, Manitoba, Winnipeg, Manitoba, Canada
| | - Eric J Bow
- Manitoba Blood and Marrow Transplant Programme, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Haematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, the University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Oncology and Haematology, CancerCare Manitoba, Winnipeg, Manitoba, Winnipeg, Manitoba, Canada
- Section of Infectious Diseases, Department of Internal Medicine, Max Rady College of Medicine, The University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
11
|
Piel FB, Rees DC, DeBaun MR, Nnodu O, Ranque B, Thompson AA, Ware RE, Abboud MR, Abraham A, Ambrose EE, Andemariam B, Colah R, Colombatti R, Conran N, Costa FF, Cronin RM, de Montalembert M, Elion J, Esrick E, Greenway AL, Idris IM, Issom DZ, Jain D, Jordan LC, Kaplan ZS, King AA, Lloyd-Puryear M, Oppong SA, Sharma A, Sung L, Tshilolo L, Wilkie DJ, Ohene-Frempong K. Defining global strategies to improve outcomes in sickle cell disease: a Lancet Haematology Commission. Lancet Haematol 2023; 10:e633-e686. [PMID: 37451304 PMCID: PMC11459696 DOI: 10.1016/s2352-3026(23)00096-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 07/18/2023]
Abstract
All over the world, people with sickle cell disease (an inherited condition) have premature deaths and preventable severe chronic complications, which considerably affect their quality of life, career progression, and financial status. In addition, these people are often affected by stigmatisation or structural racism, which can contribute to stress and poor mental health. Inequalities affecting people with sickle cell disease are also reflected in the distribution of the disease—mainly in sub-Saharan Africa, India, and the Caribbean—whereas interventions, clinical trials, and funding are mostly available in North America, Europe, and the Middle East. Although some of these characteristics also affect people with other genetic diseases, the fate of people with sickle cell disease seems to be particularly unfair. Simple, effective interventions to reduce the mortality and morbidity associated with sickle cell disease are available. The main obstacle preventing better outcomes in this condition, which is a neglected disease, is associated with inequalities impacting the patient populations. The aim of this Commission is to highlight the problems associated with sickle cell disease and to identify achievable goals to improve outcomes both in the short and long term. The ambition for the management of people with sickle cell disease is that curative treatments become available to every person with the condition. Although this would have seemed unrealistic a decade ago, developments in gene therapy make this potentially achievable, albeit in the distant future. Until these curative technologies are fully developed and become widely available, health-care professionals (with the support of policy makers, funders, etc) should make sure that a minimum standard of care (including screening, prophylaxis against infection, acute medical care, safe blood transfusion, and hydroxyurea) is available to all patients. In considering what needs to be achieved to reduce the global burden of sickle cell disease and improve the quality of life of patients, this Commission focuses on five key areas: the epidemiology of sickle cell disease (Section 1 ); screening and prevention (Section 2 ); established and emerging treatments for the management of the disease (Section 3 ); cellular therapies with curative potential (Section 4 ); and training and education needs (Section 5 ). As clinicians, researchers, and patients, our objective to reduce the global burden of sickle cell disease aligns with wider public health aims to reduce inequalities, improve health for all, and develop personalised treatment options. We have observed in the past few years some long-awaited momentum following the development of innovative point-of-care testing devices, new approved drugs, and emerging curative options. Reducing the burden of sickle cell disease will require substantial financial and political commitment, but it will impact the lives of millions of patients and families worldwide and the lessons learned in achieving this goal would unarguably benefit society as a whole.
Collapse
Affiliation(s)
- Frédéric B Piel
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| | - David C Rees
- Department of Paediatric Haematology, King's College London, King's College Hospital, London, UK
| | - Michael R DeBaun
- Department of Pediatrics, Vanderbilt-Meharry Center of Excellence for Sickle Cell Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Obiageli Nnodu
- Department of Haematology and Blood Transfusion, College of Health Sciences and Centre of Excellence for Sickle Cell Disease Research and Training, University of Abuja, Abuja, Nigeria
| | - Brigitte Ranque
- Department of Internal Medicine, Georges Pompidou European Hospital, Assistance Publique-Hopitaux de Paris Centre, University of Paris Cité, Paris, France
| | - Alexis A Thompson
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Russell E Ware
- Division of Hematology and Global Health Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Miguel R Abboud
- Department of Pediatrics and Adolescent Medicine, and Sickle Cell Program, American University of Beirut, Beirut, Lebanon
| | - Allistair Abraham
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Emmanuela E Ambrose
- Department of Paediatrics and Child Health, Bugando Medical Centre, Mwanza, Tanzania
| | - Biree Andemariam
- New England Sickle Cell Institute, University of Connecticut Health, Connecticut, USA
| | - Roshan Colah
- Department of Haematogenetics, Indian Council of Medical Research National Institute of Immunohaematology, Mumbai, India
| | - Raffaella Colombatti
- Pediatric Oncology Hematology Unit, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Nicola Conran
- Department of Clinical Medicine, School of Medical Sciences, Center of Hematology and Hemotherapy (Hemocentro), University of Campinas-UNICAMP, Campinas, Brazil
| | - Fernando F Costa
- Department of Clinical Medicine, School of Medical Sciences, Center of Hematology and Hemotherapy (Hemocentro), University of Campinas-UNICAMP, Campinas, Brazil
| | - Robert M Cronin
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Mariane de Montalembert
- Department of Pediatrics, Necker-Enfants Malades Hospital, Assistance Publique-Hopitaux de Paris Centre, Paris, France
| | - Jacques Elion
- Paris Cité University and University of the Antilles, Inserm, BIGR, Paris, France
| | - Erica Esrick
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Anthea L Greenway
- Department Clinical Haematology, Royal Children's Hospital, Parkville and Department Haematology, Monash Health, Clayton, VIC, Australia
| | - Ibrahim M Idris
- Department of Hematology, Aminu Kano Teaching Hospital/Bayero University Kano, Kano, Nigeria
| | - David-Zacharie Issom
- Department of Business Information Systems, School of Management, HES-SO University of Applied Sciences and Arts of Western Switzerland, Geneva, Switzerland
| | - Dipty Jain
- Department of Paediatrics, Government Medical College, Nagpur, India
| | - Lori C Jordan
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zane S Kaplan
- Department of Clinical Haematology, Monash Health and Monash University, Melbourne, VIC, Australia
| | - Allison A King
- Departments of Pediatrics and Internal Medicine, Divisions of Pediatric Hematology and Oncology and Hematology, Washington University School of Medicine, St Louis, MO, USA
| | - Michele Lloyd-Puryear
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Samuel A Oppong
- Department of Obstetrics and Gynecology, University of Ghana Medical School, Accra, Ghana
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lillian Sung
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Leon Tshilolo
- Institute of Biomedical Research/CEFA Monkole Hospital Centre and Official University of Mbuji-Mayi, Mbuji-Mayi, Democratic Republic of the Congo
| | - Diana J Wilkie
- Department of Biobehavioral Nursing Science, College of Nursing, University of Florida, Gainesville, FL, USA
| | - Kwaku Ohene-Frempong
- Division of Hematology, Children's Hospital of Philadelphia, Pennsylvania, USA; Sickle Cell Foundation of Ghana, Kumasi, Ghana
| |
Collapse
|
12
|
Zhang Q, Huo Y, Sun Q, Liu N, Shi H, Wang M, Xiao J, Yuan H, Tang X. Case report: Curing a rare, unstable hemoglobin variant Hb Bristol-Alesha using haploidentical hematopoietic stem cell transplantation. Front Immunol 2023; 14:1188058. [PMID: 37457725 PMCID: PMC10348747 DOI: 10.3389/fimmu.2023.1188058] [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: 03/16/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Unstable hemoglobinopathies are a rare, heterogeneous group of diseases that disrupt the stability of hemoglobin (Hb), leading to chronic hemolysis and anemia. Patients with severe phenotypes often require regular blood transfusions and iron chelation therapy. Although rare, studies have reported that hematopoietic stem cell transplantation (HSCT) seems to be an available curative approach in transfusion-dependent patients with unstable hemoglobinopathies. Here, we describe successful haploidentical HSCT for the treatment of an unstable Hb variant, Hb Bristol-Alesha, in a 6-year-old boy with severe anemia since early childhood. Two years after transplantation, he had a nearly normal hemoglobin level without evidence of hemolysis. DNA analysis showed complete chimerism of the donor cell origin, confirming full engraftment with normal erythropoiesis.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Hematology, No.971 Hospital of People's Liberation Army Navy, Qingdao, Shandong, China
| | - Yujia Huo
- Department of Traditional Chinese Medicine, Qingdao Special Service Sanatorium of People's Liberation Army Navy, Qingdao, Shandong, China
| | - Qinggang Sun
- Department of Hematology, No.971 Hospital of People's Liberation Army Navy, Qingdao, Shandong, China
| | - Nan Liu
- Department of Hematology, No.971 Hospital of People's Liberation Army Navy, Qingdao, Shandong, China
| | - Hongchuan Shi
- Department of Hematology, No.971 Hospital of People's Liberation Army Navy, Qingdao, Shandong, China
| | - Minghui Wang
- Department of Hematology, No.971 Hospital of People's Liberation Army Navy, Qingdao, Shandong, China
| | - Jinming Xiao
- Department of Hematology, No.971 Hospital of People's Liberation Army Navy, Qingdao, Shandong, China
| | - Hanzi Yuan
- Department of Pediatrics, the Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Xiangfeng Tang
- Department of Pediatrics, the Seventh Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
13
|
Algeri M, Lodi M, Locatelli F. Hematopoietic Stem Cell Transplantation in Thalassemia. Hematol Oncol Clin North Am 2023; 37:413-432. [PMID: 36907612 DOI: 10.1016/j.hoc.2022.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only consolidated, potentially curative treatment for patients with transfusion-dependent thalassemia major. In the past few decades, several new approaches have reduced the toxicity of conditioning regimens and decreased the incidence of graft-versus-host disease, improving patients' outcomes and quality of life. In addition, the progressive availability of alternative stem cell sources from unrelated or haploidentical donors or umbilical cord blood has made HSCT a feasible option for an increasing number of subjects lacking an human leukocyte antigen (HLA)-identical sibling. This review provides an overview of allogeneic hematopoietic stem cell transplantation in thalassemia, reassesses current clinical results, and discusses future perspectives.
Collapse
Affiliation(s)
- Mattia Algeri
- Department of Hematology/Oncology, Cell and Gene Therapy - IRCCS, Bambino Gesù Children's Hospital, Rome, Italy.
| | - Mariachiara Lodi
- Department of Hematology/Oncology, Cell and Gene Therapy - IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Cell and Gene Therapy - IRCCS, Bambino Gesù Children's Hospital, Rome, Italy; Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
14
|
Hu J, Gong S, Chen K, Yang R, Wang L, Yang K, Nie L, Zou L, Su T, Chen C, Xu Y, He X, Yang L, Xiao H, Fu B. Haploidentical transplant for paediatric patients with severe thalassaemia using post-transplant cyclophosphamide and methotrexate: A prospectively registered multicentre trial from the Bone Marrow Failure Working Group of Hunan Province, China. Br J Haematol 2023; 200:329-337. [PMID: 36254684 DOI: 10.1111/bjh.18520] [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: 08/25/2022] [Revised: 09/24/2022] [Accepted: 10/05/2022] [Indexed: 01/21/2023]
Abstract
Haploidentical transplantation strategies for patients with transfusion-dependent thalassaemia (TD-TM) remain to be investigated. In this study, 54 paediatric patients with TD-TM were treated with a novel approach using post-transplant cyclophosphamide (PTCy) and low-dose methotrexate (LD-MTX), following a myeloablative regimen. The incidence of neutrophil and platelet engraftment was 96.3% ± 2.6% and 94.4% ± 3.1% respectively. The cumulative incidence of grades II-III acute graft-versus-host disease (GVHD) was 13.8% ± 4.8% at 100 days. At three years, the cumulative incidence of chronic GVHD was 28.5% ± 8.5%. With a median follow-up of 520 days (132-1325 days), the overall survival (OS) and event-free survival (EFS) were 98.1% ± 1.8% and 90.7% ± 3.9% respectively. Compared with the low-dose cyclophosphamide (CTX) conditioning regimen (120 mg/kg), the high-CTX regimen (200 mg/kg) achieved a higher incidence of stable engraftment (100% vs 66.7% ± 15.7%, p = 0.003), a comparable incidence of grades II-III acute GVHD, a lower incidence of chronic GVHD (20.2% ± 8.3% vs 66.6% ± 19.2%, p = 0.011), and better overall survival (100% vs 88.9% ± 10.5%, p = 0.025) as well as EFS (95.6% ± 3.1% vs 66.7% ± 15.7%, p = 0.008). Our results using unmanipulated haploidentical grafts and PTCy with LD-MTX in TD-TM are encouraging. (chictr.org.cn ChiCTR1800017969).
Collapse
Affiliation(s)
- Jian Hu
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Susu Gong
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Keke Chen
- Department of Pediatric Hematology, Hunan Provincial People's Hospital, Changsha, China
| | - Rui Yang
- Department of Pediatric Hematology, First People's Hospital of Chenzhou, Chenzhou, China
| | - Leyuan Wang
- Department of Pediatric Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Kaitai Yang
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Lin Nie
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Lang Zou
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Tao Su
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Cong Chen
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Yajing Xu
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Changsha, China.,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Hangzhou, China
| | - Xianglin He
- Department of Pediatric Hematology, Hunan Provincial People's Hospital, Changsha, China
| | - Liangchun Yang
- Department of Pediatric Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Hong Xiao
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China
| | - Bin Fu
- Department of Hematology, Xiangya Hospital of Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Changsha, China.,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Hangzhou, China
| |
Collapse
|
15
|
Bhalla N, Bhargav A, Yadav SK, Singh AK. Allogeneic hematopoietic stem cell transplantation to cure sickle cell disease: A review. Front Med (Lausanne) 2023; 10:1036939. [PMID: 36910492 PMCID: PMC9995916 DOI: 10.3389/fmed.2023.1036939] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Sickle cell disease (SCD) had first been mentioned in the literature a century ago. Advancement in the molecular basis of the pathophysiology of the disease opens the door for various therapeutic options. Though life-extending treatments are available for treating patients with SCD, allogeneic hematopoietic stem cell transplantation (HSCT) is the only option as of yet. A major obstacle before HSCT to cure patients with SCD is the availability of donors. Matched sibling donors are available only for a small percentage of patients. To expand the donor pool, different contrasting approaches of allogeneic HSCT like T-cell replete and deplete have been tested. None of those tested approaches have been without the risk of GvHD and graft rejection. Other limitations such as transplantation-related infections and organ dysfunction caused by the harsh conditioning regimen need to be addressed on a priority basis. In this review, we will discuss available allogeneic HSCT approaches to cure SCD, as well as recent advancements to make the approach safer. The center of interest is using megadose T-cell-depleted bone marrow in conjugation with donor-derived CD8 veto T cells to achieve engraftment and tolerance across MHC barriers, under reduced intensity conditioning (RIC). This approach is in phase I/II clinical trial at the MD Anderson Cancer Centre and is open to patients with hemoglobinopathies.
Collapse
Affiliation(s)
- Nishka Bhalla
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamilnadu, India
| | - Anjali Bhargav
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamilnadu, India
| | | | - Aloukick Kumar Singh
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamilnadu, India
| |
Collapse
|
16
|
Chu Y, Talano JA, Baxter-Lowe LA, Verbsky JW, Morris E, Mahanti H, Ayello J, Keever-Taylor C, Johnson B, Weinberg RS, Shi Q, Moore TB, Fabricatore S, Grossman B, van de Ven C, Shenoy S, Cairo MS. Donor chimerism and immune reconstitution following haploidentical transplantation in sickle cell disease. Front Immunol 2022; 13:1055497. [PMID: 36569951 PMCID: PMC9780682 DOI: 10.3389/fimmu.2022.1055497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction We previously reported the initial results of a phase II multicenter transplant trial using haploidentical parental donors for children and aolescents with high-risk sickle cell disease achieving excellent survival with exceptionally low rates of graft-versus-host disease and resolution of sickle cell disease symptoms. To investigate human leukocyte antigen (HLA) sensitization, graft characteristics, donor chimerism, and immune reconstitution in these recipients. Methods CD34 cells were enriched using the CliniMACS® system with a target dose of 10 x 106 CD34+ cells/kg with a peripheral blood mononuclear cell (PBMNC) addback dose of 2x105 CD3/kg in the final product. Pre-transplant HLA antibodies were characterized. Donor chimerism was monitored 1-24 months post-transplant. Comprehensive assessment of immune reconstitution included lymphocyte subsets, plasma cytokines, complement levels, anti-viral T-cell responses, activation markers, and cytokine production. Infections were monitored. Results HLA antibodies were detected in 7 of 11 (64%) evaluable patients but rarely were against donor antigens. Myeloid engraftment was rapid (100%) at a median of 9 days. At 30 days, donor chimerism was 93-99% and natural killer cell levels were restored. By 60 days, CD19 B cells were normal. CD8 and CD4 T-cells levels were normal by 279 and 365 days, respectively. Activated CD4 and CD8 T-cells were elevated at 100-365 days post-transplant while naïve cells remained below baseline. Tregs were elevated at 100-270 days post-transplant, returning to baseline levels at one year. At one year, C3 and C4 levels were above baseline and CH50 levels were near baseline. At one year, cytokine levels were not significantly different from baseline. Discussion These results suggest that haploidentical transplantation with CD34-enriched cells and peripheral blood mononuclear cell addback results in rapid engraftment, sustained donor chimerism and broad-based immune reconstitution.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Julie-An Talano
- Department of Pediatrics, Hematology/Oncology and BMT, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lee Ann Baxter-Lowe
- Department of Pathology, Children’s Hospital of Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - James W. Verbsky
- Department of Pediatrics, Hematology/Oncology and BMT, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Erin Morris
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Harshini Mahanti
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - Carolyn Keever-Taylor
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bryon Johnson
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Qiuhu Shi
- Department of Epidemiology and Community Health, New York Medical College, Valhalla, NY, United States
| | - Theodore B. Moore
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Sandra Fabricatore
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Brenda Grossman
- Department of Pathology and Immunology, Washington University, St Louis, MO, United States
| | - Carmella van de Ven
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Shalini Shenoy
- Department of Pediatrics and Transfusion Medicine, Washington University, St Louis, MO, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, New York Medical College, Valhalla, NY, United States,Department of Medicine, New York Medical College, Valhalla, NY, United States,Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, United States,Department of Cell Biology, New York Medical College, Valhalla, NY, United States,Department of Anatomy, New York Medical College, Valhalla, NY, United States,*Correspondence: Mitchell S. Cairo,
| |
Collapse
|
17
|
Watkins B, Williams KM. Controversies and expectations for the prevention of GVHD: A biological and clinical perspective. Front Immunol 2022; 13:1057694. [PMID: 36505500 PMCID: PMC9726707 DOI: 10.3389/fimmu.2022.1057694] [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: 09/29/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Severe acute and chronic graft versus host disease (GVHD) remains a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. Historically, cord blood and matched sibling transplantation has been associated with the lowest rates of GVHD. Newer methods have modified the lymphocyte components to minimize alloimmunity, including: anti-thymocyte globulin, post-transplant cyclophosphamide, alpha/beta T cell depletion, and abatacept. These agents have shown promise in reducing severe GVHD, however, can be associated with increased risks of relapse, graft failure, infections, and delayed immune reconstitution. Nonetheless, these GVHD prophylaxis strategies have permitted expansion of donor sources, especially critical for those of non-Caucasian decent who previously lacked transplant options. This review will focus on the biologic mechanisms driving GVHD, the method by which each agent impacts these activated pathways, and the clinical consequences of these modern prophylaxis approaches. In addition, emerging novel targeted strategies will be described. These GVHD prophylaxis approaches have revolutionized our ability to increase access to transplant and have provided important insights into the biology of GVHD and immune reconstitution.
Collapse
Affiliation(s)
- Benjamin Watkins
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | | |
Collapse
|
18
|
Kassim AA, Leonard A. Debating the Future of Sickle Cell Disease Curative Therapy: Haploidentical Hematopoietic Stem Cell Transplantation vs. Gene Therapy. J Clin Med 2022; 11:jcm11164775. [PMID: 36013014 PMCID: PMC9409766 DOI: 10.3390/jcm11164775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a well-established curative therapy for patients with sickle cell disease (SCD) when using a human leukocyte antigen (HLA)-matched sibling donor. Most patients with SCD do not have a matched sibling donor, thereby significantly limiting the accessibility of this curative option to most patients. HLA-haploidentical HSCT with post-transplant cyclophosphamide expands the donor pool, with current approaches now demonstrating high overall survival, reduced toxicity, and an effective reduction in acute and chronic graft-vs.-host disease (GvHD). Alternatively, autologous genetic therapies appear promising and have the potential to overcome significant barriers associated with allogeneic HSCT, such as donor availability and GvHD. Here the authors each take a viewpoint and discuss what will be the future of curative options for patients with SCD outside of a matched sibling transplantation, specifically haploidentical HSCT vs. gene therapy.
Collapse
Affiliation(s)
- Adetola A. Kassim
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt Meharry Sickle Cell Center of Excellence, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Correspondence: (A.A.K.); or (A.L.)
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20810, USA
- Division of Hematology, Children’s National Hospital, Washington, DC 20010, USA
- Correspondence: (A.A.K.); or (A.L.)
| |
Collapse
|
19
|
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
|
20
|
Haploidentical Stem Cell Transplantation for Patients with Sickle Cell Disease: Current Status. Transfus Apher Sci 2022; 61:103534. [DOI: 10.1016/j.transci.2022.103534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
21
|
DeFilipp Z, Hefazi M, Chen YB, Blazar BR. Emerging approaches to improve allogeneic hematopoietic cell transplantation outcomes for nonmalignant diseases. Blood 2022; 139:3583-3593. [PMID: 34614174 PMCID: PMC9728560 DOI: 10.1182/blood.2020009014] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/04/2021] [Indexed: 12/14/2022] Open
Abstract
Many congenital or acquired nonmalignant diseases (NMDs) of the hematopoietic system can be potentially cured by allogeneic hematopoietic cell transplantation (HCT) with varying types of donor grafts, degrees of HLA matching, and intensity of conditioning regimens. Unique features that distinguish the use of allogeneic HCT in this population include higher rates of graft failure, immune-mediated cytopenias, and the potential to achieve long-term disease-free survival in a mixed chimerism state. Additionally, in contrast to patients with hematologic malignancies, a priority is to completely avoid graft-versus-host disease in patients with NMD because there is no theoretical beneficial graft-versus-leukemia effect that can accompany graft-versus-host responses. In this review, we discuss the current approach to each of these clinical issues and how emerging novel therapeutics hold promise to advance transplant care for patients with NMDs.
Collapse
Affiliation(s)
- Zachariah DeFilipp
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, MA
| | | | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, MA
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN
| |
Collapse
|
22
|
Abstract
Thalassaemia is a diverse group of genetic disorders with a worldwide distribution affecting globin chain synthesis. The pathogenesis of thalassaemia lies in the unbalanced globin chain production, leading to ineffective erythropoiesis, increased haemolysis, and deranged iron homoeostasis. The clinical phenotype shows heterogeneity, ranging from close to normal without complications to severe requiring lifelong transfusion support. Conservative treatment with transfusion and iron chelation has transformed the natural history of thalassaemia major into a chronic disease with a prolonged life expectancy, albeit with co-morbidities and substantial disease burden. Curative therapy with allogeneic haematopoietic stem cell transplantation is advocated for suitable patients. The understanding of the pathogenesis of the disease is guiding therapeutic advances. Novel agents have shown efficacy in improving anaemia and transfusion burden, and initial results from gene therapy approaches are promising. Despite scientific developments, worldwide inequality in the access of health resources is a major concern, because most patients live in underserved areas.
Collapse
Affiliation(s)
- Antonis Kattamis
- Division of Paediatric Haematology-Oncology, First Department of Paediatrics, National and Kapodistrian University of Athens, Athens, Greece.
| | - Janet L Kwiatkowski
- Division of Haematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Paediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yesim Aydinok
- Department of Paediatric Heamatology and Oncology, Ege University School of Medicine, Izmir, Turkey
| |
Collapse
|
23
|
Dhuyser A, Aarnink A, Pérès M, Jayaraman J, Nemat-Gorgani N, Rubio MT, Trowsdale J, Traherne J. KIR in Allogeneic Hematopoietic Stem Cell Transplantation: Need for a Unified Paradigm for Donor Selection. Front Immunol 2022; 13:821533. [PMID: 35242134 PMCID: PMC8886110 DOI: 10.3389/fimmu.2022.821533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/06/2022] [Indexed: 11/29/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (aHSCT) is a lifesaving therapy for hematological malignancies. For years, a fully matched HLA donor was a requisite for the procedure. However, new immunosuppressive strategies have enabled the recruitment of viable alternative donors, particularly haploidentical donors. Over 95% of patients have at least two potential haploidentical donors available to them. To identify the best haploidentical donor, the assessment of new immunogenetic criteria could help. To this end, the clinical benefit of KIR genotyping in aHSCT has been widely studied but remains contentious. This review aims to evaluate the importance of KIR-driven NK cell alloreactivity in the context of aHSCT and explain potential reasons for the discrepancies in the literature. Here, through a non-systematic review, we highlight how the studies in this field and their respective predictive models or scoring strategies could be conceptually opposed, explaining why the role of NK cells remains unclear in aHCST outcomes. We evaluate the limitations of each published prediction model and describe how every scoring strategy to date only partly delivers the requirements for optimally effective NK cells in aHSCT. Finally, we propose approaches toward finding the optimal use of KIR genotyping in aHSCT for a unified criterion for donor selection.
Collapse
Affiliation(s)
- Adèle Dhuyser
- Histocompatibility Laboratory, CHRU de Nancy, Vandoeuvre-les-Nancy, France
- IMoPA6, UMR7365 CNRS, Université de Lorraine, Vandoeuvre-les-Nancy, France
| | - Alice Aarnink
- Histocompatibility Laboratory, CHRU de Nancy, Vandoeuvre-les-Nancy, France
- IMoPA6, UMR7365 CNRS, Université de Lorraine, Vandoeuvre-les-Nancy, France
| | - Michaël Pérès
- Histocompatibility Laboratory, CHRU de Nancy, Vandoeuvre-les-Nancy, France
| | - Jyothi Jayaraman
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Neda Nemat-Gorgani
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Marie Thérèse Rubio
- IMoPA6, UMR7365 CNRS, Université de Lorraine, Vandoeuvre-les-Nancy, France
- Department of Hematology, CHRU de Nancy, Vandoeuvre-les-Nancy, France
| | - John Trowsdale
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - James Traherne
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
24
|
Hillier K, Harris EM, Berbert L, Pai SY, Grace RF. Characteristics and outcomes of autoimmune hemolytic anemia after pediatric allogeneic stem cell transplant. Pediatr Blood Cancer 2022; 69:e29410. [PMID: 34709706 DOI: 10.1002/pbc.29410] [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: 05/13/2021] [Accepted: 09/29/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND Autoimmune hemolytic anemia (AIHA) after allogeneic hematopoietic stem cell transplant (HSCT) is a rare but complex and serious complication. Detailed descriptions of cases and management strategies are needed due to lack of prospective trials. OBJECTIVES Describe the incidence, clinical characteristics, and management of AIHA after HSCT in a pediatric cohort. METHODS This is a retrospective cohort study of 33 pediatric patients with AIHA after HSCT at an academic tertiary care center from 2003 to 2019. RESULTS The overall incidence of AIHA after allogeneic HSCT was 3.8% (33/868). AIHA was significantly more common after transplant for nonmalignant versus malignant diagnoses (7.0% [26/370] vs. 1.4% [7/498], p < .0001). AIHA developed at a median of 4.7 months (range 1.0-29.7) after transplant. Sixteen of 33 patients (48.5%) required new AIHA-directed pharmacologic therapy; 17 (51.5%) were managed on their current immunosuppression and supportive care. Patients managed without additional therapy were significantly older, more likely to have a malignant diagnosis, and tended to develop AIHA at an earlier time point after transplant. Patients received a median of two red blood cell transfusions within the first 2 weeks of diagnosis and a median of one AIHA-directed medication (range one to four), most commonly corticosteroids and rituximab. CONCLUSIONS AIHA after HSCT is rare but occurs more commonly in patients transplanted for nonmalignant diagnoses. While some pediatric patients who develop AIHA after transplant can be managed on current immunosuppression and supportive care, many require AIHA-directed therapy including second-line medications.
Collapse
Affiliation(s)
- Kirsty Hillier
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA.,Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Emily M Harris
- Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Berbert
- Clinical Research Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sung-Yun Pai
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rachael F Grace
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Krishnamurti L. Hematopoietic cell transplantation for sickle cell disease: updates and future directions. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:181-189. [PMID: 34889368 PMCID: PMC8791142 DOI: 10.1182/hematology.2021000251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Excellent outcomes in hematopoietic cell transplantation (HCT) from HLA-identical siblings, improvements in conditioning regimens, novel graft-versus-host disease prophylaxis, and the availability of alternative donors have all contributed to the increased applicability and acceptability of HCT for sickle cell disease (SCD). In young children with symptomatic SCD with an available HLA-identical related donor, HCT should be carefully considered. HCT from alternative donors is typically undertaken only in patients with severe symptoms, causing or likely to cause organ damage, and in the context of clinical trials. Patients undergoing HCT for SCD require careful counseling and preparation. They require careful monitoring of unique organ toxicities and complications during HCT. Patients must be prospectively followed for a prolonged time to determine the long-term outcomes and late effects of HCT for SCD. Thus, there is a need for a universal, longitudinal clinical registry to follow patients after HCT for SCD in conjunction with individuals who do not receive HCT to compare outcomes. Antibody-based conditioning and ex-vivo umbilical cord blood expansion are likely to improve the availability and acceptability of HCT. In addition, new disease-modifying drugs and the emerging option of the autologous transplantation of gene-modified hematopoietic progenitor cells are likely to expand the available therapeutic options and make decision-making by patients, physicians, and caregivers even more complicated. Future efforts must also focus on determining the impact of socioeconomic status on access to and outcomes of HCT and the long-term impact of HCT on patients, families, and society.
Collapse
Affiliation(s)
- Lakshmanan Krishnamurti
- Correspondence Lakshmanan Krishnamurti, Children's Healthcare of Atlanta-Egleston, 1405 Clifton Road NE, Atlanta, GA 30322; e-mail:
| |
Collapse
|
26
|
Leonard A, Bertaina A, Bonfim C, Cohen S, Prockop S, Purtill D, Russell A, Boelens JJ, Wynn R, Ruggeri A, Abraham A. Curative therapy for hemoglobinopathies: an International Society for Cell & Gene Therapy Stem Cell Engineering Committee review comparing outcomes, accessibility and cost of ex vivo stem cell gene therapy versus allogeneic hematopoietic stem cell transplantation. Cytotherapy 2021; 24:249-261. [PMID: 34879990 DOI: 10.1016/j.jcyt.2021.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/23/2021] [Accepted: 09/04/2021] [Indexed: 12/17/2022]
Abstract
Thalassemia and sickle cell disease (SCD) are the most common monogenic diseases in the world and represent a growing global health burden. Management is limited by a paucity of disease-modifying therapies; however, allogeneic hematopoietic stem cell transplantation (HSCT) and autologous HSCT after genetic modification offer patients a curative option. Allogeneic HSCT is limited by donor selection, morbidity and mortality from transplant conditioning, graft-versus-host disease and graft rejection, whereas significant concerns regarding long-term safety, efficacy and cost limit the broad applicability of gene therapy. Here the authors review current outcomes in allogeneic and autologous HSCT for transfusion-dependent thalassemia and SCD and provide our perspective on issues surrounding accessibility and costs as barriers to offering curative therapy to patients with hereditary hemoglobinopathies.
Collapse
Affiliation(s)
- Alexis Leonard
- Division of Hematology, Children's National Hospital, Washington, DC, USA
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Carmem Bonfim
- Pediatric Bone Marrow Transplantation Division, Hospital Pequeno Principe, Curitiba, Brazil
| | - Sandra Cohen
- Université de Montréal and Maisonneuve Rosemont Hospital, Montréal, Canada
| | - Susan Prockop
- Stem Cell Transplantation and Cellular Therapies, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Duncan Purtill
- Department of Haematology, Fiona Stanley Hospital, Perth, Australia
| | - Athena Russell
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, 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
| | - Robert Wynn
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Annalisa Ruggeri
- Department of Hematology and bone marrow transplantation, IRCCS Ospedale San Raffaele, Segrate, Milan, Italy
| | - Allistair Abraham
- Center for Cancer and Immunology Research, CETI, Children's National Hospital, Washington, DC, USA.
| |
Collapse
|
27
|
HSCT remains the only cure for patients with transfusion-dependent thalassemia until gene therapy strategies are proven to be safe. Bone Marrow Transplant 2021; 56:2882-2888. [PMID: 34531544 DOI: 10.1038/s41409-021-01461-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Patients with β-thalassemia suffer from severe anemia, iron overload and multiple complications, that affect their quality of life and well-being. Allogeneic hematopoietic stem cell transplantation (HSCT) from an HLA-matched sibling donor, performed in childhood, has been the gold standard for thalassemic patients for decades. Unfortunately, siblings are available only for the minority of patients. Fully matched unrelated donors have been the second choice for cure, with equal results as far as overall survival is concerned, having though the cost of frequent and serious complications. On the other hand, haploidentical transplantation is performed more frequently during the last decade, with promising results. Gene therapy represents a novel therapeutic approach, with impressive results from clinical trials, both from gene addition strategies, as well as from the emerging gene editing tools. After reviewing current critical points of HSCT using alternative donors and assessing recently reported safety issues of gene therapy methods, we conclude that, although a breakthrough, the safety of gene therapy remains to be established.
Collapse
|
28
|
Ovsyannikova G, Balashov D, Demina I, Shelikhova L, Pshonkin A, Maschan M, Novichkova G, Maschan A, Smetanina N. Efficacy and safety of ruxolitinib in ineffective erythropoiesis suppression as a pretransplantation treatment for pediatric patients with beta-thalassemia major. Pediatr Blood Cancer 2021; 68:e29338. [PMID: 34520107 DOI: 10.1002/pbc.29338] [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: 05/23/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND Ineffective erythropoiesis (IE) is the most prominent feature of transfusion-dependent beta-thalassemia (TDT), which leads to extramedullary hemopoiesis. The rejection rate in allogeneic hematopoietic stem cell transplantation (HSCT) is high in heavily transfused patients with TDT accompanied by prominent IE. Therefore, a pretransplantation treatment bridging to HSCT is often used to reduce allosensitization and IE. Ruxolitinib is a JAK-1/JAK-2 inhibitor and has showed its efficacy in suppressing IE and the immune system. A previously published study on RUX in adult patients with TDT has revealed that this treatment significantly reduces spleen size and is well tolerated. PROCEDURE Ten patients (5-14 years old) with TDT and an enlarged spleen were enrolled. The dose of ruxolitinib was adjusted for age: for patients <11 years: 40-100 mg/m2 total daily dose and for patients >11 years: 20-30 mg/m2 total daily dose. HSCT was performed in 8 of 10 patients. RESULTS After the first 3 months of ruxolitinib therapy, spleen volume decreased in 9 of 10 cases by 9.1%-67.5% (M = 35.4%) compared with the initial size (P = 0.003). The adverse events of ruxolitinib (infectious complications, moderate thrombocytopenia, and headache) were successfully managed by reducing the dose. The outcomes of HSCT were favorable in seven of eight cases. CONCLUSION Ruxolitinib is promising as a short-term pre-HSCT treatment for pediatric patients with TDT and pronounced IE.
Collapse
Affiliation(s)
- Galina Ovsyannikova
- Department of Pediatric Hematology and Oncology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Dmitry Balashov
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Irina Demina
- Laboratory for Immunophenotyping of Hemoblastosis, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Larisa Shelikhova
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexey Pshonkin
- Department of Pediatric Hematology and Oncology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Michael Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Galina Novichkova
- Department of Pediatric Hematology and Oncology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexey Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Nataliya Smetanina
- Department of Pediatric Hematology and Oncology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
29
|
Luo XH, Zhu Y, Chen YT, Shui LP, Liu L. CMV Infection and CMV-Specific Immune Reconstitution Following Haploidentical Stem Cell Transplantation: An Update. Front Immunol 2021; 12:732826. [PMID: 34777342 PMCID: PMC8580860 DOI: 10.3389/fimmu.2021.732826] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/14/2021] [Indexed: 02/05/2023] Open
Abstract
Haploidentical stem cell transplantation (haploSCT) has advanced to a common procedure for treating patients with hematological malignancies and immunodeficiency diseases. However, cure is seriously hampered by cytomegalovirus (CMV) infections and delayed immune reconstitution for the majority of haploidentical transplant recipients compared to HLA-matched stem cell transplantation. Three major approaches, including in vivo T-cell depletion (TCD) using antithymocyte globulin for haploSCT (in vivo TCD-haploSCT), ex vivo TCD using CD34 + positive selection for haploSCT (ex vivo TCD-haploSCT), and T-cell replete haploSCT using posttransplant cyclophosphamide (PTCy-haploSCT), are currently used worldwide. We provide an update on CMV infection and CMV-specific immune recovery in this fast-evolving field. The progress made in cellular immunotherapy of CMV infection after haploSCT is also addressed. Groundwork has been prepared for the creation of personalized avenues to enhance immune reconstitution and decrease the incidence of CMV infection after haploSCT.
Collapse
Affiliation(s)
- Xiao-Hua Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zhu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu-Ting Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li-Ping Shui
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
30
|
Successful mismatched hematopoietic stem cell transplantation for pediatric hemoglobinopathy by using ATG and post-transplant cyclophosphamide. Bone Marrow Transplant 2021; 56:2203-2211. [PMID: 33941871 DOI: 10.1038/s41409-021-01302-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 02/05/2023]
Abstract
The use of HLA-mismatched (un)related donors is historically associated with a higher incidence of transplant-related complications and mortality. However, the use of such donors may overcome the limited availability of HLA-matched donors for patients with β-thalassemia major (TM) and sickle cell disease (SCD). We investigated hematopoietic stem cell transplantation (HSCT) outcomes of pediatric TM and SCD patients treated with a mismatched donor using a treosulfan-based conditioning in combination with ATG and post-transplant cyclophosphamide (PT-CY) and compared these results to the clinical outcome of patients treated by matched donor HSCT without PT-CY. Thirty-eight children (n = 24 HLA-identical or 10/10-matched donors; n = 14 HLA-mismatched donors), who received a non-depleted bone marrow graft were included. Event-free survival (EFS) and GvHD were not higher in the mismatched PT-Cy group as compared to the matched group. Moreover, despite delayed neutrophil engraftment (day +22 vs. +26, p = 0.002) and immune recovery in the mismatched PT-Cy group, this did not result in more infectious complications. Therefore, we conclude that in the absence of an HLA-identical or a matched unrelated donor, HSCT with a mismatched unrelated or haploidentical donor in combination with ATG plus PT-CY can be considered a safe and effective treatment option for pediatric hemoglobinopathy patients.
Collapse
|
31
|
αβ T-cell graft depletion for allogeneic HSCT in adults with hematological malignancies. Blood Adv 2021; 5:240-249. [PMID: 33570642 DOI: 10.1182/bloodadvances.2020002444] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
We conducted a multicenter prospective single-arm phase 1/2 study that assesses the outcome of αβ T-cell depleted allogeneic hematopoietic stem cell transplantation (allo-HSCT) of peripheral blood derived stem cells from matched related, or unrelated donors (10/10 and 9/10) in adults, with the incidence of acute graft-versus-host disease (aGVHD) as the primary end point at day 100. Thirty-five adults (median age, 59; range, 19-69 years) were enrolled. Conditioning consisted of antithymocyte globulin, busulfan, and fludarabine, followed by 28 days of mycophenolic acid after allo-HSCT. The minimal follow-up time was 24 months. The median number of infused CD34+ cells and αβ T cells were 6.1 × 106 and 16.3 × 103 cells per kg, respectively. The cumulative incidence (CI) of aGVHD grades 2-4 and 3-4 at day 100 was 26% and 14%. One secondary graft failure was observed. A prophylactic donor lymphocyte infusion (DLI) (1 × 105 CD3+ T cells per kg) was administered to 54% of the subjects, resulting in a CI of aGVHD grades 2-4 and 3-4 to 37% and 17% at 2 years. Immune monitoring revealed an early reconstitution of natural killer (NK) and γδ T cells. Cytomegalovirus reactivation associated with expansion of memory-like NK cells. The CI of relapse was 29%, and the nonrelapse mortality 32% at 2 years. The 2-year CI of chronic GVHD (cGVHD) was 23%, of which 17% was moderate. We conclude that only 26% of patients developed aGVHD 2-4 after αβ T-cell-depleted allo-HSCT within 100 days and was associated with a low incidence of cGVHD after 2 years. This trial was registered at www.trialregister.nl as #NL4767.
Collapse
|
32
|
Contemporary haploidentical stem cell transplant strategies in children with hematological malignancies. Bone Marrow Transplant 2021; 56:1518-1534. [PMID: 33674791 DOI: 10.1038/s41409-021-01246-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/01/2021] [Accepted: 02/15/2021] [Indexed: 12/14/2022]
Abstract
The barriers to HLA-mismatched or haploidentical hematopoietic stem cell transplantation (HSCT), namely GvHD and graft failure, have been overcome with novel transplant platforms. Post-transplant Cyclophosphamide (PTCy) is widely available, feasible and easy to implement. TCRαβ T and B cell depletion comes with consistent GvHD preventive benefits irrespective of age and indication. Naive T-cell depletion helps prevention of severe viral reactivations. The Beijing protocol shows promising outcomes in patients with poor remission status at the time of transplantation. For children, the toxicities and late outcomes related to these transplants are truly relevant as they suffer the most in the long run from transplant-related toxicities, especially chronic GvHD. While comparing the outcomes of different Haplo-HSCT approaches, one must understand the transplant immunobiology and factors affecting the transplant outcomes. Leukemia remission status at the time of conditioning is a consistent factor affecting the transplant outcomes using any of these platforms. Prospective comparison of these platforms lacks in a homogenous population; however, the evidence is growing, and this review highlights the areas of research gaps.
Collapse
|
33
|
Williams KM, Inamoto Y, Im A, Hamilton B, Koreth J, Arora M, Pusic I, Mays JW, Carpenter PA, Luznik L, Reddy P, Ritz J, Greinix H, Paczesny S, Blazar BR, Pidala J, Cutler C, Wolff D, Schultz KR, Pavletic SZ, Lee SJ, Martin PJ, Socie G, Sarantopoulos S. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: I. The 2020 Etiology and Prevention Working Group Report. Transplant Cell Ther 2021; 27:452-466. [PMID: 33877965 DOI: 10.1016/j.jtct.2021.02.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
Preventing chronic graft-versus-host disease (GVHD) remains challenging because the unique cellular and molecular pathways that incite chronic GVHD are poorly understood. One major point of intervention for potential prevention of chronic GVHD occurs at the time of transplantation when acute donor anti-recipient immune responses first set the events in motion that result in chronic GVHD. After transplantation, additional insults causing tissue injury can incite aberrant immune responses and loss of tolerance, further contributing to chronic GVHD. Points of intervention are actively being identified so that chronic GVHD initiation pathways can be targeted without affecting immune function. The major objective in the field is to continue basic studies and to translate what is learned about etiopathology to develop targeted prevention strategies that decrease the risk of morbid chronic GVHD without increasing the risks of cancer relapse or infection. Development of strategies to predict the risk of developing debilitating or deadly chronic GVHD is a high research priority. This working group recommends further interrogation into the mechanisms underpinning chronic GVHD development, and we highlight considerations for future trial design in prevention trials.
Collapse
Affiliation(s)
- Kirsten M Williams
- Division of Blood and Marrow Transplantation, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Yoshihiro Inamoto
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Annie Im
- Division of Hematology Oncology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Betty Hamilton
- Blood and Marrow Transplant Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - John Koreth
- Dana-Farber Cancer Institute, Division of Hematologic Malignancies, Harvard Medical School, Boston, Massachusetts
| | - Mukta Arora
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Iskra Pusic
- BMT and Leukemia Section, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Jacqueline W Mays
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Leo Luznik
- Division of Hematologic Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pavan Reddy
- Divsion of Hematology and Oncology, University of Michigan Rogel Cancer Center, Ann Arbor, Michigan
| | - Jerome Ritz
- Dana-Farber Cancer Institute, Division of Hematologic Malignancies, Harvard Medical School, Boston, Massachusetts
| | - Hildegard Greinix
- Clinical Division of Hematology, Medical University of Graz, Graz, Austria
| | - Sophie Paczesny
- Department of Microbiology and Immunology and Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Bruce R Blazar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Joseph Pidala
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Corey Cutler
- Dana-Farber Cancer Institute, Division of Hematologic Malignancies, Harvard Medical School, Boston, Massachusetts
| | - Daniel Wolff
- Department of Internal Medicine III, University Hospital of Regensburg, Regensburg, Germany
| | - Kirk R Schultz
- Pediatric Oncology, Hematology, and Bone Marrow Transplant, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Steven Z Pavletic
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Paul J Martin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Gerard Socie
- Hematology Transplantation, Saint Louis Hospital, AP-HP, and University of Paris, INSERM U976, Paris, France.
| | - Stefanie Sarantopoulos
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke Cancer Institute, Durham, North Carolina.
| |
Collapse
|
34
|
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
|
35
|
Wang X, Zhang X, Yu U, Wang C, Yang C, Li Y, Li C, Wen F, Li C, Liu S. Co-Transplantation of Haploidentical Stem Cells and a Dose of Unrelated Cord Blood in Pediatric Patients with Thalassemia Major. Cell Transplant 2021; 30:963689721994808. [PMID: 33593080 PMCID: PMC7894585 DOI: 10.1177/0963689721994808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Allogeneic stem cell transplantation is a cure for patients suffering from thalassemia major (TM). Historically, patients were limited by the selection of donors, while the advancement of haploidentical stem cell transplantation (haplo-SCT) has greatly expanded the donor pool. However, the outcomes of haplo-SCT in TM recipients vary between different programs. In this study, we retrospectively studied 73 pediatric TM patients (median age, 7 years; range, 3 to 14 years) who underwent haplo-cord transplantation. Both the estimated overall survival and transfusion-free survival were 95.26% (CI 95.77% to 96.23%). Neither primary nor secondary graft failures were observed. The median follow-up period was 811 days (range, 370 to 1433 days). Median neutrophil and platelet engraftment times were 22 days (range, 8 to 48 days) and 20 days (range, 8 to 99 days), respectively. Acute graft-versus-host disease (aGVHD) was observed in 52% of patients and of these, 25% developed grade III to IV aGVHD. Cord blood engraftment was associated with delayed immune recovery and increased aGVHD severity. Viral DNAemia occurred in a relatively high proportion of patients but only 7% of patients developed CMV disease, while another 7% of patients had post-transplantation lymphoproliferative disorder. Long-term complication outcomes were good. Only one patient developed extensive chronic GVHD. No surviving patients were reliant on blood transfusion by the time this manuscript was submitted. This is one of the largest studies on the outcomes of pediatric TM patients who received stem cell transplantations from alternative donors. The haplo-cord program is safe and practical for TM patients that do not have matched donors.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Xiaoling Zhang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Uet Yu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Chunjing Wang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Chunlan Yang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Yue Li
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Changgang Li
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Feiqiu Wen
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Chunfu Li
- Nanfang-Chunfu Children's Institute of Hematology and Oncology, Taixin Hospital, Dongguan, China.,Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sixi Liu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| |
Collapse
|
36
|
Pathak S, Meyer EH. Tregs and Mixed Chimerism as Approaches for Tolerance Induction in Islet Transplantation. Front Immunol 2021; 11:612737. [PMID: 33658995 PMCID: PMC7917336 DOI: 10.3389/fimmu.2020.612737] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/14/2020] [Indexed: 01/07/2023] Open
Abstract
Pancreatic islet transplantation is a promising method for the treatment of type 1 and type 3 diabetes whereby replacement of islets may be curative. However, long-term treatment with immunosuppressive drugs (ISDs) remains essential for islet graft survival. Current ISD regimens carry significant side-effects for transplant recipients, and are also toxic to the transplanted islets. Pre-clinical efforts to induce immune tolerance to islet allografts identify ways in which the recipient immune system may be reeducated to induce a sustained transplant tolerance and even overcome autoimmune islet destruction. The goal of these efforts is to induce tolerance to transplanted islets with minimal to no long-term immunosuppression. Two most promising cell-based therapeutic strategies for inducing immune tolerance include T regulatory cells (Tregs) and donor and recipient hematopoietic mixed chimerism. Here, we review preclinical studies which utilize Tregs for tolerance induction in islet transplantation. We also review myeloablative and non-myeloablative hematopoietic stem cell transplantation (HSCT) strategies in preclinical and clinical studies to induce sustained mixed chimerism and allograft tolerance, in particular in islet transplantation. Since Tregs play a critical role in the establishment of mixed chimerism, it follows that the combination of Treg and HSCT may be synergistic. Since the success of the Edmonton protocol, the feasibility of clinical islet transplantation has been established and nascent clinical trials testing immune tolerance strategies using Tregs and/or hematopoietic mixed chimerism are underway or being formulated.
Collapse
Affiliation(s)
- Shiva Pathak
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, United States
| | - Everett H. Meyer
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
37
|
Choudhary D, Doval D, Sharma SK, Khandelwal V, Setia R, Handoo A. T-cell replete Haplo-identical HSCT with Post transplant Cyclophosphamide for Hemoglobinopathies: A retrospective analysis from a single center. BLOOD CELL THERAPY 2021; 4:29-34. [PMID: 36712899 PMCID: PMC9847306 DOI: 10.31547/bct-2020-014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/14/2020] [Indexed: 02/01/2023]
Abstract
We report herein haplo-identical hematopoietic stem cell transplantation (haplo-HSCT) by T-cell replete graft infusion, with post-transplant cyclophosphamide (PTCy) in patients with hemoglobinopathies. Patients received a conditioning regimen consisting of either busulfan, fludarabine, cyclophosphamide, with antithymocyte globulin or Thiotepa, antithymocyte globulin, fludarabine, cyclophosphamide, and TBI. The median follow-up period was 14.3 months (range, 1-63 months). Overall survival (OS) and disease-free survival (DFS) were 80% and 62.8%, respectively. Incidence of secondary graft failure was 14%. Incidences of acute graft-versus-host disease (aGvHD) and chronic graft-versus-host disease (cGvHD) were 22.5% and 20%, respectively. Cytomegalovirus (CMV) reactivation was observed in 42.5% of cases. The 100-day mortality rate was 20%, with sepsis and aGvHD being the predominant causes of death.
Collapse
Affiliation(s)
- Dharma Choudhary
- Centre for Bone Marrow Transplant, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Divya Doval
- Centre for Bone Marrow Transplant, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Sanjeev Kumar Sharma
- Centre for Bone Marrow Transplant, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Vipin Khandelwal
- Centre for Bone Marrow Transplant, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Rasika Setia
- Centre for Bone Marrow Transplant, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| | - Anil Handoo
- Centre for Bone Marrow Transplant, BLK Superspeciality Hospital, Pusa Road, New Delhi, India
| |
Collapse
|
38
|
Krishnamurti L. Hematopoietic Cell Transplantation for Sickle Cell Disease. Front Pediatr 2021; 8:551170. [PMID: 33469520 PMCID: PMC7813811 DOI: 10.3389/fped.2020.551170] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Sickle cell disease (SCD) is a severe autosomal recessively inherited disorder of the red blood cell characterized by erythrocyte deformation caused by the polymerization of the abnormal hemoglobin, which leads to erythrocyte deformation and triggers downstream pathological changes. These include abnormal rheology, vaso-occlusion, ischemic tissue damage, and hemolysis-associated endothelial dysfunction. These acute and chronic physiologic disturbances contribute to morbidity, organ dysfunction, and diminished survival. Hematopoietic cell transplantation (HCT) from HLA-matched or unrelated donors or haploidentical related donors or genetically modified autologous hematopoietic progenitor cells is performed with the intent of cure or long-term amelioration of disease manifestations. Excellent outcomes have been observed following HLA-identical matched related donor HCT. The majority of SCD patients do not have an available HLA-identical sibling donor. Increasingly, however, they have the option of undergoing HCT from unrelated HLA matched or related haploidentical donors. The preliminary results of transplantation of autologous hematopoietic progenitor cells genetically modified by adding a non-sickling gene or by genomic editing to increase expression of fetal hemoglobin are encouraging. These approaches are being evaluated in early-phase clinical trials. In performing HCT in patients with SCD, careful consideration must be given to patient and donor selection, conditioning and graft-vs.-host disease regimen, and pre-HCT evaluation and management during and after HCT. Sociodemographic factors may also impact awareness of and access to HCT. Further, there is a substantial decisional dilemma in HCT with complex tradeoffs between the possibility of amelioration of disease manifestations and early or late complications of HCT. The performance of HCT for SCD requires careful multidisciplinary collaboration and shared decision making between the physician and informed patients and caregivers.
Collapse
Affiliation(s)
- Lakshmanan Krishnamurti
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| |
Collapse
|
39
|
Buxbaum NP, Pavletic SZ. Autoimmunity Following Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2020; 11:2017. [PMID: 32983144 PMCID: PMC7479824 DOI: 10.3389/fimmu.2020.02017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/24/2020] [Indexed: 12/28/2022] Open
Abstract
Autoimmune manifestations after allogeneic hematopoietic stem cell transplantation (AHSCT) are rare and poorly understood due to the complex interplay between the reconstituting immune system and transplant-associated factors. While autoimmune manifestations following AHSCT have been observed in children with graft-versus-host disease (GvHD), an alloimmune process, they are distinct from the latter in that they are generally restricted to the hematopoietic compartment, i.e., autoimmune hemolytic anemia, thrombocytopenia, and/or neutropenia. Autoimmune cytopenias in the setting of ASHCT represent a donor against donor immune reaction. Non-hematologic autoimmune conditions in the post-AHSCT setting have been described and do not currently fall under the GvHD diagnostic criteria, but could represent alloimmunity since they arise from the donor immune attack on the antigens that are shared by the donor and host in the thyroid, peripheral and central nervous systems, integument, liver, and kidney. As in the non-transplant setting, autoimmune conditions are primarily antibody mediated. In this article we review the incidence, risk factors, potential pathophysiology, treatment, and prognosis of hematologic and non-hematologic autoimmune manifestations in children after AHSCT.
Collapse
Affiliation(s)
- Nataliya Prokopenko Buxbaum
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Steven Z Pavletic
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
40
|
Kwon M, Bailén R, Díez-Martín JL. Evolution of the role of haploidentical stem cell transplantation: past, present, and future. Expert Rev Hematol 2020; 13:835-850. [PMID: 32749913 DOI: 10.1080/17474086.2020.1796621] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The accessibility to haplo-donors has led to an increase in the number of haplo-HSCT worldwide. A systematic search of the PubMed database between 2000 to present was performed. AREAS COVERED In this review, the authors discussed the most used approaches to perform haplo-HSCT and its results: T-cell depletion (TCD, including Perugia platform and its modifications) and T-cell repleted haplo (TCR, including the high-dose post-transplant cyclophosphamide strategy (Baltimore protocol) and the Beijing protocol). The improvements and modifications made to the different strategies have increased the indications of haplo-HSCT, including both malignant and nonmalignant disorders. Focusing on the Baltimore protocol, the authors review the results of the retrospective studies that have compared it to other donor transplants. The limitations of this strategy in terms of toxicity, graft complications, and GVHD are also discussed in detail. Finally, possible approaches to improve the outcomes of TCR haplo-HSCT are presented. EXPERT OPINION The recent advances in the field of haplo-HSCT have allowed a large number of patients with incurable diseases to benefit from this procedure despite not having a matched donor. With all available strategies, virtually no patient who needs an allogeneic transplant should be excluded by the absence of a donor.
Collapse
Affiliation(s)
- Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón , Madrid, Spain.,Departement of Translational Oncology, Institute of Health Research Gregorio Marañón , Madrid, Spain
| | - Rebeca Bailén
- Department of Hematology, Hospital General Universitario Gregorio Marañón , Madrid, Spain.,Departement of Translational Oncology, Institute of Health Research Gregorio Marañón , Madrid, Spain
| | - José Luis Díez-Martín
- Department of Hematology, Hospital General Universitario Gregorio Marañón , Madrid, Spain.,Departement of Translational Oncology, Institute of Health Research Gregorio Marañón , Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid , Madrid, Spain
| |
Collapse
|
41
|
Nazir HF, Ba Alawi FS, Al Hosni S, Al Rawas A, Dennison D. T Cell Depleted Haploidentical Hematopoietic Stem Cell Transplantation for Patients with Familial Hemophagocytic Lymphohistiocytosis Who Do Not Have Matched Family Donors: Experience in Oman. Biol Blood Marrow Transplant 2020; 26:1119-1123. [DOI: 10.1016/j.bbmt.2020.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 12/26/2022]
|
42
|
Jaiswal SR, Bhakuni P, Aiyer HM, Soni M, Bansal S, Chakrabarti S. CTLA4Ig in an Extended Schedule along with Sirolimus Improves Outcome with a Distinct Pattern of Immune Reconstitution Following Post-Transplantation Cyclophosphamide-Based Haploidentical Transplantation for Hemoglobinopathies. Biol Blood Marrow Transplant 2020; 26:1469-1476. [PMID: 32428732 DOI: 10.1016/j.bbmt.2020.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 01/24/2023]
Abstract
The major hindrances to the success of a haploidentical hematopoietic cell transplantation for hemoglobinopathies are graft failure, early post-transplant hemophagocytic syndrome (PTHPS), and graft-versus-host disease (GVHD). Following the successful incorporation of CTLA4Ig (abatacept) in post-transplantation cyclophosphamide-based haploidentical transplantation, we piloted this approach in 10 patients (aged 3 to 19 years), with thalassemia major (TM, n=5) and sickle cell disease (n = 5). Pretransplant immunosuppressive therapy (pTIST) was administered for 10 weeks. Conditioning was myeloablative. CTLA4Ig was administered every 2 weeks during pTIST and on days -1, +5, +20, and +35 and every 4 weeks thereafter for 6 months, along with sirolimus. A short course of low-dose dexamethasone was given from day +6 for 14 days. Nine patients engrafted at a median of 15 days, with 1 patient with TM dying of sepsis on day +19. None of the patients developed acute or chronic GVHD. All 9 patients are alive and disease free at a median follow-up of 28 months. Only 4 patients had cytomegalovirus reactivation. The pattern of immune reconstitution showed a prompt and sustained recovery of T cell subsets with memory phenotype, along with early and sustained increase of Tregs and NKG2C+ natural killer (NK) cells. This novel approach, targeting CD80 and CD86 on monocytes/macrophages, promoted engraftment and limited early-onset PTHPS and graft failure. The lack of GVHD and serious infections with this approach reflects an early recovery of Tregs, memory T cells, and persistence of NKG2C+ NK cells.
Collapse
Affiliation(s)
- Sarita Rani Jaiswal
- Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, Kolkata, India; Department of Blood and Marrow Transplantation, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India.
| | - Prakash Bhakuni
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Hema Malini Aiyer
- Department of Anatomic Pathology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Mayank Soni
- Department of Blood and Marrow Transplantation, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Satish Bansal
- Department of Radiology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Suparno Chakrabarti
- Cellular Therapy and Immunology, Manashi Chakrabarti Foundation, Kolkata, India; Department of Blood and Marrow Transplantation, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| |
Collapse
|
43
|
Swaminathan VV, Uppuluri R, Patel S, Ravichandran N, Ramanan KM, Vaidhyanathan L, Ramakrishnan B, Jayakumar I, Raj R. Matched Family versus Alternative Donor Hematopoietic Stem Cell Transplantation for Patients with Thalassemia Major: Experience from a Tertiary Referral Center in South India. Biol Blood Marrow Transplant 2020; 26:1326-1331. [PMID: 32200123 DOI: 10.1016/j.bbmt.2020.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 01/19/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) is the only curative option available for patients with thalassemia major in India with increasing access to alternate donor transplantation for patients with no matched family donor. We aimed to analyze the impact of family and alternate donor HSCT on morbidity and mortality post-HSCT. We conducted a retrospective study in the department between July 2007 and December 2018 where all children who underwent HSCT for thalassemia major were included. A total of 264 children were included with a median age of 6 years (male/female, 1.4:1). The graft source was matched related donor (MRD) (76%; parent 15%, sibling 85%) and matched unrelated donor (MUD) (22%). All children received a myeloablative conditioning regimen with treosulfan/thiotepa/fludarabine in 93% and busulfan/cyclophosphamide in 7%. The source of stem cells was peripheral blood in 61%, bone marrow in 38%, and umbilical cord blood in 3%. The incidence of bacteremia was 14% versus 25% in MRD versus MUD groups. There was a higher incidence of posterior reversible encephalopathy syndrome (PRES) in the MUD group (10% versus 3%). Engraftment occurred in 97% with a higher trend toward mixed chimerism in the MRD group (12% versus 2%). When indicated, whole-blood donor lymphocyte infusion was used to ensure complete chimerism in children in the MRD group. A statistically significant difference was found in the incidence of graft versus host disease (GVHD), both acute and chronic between the MUD versus MRD groups, 60% versus 20% and 41% versus 17%, respectively (P = .001). Similarly, immune cytopenia and cytomegalovirus reactivation were also significantly higher in the MUD group, 27% versus 1.4% and 25% versus 2%, respectively (P = .001). Thalassemia-free survival in our cohort was 96%, 94%, and 84% with a median follow-up of 65 months in the matched sibling donor, matched family donor, and MUD groups, respectively. Overall survival of 95% and 90% with a median follow-up of 65 months was noted in those who underwent transplantation less than and greater than 7 years of age, respectively. MUD transplantation for patients with thalassemia major involves specific challenges such as PRES and unusual manifestations of GVHD such as immune cytopenia. Early interventions to optimize supportive care and measures to reduce GVHD are required to ensure survival rates of over 90%.
Collapse
Affiliation(s)
| | - Ramya Uppuluri
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India.
| | - Shivani Patel
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | - Nikila Ravichandran
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | - Kesavan Melarcode Ramanan
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | - Lakshman Vaidhyanathan
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India; Department of Haematology, Apollo Hospitals, Chennai, India
| | - Balasubramaniam Ramakrishnan
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | - Indira Jayakumar
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India; Department of Paediatric Critical Care, Apollo Hospitals, Chennai, India
| | - Revathi Raj
- Department of Paediatric Haematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| |
Collapse
|
44
|
Foell J, Kleinschmidt K, Jakob M, Troeger A, Corbacioglu S. Alternative donor: αß/CD19 T-cell-depleted haploidentical hematopoietic stem cell transplantation for sickle cell disease. Hematol Oncol Stem Cell Ther 2020; 13:98-105. [PMID: 32202248 DOI: 10.1016/j.hemonc.2019.12.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 12/28/2019] [Indexed: 10/24/2022] Open
Abstract
Sickle cell disease (SCD) is an inherited disorder; despite significant improvements in supportive care, SCD continues to cause substantial morbidity, mortality, and reduced life expectancy. Allogeneic hematopoietic stem cell transplantation (HSCT) is currently the only widely available curative therapy for SCD, which is offered as a standard of care for patients with a matched sibling donor (MSD). Donor availability is limited to a minority of patients. Thus, αβ/CD3-depleted haploidentical HSCT, as an efficient means for depletion of graft-versus-host disease (GvHD)-mediating T cells, can be offered as an alternative curative therapy, particularly for nonmalignant diseases such as SCD. Out of 38 patients with advanced stage SCD, 25 were transplanted with CD3/CD19- or T-cell receptor αβ/CD19 T-cell-depleted peripheral stem cell grafts (T-haplo-HSCT group), whereas 13 transplanted from MSD (MSD group); both groups received an almost identical conditioning regimen. Engraftment was achieved in all. However, in the T-haplo-HSCT group, three patients succumbed to an uncontrolled cytomegalovirus pneumonitis, a macrophage activation syndrome, and a major blood group incompatibility with a late graft failure and multiorgan failure. The overall survival was 88% and 100% in T-haplo-HSCT and MSD groups, respectively. None of our patients developed a Glucksberg Grade III-IV acute GvHD. Four patients (16%) in the T-haplo-HSCT group and two patients (15%) in the MSD group developed a steroid-sensitive, mild-to-moderate chronic GvHD that resolved within 18 months posttransplant. These results are encouraging and demonstrate the feasibility, safety, and efficacy of T-haplo-HSCT in advanced stage SCD in children and adults, thus offering a curative alternative to majority of patients.
Collapse
Affiliation(s)
- Juergen Foell
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, Regensburg, Germany
| | - Katharina Kleinschmidt
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, Regensburg, Germany
| | - Marcus Jakob
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, Regensburg, Germany
| | - Anja Troeger
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, Regensburg, Germany
| | - Selim Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital of Regensburg, Regensburg, Germany.
| |
Collapse
|
45
|
Oostenbrink LVE, Jol-van der Zijde CM, Jansen-Hoogendijk AM, Pool ES, van Halteren AGS, Moes DJAR, Bredius RGM, Mohseny AB, Smiers FJW, van Tol MJD, Schilham MW, Lankester AC. Proceeding of the European Group for Blood and Marrow Transplantation (EBMT) congress on sickle cell disease, 16-17 may 2019, Regensburg, Germany: What is the impact of antithymocyte globulin pharmacokinetics on haploidentical hematopoietic stem cell transplantation? Hematol Oncol Stem Cell Ther 2020; 13:61-65. [PMID: 32202242 DOI: 10.1016/j.hemonc.2019.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/11/2019] [Indexed: 01/18/2023] Open
Abstract
Antithymocyte globulin (ATG) is a widely accepted part of the conditioning regimen applied in the setting of hematopoietic stem cell transplantation (HSCT) to prevent graft rejection and graft-versus-host disease. Although weight-based dosing of ATG has been introduced to optimize ATG dosing, substantial variance in clearance of active ATG, the actual lymphocyte binding component, remains a challenge. Therefore, further research regarding ATG pharmacokinetics and pharmacodynamics in different HSCT settings and in patients with different types of underlying diseases is required.
Collapse
Affiliation(s)
- Lisa V E Oostenbrink
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands.
| | | | | | - Emma S Pool
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Dirk Jan A R Moes
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Robbert G M Bredius
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Alex B Mohseny
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Frans J W Smiers
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten J D van Tol
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Marco W Schilham
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Arjan C Lankester
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
46
|
Ikawa Y, Miccio A, Magrin E, Kwiatkowski JL, Rivella S, Cavazzana M. Gene therapy of hemoglobinopathies: progress and future challenges. Hum Mol Genet 2020; 28:R24-R30. [PMID: 31322165 DOI: 10.1093/hmg/ddz172] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/26/2022] Open
Abstract
Recently, gene therapy clinical trials have been successfully applied to hemoglobinopathies, such as sickle cell disease (SCD) and β-thalassemia. Among the great discoveries that led to the design of genetic approaches to cure these disorders is the discovery of the β-globin locus control region and several associated transcription factors, which determine hemoglobin switching as well as high-level, erythroid-specific expression of genes at the ß-globin locus. Moreover, increasing evidence shows that lentiviral vectors are efficient tools to insert large DNA elements into nondividing hematopoietic stem cells, showing reassuring safe integration profiles. Alternatively, genome editing could restore expression of fetal hemoglobin or target specific mutations to restore expression of the wild-type β-globin gene. The most recent clinical trials for β-thalassemia and SCD are showing promising outcomes: patients were able to discontinue transfusions or had reduced transfusion requirements. However, toxic myeloablation and the high cost of current ex vivo hematopoietic stem cell gene therapy platforms represent a barrier to a widespread application of these approaches. In this review, we summarize these gene therapy strategies and ongoing clinical trials. Finally, we discuss possible strategies to improve outcomes, reduce myeloablative regimens and future challenges to reduce the cost of gene therapy platform.
Collapse
Affiliation(s)
- Yasuhiro Ikawa
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Annarita Miccio
- Laboratory of chromatin and gene regulation during development, INSERM UMR1163, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Elisa Magrin
- Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Janet L Kwiatkowski
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA.,University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Stefano Rivella
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA.,University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.,Cell and Molecular Biology affinity group (CAMB), University of Pennsylvania.,Penn Center for Musculoskeletal Disorders, CHOP, Philadelphia, PA, USA
| | - Marina Cavazzana
- Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France.,INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis, Paris, France
| |
Collapse
|
47
|
Levine JE, Antin JH, Allen CE, Burroughs LM, Cooke KR, Devine S, Heslop H, Nakamura R, Talano JA, Yanik G, DiFronzo N. Priorities for Improving Outcomes for Nonmalignant Blood Diseases: A Report from the Blood and Marrow Transplant Clinical Trials Network. Biol Blood Marrow Transplant 2020; 26:e94-e100. [PMID: 32035274 DOI: 10.1016/j.bbmt.2020.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/24/2019] [Accepted: 01/28/2020] [Indexed: 01/19/2023]
Abstract
Nonmalignant blood diseases such as bone marrow failure disorders, immune dysregulation disorders, and hemoglobinopathies often lead to shortened life spans and poor quality of life. Many of these diseases can be cured with allogeneic hematopoietic cell transplantation, but patients are often not offered the procedure because of perceived insufficient efficacy and/or excess toxicity. In 2018, the Blood and Marrow Transplant Clinical Trials Network convened a task force to identify the most urgently needed yet feasible clinical trials with potential to improve the outcomes for patients with nonmalignant diseases. This report summarizes the task force discussions and specifies the network plans for clinical trial development for nonmalignant blood diseases.
Collapse
Affiliation(s)
- John E Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Joseph H Antin
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Carl E Allen
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Lauri M Burroughs
- Fred Hutchinson Cancer Research Center, University of Washington School of Medicine, Seattle, Washington; Seattle Children's Hospital, Seattle, Washington
| | - Kenneth R Cooke
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven Devine
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | - Helen Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | | | - Julie An Talano
- Department of Pediatric Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gregory Yanik
- Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Nancy DiFronzo
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
48
|
Leonard A, Tisdale J, Abraham A. Curative options for sickle cell disease: haploidentical stem cell transplantation or gene therapy? Br J Haematol 2020; 189:408-423. [PMID: 32034776 DOI: 10.1111/bjh.16437] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Haematopoietic stem cell transplantation (HSCT) is curative in sickle cell disease (SCD); however, the lack of available matched donors makes this therapy out of reach for the majority of patients with SCD. Alternative donor sources such as haploidentical HSCT expand the donor pool to nearly all patients with SCD, with recent data showing high overall survival, limited toxicities, and effective reduction in acute and chronic graft-versus-host disease (GVHD). Simultaneously, multiple gene therapy strategies are entering clinical trials with preliminary data showing their success, theoretically offering all patients yet another curative strategy without the morbidity and mortality of GVHD. As improvements are made for alternative donors in the allogeneic setting and as data emerge from gene therapy trials, the optimal curative strategy for any individual patient with SCD will be determined by many critical factors including efficacy, transplant morbidity and mortality, safety, patient disease status and preference, cost and applicability. Haploidentical may be the preferred choice now based mostly on availability of data; however, gene therapy is closing the gap and may ultimately prove to be the better option. Progress in both strategies, however, makes cure more attainable for the individual with SCD.
Collapse
Affiliation(s)
- Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) and National Institute of Diabetes, Digestive, and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.,Division of Hematology, Center for Cancer and Blood Disorders, Children's National Health System, Washington, DC, USA.,Blood and Marrow Transplantation, Center for Cancer and Blood Disorders, Children's National Health System, Washington, DC, USA
| | - John Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) and National Institute of Diabetes, Digestive, and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Allistair Abraham
- Blood and Marrow Transplantation, Center for Cancer and Blood Disorders, Children's National Health System, Washington, DC, USA
| |
Collapse
|
49
|
Anurathapan U, Hongeng S, Pakakasama S, Songdej D, Sirachainan N, Pongphitcha P, Chuansumrit A, Charoenkwan P, Jetsrisuparb A, Sanpakit K, Rujkijyanont P, Meekaewkunchorn A, Lektrakul Y, Iamsirirak P, Surapolchai P, Sirireung S, Sruamsiri R, Wahidiyat PA, Andersson BS. Hematopoietic Stem Cell Transplantation for Severe Thalassemia Patients from Haploidentical Donors Using a Novel Conditioning Regimen. Biol Blood Marrow Transplant 2020; 26:1106-1112. [PMID: 31931116 DOI: 10.1016/j.bbmt.2020.01.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Abstract
Patients with severe thalassemia commonly have a survival that is significantly shorter than that of the general population. Allogeneic hematopoietic stem cell transplantation (allo-SCT) is the only established treatment that is potentially curative, but it is limited by the availability of donors and the medical condition of the patient. To expand the donor pool to include haploidentical related donors, we introduced a program consisting of a pharmacologic pretransplant immune suppression phase (PTIS) and 2 courses of dexamethasone and fludarabine, followed by pretransplant conditioning with fludarabine-i.v. busulfan and post-transplant graft-versus-host disease (GVHD) prophylaxis with cyclophosphamide, tacrolimus, and mycophenolate mofetil. We transplanted 83 consecutive transfusion-dependent patients with thalassemia (median age, 12 years; range, 1 to 28 years) with a minimum follow-up of 6 months (median, 15 months; range, 7 to 53 months); the 3-year projected overall and event-free survival is over 96%, and there have been no secondary graft failures. Of the first 31 patients, we had 2 graft failures, both of them occurring in patients with extremely high titers of anti-donor-specific HLA antibodies (anti-DSAs), but after adjusting the PTIS to include bortezomib and rituximab for patients with high titers of anti-DSAs and using pharmacologic dose guidance for busulfan, we had no graft failures in the last 52 patients. Six (7%) of 83 patients developed severe GVHD. We conclude that this is a safe and efficacious approach to allogeneic SCT in thalassemia, yielding results comparable to those available for patients with fully matched donors.
Collapse
Affiliation(s)
- Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Samart Pakakasama
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duantida Songdej
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nongnuch Sirachainan
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pongpak Pongphitcha
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Ampaiwan Chuansumrit
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pimlak Charoenkwan
- Department of Pediatrics, Chiangmai University Hospital, Chiangmai, Thailand
| | | | - Kleebsabai Sanpakit
- Department of Pediatrics, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Piya Rujkijyanont
- Department of Pediatrics, Phramongkutklao Hospital, Bangkok, Thailand
| | | | | | | | | | - Somtawin Sirireung
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rosarin Sruamsiri
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pustika Amalia Wahidiyat
- Department of Child Health, Cipto Mangunkusumo Hospital, Universitas Indonesia, Jakarta, Indonesia
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
50
|
Patel DA, Dhedin N, Chen H, Karnik L, Gatwood K, Culos K, Mohan S, Engelhardt BG, Kitko C, Connelly J, Satyanarayana G, Jagasia M, De La Fuente J, Kassim A. Early viral reactivation despite excellent immune reconstitution following haploidentical Bone marrow transplant with post‐transplant cytoxan for sickle cell disease. Transpl Infect Dis 2019; 22:e13222. [DOI: 10.1111/tid.13222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/11/2019] [Accepted: 11/24/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Dilan A. Patel
- Department of Hematology and Bone Marrow Transplant Vanderbilt University Medical Center Nashville TN USA
| | - Nathalie Dhedin
- Department of Haematology for Adolescents and Young Adults Saint‐Louis Hospital Paris France
| | - Heidi Chen
- Department of Biostatistics Vanderbilt University School of Medicine Nashville TN USA
| | - Leena Karnik
- Department of Paediatrics St. Mary’s Hospital Imperial College London UK
| | - Katie Gatwood
- Department of Pharmaceutical Services Vanderbilt University Medical Center Nashville TN USA
| | - Katie Culos
- Department of Pharmaceutical Services Vanderbilt University Medical Center Nashville TN USA
| | - Sanjay Mohan
- Department of Hematology and Bone Marrow Transplant Vanderbilt University Medical Center Nashville TN USA
| | - Brian G. Engelhardt
- Department of Hematology and Bone Marrow Transplant Vanderbilt University Medical Center Nashville TN USA
| | - Carrie Kitko
- Department of Pediatrics Pediatric Hematology and Bone Marrow Transplant Vanderbilt Childrens Hospital Nashville TN USA
| | - Jim Connelly
- Department of Pediatrics Pediatric Hematology and Bone Marrow Transplant Vanderbilt Childrens Hospital Nashville TN USA
| | - Gowri Satyanarayana
- Department of Infectious Disease Vanderbilt University Medical Center Nashville TN USA
| | - Madan Jagasia
- Department of Hematology and Bone Marrow Transplant Vanderbilt University Medical Center Nashville TN USA
| | - Josu De La Fuente
- Department of Paediatrics St. Mary’s Hospital Imperial College London UK
| | - Adetola Kassim
- Department of Hematology and Bone Marrow Transplant Vanderbilt University Medical Center Nashville TN USA
| |
Collapse
|