1
|
Walters MC, Eapen M, Liu Y, El Rassi F, Waller EK, Levine JE, Strouse JJ, Antin JH, Parikh SH, Bakshi N, Dampier C, Jaroscak JJ, Bergmann S, Wong T, Kota V, Pace B, Lekakis LJ, Lulla P, Nickel RS, Kasow KA, Popat U, Smith W, Yu L, DiFronzo N, Geller N, Kamani N, Klings ES, Hassell K, Mendizabal A, Sullivan K, Neuberg D, Krishnamurti L. Hematopoietic cell transplant compared with standard care in adolescents and young adults with sickle cell disease. Blood Adv 2025; 9:955-965. [PMID: 39471440 PMCID: PMC11907447 DOI: 10.1182/bloodadvances.2024013926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 11/01/2024] Open
Abstract
ABSTRACT Disease-modifying therapies are standard of care (SOC) for sickle cell disease (SCD), but hematopoietic cell transplantation (HCT) has curative potential. We compared outcomes prospectively through 2 years after biologic assignment to a donor or no donor (SOC) arm based on the availability of an HLA-matched sibling or unrelated donor (BMT CTN 1503). A donor search was commenced after eligibility confirmation. The primary end point was a comparison of survival between the treatment arms 2 years after biologic assignment. Power calculations required 60 participants in the donor arm and 140 in the no donor arm to determine if early transplant-related mortality might be balanced by disease-related mortality over a longer period of follow-up. Secondary objectives were a comparison of the changes in SCD-related events, functional outcomes, and organ function. The data were analyzed according to the intent-to-treat principle. A total of 113 participants were enrolled with 28 in the donor arm and 85 in the no donor arm. The 2-year probabilities of survival were 89% and 93%, in the donor vs no donor arms. Vaso-occlusive pain (VOC) was less frequent in the donor arm in the second year after biologic assignment (P < .001). Based on PROMIS-57 surveys, there was a decrease in fatigue (P = .003) and an increase in the ability to participate in social roles and activities (P = .003) in the donor arm 2 years after biologic assignment. Differences in other secondary outcomes did not reach statistical significance. Barriers to accrual prevented an objective comparison of survival. Assignment to the donor arm led to improvements in VOC, fatigue, and social function. This trial was registered at www.clinicaltrials.gov as #NCT02766465.
Collapse
Affiliation(s)
- Mark C. Walters
- Department of Pediatrics, Division of Hematology, University of California San Francisco, San Francisco, CA
| | - Mary Eapen
- Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Yiwen Liu
- Department of Data Science, Division of Biostatistics, Dana-Farber Cancer Institute, Boston, MA
| | - Fuad El Rassi
- Department of Hematology and Medical Oncology, Division of Hematology, Emory University, Atlanta, GA
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Division of Hematology, Emory University, Atlanta, GA
| | - John E. Levine
- Department of Pediatrics, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John J. Strouse
- Department of Medicine, Division of Hematology Oncology, Duke University Medical Center, Durham, NC
| | - Joseph H. Antin
- Hematologic Oncology Treatment Program, Dana-Farber Cancer Institute, Boston, MA
| | - Suhag H. Parikh
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA
| | - Nitya Bakshi
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA
- Department of Pediatrics, Division of Hematology/Oncology/BMT, Yale School of Medicine, New Haven, CT
| | - Carlton Dampier
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA
| | - Jennifer J. Jaroscak
- Department of Pediatrics, Division of Hematology/Oncology/BMT, Medical University of South Carolina, Charleston, SC
| | - Shayla Bergmann
- Department of Pediatrics, Division of Hematology/Oncology/BMT, Medical University of South Carolina, Charleston, SC
| | - Trisha Wong
- Division of Hematology and Oncology, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Vamsi Kota
- Department of Medicine, Division of Hematology and Oncology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Betty Pace
- Department of Pediatrics, Division of Hematology and Oncology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Lazaros J. Lekakis
- Department of Medicine, Division of Hematology/Oncology/BMT, University of Miami, Miami, FL
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Robert S. Nickel
- Department of Pediatrics, Division of Hematology, Children’s National Hospital, Washington, DC
| | - Kimberly A. Kasow
- Department of Pediatrics, Division of Hematology/Oncology/BMT, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Uday Popat
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wally Smith
- Department of Medicine, Division of General Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Lolie Yu
- Department of Pediatrics, Division of Hematology/Oncology/BMT, Children’s Hospital of New Orleans, New Orleans, LA
| | - Nancy DiFronzo
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Nancy Geller
- National Heart, Lung, and Blood Institute, Division of Intramural Research, National Institutes of Health, Bethesda, MD
| | - Naynesh Kamani
- Department of Pediatrics, Division of Immunology, Children’s National Hospital, Washington, DC
| | - Elizabeth S. Klings
- Department of Medicine, Division of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Kathryn Hassell
- Department of Medicine, Division of Hematology, University of Colorado, Denver, CO
| | | | - Keith Sullivan
- Department of Medicine, Division of Hematologic Malignanices and Cellular Therapy, Duke University Medical Center, Durham, NC
| | - Donna Neuberg
- Department of Data Science, Division of Biostatistics, Dana-Farber Cancer Institute, Boston, MA
| | - Lakshmanan Krishnamurti
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA
- Department of Pediatrics, Division of Hematology/Oncology/BMT, Yale School of Medicine, New Haven, CT
| |
Collapse
|
2
|
Jones RJ, Kassim AA, Brodsky RA, DeBaun MR. Is allogeneic transplantation for sickle cell disease still relevant in the era of gene therapy? Blood Adv 2025; 9:877-883. [PMID: 39602668 PMCID: PMC11875129 DOI: 10.1182/bloodadvances.2024013693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
ABSTRACT Sickle cell disease (SCD) is the most common inherited blood disease. Disease-modifying therapy and supportive care have improved the survival of children with SCD in the United States and Europe. Yet, adults with SCD continue to have high risks of morbidity and early death. Recently, 2 US Food and Drug Administration-approved genetic therapies offer the potential for a short-term decrease in acute vaso-occlusive pain events if not cure. Allogeneic hematopoietic cell transplantation (allo-HCT) is also curative but, until recently, was constrained by limited donor availability and the risks of graft-versus-host disease, graft rejection, and death. Importantly, recent advances have attenuated these barriers. Here, we discuss the current state of therapies with curative intent for SCD. Both genetic therapy and allo-HCT offer the potential for cure for most with SCD. However, the cost (∼5 times higher), the current need for myeloablation, and associated late-health effects may make genetic therapies less favorable choices than allo-HCT.
Collapse
Affiliation(s)
- Richard J. Jones
- Departments of Oncology and Medicine, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD
| | - Adetola A. Kassim
- Department of Medicine (Hematology/Oncology), Vanderbilt University School of Medicine, Nashville, TN
| | - Robert A. Brodsky
- Departments of Oncology and Medicine, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD
| | - Michael R. DeBaun
- Departments of Pediatrics and Medicine, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
3
|
Kostamo Z, Ortega MA, Xu C, Feliciano PR, Budak E, Lam D, Winton V, Jenkins R, Venugopal A, Zhang M, Jamieson J, Coisman B, Goldsborough K, Hernandez B, Kanne CK, Evans EN, Zgodny J, Zhang Y, Darazim J, Patel A, Pendergast MA, Manis J, Hartigan AJ, Ciaramella G, Lee SJ, Chu SH, Sheehan VA. Base editing HbS to HbG-Makassar improves hemoglobin function supporting its use in sickle cell disease. Nat Commun 2025; 16:1441. [PMID: 39920120 PMCID: PMC11806015 DOI: 10.1038/s41467-025-56578-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Adenine base editing can convert sickle hemoglobin (HbS, βΕ6V) to G-Makassar hemoglobin (HbG, βE6A), a naturally occurring variant that is clinically asymptomatic. However, the quality and functionality of purified HbG and of mature HbGG and HbGS red blood cells (RBC) has not been assessed. Here, we develop a mouse model to characterize HbG. Purified HbG appears normal and does not polymerize under hypoxia. The topology of the hemoglobin fold with the βΕ6Α mutation is similar to HbA in the oxy and deoxy states. However, RBC containing HbGS are dehydrated, showing altered function and increased sickling under hypoxia. Blood counts and mitochondrial retention measures place HbGS RBCs as intermediate in severity between HbAS and HbSS, while organ function is comparable to HbAS. HbGG resembles HbAA for most metrics. Our results highlight the importance of functionally assessing the mature red cell environment when evaluating novel gene editing strategies for hematologic disorders.
Collapse
Affiliation(s)
- Zachary Kostamo
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | | | | | | | | | - Daisy Lam
- Beam Therapeutics, Cambridge, MA, USA
| | | | | | | | | | | | | | | | - Britney Hernandez
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | - Celeste K Kanne
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | - Erica N Evans
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | - Jordan Zgodny
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | - Yankai Zhang
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | - Jawa Darazim
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | - Ashwin Patel
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA
| | | | - John Manis
- Joint Program in Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Vivien A Sheehan
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA, USA.
| |
Collapse
|
4
|
Jacobsohn DA. Dare mighty things for SCD. Blood 2024; 143:2569-2570. [PMID: 38900474 DOI: 10.1182/blood.2024024273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Affiliation(s)
- David Alex Jacobsohn
- Children's National Hospital
- George Washington University School of Medicine and Health Sciences
| |
Collapse
|
5
|
Mohrez M, Troeger A, Kleinschmidt K, Alali TH, Jakob M, Brosig A, Hähnel V, Kietz S, Offner R, Burkhardt R, Corbacioglu S, Ahrens N, Foell J. Feasibility of peripheral blood stem cell collection from sickle cell trait donors with an intensified G-CSF regimen. Eur J Haematol 2023; 111:824-830. [PMID: 37635081 DOI: 10.1111/ejh.14083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVES Allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative treatment for SCD and bone marrow from an HLA-matched sibling is currently the standard of care. Haploidentical HSCT from a family donor with a TCR αβ/CD19 depleted graft (T-haplo) is an increasingly successful alternative, which requires the generation of G-CSF stimulated peripheral stem cell (PBSC) from haploidentical relatives. These sickle cell trait (SCT) donors reported to develop SCD-related complications in conditions of severe stress. METHODS In this retrospective analysis, we compared the safety and efficacy of PBSC mobilization with a G-CSF intensified mobilization regimen in SCT donors with a conventional G-CSF mobilization regimen in healthy donors. RESULTS The reported adverse events were similar during intensified G-CSF mobilization, apheresis, and shortly after stem cell apheresis in SCT and control donors. In SCT and control donors, we were able to mobilize high yields of CD34+ stem cells and the harvested CD34+ cell count was comparable with control donors. CONCLUSIONS Peripheral stem cell mobilization using an intensified G-CSF regimen is safe, and well tolerated among SCT donors. SCT donors are a valid alternative for collection of peripheral CD34+ stem cells for T-cell-depleted haploidentical stem cell transplantation.
Collapse
Affiliation(s)
- Morad Mohrez
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Anja Troeger
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital Regensburg, Regensburg, Germany
| | - Katharina Kleinschmidt
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital Regensburg, Regensburg, Germany
| | - Tarek Hanafee Alali
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital Regensburg, Regensburg, Germany
| | - Marcus Jakob
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital Regensburg, Regensburg, Germany
| | - Andreas Brosig
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Viola Hähnel
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Silke Kietz
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital Regensburg, Regensburg, Germany
| | - Robert Offner
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ralph Burkhardt
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Selim Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital Regensburg, Regensburg, Germany
| | - Norbert Ahrens
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Juergen Foell
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children's Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
6
|
Gandhi AP, Newell LF, Maziarz RT. A new beginning: can omidubicel emerge as the next, viable alternative donor source? Ther Adv Hematol 2023; 14:20406207231192146. [PMID: 37664800 PMCID: PMC10469227 DOI: 10.1177/20406207231192146] [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: 03/10/2023] [Accepted: 07/19/2023] [Indexed: 09/05/2023] Open
Abstract
Umbilical cord blood (UCB) transplantation (CBT) has been an important alternative donor option for patients lacking matched related donor (MRD) or unrelated donor (URD) grafts. Only 30% of patients with high-risk hematologic malignancies have a human leukocyte antigen (HLA)-identical sibling; subjects without a MRD option are referred for HLA-matched URD selection, or utilize alternative donor sources such as HLA-mismatched URD, UCB, or haploidentical donor grafts. While CBT demonstrates an excellent graft-versus-leukemia (GVL) effect, use of UCB as a graft source is limited due to a lower cell dose that can result in delayed engraftment and an immature immune system with increased infectious risk as a consequence. Together, increased transplant related mortality (TRM) has been associated with UCB allografts. Omidubicel is an ex vivo expanded single cord blood product that has demonstrated rapid engraftment, improved immune reconstitution, and reduced infectious complications in clinical trials. Omidubicel has now been granted U.S. Food & Drug Administration approval to enhance neutrophil recovery and decrease infectious risk. This review will focus on CBT, benefits and barriers to using this alternative donor source, and finally the potential advancements with incorporation of omidubicel in the transplant setting for malignant and non-malignant diseases.
Collapse
Affiliation(s)
- Arpita P. Gandhi
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Laura F. Newell
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Richard T. Maziarz
- Knight Cancer Institute, Oregon Health and Science University, Mail code: OC14HO, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
7
|
Klein OR, Bonfim C, Abraham A, Ruggeri A, Purtill D, Cohen S, Wynn R, Russell A, Sharma A, Ciccocioppo R, Prockop S, Boelens JJ, Bertaina A. Transplant for non-malignant disorders: an International Society for Cell & Gene Therapy Stem Cell Engineering Committee report on the role of alternative donors, stem cell sources and graft engineering. Cytotherapy 2023; 25:463-471. [PMID: 36710227 DOI: 10.1016/j.jcyt.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 01/30/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) is curative for many non-malignant disorders. As HSCT and supportive care technologies improve, this life-saving treatment may be offered to more and more patients. With the development of new preparative regimens, expanded alternative donor availability, and graft manipulation techniques, there are many options when choosing the best regimen for patients. Herein the authors review transplant considerations, transplant goals, conditioning regimens, donor choice, and graft manipulation strategies for patients with non-malignant disorders undergoing HSCT.
Collapse
Affiliation(s)
- Orly R Klein
- Division of Hematology, Oncology and Stem Cell Transplant and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA.
| | - Carmem Bonfim
- Pediatric Blood and Marrow Transplantation Division and Pele Pequeno Principe Research Institute, Hospital Pequeno Principe, Curitiba, Brazil
| | - Allistair Abraham
- Center for Cancer and Immunology Research, Cell Enhancement and Technologies for Immunotherapy, Children's National Hospital, Washington, DC, USA
| | - Annalisa Ruggeri
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Duncan Purtill
- Department of Hematology, Fiona Stanley Hospital, Perth, Australia
| | - Sandra Cohen
- Université de Montréal and Maisonneuve Rosemont Hospital, Montréal, Canada
| | - Robert Wynn
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Athena Russell
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy
| | - Susan Prockop
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Alice Bertaina
- Division of Hematology, Oncology and Stem Cell Transplant and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
8
|
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
|
9
|
Allogeneic stem cell transplantation with omidubicel in sickle cell disease. Blood Adv 2021; 5:843-852. [PMID: 33560399 DOI: 10.1182/bloodadvances.2020003248] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
Many patients with sickle cell disease (SCD) do not have HLA-matched related donors for hematopoietic stem cell transplantation (HSCT). Unrelated cord blood (UCB) is an alternative graft option but is historically associated with high graft failure rates, with inadequate cell dose a major limitation. Omidubicel is a nicotinamide-based, ex vivo-expanded UCB product associated with rapid engraftment in adults with hematologic malignancies. We hypothesized that increasing the UCB cell dose with this strategy would lead to improved engraftment in pediatric patients undergoing myeloablative HSCT for SCD. We report the outcomes of a phase 1/2 study in 13 patients with severe SCD who received omidubicel in combination with an unmanipulated UCB graft and 3 who received a single omidubicel graft. Grafts were minimally matched with patients at 4 of 6 HLA alleles. Median age at transplant was 13 years. A median CD34+ expansion of ∼80-fold was observed in omidubicel and led to rapid neutrophil engraftment (median, 7 days). Long-term engraftment was derived from the unmanipulated graft in most of the double cord blood recipients. Two of the 3 single omidubicel recipients also had sustained engraftment. Incidence of acute graft-versus-host disease (GVHD) was high, but resolved in all surviving patients. Event-free survival in the double cord group was 85% (median follow-up 4 years). All 3 patients in the single cord group were alive at 1 year after transplantation. Ex vivo expansion of UCB with omidubicel supports engraftment in patients with SCD. This approach to decreasing the incidence of GVHD should be optimized for general use in patients with SCD. This study was registered at www.clinicaltrials.gov as #NCT01590628.
Collapse
|
10
|
Kogel F, Hakimeh D, Sodani P, Lang P, Kühl JS, Hundsdoerfer P, Künkele A, Eggert A, Oevermann L, Schulte JH. Allogeneic hematopoietic stem cell transplantation from sibling and unrelated donors in pediatric patients with sickle cell disease-A single center experience. Pediatr Transplant 2021; 25:e13892. [PMID: 33098344 DOI: 10.1111/petr.13892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/21/2020] [Accepted: 09/27/2020] [Indexed: 11/27/2022]
Abstract
HSCT is curative in SCD. Patients with HLA-identical sibling donor have an excellent outcome ranging from 90%-100% overall and event-free survival. However, due to the lack of matched sibling donors this option is out of reach for 70% of patients with SCD. The pool of potential donors needs to be extended. Transplantations from HLA-matched unrelated donors were reported to be less successful with shorter event-free survival and higher incidences of complications including graft-vs-host disease, especially in patients with advanced stage SCD. Here we report transplantation outcomes for 25 children with SCD transplanted using HLA-matched grafts from related or unrelated donors. Overall survival was 100% with no severe (grade III-IV) graft-vs-host disease and a 12% rejection rate. Mixed donor chimerisms only occurred in transplantations from siblings, while transplantations from unrelated donors resulted in either complete donor chimerism or rejection. Despite the small patient number, overall and disease-free survival for unrelated donor transplantations is excellent in this cohort. The advanced disease state, higher alloreactive effect and stronger immunosuppression in unrelated donor transplantations raises patient risk, for which possible solutions could be found in optimization of transplant preparation, graft manipulation or haploidentical transplantation using T cell receptor α/β-depleted grafts.
Collapse
Affiliation(s)
- Friederike Kogel
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dani Hakimeh
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pietro Sodani
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Lang
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Pediatric Hematology and Oncology, University Hospital, Tübingen, Germany
| | - Jörn-Sven Kühl
- Department of Pediatric Oncology, Hematology, and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Patrick Hundsdoerfer
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Department of Pediatrics, Helios-Klinikum Berlin-Buch, Berlin, Germany
| | - Annette Künkele
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angelika Eggert
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena Oevermann
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Johannes H Schulte
- Department of Pediatric Hematology and Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
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
|
12
|
Iqbal M, Reljic T, Corbacioglu S, de la Fuente J, Gluckman E, Kumar A, Yassine F, Ayala E, El-Jawahri A, Murthy H, Almohareb F, Hashmi SK, Cappelli B, Alahmari A, Scigliuolo GM, Kassim A, Aljurf M, Kharfan-Dabaja MA. Systematic Review/Meta-Analysis on Efficacy of Allogeneic Hematopoietic Cell Transplantation in Sickle Cell Disease: An International Effort on Behalf of the Pediatric Diseases Working Party of European Society for Blood and Marrow Transplantation and the Sickle Cell Transplantation International Consortium. Transplant Cell Ther 2020; 27:167.e1-167.e12. [PMID: 33830027 DOI: 10.1016/j.jtct.2020.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/28/2020] [Accepted: 10/21/2020] [Indexed: 11/28/2022]
Abstract
Sickle cell disease (SCD) affects more than 300,000 children annually worldwide. Despite improved supportive care, long-term prognosis remains poor. Allogeneic hematopoietic cell transplantation (allo-HCT) is the sole validated curative option, resulting in sustained resolution of the clinical phenotype. The medical literature on allo-HCT for SCD is largely limited to children. Recent studies have evaluated allo-HCT efficacy in adults. Here, we conducted a systematic review/meta-analysis to assess the totality of evidence on the efficacy, or lack thereof, of allo-HCT in treating SCD. We performed a comprehensive literature search using PubMed/Medline, Embase, and Cochrane library databases on November 13, 2019. Four authors independently extracted data on clinical outcomes related to benefits (overall survival [OS] and disease-free survival [DFS]) and harms (acute graft-versus-host disease [aGVHD], chronic graft-versus-host disease [cGVHD], nonrelapse mortality [NRM], and graft failure [GF]). Our search identified a total of 1906 references. Only 33 studies (n= 2853 patients) met our inclusion criteria. We also performed a subset analysis by age. Analyses of all-age groups showed pooled rates of 96% for OS, 90% for DFS, 20% for aGVHD, 10% for cGVHD, 4% for NRM, and 5% for GF. In the pediatric population, pooled rates for OS, DFS, aGVHD, cGVHD, NRM, and GF were 97%, 91%, 26%, 11%, 5%, and 3%, respectively. In adults, pooled rates for OS, DFS, aGVHD, cGVHD, NRM, and GF were 98%, 90%, 7%, 1%, 0%, and 14%, respectively. Our data show that allo-HCT is safe and effective, yielding pooled OS rates exceeding 90%. The high GF rate of 14% in adults is concerning and emphasizes the need to evaluate new strategies.
Collapse
Affiliation(s)
- Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, Florida
| | - Tea Reljic
- Program for Comparative Effectiveness Research, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Selim Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Germany
| | - Josu de la Fuente
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Eliane Gluckman
- Eurocord, Paris-Diderot University Equipe d'Accueil 3518, Hospital Saint Louis, Paris, France; Monacord, Centre Scientifique de Monaco, Monaco
| | - Ambuj Kumar
- Program for Comparative Effectiveness Research, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Farah Yassine
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, Florida
| | - Ernesto Ayala
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, Florida
| | - Areej El-Jawahri
- Department of Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Hemant Murthy
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, Florida
| | - Fahad Almohareb
- Adult Hematology/Bone Marrow Transplantation, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Shahrukh K Hashmi
- Adult Hematology/Bone Marrow Transplantation, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; Department of Hematology, Mayo Clinic, Rochester, Minnesota
| | - Barbara Cappelli
- Eurocord, Paris-Diderot University Equipe d'Accueil 3518, Hospital Saint Louis, Paris, France; Monacord, Centre Scientifique de Monaco, Monaco
| | - Ali Alahmari
- Adult Hematology/Bone Marrow Transplantation, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Graziana Maria Scigliuolo
- Eurocord, Paris-Diderot University Equipe d'Accueil 3518, Hospital Saint Louis, Paris, France; Monacord, Centre Scientifique de Monaco, Monaco
| | - Adetola Kassim
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Mahmoud Aljurf
- Adult Hematology/Bone Marrow Transplantation, Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation and Cellular Therapy Program, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
13
|
Brodsky RA, DeBaun MR. Are genetic approaches still needed to cure sickle cell disease? J Clin Invest 2020; 130:7-9. [PMID: 31738187 DOI: 10.1172/jci133856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Robert A Brodsky
- Division of Hematology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Michael R DeBaun
- Vanderbilt-Meharry Sickle Cell Disease Center of Excellence, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Tozatto-Maio K, Torres MA, Degaide NHS, Cardoso JF, Volt F, Pinto ACS, Oliveira D, Elayoubi H, Kashima S, Loiseau P, Veelken H, Ferster A, Cappelli B, Rodrigues ES, Scigliuolo GM, Kenzey C, Ruggeri A, Rocha V, Simões BP, Tamouza R, Gluckman E. HLA-Matched Unrelated Donors for Patients with Sickle Cell Disease: Results of International Donor Searches. Biol Blood Marrow Transplant 2020; 26:2034-2039. [PMID: 32712327 DOI: 10.1016/j.bbmt.2020.07.015] [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: 05/04/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 01/31/2023]
Abstract
Sickle cell disease (SCD) is the most common inherited hemoglobinopathy. Hematopoietic stem cell transplantation (HCT) is the sole curative therapy for SCD, but few patients will have a matched sibling donor. Patients with SCD are mostly of African origin and thus are less likely to find a matched unrelated donor in international registries. Using HaploStats, we estimated HLA haplotypes for 185 patients with SCD (116 from a Brazilian center and 69 from European Society for Blood and Marrow Transplantation [EBMT] centers) and classified the ethnic origin of haplotypes. Then we assessed the probability of finding an HLA-matched unrelated adult donor (MUD), considering loci A, B, and DRB1 (6/6), in international registries. Most haplotypes were African, but Brazilians showed a greater ethnic admixture than EBMT patients. Nevertheless, the chance of finding at least one 6/6 potential allelic donor was 47% for both groups. Most potential allelic donors were from the US National Marrow Donor Program registry and from the Brazilian REDOME donor registry. Although the probability of finding a donor is higher than previously reported, strategies are needed to improve ethnic diversity in registries. Moreover, predicting the likelihood of having an MUD might influence SCD management.
Collapse
Affiliation(s)
- Karina Tozatto-Maio
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Center for Cell-Based Therapy, Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Hematology, Clinics Hospital, University of São Paulo Medical School, São Paulo, Brazil.
| | | | - Neifi Hassan Saloum Degaide
- Center for Cell-Based Therapy, Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Juliana Fernandes Cardoso
- Brazilian Bone Marrow Registry (REDOME), Instituto Nacional do Câncer, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Fernanda Volt
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Ana Cristina Silva Pinto
- Center for Cell-Based Therapy, Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Danielli Oliveira
- Brazilian Bone Marrow Registry (REDOME), Instituto Nacional do Câncer, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Hanadi Elayoubi
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Simone Kashima
- Center for Cell-Based Therapy, Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Pascale Loiseau
- Saint Louis Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Hendrik Veelken
- Bone Marrow Transplantation Centre Leiden, Leiden University Hospital, Leiden, The Netherlands
| | - Alina Ferster
- Department of Hematology/Oncology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, B-1020 Brussels, Belgium
| | - Barbara Cappelli
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Evandra Strazza Rodrigues
- Center for Cell-Based Therapy, Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Graziana Maria Scigliuolo
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Chantal Kenzey
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Annalisa Ruggeri
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Haematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vanderson Rocha
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Department of Hematology, Clinics Hospital, University of São Paulo Medical School, São Paulo, Brazil; Department of Hematology, Churchill Hospital, University of Oxford, Oxford, United Kingdom
| | - Belinda Pinto Simões
- Center for Cell-Based Therapy, Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ryad Tamouza
- INSERM U955, Centre Hospitalier Universitaire Henri Mondor, Créteil, France
| | - Eliane Gluckman
- Monacord, Centre Scientifique de Monaco, Principauté de Monaco, Monaco; Eurocord, Hôpital Saint Louis, Institut de recherche Saint Louis, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
15
|
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
|
16
|
Stenger EO, Shenoy S, Krishnamurti L. How I treat sickle cell disease with hematopoietic cell transplantation. Blood 2019; 134:2249-2260. [PMID: 31697818 PMCID: PMC6923666 DOI: 10.1182/blood.2019000821] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Sickle cell disease (SCD) leads to significant morbidity and early mortality, and hematopoietic cell transplantation (HCT) is the only widely available cure, with impacts seen on SCD-related organ dysfunction. Outcomes are excellent following matched-related donor (MRD) HCT, leading to significantly expanded application of this treatment over the past decade. The majority of SCD patients lack an MRD, but outcomes following alternative donor HCT continue to improve on clinical trials. Within this framework, we aim to provide our perspective on how to apply research findings to clinical practice, for an individual patient. We also emphasize that the preparation of SCD recipients for HCT and supporting them through HCT have special nuances that require awareness and close attention. Through the use of clinical vignettes, we provide our perpsective on the complex decision-making process in HCT for SCD as well as recommendations for the evaluation and support of these patients through HCT.
Collapse
Affiliation(s)
- Elizabeth O Stenger
- Division of Blood and Marrow Transplantation and Cellular Therapies, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Shalini Shenoy
- Division of Hematology/Oncology, Children's Hospital St. Louis, St. Louis, MO; and
| | | |
Collapse
|
17
|
Morgan RA, Unti MJ, Aleshe B, Brown D, Osborne KS, Koziol C, Ayoub PG, Smith OB, O'Brien R, Tam C, Miyahira E, Ruiz M, Quintos JP, Senadheera S, Hollis RP, Kohn DB. Improved Titer and Gene Transfer by Lentiviral Vectors Using Novel, Small β-Globin Locus Control Region Elements. Mol Ther 2019; 28:328-340. [PMID: 31628051 DOI: 10.1016/j.ymthe.2019.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/11/2019] [Accepted: 09/19/2019] [Indexed: 01/11/2023] Open
Abstract
β-globin lentiviral vectors (β-LV) have faced challenges in clinical translation for gene therapy of sickle cell disease (SCD) due to low titer and sub-optimal gene transfer to hematopoietic stem and progenitor cells (HSPCs). To overcome the challenge of preserving efficacious expression while increasing vector performance, we used published genomic and epigenomic data available through ENCODE to redefine enhancer element boundaries of the β-globin locus control region (LCR) to construct novel ENCODE core sequences. These novel LCR elements were used to design a β-LV of reduced proviral length, termed CoreGA-AS3-FB, produced at higher titers and possessing superior gene transfer to HSPCs when compared to the full-length parental β-LV at equal MOI. At low vector copy number, vectors containing the ENCODE core sequences were capable of reversing the sickle phenotype in a mouse model of SCD. These studies provide a β-LV that will be beneficial for gene therapy of SCD by significantly reducing the cost of vector production and extending the vector supply.
Collapse
Affiliation(s)
- Richard A Morgan
- Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mildred J Unti
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bamidele Aleshe
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Devin Brown
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kyle S Osborne
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Colin Koziol
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paul G Ayoub
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Oliver B Smith
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rachel O'Brien
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Curtis Tam
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric Miyahira
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marlene Ruiz
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jason P Quintos
- CSUN-UCLA Stem Cell Scientist Training Program, California State University, Northridge, Northridge, CA 91330, USA
| | - Shantha Senadheera
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Roger P Hollis
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Donald B Kohn
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; The Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Haploidentical CD3 or α/β T-cell depleted HSCT in advanced stage sickle cell disease. Bone Marrow Transplant 2019; 54:1859-1867. [DOI: 10.1038/s41409-019-0550-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/16/2019] [Accepted: 04/25/2019] [Indexed: 12/21/2022]
|
19
|
Effect of increased dose of total body irradiation on graft failure associated with HLA-haploidentical transplantation in patients with severe haemoglobinopathies: a prospective clinical trial. LANCET HAEMATOLOGY 2019; 6:e183-e193. [PMID: 30878319 DOI: 10.1016/s2352-3026(19)30031-6] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Although severe haemoglobinopathies can be cured with allogeneic blood or bone marrow transplantation, availability of matched donors and toxic effects can be problematic. We previously found that non-myeloablative haploidentical related bone marrow transplantation with post-transplantation cyclophosphamide expanded the donor pool while limiting graft-versus-host disease (GVHD). However, graft failure-albeit with full host haemopoietic recovery-occurred in 50% of patients. In this study, we investigated whether increasing total body irradiation from 200 cGy to 400 cGy would improve engraftment while maintaining the safety profile. METHODS This study was done at Johns Hopkins Hospital (Baltimore, MD, USA). Patients aged 2-70 years receiving their first bone marrow transplant were eligible for inclusion in the study. Patients received rabbit-derived intravenous anti-thymocyte globulin 0·5 mg/kg on day -9 and 2 mg/kg on days -8 and -7, intravenous fludarabine 30 mg/m2 on days -6 to -2, intravenous cyclophosphamide 14·5 mg/kg on days -6 and -5, and total body irradiation 400 cGy administered as a single fraction on day -1. We collected unmanipulated bone marrow and infused on day 0. GVHD prophylaxis comprised intravenous cyclophosphamide 50 mg/kg per day on days 3 and 4 after transplantation, oral mycophenolate mofetil 15 mg/kg per dose (maximum 1 g) every 8 h on days 5 to 35, and oral sirolimus to maintain a level of 5-15 ng/dL for at least 1 year starting on day 5. The original planned primary objectives of this phase 2 clinical trial were transplant-related mortality and progression-free survival. However, the coverage decision by the Centers for Medicare and Medicaid Services to only provide payment for allogeneic bone marrow transplantation for patients with sickle cell disease on a clinical trial that had a comparison arm with patients not receiving bone marrow transplantation prompted the closure of this trial to accrual in 2017. Therefore, as we were unable to perform our planned statistical analysis, the primary objective was modified to evaluate engraftment, assessed by chimerism. This trial is registered with ClinicalTrials.gov, number NCT00489281. The study is closed to new participants and this is the primary analysis. FINDINGS Between Sept 24, 2014, and Aug 1, 2017, we enrolled 17 consecutive patients: 12 (71%) with sickle cell disease and 5 (29%) with β-thalassaemia major. The median patient age was 16 years (range 6-31, IQR 7·7-27·5). One (6%) of 17 patients had primary graft failure with recovery of host haemopoiesis. 13 (76%) of 17 patients achieved full donor chimerism and three (18%) had mixed donor-host chimerism. Five (29%) of 17 patients developed grade 2-4 acute GVHD, including four (24%) with maximal grade 2 GVHD and one (6%) with grade 3 GVHD. Chronic GVHD developed in three (18%) patients. As of their last follow-up visit, GVHD had resolved in all patients and no patients were receiving systemic GVHD therapy. All patients remained alive as of Aug 4, 2019, and the median follow-up duration was 705 days (range 355-1294; IQR 398-943). Only one (6%) of the 16 engrafted patients remained transfusion dependent, and 14 (88%) discontinued immunosuppression. INTERPRETATION Increasing total body irradiation to 400 cGy substantially reduced graft failure while maintaining the safety of haploidentical bone marrow transplantation with post-transplantation cyclophosphamide. These results suggest that engraftment after haploidentical bone marrow transplantation for haemoglobinopathies is possible, and primary graft failure-the main problem previously reported-might be addressed by this strategy. Therefore, this curative approach should no longer be restricted to patients with HLA-matched donors. FUNDING Maryland Stem Cell Research Fund and US National Institutes of Health.
Collapse
|
20
|
Darbari I, O'Brien JE, Hardy SJ, Speller-Brown B, Thaniel L, Martin B, Darbari DS, Nickel RS. Views of parents of children with sickle cell disease on pre-implantation genetic diagnosis. Pediatr Blood Cancer 2018; 65:e27102. [PMID: 29667775 DOI: 10.1002/pbc.27102] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 01/10/2023]
Abstract
Pre-implantation genetic diagnosis (PGD) is an option for parents who have a child with sickle cell disease (SCD) to have another child without SCD. We conducted a survey of 19 parents with at least one child with SCD to investigate views on PGD. Before education, 44% of parents were aware of PGD. All parents rated PGD education as important. All parents considering another child also reported interest in using PGD if insurance covered its costs. Parents who have a child with SCD appear to be interested in PGD and educational tools informing this group about PGD should be developed.
Collapse
Affiliation(s)
- Isha Darbari
- Division of Hematology, Children's National Health System, Washington, District of Columbia
| | | | - Steven J Hardy
- Division of Hematology, Children's National Health System, Washington, District of Columbia
| | - Barbara Speller-Brown
- Division of Hematology, Children's National Health System, Washington, District of Columbia
| | - Lisa Thaniel
- Division of Hematology, Children's National Health System, Washington, District of Columbia
| | - Brenda Martin
- Division of Hematology, Children's National Health System, Washington, District of Columbia
| | - Deepika S Darbari
- Division of Hematology, Children's National Health System, Washington, District of Columbia
| | - Robert S Nickel
- Division of Hematology, Children's National Health System, Washington, District of Columbia
| |
Collapse
|
21
|
Abstract
Sickle cell disease (SCD) complications begin with the polymerization of sickle hemoglobin (HbS). Thus, SCD therapies are focused on preventing HbS production or reducing the circulating amount of HbS. Hydroxyurea treatment has become more widespread, whereas the number of evidence-based indications for erythrocyte transfusion is small. Hematopoietic stem cell transplant is a curative option for SCD but less than 25% of patients have a suitable donor. This article focuses on supportive and preventive care improvements and the benefits of hydroxyurea. Indications for erythrocyte transfusion, hematopoietic stem cell transplant, and gene therapy trials are also summarized.
Collapse
Affiliation(s)
- Emily Riehm Meier
- Sickle Cell Research, Indiana Hemophilia and Thrombosis Center, 8326 Naab Road, Indianapolis, IN 46260, USA.
| |
Collapse
|
22
|
Unrelated Umbilical Cord Blood Transplantation for Sickle Cell Disease Following Reduced-Intensity Conditioning: Results of a Phase I Trial. Biol Blood Marrow Transplant 2017; 23:1587-1592. [DOI: 10.1016/j.bbmt.2017.05.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/28/2017] [Indexed: 01/08/2023]
|
23
|
Ohemeng A, Boadu I. The role of nutrition in the pathophysiology and management of sickle cell disease among children: A review of literature. Crit Rev Food Sci Nutr 2017; 58:2299-2305. [PMID: 28686043 DOI: 10.1080/10408398.2017.1319794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Sickle cell disease (SCD) is one of the common inherited blood disorders in humans and has been associated with decreased dietary intake which results in poor nutritional status and impaired growth. Nutrition is one of the most important but often forgotten aspect of care of patients with chronic disorders and there have been emerging concern in literature on increased nutritional needs of SCD patients. This paper sought to review the available literature on the roles of individual nutrients in the pathophysiology and management of SCD among children. Children with SCD have been shown to exhibit suboptimal status with respect to both macronutrients and micronutrients. Thus, nutrition could play an important role in the management of SCD. However, there is paucity of evidence coming from trials with large sample sizes to support the suggestion that supplementation with various nutrients that have been considered in this review will be helpful.
Collapse
Affiliation(s)
- Agartha Ohemeng
- a Department of Nutrition and Food Science , School of Biological Sciences, University of Ghana , Legon , Accra , Ghana
| | - Isaac Boadu
- a Department of Nutrition and Food Science , School of Biological Sciences, University of Ghana , Legon , Accra , Ghana
| |
Collapse
|
24
|
da Silva-Malta MCF, Rodrigues PS, Zuccherato LW, de Souza FCB, Domingues EMFL, Souza VR, Tarazona-Santos E, Martins ML. Human leukocyte antigen distribution and genomic ancestry in Brazilian patients with sickle cell disease. HLA 2017; 90:211-218. [DOI: 10.1111/tan.13102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/13/2017] [Accepted: 07/18/2017] [Indexed: 12/11/2022]
Affiliation(s)
- M. C. F. da Silva-Malta
- Fundação Centro de Hematologia e Hemoterapia de Minas Gerais - Hemominas; Belo Horizonte Brazil
| | - P. S. Rodrigues
- Fundação Centro de Hematologia e Hemoterapia de Minas Gerais - Hemominas; Belo Horizonte Brazil
| | - L. W. Zuccherato
- Departamento de Biologia Geral, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - F. C. B. de Souza
- Fundação Centro de Hematologia e Hemoterapia de Minas Gerais - Hemominas; Belo Horizonte Brazil
| | - E. M. F. L. Domingues
- Fundação Centro de Hematologia e Hemoterapia de Minas Gerais - Hemominas; Belo Horizonte Brazil
| | - V. R. Souza
- Fundação Centro de Hematologia e Hemoterapia de Minas Gerais - Hemominas; Belo Horizonte Brazil
| | - E. Tarazona-Santos
- Departamento de Biologia Geral, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - M. L. Martins
- Fundação Centro de Hematologia e Hemoterapia de Minas Gerais - Hemominas; Belo Horizonte Brazil
| |
Collapse
|
25
|
Alternative donor hematopoietic stem cell transplantation for sickle cell disease. Blood Adv 2017; 1:1215-1223. [PMID: 29296761 DOI: 10.1182/bloodadvances.2017005462] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/15/2017] [Indexed: 11/20/2022] Open
Abstract
Most patients who could be cured of sickle cell disease (SCD) with stem cell transplantation do not have a matched sibling donor. Successful use of alternative donors, including mismatched family members, could provide a donor for almost all patients with SCD. The use of a reduced-intensity conditioning regimen may decrease late adverse effects. Ten patients with symptomatic SCD underwent CD34+ cell-selected, T-cell-depleted peripheral blood stem cell transplantation from a mismatched family member or unrelated donor. A reduced-intensity conditioning regimen including melphalan, thiotepa, fludarabine, and rabbit anti-thymocyte globulin was used. Patients were screened for a companion study for immune reconstitution that included a donor lymphocyte infusion given 30-42 days after transplant with intravenous methotrexate as graft-versus-host disease (GVHD) prophylaxis. Seven eligible patients were treated on the companion study. Nine of 10 patients are alive with a median follow-up of 49 months (range, 14-60 months). Surviving patients have stable donor hematopoietic engraftment (mean donor chimerism, 99.1% ± 0.7%). There were no sickle cell complications after transplant. Two patients had grade II-IV acute GVHD. One patient had chronic GVHD. Epstein-Barr virus-related posttransplant lymphoproliferative disorder (PTLD) occurred in 3 patients, and 1 patient died as a consequence of treatment of PTLD. Two-year overall survival was 90%, and event-free survival was 80%. A reduced-intensity conditioning regimen followed by CD34+ cell-selected, T-cell-depleted alternative donor peripheral blood stem cell transplantation achieved primary engraftment in all patients with a low incidence of GVHD, although PTLD was problematic. This trial was registered at clinicaltrials.gov as #NCT00968864.
Collapse
|
26
|
Reduced toxicity, myeloablative HLA-haploidentical hematopoietic stem cell transplantation with post-transplantation cyclophosphamide for sickle cell disease. Ann Hematol 2017; 96:1373-1377. [DOI: 10.1007/s00277-017-3030-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023]
|
27
|
Treosulfan-Based Conditioning Regimen in Sibling and Alternative Donor Hematopoietic Stem Cell Transplantation for Children with Sickle Cell Disease. Mediterr J Hematol Infect Dis 2017; 9:e2017014. [PMID: 28293402 PMCID: PMC5333731 DOI: 10.4084/mjhid.2017.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/12/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Lack of suitable donors and regimen related toxicity are major barriers for hematopoietic stem cell transplantation (HSCT) in patients with sickle cell disease (SCD). The aim of the study is the assessment of efficacy and toxicity of Treosulfan-based conditioning regimen for SCD also when alternative donors such as mismatched unrelated donor and haploidentical donor are employed. METHODS We report our single-center experience: 11 patients with SCD received HSCT with a Treosulfan/Thiotepa/Fludarabine/Anti-thymoglobulin conditioning regimen between 2010 and 2015. The donor was a matched sibling donor (n= 7), a haploidentical parent (n= 2), a matched unrelated donor (n= 1) or a mismatched unrelated donor (n=1). The haploidentical and mismatched unrelated donor grafts were manipulated by removing TCRαβ and CD19 positive cells. RESULTS All patients survived the procedure and achieved stable engraftment. Stable mixed chimerism was observed in 5/11 patients. Grade III-IV regimen related toxicity was limited to mucositis and no grade III-IV graft-versus-host disease (GvHD) occurred. No SCD manifestation was observed post transplant and cerebral vasculopathy improved in 3/5 evaluable patients. Organ function evaluation showed no pulmonary, cardiac or renal toxicity but gonadal failure occurred in 1/4 evaluable patients. CONCLUSION Our data suggest that Treosulfan is associated with low toxicity and may be employed also for unrelated and haploidentical donor HSCT.
Collapse
|
28
|
Abraham A, Jacobsohn DA, Bollard CM. Cellular therapy for sickle cell disease. Cytotherapy 2016; 18:1360-1369. [PMID: 27421743 DOI: 10.1016/j.jcyt.2016.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/25/2016] [Accepted: 06/16/2016] [Indexed: 02/02/2023]
Abstract
Sickle cell disease (SCD) is a monogenic red cell disorder affecting more than 300 000 annual births worldwide and leading to significant organ toxicity and premature mortality. Although chronic therapies such as hydroxyurea have improved outcomes, more durable therapeutic and curative options are still being investigated. Newer understanding of the disease has implicated invariant natural killer T cells as a critical immune profile that potentiates SCD. Hence, targeting this cell population may offer a new approach to disease management. Hematopoietic stem cell transplant is a curative option for patients with SCD, but the under-representation of minorities on the unrelated donor registry means that this is not a feasible option for more than 75% of patients. Work in this area has therefore focused on increasing the donor pool and decreasing transplant-related toxicities to make this a treatment option for the majority of patients with SCD. This review focuses on the currently available cell and gene therapies for patients with SCD and acknowledges that newer gene-editing approaches to improve gene therapy efficiency and safety are the next wave of potentially curative approaches.
Collapse
Affiliation(s)
- Allistair Abraham
- Division of Blood and Marrow Transplant, Children's National Health System and The George Washington University, Washington, DC, USA; Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Health System and The George Washington University, Washington, DC, USA.
| | - David A Jacobsohn
- Division of Blood and Marrow Transplant, Children's National Health System and The George Washington University, Washington, DC, USA
| | - Catherine M Bollard
- Division of Blood and Marrow Transplant, Children's National Health System and The George Washington University, Washington, DC, USA; Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Health System and The George Washington University, Washington, DC, USA
| |
Collapse
|
29
|
Alfraih F, Aljurf M, Fitzhugh CD, Kassim AA. Alternative donor allogeneic hematopoietic cell transplantation for hemoglobinopathies. Semin Hematol 2016; 53:120-8. [PMID: 27000737 DOI: 10.1053/j.seminhematol.2016.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) offers a curative therapy for patients with hemoglobinopathies, mainly severe sickle cell disease (SCD) and thalassemia (TM). However, the applicability of HSCT has been limited mainly by donor availability, with a less than 25%-30% of eligible patients having human leukocyte antigen (HLA)-matched sibling donors. Previous outcomes using alternate donor options have been markedly inferior due to increased regimen-related toxicity, transplant-related mortality, graft failure, and graft-versus-host disease (GVHD). Advances in transplant technology, including high-resolution HLA typing, improved GVHD prophylactic approaches with tolerance induction, and better supportive care over the last decade, are addressing these historical challenges, resulting in increasing donor options. Herein, we review alternate donor HSCT approaches for severe SCD and TM using unrelated donors, umbilical cord blood units, or related haploidentical donors. Though this is an emerging field, early results are promising and in selected patients, this may be the preferred option to mitigate against the age-related morbidity and early mortality associated with these disorders.
Collapse
Affiliation(s)
- Feras Alfraih
- Adult Hematology and Hematopoietic Stem Cell Transplantation, King Faisal Hospital and Research Centre, Riyadh, Saudi Arabia.
| | - Mahmoud Aljurf
- Molecular and Clinical Hematology Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Courtney D Fitzhugh
- Division of Hematology and Oncology, Department of Medicine and Vanderbilt- Meharry Center for Excellence in Sickle Cell Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adetola A Kassim
- Division of Hematology and Oncology, Department of Medicine and Vanderbilt- Meharry Center for Excellence in Sickle Cell Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
30
|
Saraf SL, Oh AL, Patel PR, Jalundhwala Y, Sweiss K, Koshy M, Campbell-Lee S, Gowhari M, Hassan J, Peace D, Quigley JG, Khan I, Molokie RE, Hsu LL, Mahmud N, Levinson DJ, Pickard AS, Garcia JGN, Gordeuk VR, Rondelli D. Nonmyeloablative Stem Cell Transplantation with Alemtuzumab/Low-Dose Irradiation to Cure and Improve the Quality of Life of Adults with Sickle Cell Disease. Biol Blood Marrow Transplant 2015; 22:441-8. [PMID: 26348889 DOI: 10.1016/j.bbmt.2015.08.036] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/31/2015] [Indexed: 11/25/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is rarely performed in adult patients with sickle cell disease (SCD). We utilized the chemotherapy-free, alemtuzumab/total body irradiation 300 cGy regimen with sirolimus as post-transplantation immunosuppression in 13 high-risk SCD adult patients between November 2011 and June 2014. Patients received matched related donor (MRD) granulocyte colony-stimulating factor-mobilized peripheral blood stem cells, including 2 cases that were ABO incompatible. Quality-of-life (QoL) measurements were performed at different time points after HSCT. All 13 patients initially engrafted. A stable mixed donor/recipient chimerism was maintained in 12 patients (92%), whereas 1 patient not compliant with sirolimus experienced secondary graft failure. With a median follow-up of 22 months (range, 12 to 44 months) there was no mortality, no acute or chronic graft-versus-host disease (GVHD), and no grades 3 or 4 extramedullary toxicities. At 1 year after transplantation, patients with stable donor chimerism have normalized hemoglobin concentrations and improved cardiopulmonary and QoL parameters including bodily pain, general health, and vitality. In 4 patients, sirolimus was stopped without rejection or SCD-related complications. These results underscore the successful use of a chemotherapy-free regimen in MRD HSCT for high-risk adult SCD patients and demonstrates a high cure rate, absence of GVHD or mortality, and improvement in QoL including the applicability of this regimen in ABO mismatched cases (NCT number 01499888).
Collapse
Affiliation(s)
- Santosh L Saraf
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Sickle Cell Center, Department of Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois.
| | - Annie L Oh
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - Pritesh R Patel
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois
| | - Yash Jalundhwala
- College of Pharmacy, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - Karen Sweiss
- College of Pharmacy, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - Matthew Koshy
- University of Illinois Cancer Center, Chicago, Illinois; Department of Radiation Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - Sally Campbell-Lee
- Institute for Transfusion Medicine & Department of Pathology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - Michel Gowhari
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Sickle Cell Center, Department of Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Johara Hassan
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Sickle Cell Center, Department of Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - David Peace
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois
| | - John G Quigley
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois
| | - Irum Khan
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois
| | - Robert E Molokie
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Sickle Cell Center, Department of Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Lewis L Hsu
- University of Illinois Sickle Cell Center, Department of Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois; Division of Pediatric Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - Nadim Mahmud
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois
| | - Dennis J Levinson
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - A Simon Pickard
- College of Pharmacy, University of Illinois Hospital and Health Sciences System, Chicago, Illinois
| | - Joe G N Garcia
- Arizona Respiratory Center and Department of Medicine, University of Arizona, Tucson, Arizona
| | - Victor R Gordeuk
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Sickle Cell Center, Department of Medicine, University of Illinois Hospital & Health Sciences System, Chicago, Illinois
| | - Damiano Rondelli
- Division of Hematology/Oncology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois; University of Illinois Center for Global Health, Chicago, Illinois.
| |
Collapse
|
31
|
Lipton JM. Curing sickle cell disease: Mission accomplished? Pediatr Blood Cancer 2015; 62:1129-30. [PMID: 25788216 DOI: 10.1002/pbc.25497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 02/06/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Jeffrey M Lipton
- Steven and Alexandra Cohen Children's Medical Center of New York, New Hyde Park, New York.,Feinstein Institute for Medical Research, Manhasset, New York.,Hofstra North Shore-LIJ School of Medicine, Hempstead, New York
| |
Collapse
|