1
|
Chok R, Girgulis K, Nickel RS, Guilcher GMT. Systemic Inflammatory Response Post Alemtuzumab and Low-Dose Total Body Irradiation in Pediatric Patients With Sickle Cell Disease: A Case Series. Pediatr Blood Cancer 2025; 72:e31448. [PMID: 39568174 DOI: 10.1002/pbc.31448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Affiliation(s)
- Rozalyn Chok
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Lucile Packard Children's Hospital, Stanford University, Stanford, California, USA
| | - Katherine Girgulis
- Section of Oncology/BMT, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert S Nickel
- Division of Hematology and Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, District of Columbia, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Gregory M T Guilcher
- Section of Oncology/BMT, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
2
|
Chevillon F, Rebotier M, Dhédin N, Bruno B, Cacciatore C, Charbonnier A, Joseph L, Le Bourgeois A, Talouarn M, Magro L, Barraud Lange V. [Fertility preservation and hematopoietic stem cell transplantation (SFGM-TC)]. Bull Cancer 2025; 112:S24-S35. [PMID: 38918137 DOI: 10.1016/j.bulcan.2024.04.009] [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: 02/12/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 06/27/2024]
Abstract
Conditioning regimen prior to hematopoietic stem cell transplantation have an impact on patient fertility through the use of gonadal irradiation and/or bifunctional alkylating agents. Their impact on fertility depends mainly on the dose used and, in women, on age at the time of treatment. All patients should benefit before treatment from a consultation informing them of the potential impact on fertility and of fertility preservation techniques. In the absence of contraindications, the major toxicity of myeloablative conditioning regimen justifies fertility preservation. There are few data concerning fertility after reduced-intensity conditioning. Despite lower theoretical gonadotoxicity, we also recommend fertility preservation, if possible before transplantation. The fertility preservation techniques used depend on the patient's age, pathology and conditioning. In the event of subsequent use of harvested gonadal tissue in the context of acute leukemia or aggressive lymphoma, it is advisable to assess the risk of reintroduction of tumor cells. Finally, it is recommended to assess gonadal function after transplant, especially after reduced conditioning. If there is persistent residual gonadal function, post-treatment fertility preservation should be discuss.
Collapse
Affiliation(s)
- Florian Chevillon
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France.
| | - Marine Rebotier
- Service oncogynécologie, centre Leon-Berard et IHOPe, 28, promenade Léa et Napoléon Bullukian, 69008 Lyon, France
| | - Nathalie Dhédin
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - Bénédicte Bruno
- Service hématologie pédiatrique, hôpital Jeanne-de-Flandre, avenue Eugène-Avinée, 59037 Lille, France
| | - Carlotta Cacciatore
- Service de médecine interne, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | | | - Laure Joseph
- Service hématologie, département de biothérapie, hôpital Necker-enfants malades, AP-HP, 149, rue de Sèvres, 75015 Paris, France
| | - Amandine Le Bourgeois
- Service d'hématologie, CHU de Nantes, 1, place Alexis-Ricordeau, 44000 Nantes, France
| | - Marie Talouarn
- Service d'hématologie, hôpital Saint-Antoine, AP-HP, 184, rue du faubourg Saint-Antoine, 75012 Paris, France
| | - Leonardo Magro
- Service d'hématologie, CHU de Lille, 2, avenue Oscar-Lambret, 59000 Lille, France
| | - Virginie Barraud Lange
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service biologie de la reproduction, hôpital Cochin Port Royal, AP-HP, 123, boulevard de Port Royal, 75014 Paris, France
| |
Collapse
|
3
|
Queen J, Limerick E, Jeffries N, Hsieh MM, Shamburek RD, Fitzhugh CD. Lipid Levels Increase to the Normal Range After Nonmyeloablative Hematopoietic Cell Transplantation for Sickle Cell Disease. Transplant Cell Ther 2024:S2666-6367(24)00808-X. [PMID: 39701291 DOI: 10.1016/j.jtct.2024.12.008] [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: 08/05/2024] [Revised: 11/15/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Individuals with sickle cell disease (SCD) have a unique type of dyslipidemia characterized by low total cholesterol (TC), low low-density lipoprotein cholesterol (LDL-c), low high-density lipoprotein cholesterol (HDL-c), and normal triglycerides (TG). This lipid state is theorized to be cardioprotective against atherosclerosis. In SCD, hematopoietic cell transplant (HCT) offers a potentially curative therapy. Long-term survivors of HCT for hematologic malignancies are at increased risk for dyslipidemia and atherosclerosis long-term. The effects of HCT on SCD dyslipidemia are unknown. This retrospective cohort study characterizes lipid profiles at baseline and after nonmyeloablative allogeneic HCT for SCD. We analyzed data from 116 patients after nonmyeloablative HLA-matched sibling or haploidentical HCT for SCD at the NIH from 2009 to 2021. TC, HDL-c, LDL-c, and TG were collected pre-HCT, 1-year post-HCT, and annually thereafter. Data were analyzed using linear generalized estimating equation regression modeling. Successful HCT was associated with a rise in TC, LDL-c, and HDL-c and a decline in TG post-HCT. After HCT, previously low lipid levels increased to the normal range. These changes occurred within the first year of HCT and were maintained thereafter. In patients with graft failure, TC and LDL-c levels remain unchanged from their pre-HCT baseline. Sirolimus use for graft versus host disease prophylaxis was associated with higher TG levels. These findings suggest that SCD dyslipidemia resolves with reversal of the SCD phenotype. The normalization of lipid parameters suggests SCD patients are not at increased risk for atherosclerosis after successful HCT compared to their peers; further studies with longer follow-up are required.
Collapse
Affiliation(s)
- Jackie Queen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Limerick
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Neal Jeffries
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Matthew M Hsieh
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert D Shamburek
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Courtney D Fitzhugh
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
4
|
Goldenberg M, Lanzkron S, Pecker LH. Late effects of hemopoietic stem cell transplant for sickle cell disease: monitoring and management. Expert Rev Hematol 2024; 17:891-905. [PMID: 39499235 PMCID: PMC11669372 DOI: 10.1080/17474086.2024.2423368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/27/2024] [Indexed: 11/07/2024]
Abstract
INTRODUCTION Allogeneic hemopoietic stem cell transplantation (HSCT) is a curative therapy for sickle cell disease (SCD). Exposure to both SCD and HSCT conditioning regimens is associated with late health effects. AREAS COVERED This review addresses post-HSCT outcomes and late health effects among individuals with SCD exposed to allogeneic HSCT regimens, summarizes recommendations for long-term care, and identifies future survivorship research needs. EXPERT OPINION Individuals with SCD exposed to HSCT and gene therapy require multidisciplinary care to monitor late health effects. To optimize care, multi-disciplinary clinics that include experts in late effects of HSCT exposure, SCD, complex chronic pain, mental health, and social work are needed. Research defining the late effects of exposure is needed to inform patient management and build clinical care infrastructure.
Collapse
Affiliation(s)
- Marti Goldenberg
- Pediatric Hematology Program, Division of Pediatric Hematology, Bloomberg Children’s Center at John’s Hopkins Hospital, Johns Hopkins University School of Medicine, United States
| | - Sophie Lanzkron
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lydia H Pecker
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Saib I, Alahmari B, Alsadi H, Alaskar A, Hejazi A, Salama H, Al Raiza A, Saleh ASA, Ibrahim A, Bakkar M, Ghori A, Alsuhaibani A, Alharbi A, Alanazi T, Ahmed R, Shehabeddine I, Alkhraisat S, Alharbi A, Mahasneh I, Ballili M, Aljubour Z, Ahmed M, Alzahrani M. Effect of ABO Mismatch and Red Blood Cell Alloimmunization on the Outcome of Hematopoietic Cell Transplantation for Sickle Cell Disease. Transplant Cell Ther 2024:S2666-6367(24)00747-4. [PMID: 39522720 DOI: 10.1016/j.jtct.2024.11.003] [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: 06/06/2024] [Revised: 09/21/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Hematopoietic stem cell transplantation (HCT) remains the sole well-established curative option for patients with sickle cell disease (SCD). Nonmyeloablative conditioning has been used to improve outcomes and reduce toxicities in adult SCD patients. However, recipient-donor ABO incompatibility and alloimmunization may be significant impediments to successful outcomes of HSCT in SCD patients owing to risks of hemolysis, delayed engraftment, poor graft function, and graft failure (GF). Here we report our experience with allogeneic HCT for SCD and the effects of RBC group mismatch and alloimmunization on the outcome of transplant recipients. We conducted a retrospective analysis of all SCD patients age >14 years who underwent HCT between January 2015 and February 2022 at our center. All patients received i.v. alemtuzumab (1 mg/kg divided over 5 days on days -7 to -3) and 300 cGy total body irradiation on day -2 or -1 for conditioning. Pretransplantation preparation consisted of hydroxyurea at the maximum tolerated dose for 2 to 3 months and 1 session of exchange transfusion. Peripherally mobilized CD34 stem cells targeting 10 × 106 /kg of recipient weight were used. For graft-versus-host disease prophylaxis, sirolimus was started on day -1 and continued for 1 year with tapering if lymphoid chimerism was >50%. For patients with major ABO incompatibility, we administered 2 doses of rituximab (375 mg/m2) and 3 sessions of plasmapheresis before starting the conditioning regimen, targeting an isohemagglutinin titer <1/32. The primary objective was to determine the impact of RBC group mismatch and alloimmunization on the outcomes of the HCT recipients. The secondary objective was to assess the impact of GF on overall survival (OS). Logistic regression was done to evaluate predictors for GF. The Kaplan-Meier method was used for survival analysis. A total of 194 patients were included, with a median age of 26 years. Their median baseline hemoglobin and hemoglobin S values were 93 g/L and 71.3%, respectively. Indications for HCT included recurrent vaso-occlusive crisis in 52.5% of the patients, central nervous system events in 19.6%, and acute chest syndrome in 17%. After a median follow-up of 28.8 months (range, 5 to 83 months), 16 patients (8%) experienced GF (3 with primary GF and 13 with secondary GF). On univariate analysis, ABO minor incompatibility and RBC alloantibodies against donor non-ABO antigens were predictive of GF. On multivariate analysis, RBC alloantibodies against donor non-ABO antigens (odds ratio, 8.29; 95% confidence interval, 2.01 to 34.05; P = .0033) was the sole factor predictive of GF. None of the 16 patients with major ABO incompatibility developed GF. The 2-year OS for all patients was 95%. The 2-year OS was 98% in patients without GF, compared to 74% in patients with GF (P < .0001). In our SCD patients who underwent HSCT, the presence of RBC alloantibodies against donor non-ABO antigens was an independent risk factor for GF. Patients with GF had inferior survival, and strategies for decreasing the risk of GF are needed.
Collapse
Affiliation(s)
- Israa Saib
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Bader Alahmari
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Husam Alsadi
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmed Alaskar
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; Saudi Scientific Society of Blood and Marrow Transplantation, Jeddah, Saudi Arabia
| | - Ayman Hejazi
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; Saudi Scientific Society of Blood and Marrow Transplantation, Jeddah, Saudi Arabia
| | - Hind Salama
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abdulrahman Al Raiza
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abdullah S Al Saleh
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ayman Ibrahim
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mohammed Bakkar
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abdulrauf Ghori
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmed Alsuhaibani
- Pathology and Laboratory Medicine Department, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmed Alharbi
- Pathology and Laboratory Medicine Department, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Tahani Alanazi
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Rasha Ahmed
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Inaam Shehabeddine
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Suha Alkhraisat
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Amani Alharbi
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Isam Mahasneh
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Maybelle Ballili
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Zied Aljubour
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mazen Ahmed
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mohsen Alzahrani
- Division of Hematology & HSCT, Department of Oncology, King Abdulaziz Medical City, Ministry of National Guards-Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; Saudi Scientific Society of Blood and Marrow Transplantation, Jeddah, Saudi Arabia.
| |
Collapse
|
6
|
Ruhl AP, Shalhoub R, Jeffries N, Limerick EM, Leonard A, Barochia AV, Tisdale JF, Fitzhugh CD, Hsieh MM. Pulmonary Function after Nonmyeloablative Hematopoietic Cell Transplant for Sickle Cell Disease. Ann Am Thorac Soc 2024; 21:1398-1406. [PMID: 39189784 PMCID: PMC11451896 DOI: 10.1513/annalsats.202309-771oc] [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: 09/06/2023] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
Rationale: Sickle cell disease (SCD) is a monogenetic condition with recurring vasoocclusive events causing lifelong pulmonary morbidity and mortality. There is increasing access to curative therapies, such as hematopoietic cell transplant (HCT), for people living with SCD. However, more information on pulmonary function in adults with SCD after HCT is needed to best guide decisions for HCT and post-HCT care. Objectives: To test the hypothesis that forced expiratory volume in 1 second (FEV1) and other pulmonary function testing (PFT) parameters remain stable 3 years after HCT. Methods: People living with SCD undergoing nonmyeloablative HCT in a prospective cohort at the NIH Clinical Center from 2004 to 2019 were evaluated for enrollment. Global Lung Function Initiative reference equations and descriptive statistics were calculated before HCT and annually for 3 years. Six-minute-walk distance (6MWD) testing was performed. Generalized estimating equations were employed to evaluate interindividual changes in PFT parameters and 6MWD. Results: Of 97 patients with SCD undergoing HCT, 41 (42%) were female with median (25th, 75th percentile) age 31.8 (24.8, 38.0) years. Each year of measurement included the following numbers of subjects available for analysis with PFTs: baseline (n = 97), Year 1 (n = 91), Year 2 (n = 72), and Year 3 (n = 55); and the following numbers of subjects available for analysis with 6MWD: baseline (n = 79), Year 1 (n = 73), Year 2 (n = 57), and Year 3 (n = 41). Pre-HCT FEV1 was median (25th, 75th percentile) 68.3% (61.3%, 80.3%) and 69.2% (60.8%, 77.7%) 3 years after HCT, and pre-HCT diffusing capacity of the lung for carbon monoxide (DlCO) was 60.5% (53.0%, 66.3%) and 64.6% (55.1%, 73.4%) 3 years after HCT. Generalized estimating equations estimated that DlCO percent predicted increased significantly by 3.7% (95% confidence interval, 1.0%, 6.3%), and the 6MWD significantly increased by 25.9 (6.6, 45.2) meters 3 years after HCT, whereas there was no significant change in percent predicted FEV1 or FVC compared with before HCT. Conclusions: Overall, PFT results remained stable and there was an improvement in DlCO and 6MWD in this predominantly adult cohort undergoing nonmyeloablative HCT for SCD. Allogeneic HCT for SCD may cease the cycle of vasoocclusive pulmonary injury and prevent continued damage. Multicenter studies are needed to evaluate the long-term lung health effects of HCT for SCD in adults and children.
Collapse
Affiliation(s)
- A. Parker Ruhl
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases
- Pulmonary Branch
| | | | | | - Emily M. Limerick
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | | | - John F. Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Courtney D. Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Matthew M. Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
7
|
Dovern E, Aydin M, Hazenberg MD, Tang MW, Suijk EM, Hoogendoorn GM, Van Tuijn CFJ, Kerkhoffs JL, Rutten CE, Zeerleder SS, de la Fuente J, Biemond BJ, Nur E. Azathioprine/hydroxyurea preconditioning prior to nonmyeloablative matched sibling donor hematopoietic stem cell transplantation in adults with sickle cell disease: A prospective observational cohort study. Am J Hematol 2024; 99:1523-1531. [PMID: 38733340 DOI: 10.1002/ajh.27360] [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: 03/13/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
Nonmyeloablative, matched sibling donor hematopoietic stem cell transplantation with alemtuzumab/total body irradiation (TBI) conditioning is a curative therapy with low toxicity for adults with sickle cell disease (SCD). However, relatively low donor chimerism levels and graft rejection remain important challenges. We hypothesized that adding azathioprine/hydroxyurea preconditioning will improve donor chimerism levels and reduce graft failure rate. In this prospective cohort study, we enrolled consecutive adult patients with SCD undergoing matched sibling donor transplantation at the Amsterdam UMC. Patients received azathioprine 150 mg/day and hydroxyurea 25 mg/kg/day for 3 months prior to alemtuzumab 1 mg/kg and 300 cGy TBI conditioning. Twenty patients with SCD (median age 26 years [range 19-49], 13 females) were transplanted. Median follow-up was 46.0 months (IQR 21.8-57.9). One-year overall survival and event-free survival (graft failure or death) were both 95% (95% confidence interval 86-100). Mean donor myeloid and T-cell chimerism 1-year post-transplant were 95.2% (SD ±10.6) and 67.3% (±15.3), respectively. One patient (5%) experienced graft failure without autologous regeneration, resulting in infections and death. All other patients had a corrected SCD phenotype and were able to discontinue sirolimus. Three patients were successfully treated with alemtuzumab (1 mg/kg) after the transplant because of declining donor chimerism and cytopenias to revert impending graft rejection. Toxicity was mostly related to sirolimus and alemtuzumab. One patient developed steroid-responsive grade II intestinal acute graft-versus-host disease. Collectively, preconditioning with azathioprine/hydroxyurea prior to nonmyeloablative matched sibling donor transplantation resulted in excellent event-free survival and robust donor T-cell chimerism, enabling the successful withdrawal of sirolimus. ClinicalTrials.gov: NCT05249452.
Collapse
Affiliation(s)
- Elisabeth Dovern
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Mesire Aydin
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Man Wai Tang
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Elisabeth M Suijk
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Gerianne M Hoogendoorn
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Charlotte F J Van Tuijn
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Caroline E Rutten
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Sacha S Zeerleder
- Department of Hematology, Division of Internal Medicine, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Josu de la Fuente
- Department of Paediatrics, St. Mary's Hospital, Imperial Healthcare NHS Trust, London, UK
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Bart J Biemond
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Erfan Nur
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Blood Cell Research, Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Alsuhebany N, Alshehri B, Aldairem A, Aleissa MM, AlQahtani H, Albarqi K, Almotairi B, AlAmri M, Alanazi W, Alay S, Alahmari B, Alzahrani M. Herpes simplex virus reactivation among hematopoietic stem cell transplant recipients: re-evaluating acyclovir dosage. Bone Marrow Transplant 2024; 59:1043-1045. [PMID: 38627451 DOI: 10.1038/s41409-024-02273-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 07/07/2024]
Affiliation(s)
- Nada Alsuhebany
- Department of Pharmaceutical Care, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Bashayer Alshehri
- Department of Pharmaceutical Care, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Atheer Aldairem
- Department of Pharmaceutical Care, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Muneerah M Aleissa
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Hajar AlQahtani
- Department of Pharmaceutical Care, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Khalid Albarqi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Basil Almotairi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mohammed AlAmri
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Walid Alanazi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Saeed Alay
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Bader Alahmari
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Oncology, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Mohsen Alzahrani
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Oncology, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Dovern E, Aydin M, DeBaun MR, Alizade K, Biemond BJ, Nur E. Effect of allogeneic hematopoietic stem cell transplantation on sickle cell disease-related organ complications: A systematic review and meta-analysis. Am J Hematol 2024; 99:1129-1141. [PMID: 38517255 DOI: 10.1002/ajh.27297] [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: 11/23/2023] [Revised: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
Sickle cell disease (SCD)-related organ complications are a major cause of morbidity and mortality in patients with SCD. We sought to assess whether hematopoietic stem cell transplantation (HSCT) stabilizes, attenuates, or exacerbates organ decline. We performed a systematic review and meta-analysis of trials investigating organ function before and after HSCT in patients with SCD. We searched MEDLINE/PubMed and EMBASE up to September 21, 2023. Continuous data were expressed as standardized mean difference (SMD) and pooled in a weighted inverse-variance random-effects model; binomial data were expressed as risk ratio (RR) using the Mantel-Haenszel random-effects meta-analyses. Of 823 screened studies, 34 were included in this review. Of these, 17 (774 patients, 23.6% adults, 86.3% HLA-identical sibling donor, 56.7% myeloablative conditioning regimen) were included in the meta-analyses. Pulmonary function remained stable. Mean tricuspid regurgitant jet velocity decreased but did not reach statistical significance. In children, estimated glomerular filtration rate decreased (SMD -0.80, p = .01), and the presence of proteinuria increased (RR 2.00, p = <.01), while splenic uptake and phagocytic function improved (RR 0.31, p = <.01; RR 0.23, p = <.01). Cerebral blood flow improved (SMD -1.39, p = <.01), and a low incidence of stroke after transplantation in high-risk patients was found. Retinopathy and avascular osteonecrosis were investigated in only one study, showing no significant changes. While HSCT can improve some SCD-related organ dysfunctions, transplantation-related toxicity may have an adverse effect on others. Future research should focus on identifying individuals with SCD who might benefit most from HSCT and which forms of organ damage are more likely to exacerbate post-transplantation.
Collapse
Affiliation(s)
- Elisabeth Dovern
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, The Netherlands
| | - Mesire Aydin
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, The Netherlands
| | - Michael R DeBaun
- Department of Pediatrics, Vanderbilt-Meharry Center of Excellence in Sickle Cell Disease, Vanderbilt University Medical Center, Nashville, USA
| | - Komeil Alizade
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, The Netherlands
| | - Bart J Biemond
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, The Netherlands
| | - Erfan Nur
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, The Netherlands
- Department of Blood Cell Research, Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Damlaj M, Alahmari B, Alaskar A, Alhejazi A, Alsadi H, Ahmed M, Alanazi T, Ahmed R, Alharbi A, Shehabeddine I, Alzaidi A, Alkhuraisat S, Mahassnah I, Alquraan H, Ballili M, Alzahrani M. Favorable outcome of non-myeloablative allogeneic transplantation in adult patients with severe sickle cell disease: A single center experience of 200 patients. Am J Hematol 2024; 99:1023-1030. [PMID: 38488686 DOI: 10.1002/ajh.27295] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 05/16/2024]
Abstract
Allogeneic hematopoietic stem cell transplant (HSCT) for adults with severe sickle cell disease (SCD) is potentially curative but not commonly utilized therapy due to complications such as graft failure (GF) and organ toxicity. Herein, we are reporting our long-term outcome data of non-myeloablative (NMA) HSCT in adults with severe SCD with emphasis on factors predicting event free survival (EFS). Adults with severe SCD undergoing NMA match-related donor allogeneic HSCT from 2015 to 2021 with at least 12 months of follow-up were included. A total of 200 patients were included with a median age of 26 years (14-43) and 56% were male. The median infused CD34 dose was 13.7 (5.07-25.8), respectively. Median absolute neutrophil count engraftment was 19 (13-39) days with 51% of patients receiving GCSF to expedite recovery. A total of 17 patients experienced GF; 3 as primary and 14 as secondary within a median time of 204 days (40-905). A 76% successfully discontinued sirolimus at the last follow-up. Median follow-up for the cohort is 29.2 (2.1-71.4) months. Estimated 3-year EFS and OS were 88.2% (81.9-92.5) and 94.6% (89.2-97.3). At multivariable analysis, minor ABC incompatibility hazard ratio (HR) 4 (1.3-12.1; 0.014) and allo-antibody against non-ABO donor antigens HR 4.3 (1.3-14.1; 0.016) were significant for EFS. No clonal evolution or myeloid malignancies were seen. This largest single-center report of NMA HSCT in adults with severe SCD further delineated its feasibility, potential toxicities, and fertility outcomes. GF remains a major impediment and appears dependent on ABO matching and non-ABO antibodies.
Collapse
Affiliation(s)
- Moussab Damlaj
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Blood and Cancer Research Unit, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Division of Hematology, Sheikh Shakhbout Medical City, Abu Dhabi, UAE
- College of Medicine, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, UAE
| | - Bader Alahmari
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Blood and Cancer Research Unit, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmed Alaskar
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Blood and Cancer Research Unit, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ayman Alhejazi
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Blood and Cancer Research Unit, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Husam Alsadi
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mazin Ahmed
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Tahani Alanazi
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Rasha Ahmed
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Amani Alharbi
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Inaam Shehabeddine
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Afnan Alzaidi
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Suha Alkhuraisat
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Isam Mahassnah
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Hamza Alquraan
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Maybelle Ballili
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohsen Alzahrani
- Division of Hematology, King Abdulaziz Medical City - Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Blood and Cancer Research Unit, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Spencer Chapman M, Cull AH, Ciuculescu MF, Esrick EB, Mitchell E, Jung H, O'Neill L, Roberts K, Fabre MA, Williams N, Nangalia J, Quinton J, Fox JM, Pellin D, Makani J, Armant M, Williams DA, Campbell PJ, Kent DG. Clonal selection of hematopoietic stem cells after gene therapy for sickle cell disease. Nat Med 2023; 29:3175-3183. [PMID: 37973947 PMCID: PMC10719109 DOI: 10.1038/s41591-023-02636-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023]
Abstract
Gene therapy (GT) provides a potentially curative treatment option for patients with sickle cell disease (SCD); however, the occurrence of myeloid malignancies in GT clinical trials has prompted concern, with several postulated mechanisms. Here, we used whole-genome sequencing to track hematopoietic stem cells (HSCs) from six patients with SCD at pre- and post-GT time points to map the somatic mutation and clonal landscape of gene-modified and unmodified HSCs. Pre-GT, phylogenetic trees were highly polyclonal and mutation burdens per cell were elevated in some, but not all, patients. Post-GT, no clonal expansions were identified among gene-modified or unmodified cells; however, an increased frequency of potential driver mutations associated with myeloid neoplasms or clonal hematopoiesis (DNMT3A- and EZH2-mutated clones in particular) was observed in both genetically modified and unmodified cells, suggesting positive selection of mutant clones during GT. This work sheds light on HSC clonal dynamics and the mutational landscape after GT in SCD, highlighting the enhanced fitness of some HSCs harboring pre-existing driver mutations. Future studies should define the long-term fate of mutant clones, including any contribution to expansions associated with myeloid neoplasms.
Collapse
Affiliation(s)
- Michael Spencer Chapman
- Wellcome Sanger Institute, Hinxton, UK
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Alyssa H Cull
- York Biomedical Research Institute, University of York, York, UK
| | | | - Erica B Esrick
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Emily Mitchell
- Wellcome Sanger Institute, Hinxton, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | | | | | | | - Margarete A Fabre
- Wellcome Sanger Institute, Hinxton, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Jyoti Nangalia
- Wellcome Sanger Institute, Hinxton, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - Joanne Quinton
- York Biomedical Research Institute, University of York, York, UK
| | - James M Fox
- York Biomedical Research Institute, University of York, York, UK
| | - Danilo Pellin
- Harvard Medical School, Boston, MA, USA
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Julie Makani
- Muhimbili University of Health and Allied Sciences (MUHAS), Dar-es-Salaam, Tanzania
- SickleInAfrica Clinical Coordinating Center, MUHAS, Dar-es-Salaam, Tanzania
- Imperial College London, London, UK
| | - Myriam Armant
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - David A Williams
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Peter J Campbell
- Wellcome Sanger Institute, Hinxton, UK.
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK.
| | - David G Kent
- York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
12
|
Duvoux C, Blaise L, Matimbo JJ, Mubenga F, Ngongang N, Hurtova M, Laurent A, Augustin J, Calderaro J, Reizine E, Luciani A, Habibi A, Bachir D, Vole G, Gellen-Dautremer J, Leroy V, Levesque E, Bartolucci P. The liver in sickle cell disease. Presse Med 2023; 52:104212. [PMID: 37981193 DOI: 10.1016/j.lpm.2023.104212] [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] [Indexed: 11/21/2023] Open
Abstract
Liver involvement in SCD patients is frequent but often misdiagnosed or underestimated, except in case of advanced liver diseases. Because of so far poorly recognized forms of chronic SCD-related vascular injury that can silently evolved towards end stages or facilitate ACLF, any persisting liver function tests abnormalities should be carefully investigated, following the above proposed algorithm. Work up and management must be considered multidisciplinary in relationship with a Hepatologist. Early SCD hepatopathy should prompt revision of SCD management to prevent further liver injury and decompensation, discussing transfusion exchanges and hydro urea when not yet initiated, and control for any cofactor of liver injury. The role of HSCT in early SCD hepatopathies also deserves evaluation. In advanced SCD hepatopathies, liver transplantation, which has been rarely performed so far, is the only therapeutic option associated with improved survival. It should definitely be discussed- either electively in case of decompensation in SCD cirrhosis or jaundice/recurrent cholangitis in cholestatic diseases, with excellent outcome, - or emergently in case of ALF or ACLF with more mitigate results. To improve knowledge and management of SCD liver diseases, creation of national and international registries, as well as longitudinal observational cohorts are encouraged.
Collapse
Affiliation(s)
- Christophe Duvoux
- Department of Hepatology and Medical Liver Transplant Unit, Henri Mondor Hospital-APHP, University Paris Est Créteil, France.
| | - Lorraine Blaise
- Department of Hepatology and Liver Oncology, Avicenne Hospital-APHP, Bobigny, France
| | - Jean-Jacques Matimbo
- Department of Hepatology and Medical Liver Transplant Unit, Henri Mondor Hospital-APHP, University Paris Est Créteil, France; Department of Hepatology and Liver Oncology, Avicenne Hospital-APHP, Bobigny, France; Department of Hepatology and Gastroenterology, Clinique Universitaire Kinshasa, Democratic Republic of Congo
| | | | - Norbert Ngongang
- Department of Hepatology and Medical Liver Transplant Unit, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Monika Hurtova
- Department of Hepatology and Medical Liver Transplant Unit, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Alexis Laurent
- Department of Digestive & HPB Surgery, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Jérémy Augustin
- Department of Pathology, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Julien Calderaro
- Department of Pathology, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Edouard Reizine
- Department of Radiology, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Alain Luciani
- Department of Radiology, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Anoosha Habibi
- UMGGR, Department of Internal Medicine, Sickle Cell National Referral Center, Henri Mondor-Hospital APHP, University Paris Est Créteil, France; IMRB, UPEC, INSERM, EFS, Team Pirenne. University Paris Est Créteil, France
| | - Dora Bachir
- UMGGR, Department of Internal Medicine, Sickle Cell National Referral Center, Henri Mondor-Hospital APHP, University Paris Est Créteil, France
| | - Geoffroy Vole
- UMGGR, Department of Internal Medicine, Sickle Cell National Referral Center, Henri Mondor-Hospital APHP, University Paris Est Créteil, France; IMRB, UPEC, INSERM, EFS, Team Pirenne. University Paris Est Créteil, France
| | | | - Vincent Leroy
- Department of Hepatology and Medical Liver Transplant Unit, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Eric Levesque
- Department of Anesthesia and Surgical Intensive Care-liver ICU, Henri Mondor Hospital-APHP, University Paris Est Créteil, France
| | - Pablo Bartolucci
- UMGGR, Department of Internal Medicine, Sickle Cell National Referral Center, Henri Mondor-Hospital APHP, University Paris Est Créteil, France; IMRB, UPEC, INSERM, EFS, Team Pirenne. University Paris Est Créteil, France
| |
Collapse
|
13
|
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
|
14
|
Ayuk-Arrey AT, Martin OY, Darbari I, Abraham A, Nickel RS. Disparity in HLA typing rates among hospitalized pediatric patients with sickle cell disease. Transplant Cell Ther 2023; 30:S2666-6367(23)01641-X. [PMID: 39492481 DOI: 10.1016/j.jtct.2023.10.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Hematopoietic stem cell transplantation (HSCT) using an HLA-identical sibling donor is a well-established cure for sickle cell disease (SCD). The proportion of patients with SCD who have completed HLA typing, a key first step in considering HSCT, is unknown. We sought to determine the prevalence of HLA typing among patients with SCD hospitalized at our institution, identify characteristics associated with having had typing, and describe the acceptability of this testing. Area deprivation index (ADI) was studied as a measure of family socioeconomic status. Among 291 hospitalized patients with SCD, 71 (24%) had HLA typing at study baseline. On multivariate analysis, HLA typing at baseline was associated with hydroxyurea (OR 3.10, p=0.003) and chronic transfusion therapy (OR 7.04, p<0.001), prior intensive care unit admission (OR: 1.89, p=0.04), and ADI score (OR 0.74, p=0.004). Patients who lived in more disadvantaged neighborhoods were less likely to have had HLA typing. Among patients with healthy full siblings, 69% of families without typing at baseline were interested in obtaining testing when explictly offered. These findings suggest that continued work is needed to raise family awareness of HLA typing and ensure equal access to curative therapy for SCD.
Collapse
Affiliation(s)
| | - Olufunke Y Martin
- Children's National Hospital, Washington, DC; University of Texas Southwestern Medical Center, Dallas, TX
| | - Isha Darbari
- University of Texas Southwestern Medical Center, Dallas, TX
| | - Allistair Abraham
- Children's National Hospital, Washington, DC; George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Robert Sheppard Nickel
- Children's National Hospital, Washington, DC; George Washington University School of Medicine and Health Sciences, Washington, DC.
| |
Collapse
|
15
|
Miller YM, Bakhtary S, Chou ST, Hailu B, Reik RA, Richard RH, Spencer BR, Witherspoon R, Delaney M. Involvement of Diverse Populations in Transfusion Medicine Research. Transfus Med Rev 2023; 37:150766. [PMID: 37993382 PMCID: PMC11032214 DOI: 10.1016/j.tmrv.2023.150766] [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: 05/19/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 11/24/2023]
Abstract
Communities of color and diverse communities (eg, race, socioeconomic status, language, sexual orientation etc.) have not been recruited and enrolled equitably to participate in research studies in transfusion medicine. The exclusion of diverse communities in transfusion research can lead to health disparities lack of access to approved therapeutics and unequal allocation of interventions, resulting in missed opportunities to optimize health for individuals and communities. Involvement of diverse populations in research goes beyond inclusion as research subjects. Strategies should include specific studies on health conditions of importance to diverse communities with stable funding sources and specific funding announcements to develop projects led by diverse researchers, mentorship of diverse researchers, and openness to various ways of communicating research plans. Qualitative approaches and interdisciplinary collaboration should be supported to enhance inclusivity.
Collapse
Affiliation(s)
| | - Sara Bakhtary
- Transfusion Medicine, Department of Laboratory Medicine, University of California San Francisco, USA
| | - Stella T Chou
- Division of Transfusion Medicine, The Children's Hospital of Philadelphia University of Pennsylvania School of Medicine, USA
| | | | | | - Raven Hardy Richard
- National Institutes of Health (NIH) National Human Genome Research Institute (NHGRI), USA
| | | | | | - Meghan Delaney
- Division of Pathology & Laboratory Medicine Children's National Hospital, The George Washington University, USA
| |
Collapse
|
16
|
Nickel RS, Maher JY. RIC does not do the trick to prevent the high infertility risk in females with sickle cell disease after hematopoietic cell transplantation. Transplant Cell Ther 2023; 29:537-538. [PMID: 37625886 DOI: 10.1016/j.jtct.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Affiliation(s)
- Robert Sheppard Nickel
- Children's National Hospital, Division of Hematology, Washington, DC; Children's National Hospital, Division of Blood and Marrow Transplantation, Washington, DC; The George Washington University School of Medicine and Health Sciences, Washington, DC.
| | - Jacqueline Y Maher
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Pediatric and Adolescent Gynecology, National Institutes of Health, Bethesda, MD; Children's National Hospital, Pediatric and Adolescent Gynecology Program, Washington, DC
| |
Collapse
|
17
|
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: 32] [Impact Index Per Article: 16.0] [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
|
18
|
Sachdev V, Limerick E, Nguyen ML, Li W, Jeffries N, Ramachandra S, Tofovic D, Rondelli D, Al Zahrani M, Aljizeeri A, Saraf S, Hsieh M, Fitzhugh CD. Cardiac effects 2 years after successful non-myeloablative human leukocyte antigen-matched related donor hematopoietic cell transplants in sickle cell disease. Am J Hematol 2023; 98:E219-E221. [PMID: 37282828 PMCID: PMC10482363 DOI: 10.1002/ajh.26985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/08/2023]
Affiliation(s)
- Vandana Sachdev
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - Emily Limerick
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - My-Le Nguyen
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - Wen Li
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | - Neal Jeffries
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | | | - David Tofovic
- University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Mohsen Al Zahrani
- King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmed Aljizeeri
- King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Santosh Saraf
- University of Illinois at Chicago, Chicago, Illinois, USA
| | - Matthew Hsieh
- NHLBI, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
19
|
Inam Z, Tisdale JF, Leonard A. Outcomes and long-term effects of hematopoietic stem cell transplant in sickle cell disease. Expert Rev Hematol 2023; 16:879-903. [PMID: 37800996 DOI: 10.1080/17474086.2023.2268271] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Hematopoietic stem cell transplant (HSCT) is the only readily available curative option for sickle cell disease (SCD). Cure rates following human leukocyte antigen (HLA)-matched related donor HSCT with myeloablative or non-myeloablative conditioning are >90%. Alternative donor sources, including haploidentical donor and autologous with gene therapy, expand donor options but are limited by inferior outcomes, limited data, and/or shorter follow-up and therefore remain experimental. AREAS COVERED Outcomes are improving with time, with donor type and conditioning regimens having the greatest impact on long-term complications. Patients with stable donor engraftment do not experience SCD-related symptoms and have stabilization or improvement of end-organ pathology; however, the long-term effects of curative strategies remain to be fully established and have significant implications in a patient's decision to seek therapy. This review covers currently published literature on HSCT outcomes, including organ-specific outcomes implicated in SCD, as well as long-term effects. EXPERT OPINION HSCT, both allogeneic and autologous gene therapy, in the SCD population reverses the sickle phenotype, prevents further organ damage, can resolve prior organ dysfunction in both pediatric and adult patients. Data support greater success with HSCT at a younger age, thus, curative therapies should be discussed early in the patient's life.
Collapse
Affiliation(s)
- Zaina Inam
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
20
|
Dovern E, Biemond BJ, Nur E. Case report: Successful treatment with daratumumab for pure red cell aplasia in a patient with mixed lymphoid chimerism after ABO-mismatched stem cell transplant for sickle cell disease. Front Immunol 2023; 14:1212007. [PMID: 37426651 PMCID: PMC10326381 DOI: 10.3389/fimmu.2023.1212007] [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: 04/25/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023] Open
Abstract
Pure red cell aplasia (PRCA) is a serious complication after ABO-mismatched allogeneic hematopoietic stem cell transplantation (HSCT). Following HSCT, persistent anti-donor isohemagglutinins against donor ABO antigens are considered the immunological cause of PRCA. Patients with post-transplant PRCA are at risk for graft rejection and prolonged red blood cell transfusion dependency. No standard treatment exists. Recently however, the anti-CD38 monoclonal antibody daratumumab has been reported to be an effective treatment for post-transplant PRCA in patients with complete donor chimerism. Here, we describe the first case of PRCA in a patient with mixed lymphoid patient/donor chimerism that was successfully treated with daratumumab. This is also the first report of a transplant recipient with sickle cell disease who was treated with this relatively new approach. Fourteen months post-transplantation and twelve months after treatment with daratumumab, our patient has a normal complete blood count and the anti-donor isohemagglutinins remain undetectable despite mixed lymphoid chimerism. Mixed chimerism is a common manifestation in adult patients with sickle cell disease transplanted with non-myeloablative conditioning and a matched sibling donor. The application of non-myeloablative HSCT for patients with sickle cell disease is steadily increasing. Therefore, the incidence of PRCA in this setting might also increase. As the risk of graft rejection due to PRCA can be especially high in patients with mixed chimerism, clinicians should be aware that daratumumab can be an effective treatment in the setting of mixed chimerism.
Collapse
Affiliation(s)
- E. Dovern
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, Netherlands
| | - B. J. Biemond
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, Netherlands
| | - E. Nur
- Department of Hematology, Amsterdam University Medical Centers, location University of Amsterdam, Amsterdam, Netherlands
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| |
Collapse
|
21
|
Dovern E, Nijland SJAM, van Muilekom MM, Suijk LMJ, Hoogendoorn GM, Mekelenkamp H, Biemond BJ, Haverman L, Nur E. Physical, Mental, and Social Health of Adult Patients with Sickle Cell Disease after Allogeneic Hematopoietic Stem Cell Transplantation: A Mixed-Methods Study. Transplant Cell Ther 2023; 29:283.e1-283.e9. [PMID: 36634737 DOI: 10.1016/j.jtct.2023.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/02/2023] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
Patients with sickle cell disease (SCD) experience a considerable physical and psychosocial disease burden. In recent years, the application of allogeneic hematopoietic stem cell transplantation (HSCT) to treat adults with SCD has increased. A thorough understanding of patients' physical, mental, and social health before and after cure is needed to meet the needs of this growing group of patients. We aimed to explore the perspectives of adult SCD patients on the changes in their experienced health and personal life goals after being cured. A mixed-methods approach was used, comprising a semistructured interview and a set of 9 Patient Reported Outcomes Measurement Information System (PROMIS) measures. Adult SCD patients who underwent HSCT at least 1 year earlier were eligible to participate in the study. Interviews were thematically analyzed using MAXQDA software. PROMIS T scores were compared with reference scores of the general population using SPSS Statistics. Ten patients participated in the study; their median age was 29.5 years (range, 19 to 49 years), and their median time since HSCT was 2.7 years (range, 1.0 to 3.5 years). Themes from the interviews were (1) pain/living pain free, (2) physical well-being, (3) mental well-being, (4) perspective/ outlook, (5) education/work, (6) family/friends, and (7) activities/participation. Following the PROMIS framework, we described these themes in a narrative synthesis according to health domain and categorized in 4 chronological time phases: before HSCT, first year post-transplantation, current situation, and future expectations. Physical health improved greatly, but transplantation-related toxicity, ongoing pain from avascular osteonecrosis, and fatigue negatively impacted quality of life in some patients. Furthermore, emotional struggles during the post-transplantation period were common, and patients expressed a need for psychological help. Patients reported improvements in social health and the ability to pursue personal life goals. The mean T scores of all PROMIS measures fell within the normal symptom limits compared with reference data of the general population, although, large variations were observed among the participants, matching our qualitative findings. In general, adult SCD patients experienced improved physical, mental, and social health after cure by HSCT and were able to pursue personal life goals. Yet they found themselves confronted with a new and unfamiliar reality that brought different challenges. Pain due to irreversible avascular osteonecrosis continued to have a negative impact. Clinicians should aim to help patients have realistic expectations before transplantation and offer timely psychological care.
Collapse
Affiliation(s)
- Elisabeth Dovern
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands.
| | - Sterre J A M Nijland
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Maud M van Muilekom
- Department of Child and Adolescent Psychiatry & Psychosocial Care, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Mental Health and Personalized Medicine, Amsterdam Public Health, Amsterdam, The Netherlands; Child Development, Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Liesbeth M J Suijk
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Gerianne M Hoogendoorn
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Hilda Mekelenkamp
- Division of Stem Cell Transplantation, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart J Biemond
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Lotte Haverman
- Department of Child and Adolescent Psychiatry & Psychosocial Care, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Mental Health and Personalized Medicine, Amsterdam Public Health, Amsterdam, The Netherlands; Child Development, Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Erfan Nur
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Department of Blood Cell Research, Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Redjoul R, Beckerich F, de Luna G, Leclerc M, Hebert N, Sloma I, Menouche D, Bartolucci P, Maury S. ABO blood barrier to engraftment after allogeneic stem cell transplantation in sickle cell disease: A case-story with two successive HLA-matched sibling donors. Am J Hematol 2023; 98:692-696. [PMID: 36510370 DOI: 10.1002/ajh.26808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Rabah Redjoul
- Hematology Department, AP-HP, Hôpital Henri Mondor, Créteil, France
| | | | - Gonzalo de Luna
- AP-HP, Hôpital Henri Mondor, Unité des maladies génétiques du globule rouge and Etablissement Français du Sang, Créteil, France
| | - Mathieu Leclerc
- Hematology Department, AP-HP, Hôpital Henri Mondor, Créteil, France
- IMRB, INSERM U955, Paris Est Créteil University UPEC, Créteil, France
| | - Nicolas Hebert
- AP-HP, Hôpital Henri Mondor, Unité des maladies génétiques du globule rouge and Etablissement Français du Sang, Créteil, France
| | - Ivan Sloma
- IMRB, INSERM U955, Paris Est Créteil University UPEC, Créteil, France
- Laboratoire d'onco-hématologie Moléculaire, AP-HP, Hôpital Henri Mondor, Créteil, France
| | - Dehbia Menouche
- Hematology Department, AP-HP, Hôpital Henri Mondor, Créteil, France
| | - Pablo Bartolucci
- AP-HP, Hôpital Henri Mondor, Unité des maladies génétiques du globule rouge and Etablissement Français du Sang, Créteil, France
- IMRB, INSERM U955, Paris Est Créteil University UPEC, Créteil, France
| | - Sébastien Maury
- Hematology Department, AP-HP, Hôpital Henri Mondor, Créteil, France
- IMRB, INSERM U955, Paris Est Créteil University UPEC, Créteil, France
| |
Collapse
|
23
|
Sharpe CC, Suddle A, Stuart-Smith S. An Overview of Solid Organ Transplantation in Patients With Sickle Cell Disease. Transplantation 2023; 107:596-604. [PMID: 36210501 DOI: 10.1097/tp.0000000000004305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Sickle cell disease is a common genetic disorder affecting >300 000 people across the world. The vast majority of patients cared for in high-resource settings live well into adulthood, but many develop a high burden of disease complications. Good standard of care including disease-modifying agents and transfusion programs limits the number of patients who develop end-stage organ disease, but for those that do, the prognosis can be very poor. Solid organ transplantation is a well-established mode of treatment for patients with sickle cell disease and kidney or liver failure, but appropriate patient selection and perioperative management are important for achieving good outcomes. Hematopoietic stem cell transplantation and gene therapy may offer novel treatment options for adult patients with chronic organ damage in the future, but these are not yet widely available. For now, good, holistic care and early intervention of end-organ complications can minimize the number of patients requiring solid organ transplantation later in life.
Collapse
Affiliation(s)
- Claire C Sharpe
- Department of Inflammation Biology, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Abid Suddle
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Sara Stuart-Smith
- Department of Haematology, Kings College Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
24
|
Leonard A, Furstenau D, Abraham A, Darbari DS, Nickel RS, Limerick E, Fitzhugh C, Hsieh M, Tisdale JF. Reduction in vaso-occlusive events following stem cell transplantation in patients with sickle cell disease. Blood Adv 2023; 7:227-234. [PMID: 36240296 PMCID: PMC9860452 DOI: 10.1182/bloodadvances.2022008137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 01/29/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is potentially curative for patients with sickle cell disease (SCD). Patients with stable donor engraftment after allogeneic HSCT generally do not experience SCD-related complications; however, there are no published data specifically reporting the change in vaso-occlusive events (VOE) after HSCT. Data regarding the number of VOEs requiring medical attention in the 2 years before allogeneic HSCT were compared with the number of VOEs in the 2 years (0-12 months and 12-24 months) after allogeneic HSCT in patients with SCD. One-hundred sixty-three patients with SCD underwent allogeneic HSCT between 2005 and 2019. The average age at the time of HSCT was 21 years (range, 7 months - 64 years). Most patients underwent nonmyeloablative conditioning (75% [N = 123]) and had a matched sibling donor (72% [N = 118]). The mean number of VOEs was reduced from 5.6 (range, 0-52) in the 2 years before HSCT to 0.9 (range, 0-12) in the 2 years after HSCT (P < .001). Among the post-HSCT events, VOE was more frequent during the first 12 months (0.8 [range, 0-12]) than at 12 to 24 months after HSCT (0.1 [range, 0-8) (P < .001)). In patients who had graft rejection (12%, N = 20), VOEs were reduced from 6.6 (range, 0-24) before HSCT to 1.1 (range, 0-6) and 0.8 (range, 0-8) at 0 to 12 months and 12 to 24 months after HSCT, respectively (P < .001). VOEs requiring medical care were significantly reduced after allogeneic HSCT for patients with SCD. These data will inform the development of novel autologous HSCT gene therapy approaches.
Collapse
Affiliation(s)
- Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
- Division of Hematology, Children's National Hospital, Washington, DC
| | - Dana Furstenau
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Allistair Abraham
- Center for Cancer and Immunology Research, CETI, Children’s National Hospital, Washington, DC
| | | | - Robert S. Nickel
- Division of Hematology, Children's National Hospital, Washington, DC
| | - Emily Limerick
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Courtney Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Matt Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - John F. Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
25
|
Hulbert ML, King AA, Shenoy S. Organ function indications and potential improvements following curative therapy for sickle cell disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:277-282. [PMID: 36485131 PMCID: PMC9820741 DOI: 10.1182/hematology.2022000372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Curative therapies for sickle cell disease include allogeneic hematopoietic stem cell transplantation (HSCT) and gene-modified autologous stem cell transplantation. HSCT has been used for 30 years with success measured by engraftment, symptom control, graft-vs-host disease (GVHD) risk, organ toxicity, and immune reconstitution. While human leukocyte antigen-matched sibling donor (MSD) transplants have excellent outcomes, alternate donor transplants (unrelated/haploidentical) are just beginning to overcome GVHD and engraftment hurdles to match MSD. Gene therapy, a newly developed treatment, is undergoing careful evaluation in many trials with varying approaches. The risk/benefit ratio to the patient in relation to outcomes, toxicities, and mortality risk drives eligibility for curative interventions. Consequently, eligibility criteria for MSD transplants can be less stringent, especially in the young. Posttransplant outcome analysis after the "cure" with respect to organ function recovery is essential. While established damage such as stroke is irreversible, transplant can help stabilize (pulmonary function), prevent further deterioration (stroke), improve (neurocognition), and protect unaffected organs. Tracking organ functions postintervention uniformly between clinical trials and for adequate duration is essential to answer safety and efficacy questions related to curative therapies. Age-appropriate application/outcome analyses of such therapies will be the ultimate goal in overcoming this disease.
Collapse
|
26
|
Long-term health outcomes following curative therapies for sickle cell disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:272-276. [PMID: 36485115 PMCID: PMC9820909 DOI: 10.1182/hematology.2022000373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Treatment options for patients with sickle cell disease (SCD) continue to rapidly expand and evolve. The goal of therapies such as an allogeneic hematopoietic stem cell transplant (HSCT), gene therapy, and gene editing is to cure rather than control SCD. The benefits of these therapies must be accompanied by minimizing long-term adverse health outcomes from SCD and its treatment. SCD can have adverse effects on a variety of organ systems, including the heart, lung, kidney, and reproductive system, leading to high disease burden, morbidity, and premature mortality in both pediatric and adult patients. While curative therapies are being increasingly used, there remains a paucity of data on the long-term health outcomes associated with these treatments in children and adults with SCD. There are data available regarding the effects of HSCT performed largely for malignant diseases, from which data on SCD outcomes may be extrapolated. However, given the significant differences between these 2 populations of patients who undergo HSCT, such extrapolation is imprecise at best. Furthermore, there are currently no published data on long-term health outcomes following gene therapy for SCD due to current short follow-up times. We summarize the limited data reported on health outcomes following HSCT for SCD and emphasize the need for more research within this area.
Collapse
|
27
|
Fitzhugh CD. Knowledge to date on secondary malignancy following hematopoietic cell transplantation for sickle cell disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:266-271. [PMID: 36485129 PMCID: PMC9820448 DOI: 10.1182/hematology.2022000371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Allogeneic hematopoietic cell transplantation, gene therapy, and gene editing offer a potential cure for sickle cell disease (SCD). Unfortunately, myelodysplastic syndrome and acute myeloid leukemia development have been higher than expected after graft rejection following nonmyeloablative conditioning and lentivirus-based gene therapy employing myeloablative busulfan for SCD. Somatic mutations discovered in 2 of 76 patients who rejected their grafts were identified at baseline at much lower levels. While a whole-genome sequencing analysis reported no difference between patients with SCD and controls, a study including whole-exome sequencing revealed a higher prevalence of clonal hematopoiesis in individuals with SCD compared with controls. Genetic risk factors for myeloid malignancy development after curative therapy for SCD are currently being explored. Once discovered, decisions could be made about whether gene therapy may be feasible vs allogeneic hematopoietic cell transplant, which results in full donor chimerism. In the meantime, care should be taken to perform a benefit/risk assessment to help patients identify the best curative approach for them. Long-term follow-up is necessary to monitor for myeloid malignancies and other adverse effects of curative therapies for SCD.
Collapse
Affiliation(s)
- Courtney D. Fitzhugh
- Correspondence Courtney D. Fitzhugh, Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, MSC 1589, Building 10, room 6N240A, Bethesda, MD 20814; e-mail:
| |
Collapse
|
28
|
Increased incidence of hematologic malignancies in SCD after HCT in adults with graft failure and mixed chimerism. Blood 2022; 140:2514-2518. [PMID: 36044658 PMCID: PMC9837433 DOI: 10.1182/blood.2022017960] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 01/21/2023] Open
Abstract
Lawal et al report on a 45-fold increase in secondary hematologic malignancy in 120 patients following hematopoietic stem cell transplantation (HSCT) for sickle cell disease (SCD), comparable to what has been reported following gene therapy. Notably, the cohort is enriched for older patients and for haploidentical transplant recipients with mixed chimerism following HSCT. These data further support the idea that pre-existing premalignant myeloid clones undergo clonal selection in the setting of nonmyeloablative HSCT and contribute to secondary malignancy.
Collapse
|
29
|
Lawal RA, Walters MC, Fitzhugh CD. Allogeneic Transplant and Gene Therapy: Evolving Toward a Cure. Hematol Oncol Clin North Am 2022; 36:1313-1335. [PMID: 36400545 PMCID: PMC9681017 DOI: 10.1016/j.hoc.2022.06.007] [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] [Indexed: 11/17/2022]
Abstract
Curative therapies for sickle cell disease (SCD) include allogeneic human leukocyte antigen (HLA)- matched sibling and haploidentical hematopoietic cell transplant (HCT), gene therapy, and gene editing. However, comparative trial data that might facilitate selecting one curative therapy over another are unavailable. New strategies to decrease graft rejection and graft-versus-host disease (GVHD) risks are needed to expand haploidentical HCT. Myeloablative gene therapy and gene editing also has limitations. Herein, we review recent studies on curative therapies for SCD in the past 5 years.
Collapse
Affiliation(s)
- R AdeBisi Lawal
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Building 10, Room 4-5140, Bethesda, MD 20892, USA; Hematology Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark C Walters
- University of California San Francisco Benioff Children's Hospital, 747 52nd Street, Oakland CA 94609, USA
| | - Courtney D Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Building 10, Room 6N240A, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Waldner RC, Guilcher GMT, Monagel DA, Ho J, Desai S, Rosolowsky ET. Gonadal health in recipients of nonmyeloablative hematopoietic stem cell transplantation in children and adolescents with sickle cell disease. Pediatr Hematol Oncol 2022; 40:682-687. [PMID: 36382784 DOI: 10.1080/08880018.2022.2138659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Richelle C Waldner
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Gregory M T Guilcher
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Dania A Monagel
- King Saud bin Abdulaziz University for Health Sciences/KAIMRC, Jeddah, Saudi Arabia
| | - Josephine Ho
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sunil Desai
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
31
|
John T, Namazzi R, Chirande L, Tubman VN. Global perspectives on cellular therapy for children with sickle cell disease. Curr Opin Hematol 2022; 29:275-280. [PMID: 36206076 PMCID: PMC10107365 DOI: 10.1097/moh.0000000000000738] [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] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Low-income and middle-income countries (LMICs), primarily in sub-Saharan Africa (SSA), predominantly experience the burden of sickle cell disease (SCD). High frequency of acute and chronic complications leads to increased utilization of healthcare, which burdens fragile health systems. Mortality for children with limited healthcare access remains alarmingly high. Cellular based therapies such as allogeneic hematopoietic stem cell transplant (HSCT) are increasingly used in resource-rich settings as curative therapy for SCD. Broad access to curative therapies for SCD in SSA would dramatically alter the global impact of the disease. RECENT FINDINGS Currently, application of cellular based therapies in LMICs is limited by cost, personnel, and availability of HSCT-specific technologies and supportive care. Despite the challenges, HSCT for SCD is moving forward in LMICs. Highly anticipated gene modification therapies have recently proven well tolerated and feasible in clinical trials in resource-rich countries, but access remains extremely limited. SUMMARY Translation of curative cellular based therapies for SCD should be prioritized to LMICs where the disease burden and cost of noncurative treatments is high, and long-term quality of life is poor. Focus on thoughtful modifications of current and future therapies to meet the need in LMICs, especially in SSA, will be especially impactful.
Collapse
Affiliation(s)
- Tami John
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Texas Children’s Cancer and Hematology Centers, Texas Children’s Hospital, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Ruth Namazzi
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Lulu Chirande
- School of Medicine, The Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Venée N. Tubman
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
32
|
Alsultan A, Abujoub R, Elbashir E, Essa MF. The effect of intensity of conditioning regimen on the outcome of HSCT in children with sickle cell disease. Clin Transplant 2022; 36:e14787. [PMID: 35929611 DOI: 10.1111/ctr.14787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (HSCT) provides a cure for patients with sickle cell disease (SCD). This study describes the effect of conditioning regimen intensity on HSCT outcomes among children younger than 14 years with SCD. METHODS Transplants from HLA-matched related donors (MRD) and unrelated donors (MUD) using either myeloablative conditioning (MAC) regimens or reduced intensity conditioning (RIC) regimens were considered. Event-free survival (EFS) was the primary endpoint. Secondary endpoints included overall survival (OS) and occurrence of GVHD. RESULTS 48 SCD patients underwent HSCT, 45 (93.8%) patients had MRD, 1 (2.1%) had 9/10 related donor, and 2 (4.1%) had MUD. The median age at transplant was 8.6 years (range, 3.1-13.8). Conditioning regimens were myeloablative (MAC) in 41 (85.4%) patients and of reduced intensity in 7 (14.6%) patients. EFS at 2 years was 100% among MAC group compared to 29% in the RIC group (p < .001). The median follow-up was 43.4 months (range 26.8-134). All events in the RIC group were secondary graft failure. However, OS was 100% in both groups at 2 years. Acute GVHD II-IV was diagnosed in 2 (4.1%) patients. Chronic GVHD occurred in 2 (4.1%) patients. GVHD did not occur in patients who underwent MUD HSCT. CONCLUSIONS MAC in children with SCD is well tolerated and associated with an excellent outcome for HLA-matched HSCT in SCD. There was a high rate of secondary graft failure with the use of RIC. Future studies are needed to optimize RIC regimens in HSCT of children with SCD.
Collapse
Affiliation(s)
- Abdulrahman Alsultan
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Rodina Abujoub
- Department of Nursing, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Enas Elbashir
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammed F Essa
- Department of Pediatric Hematology/Oncology, King Abdullah Specialist Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
33
|
Bedrick BS, Kohn TP, Pecker LH, Christianson MS. Fertility preservation for pediatric patients with hemoglobinopathies: Multidisciplinary counseling needed to optimize outcomes. Front Endocrinol (Lausanne) 2022; 13:985525. [PMID: 36353243 PMCID: PMC9638952 DOI: 10.3389/fendo.2022.985525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/30/2022] [Indexed: 01/19/2023] Open
Abstract
Hemoglobinopathies are autosomal recessive disorders that occur when genetic mutations negatively impact the function of hemoglobin. Common hemoglobinopathies that are clinically significant include sickle cell disease, alpha thalassemia, and beta thalassemia. Advancements in disease-modifying and curative treatments for the common hemoglobinopathies over the past thirty years have led to improvements in patient quality of life and longevity for those who are affected. However, the diseases, their treatments and cures pose infertility risks, making fertility preservation counseling and treatment an important part of the contemporary comprehensive patient care. Sickle cell disease negatively impacts both male and female infertility, primarily by testicular failure and decreased ovarian reserve, respectively. Fertility in both males and females with beta thalassemia major are negatively impacted by iron deposition due to chronic blood transfusions. Hematopoietic stem cell transplant (HSCT) is currently the only curative treatment for SCD and transfusion dependent beta thalassemia. Many of the conditioning regimens for HSCT contain chemotherapeutic agents with known gonadotoxicity and whole-body radiation. Although most clinical studies on toxicity and impact of HSCT on long-term health do not evaluate fertility, gonadal failure is common. Male fertility preservation modalities that exist prior to gonadotoxic treatment include sperm banking for pubertal males and testicular cryopreservation for pre-pubertal boys. For female patients, fertility preservation options include oocyte cryopreservation and ovarian tissue cryopreservation. Oocyte cryopreservation requires controlled ovarian hyperstimulation (COH) with ten to fourteen days of intensive monitoring and medication administration. This is feasible once the patient has undergone menarche. Follicular growth is monitored via transvaginal or transabdominal ultrasound, and hormone levels are monitored through frequent blood work. Oocytes are then harvested via a minimally invasive approach under anesthesia. Complications of COH are more common in patients with hemoglobinopathies. Ovarian hyperstimulation syndrome creates a greater risk to patients with underlying vascular, pulmonary, and renal injury, as they may be less able to tolerate fluids shifts. Thus, it is critical to monitor patients undergoing COH closely with close collaboration between the hematology team and the reproductive endocrinology team. Counseling patients and families about future fertility must take into consideration the patient's disease, treatment history, and planned treatment, acknowledging current knowledge gaps.
Collapse
Affiliation(s)
- Bronwyn S. Bedrick
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Taylor P. Kohn
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lydia H. Pecker
- Department of Medicine, Division of Adult Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mindy S. Christianson
- Department of Gynecology and Obstetrics, Division of Reproductive Endocrinology and Infertility, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
34
|
Dhedin N, Chevillon F, Castelle M, Lavoipière V, Vasseur L, Dalle JH, Joseph L, Beckerich F, Buchbinder N, Coman T, Garban F, Ferster A, Nguyen S, Boissel N, Arlet JB, Pondarre C. HLA-matched related-donor hematopoietic stem cell transplantation is a suitable treatment in adolescents and adults with sickle cell disease: comparison of myeloablative and non-myeloablative approaches. Am J Hematol 2022; 97:E359-E362. [PMID: 35802796 DOI: 10.1002/ajh.26656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Nathalie Dhedin
- Hematology Unit for Adolescents and Young Adults, Saint Louis Hospital, Paris, France
| | - Florian Chevillon
- Hematology Unit for Adolescents and Young Adults, Saint Louis Hospital, Paris, France
| | | | - Virginie Lavoipière
- Internal Medicine and Clinical Immunology, Conception Hospital, Marseille. France; Internal Medicine, Martigues Hospital, France
| | - Loic Vasseur
- Hematology Unit for Adolescents and Young Adults, Saint Louis Hospital, Paris, France
| | | | | | | | | | - Tereza Coman
- Department of Hematology, Gustave Roussy Hospital, Villejuif, France
| | | | - Alina Ferster
- Hemato-Oncology and Transplantation Unit, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - S Nguyen
- Department of Hematology, Pitié Salpetrière Hospital, Sorbonne University, Paris, France
| | - Nicolas Boissel
- Hematology Unit for Adolescents and Young Adults, Saint Louis Hospital, Paris, France
| | - Jean-Benoit Arlet
- French Sickle Cell Referral Center, Internal Medicine Department, Georges Pompidou European Hospital, and Université de Paris, Paris, France
| | - Corinne Pondarre
- Pediatric Department, Sickle Cell Referral Center, Centre Hospitalier Intercommunal de Créteil, France and Inserm U955, Université Paris XII, Créteil, France
| | | |
Collapse
|
35
|
CD34-selected stem cell boost as therapy for late graft rejection following allogeneic transplantation for sickle cell disease. Bone Marrow Transplant 2022; 57:1592-1594. [PMID: 35798858 DOI: 10.1038/s41409-022-01749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 11/08/2022]
|
36
|
Across the Myeloablative Spectrum: Hematopoietic Cell Transplant Conditioning Regimens for Pediatric Patients with Sickle Cell Disease. J Clin Med 2022; 11:jcm11133856. [PMID: 35807140 PMCID: PMC9267729 DOI: 10.3390/jcm11133856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
One out of every five hundred African American children in the United States has sickle cell disease (SCD). While multiple disease-modifying therapies are available, hematopoietic cell transplantation (HCT) remains the only curative option for children with SCD. HLA-matched sibling HCT has demonstrated excellent efficacy, but its availability remains limited; alternative donor strategies are increasingly explored. While Busulfan-Cyclophosphamide has become the most widespread conditioning regimen employed in HCT for pediatric SCD, many other regimens have been examined. This review explores different conditioning regimens across the intensity spectrum: from myeloablative to non-myeloablative. We describe survival and organ function outcomes in pediatric SCD patients who have received HCT and discuss the strengths and weaknesses of the various conditioning intensities. Finally, we posit novel directions in allogeneic HCT for SCD.
Collapse
|
37
|
Clonal Hematopoiesis and the Risk of Hematologic Malignancies after Curative Therapies for Sickle Cell Disease. J Clin Med 2022; 11:jcm11113160. [PMID: 35683547 PMCID: PMC9181510 DOI: 10.3390/jcm11113160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 12/30/2022] Open
Abstract
Sickle cell disease (SCD) is associated with severe morbidity and early mortality. Two large population studies found an increased risk for leukemia in individuals with SCD. Notably, while the relative risk of leukemia development is high, the absolute risk is low in individuals with SCD who do not receive cell-based therapies. However, the risk of leukemia in SCD is high after graft rejection and with gene therapy. Clonal hematopoiesis (CH) is a well-recognized premalignant condition in the general population and in patients after high-dose myelotoxic therapies. Recent studies suggest that CH may be more common in SCD than in the general population, outside the cell-based therapy setting. Here, we review risk factors for CH and progression to leukemia in SCD. We surmise why patients with SCD are at an increased risk for CH and why leukemia incidence is unexpectedly high after graft rejection and gene therapy for SCD. Currently, we are unable to reliably assess genetic risk factors for leukemia development after curative therapies for SCD. Given our current knowledge, we recommend counseling patients about leukemia risk and discussing the importance of an individualized benefit/risk assessment that incorporates leukemia risk in patients undergoing curative therapies for SCD.
Collapse
|
38
|
Long-Term Health Effects of Curative Therapies on Heart, Lungs, and Kidneys for Individuals with Sickle Cell Disease Compared to Those with Hematologic Malignancies. J Clin Med 2022; 11:jcm11113118. [PMID: 35683502 PMCID: PMC9181610 DOI: 10.3390/jcm11113118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/30/2022] Open
Abstract
The goal of curing children and adults with sickle cell disease (SCD) is to maximize benefits and minimize intermediate and long-term adverse outcomes so that individuals can live an average life span with a high quality of life. While greater than 2000 individuals with SCD have been treated with curative therapy, systematic studies have not been performed to evaluate the long-term health effects of hematopoietic stem cell transplant (HSCT) in this population. Individuals with SCD suffer progressive heart, lung, and kidney disease prior to curative therapy. In adults, these sequalae are associated with earlier death. In comparison, individuals who undergo HSCT for cancer are heavily pretreated with chemotherapy, resulting in potential acute and chronic heart, lung, and kidney disease. The long-term health effects on the heart, lung, and kidney for children and adults undergoing HSCT for cancer have been extensively investigated. These studies provide the best available data to extrapolate the possible late health effects after curative therapy for SCD. Future research is needed to evaluate whether HSCT abates, stabilizes, or exacerbates heart, lung, kidney, and other diseases in children and adults with SCD receiving myeloablative and non-myeloablative conditioning regimens for curative therapy.
Collapse
|
39
|
Nickel RS, Maher JY, Hsieh MH, Davis MF, Hsieh MM, Pecker LH. Fertility after Curative Therapy for Sickle Cell Disease: A Comprehensive Review to Guide Care. J Clin Med 2022; 11:2318. [PMID: 35566443 PMCID: PMC9105328 DOI: 10.3390/jcm11092318] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/30/2022] Open
Abstract
Curative therapy for sickle cell disease (SCD) currently requires gonadotoxic conditioning that can impair future fertility. Fertility outcomes after curative therapy are likely affected by pre-transplant ovarian reserve or semen analysis parameters that may already be abnormal from SCD-related damage or hydroxyurea treatment. Outcomes are also likely affected by the conditioning regimen. Conditioning with myeloablative busulfan and cyclophosphamide causes serious gonadotoxicity particularly among post-pubertal females. Reduced-intensity and non-myeloablative conditioning may be acutely less gonadotoxic, but more short and long-term fertility outcome data after these approaches is needed. Fertility preservation including oocyte/embryo, ovarian tissue, sperm, and experimental testicular tissue cryopreservation should be offered to patients with SCD pursing curative therapy. Regardless of HSCT outcome, longitudinal post-HSCT fertility care is required.
Collapse
Affiliation(s)
- Robert Sheppard Nickel
- Children’s National Hospital, Division of Hematology, Washington, DC 20001, USA;
- Children’s National Hospital, Division of Blood and Marrow Transplantation, Washington, DC 20001, USA
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20001, USA;
| | - Jacqueline Y. Maher
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Pediatric and Adolescent Gynecology, National Institutes of Health, Bethesda, MD 20810, USA;
- Children’s National Hospital, Pediatric and Adolescent Gynecology Program, Washington, DC 20001, USA
| | - Michael H. Hsieh
- School of Medicine and Health Sciences, The George Washington University, Washington, DC 20001, USA;
- Children’s National Hospital, Division of Urology, Washington, DC 20001, USA
| | - Meghan F. Davis
- Department of Urology, MedStar Georgetown University Hospital, Washington, DC 20001, USA;
| | - Matthew M. Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20810, USA;
| | - Lydia H. Pecker
- Division of Hematology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 20810, USA
| |
Collapse
|
40
|
Feasibility of early sirolimus cessation post non-myeloablative transplantation in adult patients with severe sickle cell disease. Bone Marrow Transplant 2022; 57:319-322. [DOI: 10.1038/s41409-021-01548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/18/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
|
41
|
Shah NC, Bhoopatiraju S, Abraham A, Anderson E, Andreansky M, Bhatia M, Chaudhury S, Cuvelier GDE, Godder K, Grimley M, Hale G, Kamani N, Jacobsohn D, Ngwube A, Gilman AL, Skiles J, Yu LC, Shenoy S. Granulocyte Colony-Stimulating Factor is Safe and Well Tolerated following Allogeneic Transplantation in Patients with Sickle Cell Disease. Transplant Cell Ther 2021; 28:174.e1-174.e5. [PMID: 34958973 DOI: 10.1016/j.jtct.2021.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) used after hematopoietic stem cell transplantation (HSCT) can enhance neutrophil recovery in patients rendered neutropenic by the preparative regimen. G-CSF is contraindicated in patients with sickle cell disease (SCD) as life-threatening complications can ensue in the presence of sickle vasculopathy. However, the safety profile of G-CSF after HSCT for SCD has not been previously described. We report clinical outcomes in the first 100 days post-HSCT in patients supported with G-CSF until neutrophil recovery on a clinical trial of reduced intensity transplantation for SCD. Patients (n=62) received G-CSF for a median of 9 days (range, 5-33) following transplant from the best available stem cell source. Preparation for transplant included a target hemoglobin S level of ≤45%. Neutrophil engraftment (ANC >0.5 × 103/mL) was achieved at a median of 13 days (range,10-34) and platelet engraftment (>50 × 103/mL) at a median of 19 days (range, 12-71). The median duration of inpatient hospitalization following stem cell infusion (day 0) was 21.5 days (range 11-33). No patient developed SCD related complications following G-CSF use. The most common organ toxicities encountered between G-CSF commencement (on day +7) and day +100 were anorexia (14), hypertension (11) and electrolyte imbalance requiring correction (9). Central nervous system related events were noted in 5 patients, all with pre-existing cerebral vasculopathy/moyamoya disease and attributed to reversible posterior leukoencephalopathy syndrome (RPLS) in the presence of calcineurin inhibitor therapy and hypertension. We conclude that G-CSF does not adversely impact SCD transplant recipients and can be safely used post-HSCT to enhance neutrophil recovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Geoff D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, Cancer Care Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | | | | | | - Lolie C Yu
- Children's Hospital/LSUHSC, New Orleans, LA
| | | |
Collapse
|
42
|
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: 3] [Impact Index Per Article: 0.8] [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
|
43
|
Furstenau D, Peer CJ, Hughes TE, Uchida N, Tisdale J, Hall OM, Figg WD, Hsieh M. Alemtuzumab clearance, lymphocyte count, and T-cell chimerism after hematopoietic stem cell transplant in sickle cell disease. Pharmacotherapy 2021; 42:14-22. [PMID: 34669981 DOI: 10.1002/phar.2641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 11/06/2022]
Abstract
STUDY OBJECTIVE Alemtuzumab is a monoclonal antibody that targets the cell surface antigen CD52 on lymphocytes. Although it is used for the treatment of hematologic malignancies, such as chronic lymphocytic leukemia, and incorporated into many hematopoietic stem cell transplant (HSCT) conditioning regimens, few studies have evaluated the pharmacology of alemtuzumab in adult patients with sickle cell disease (SCD). We therefore examined the pharmacokinetics (PK) and pharmacodynamics (PD) of alemtuzumab in adults with SCD who received a matched related donor HSCT to determine if the clearance of alemtuzumab affects transplant outcomes. DESIGN PK and PD analysis of patient data from a single-center clinical trial. SETTING Clinical research center. PATIENTS Twenty-two adult patients with SCD who received one of two nonmyeloablative allogeneic HSCT regimens: alemtuzumab and total body irradiation (Alem-TBI) or pentostatin, cyclophosphamide, alemtuzumab, and total body irradiation (Pento-Cy-Alem-TBI). MEASUREMENTS AND MAIN RESULTS Alemtuzumab serum concentrations, absolute lymphocyte counts, T-cell (CD3), and myeloid (CD14/15) chimerism were collected at distinct time points and analyzed. A semi-mechanistic PK population model was built to understand inter-individual differences in pharmacology. Alemtuzumab was detectable up to 28 days post-HSCT. The mean alemtuzumab level 7 days after transplant for patients on Alem-TBI was 818 ng/ml, significantly lower than the mean level of 1502 ng/ml for patients on Pento-Cy-Alem-TBI (p < 0.001), but this difference decreased as time progressed. The clearance of alemtuzumab was linear, and the half-life was longer in the Pento-Cy-Alem-TBI group (average half-life = 61.1 h) compared to the Alem-TBI group (average half-life = 44.1 h) (p < 0.001). The CD3 chimerism at 2 and 4 months after transplant positively correlated with alemtuzumab levels collected on day 14 after transplant (R2 = 0.40 and p = 0.004 at 2 months, R2 = 0.36 and p = 0.005 at 4 months), but this significance was lost by 6 months after HSCT. No correlation was seen between alemtuzumab levels and CD14/15 chimerism. CONCLUSION Between 2 and 4 months after transplant, higher alemtuzumab levels measured 14 days after transplant correlated with patients having better engraftment, suggesting more lymphodepletion may be needed to reduce graft failure in these two non-myeloablative matched related donor HSCT regimens.
Collapse
Affiliation(s)
- Dana Furstenau
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.,Division of Hematology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cody J Peer
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas E Hughes
- Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, Maryland, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Oliver Morgan Hall
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - William D Figg
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
44
|
Infectious complications in adult sickle cell anemia patients undergoing hematopoietic stem cell transplantation. Bone Marrow Transplant 2021; 56:3078-3080. [PMID: 34508177 DOI: 10.1038/s41409-021-01457-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/09/2021] [Accepted: 08/27/2021] [Indexed: 11/08/2022]
|
45
|
John TD, Friend B, Yassine K, Sasa G, Bhar S, Salem B, Omer B, Craddock J, Doherty E, Martinez C, Heslop HE, Krance RA, Leung K. Matched related hematopoietic cell transplant for sickle cell disease with alemtuzumab: the Texas Children's Hospital experience. Bone Marrow Transplant 2021; 56:2797-2803. [PMID: 34274957 DOI: 10.1038/s41409-021-01415-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 11/09/2022]
Abstract
Serotherapeutic agents facilitate engraftment and prevent graft-versus-host disease (GVHD) following hematopoietic stem cell transplant. Anti-thymocyte globulin is generally added to conditioning chemotherapy for matched related donor transplant (MRD-HCT) for sickle cell disease (SCD). Alemtuzumab, however, is appealing due to its broad lymphocyte killing that may achieve very low rejection and GVHD rates. To assess the impact of alemtuzumab in MRD-HCT for SCD, we retrospectively reviewed transplant-related outcomes and markers of immunity in 38 consecutive patients at Texas Children's Hospital having received myeloablative conditioning with alemtuzumab. Median follow-up was 4.8 years (range: 0.2-17). All patients engrafted. Donor chimerism was mixed in 47.1% of patients at ≥2-years. Donor chimerism <50% was uncommon (n = 2). One patient with low myeloid chimerism (19%) had sickle-related hemolysis at 10-years. Incidence of acute GVHD grade II-IV (5.3%) and extensive chronic GVHD (2.8%) was very low. Five-year event-free survival (EFS) and composite chronic GVHD-EFS were excellent at 94.7% (95% CI: 80.3, 98.6) and 89.2% (95% CI: 73.7, 95.8), respectively. Infections did not contribute to mortality although cytomegalovirus reactivation occurred commonly in the first 3 months after transplant. Our data suggest potential for alemtuzumab in myeloablative transplant for children with SCD although further evaluation in older patients and with unrelated donors is warranted.
Collapse
Affiliation(s)
- Tami D John
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
| | - Brian Friend
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Khaled Yassine
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Ghadir Sasa
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Saleh Bhar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Baheyeldin Salem
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - John Craddock
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Erin Doherty
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Caridad Martinez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Robert A Krance
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Kathryn Leung
- Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|