1
|
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
|
2
|
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: 12.0] [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
|
3
|
Uchida N, Stasula U, Demirci S, Germino-Watnick P, Hinds M, Le A, Chu R, Berg A, Liu X, Su L, Wu X, Krouse AE, Linde NS, Bonifacino A, Hong SG, Dunbar CE, Lanieri L, Bhat A, Palchaudhuri R, Bennet B, Hoban M, Bertelsen K, Olson LM, Donahue RE, Tisdale JF. Fertility-preserving myeloablative conditioning using single-dose CD117 antibody-drug conjugate in a rhesus gene therapy model. Nat Commun 2023; 14:6291. [PMID: 37828021 PMCID: PMC10570335 DOI: 10.1038/s41467-023-41153-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/23/2023] [Indexed: 10/14/2023] Open
Abstract
Hematopoietic stem cell (HSC) gene therapy has curative potential; however, its use is limited by the morbidity and mortality associated with current chemotherapy-based conditioning. Targeted conditioning using antibody-drug conjugates (ADC) holds promise for reduced toxicity in HSC gene therapy. Here we test the ability of an antibody-drug conjugate targeting CD117 (CD117-ADC) to enable engraftment in a non-human primate lentiviral gene therapy model of hemoglobinopathies. Following single-dose CD117-ADC, a >99% depletion of bone marrow CD34 + CD90 + CD45RA- cells without lymphocyte reduction is observed, which results are not inferior to multi-day myeloablative busulfan conditioning. CD117-ADC, similarly to busulfan, allows efficient engraftment, gene marking, and vector-derived fetal hemoglobin induction. Importantly, ADC treatment is associated with minimal toxicity, and CD117-ADC-conditioned animals maintain fertility. In contrast, busulfan treatment commonly causes severe toxicities and infertility in humans. Thus, the myeloablative capacity of single-dose CD117-ADC is sufficient for efficient engraftment of gene-modified HSCs while preserving fertility and reducing adverse effects related to toxicity in non-human primates. This targeted conditioning approach thus provides the proof-of-principle to improve risk-benefit ratio in a variety of HSC-based gene therapy products in humans.
Collapse
Affiliation(s)
- Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA.
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Ulana Stasula
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Paula Germino-Watnick
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Malikiya Hinds
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Anh Le
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Rebecca Chu
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Alexander Berg
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Xiong Liu
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - Ling Su
- Genomics Technology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Xiaolin Wu
- Genomics Technology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Allen E Krouse
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD, USA
| | - N Seth Linde
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Aylin Bonifacino
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD, USA
| | - So Gun Hong
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD, USA
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD, USA
| | | | | | | | | | | | | | | | - Robert E Donahue
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI) / National Institute of Diabetes, and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, MD, USA
| |
Collapse
|
4
|
Cannas G, Poutrel S, Heiblig M, Labussière H, Larcher MV, Thomas X, Hot A. Sickle cell disease and acute leukemia: one case report and an extensive review. Ann Hematol 2023; 102:1657-1667. [PMID: 37269388 PMCID: PMC10239223 DOI: 10.1007/s00277-023-05294-3] [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: 03/20/2023] [Accepted: 05/22/2023] [Indexed: 06/05/2023]
Abstract
Population-based studies and case reports suggest that there may be an increased risk of acute leukemia associated with sickle cell disease (SCD). Following the description of a new case report, an extensive review of the literature identified 51 previously described cases. Most cases study showed myelodysplastic features confirmed, when available, by genetic markers such as chromosome 5 and/or chromosome 7 abnormalities and TP53 gene mutations. The increased risk of leukemogenesis is certainly multifactorial and related to the pathophysiologic mechanisms of the clinical manifestations of SCD. Chronic hemolysis and secondary hemochromatosis may cause increased chronic inflammation, resulting in persistent marrow stress, which could potentially compromise the genomic stability of the hematopoietic stem cells generating genomic damage and somatic mutations over the course of SCD and its treatment, resulting in a clone that led to acute myeloid leukemia.
Collapse
Affiliation(s)
- Giovanna Cannas
- Internal Medicine, Hospices Civils de Lyon, Edouard Herriot Hospital, 5, place d'Arsonval, Lyon cedex 03, 69437, Lyon, France.
- Constitutive reference center: Major sickle cell syndromes, thalassemias and other rare pathologies of red blood cell and erythropoiesis, Edouard Herriot Hospital, Lyon, France.
| | - Solène Poutrel
- Internal Medicine, Hospices Civils de Lyon, Edouard Herriot Hospital, 5, place d'Arsonval, Lyon cedex 03, 69437, Lyon, France
- Constitutive reference center: Major sickle cell syndromes, thalassemias and other rare pathologies of red blood cell and erythropoiesis, Edouard Herriot Hospital, Lyon, France
| | - Maël Heiblig
- Hematology, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Hélène Labussière
- Hematology, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
| | | | - Xavier Thomas
- Constitutive reference center: Major sickle cell syndromes, thalassemias and other rare pathologies of red blood cell and erythropoiesis, Edouard Herriot Hospital, Lyon, France
| | - Arnaud Hot
- Internal Medicine, Hospices Civils de Lyon, Edouard Herriot Hospital, 5, place d'Arsonval, Lyon cedex 03, 69437, Lyon, France
- Constitutive reference center: Major sickle cell syndromes, thalassemias and other rare pathologies of red blood cell and erythropoiesis, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
5
|
Nash A, Lokhorst N, Veiseh O. Localized immunomodulation technologies to enable cellular and organoid transplantation. Trends Mol Med 2023:S1471-4914(23)00097-7. [PMID: 37301656 DOI: 10.1016/j.molmed.2023.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Abstract
Localized immunomodulation technologies are rapidly emerging as a new modality with the potential to revolutionize transplantation of cells and organs. In the past decade, cell-based immunomodulation therapies saw clinical success in the treatment of cancer and autoimmune diseases. In this review, we describe recent advances in engineering solutions for the development of localized immunomodulation techniques focusing on cellular and organoid transplantation. We begin by describing cell transplantation and highlighting notable clinical successes, particularly in the areas of stem cell therapy, chimeric antigen receptor (CAR)-T cell therapy, and islet transplantation. Next, we detail recent preclinical studies centered on genome editing and biomaterials to enhance localized immunomodulation. We close by discussing future opportunities to improve clinical and commercial success using these approaches to facilitate long-term immunomodulation technologies.
Collapse
Affiliation(s)
- Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Nienke Lokhorst
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht 3584, CG, The Netherlands
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
6
|
Eapen M, Brazauskas R, Williams DA, Walters MC, St Martin A, Jacobs BL, Antin JH, Bona K, Chaudhury S, Coleman-Cowger VH, DiFronzo NL, Esrick EB, Field JJ, Fitzhugh CD, Kanter J, Kapoor N, Kohn DB, Krishnamurti L, London WB, Pulsipher MA, Talib S, Thompson AA, Waller EK, Wun T, Horowitz MM. Secondary Neoplasms After Hematopoietic Cell Transplant for Sickle Cell Disease. J Clin Oncol 2023; 41:2227-2237. [PMID: 36623245 PMCID: PMC10448940 DOI: 10.1200/jco.22.01203] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/04/2022] [Accepted: 12/02/2022] [Indexed: 01/11/2023] Open
Abstract
PURPOSE To report the incidence and risk factors for secondary neoplasm after transplantation for sickle cell disease. METHODS Included are 1,096 transplants for sickle cell disease between 1991 and 2016. There were 22 secondary neoplasms. Types included leukemia/myelodysplastic syndrome (MDS; n = 15) and solid tumor (n = 7). Fine-Gray regression models examined for risk factors for leukemia/MDS and any secondary neoplasm. RESULTS The 10-year incidence of leukemia/MDS was 1.7% (95% CI, 0.90 to 2.9) and of any secondary neoplasm was 2.4% (95% CI, 1.4 to 3.8). After adjusting for other risk factors, risks for leukemia/MDS (hazard ratio, 22.69; 95% CI, 4.34 to 118.66; P = .0002) or any secondary neoplasm (hazard ratio, 7.78; 95% CI, 2.20 to 27.53; P = .0015) were higher with low-intensity (nonmyeloablative) regimens compared with more intense regimens. All low-intensity regimens included total-body irradiation (TBI 300 or 400 cGy with alemtuzumab, TBI 300 or 400 cGy with cyclophosphamide, TBI 200, 300, or 400 cGy with cyclophosphamide and fludarabine, or TBI 200 cGy with fludarabine). None of the patients receiving myeloablative and only 23% of those receiving reduced-intensity regimens received TBI. CONCLUSION Low-intensity regimens rely on tolerance induction and establishment of mixed-donor chimerism. Persistence of host cells exposed to low-dose radiation triggering myeloid malignancy is one plausible etiology. Pre-existing myeloid mutations and prior inflammation may also contribute but could not be studied using our data source. Choosing conditioning regimens likely to result in full-donor chimerism may in part mitigate the higher risk for leukemia/MDS.
Collapse
Affiliation(s)
- Mary Eapen
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Ruta Brazauskas
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | - David A. Williams
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Mark C. Walters
- University of California San Francisco Benioff Children's Hospital, Oakland, CA
| | - Andrew St Martin
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Benjamin L. Jacobs
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Joseph H. Antin
- Dana-Farber Cancer Center, Harvard Medical School, Boston, MA
| | - Kira Bona
- Dana-Farber Cancer Center, Harvard Medical School, Boston, MA
| | | | | | | | - Erica B. Esrick
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Joshua J. Field
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Courtney D. Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Julie Kanter
- University of Alabama Birmingham, Birmingham, AL
| | - Neena Kapoor
- Children's Hospital of Los Angeles, Los Angeles, CA
| | - Donald B. Kohn
- David Geffen School of Medicine, University of California, Los Angeles, CA
| | | | - Wendy B. London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | | | - Sohel Talib
- California Institute for Regenerative Medicine, San Francisco, CA
| | | | | | - Ted Wun
- University of California Davis School of Medicine, Davis, CA
| | - Mary M. Horowitz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
7
|
Flevari P, Voskaridou E, Galactéros F, Cannas G, Loko G, Joseph L, Bartolucci P, Gellen-Dautremer J, Bernit E, Charneau C, Habibi A. Case Report of Myelodysplastic Syndrome in a Sickle-Cell Disease Patient Treated with Hydroxyurea and Literature Review. Biomedicines 2022; 10:biomedicines10123201. [PMID: 36551957 PMCID: PMC9775156 DOI: 10.3390/biomedicines10123201] [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: 11/03/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The safety profile of hydroxyurea (HU) in patients with sickle-cell disease (SCD) is relatively well known. However, despite the suspected association of HU with myeloid neoplasms in myeloproliferative neoplasms (MPN), and the publication of sporadic reports of myeloid malignancies in SCD patients treated with HU, the possible excess risk imparted by HU in this population having an increasing life expectancy has failed to be demonstrated. Herein, we report one case of myelodysplastic syndrome emanating from the results on safety and effectiveness of HU on the largest European cohort of 1903 HU-treated adults and children who were followed-up prospectively in an observational setting over 10 years, accounting for a total exposure of 7309.5 patient-years. A comparison of this single case with previously published similar cases did not allow us to draw any significant conclusions due to the paucity of these events.
Collapse
Affiliation(s)
- Pagona Flevari
- Centre of Excellence in Rare Hematological Disease-Hemoglobinopathies, Laiko General Hospital, 11527 Athens, Greece
- Correspondence: (P.F.); (E.V.)
| | - Ersi Voskaridou
- Centre of Excellence in Rare Hematological Disease-Hemoglobinopathies, Laiko General Hospital, 11527 Athens, Greece
- Correspondence: (P.F.); (E.V.)
| | - Frédéric Galactéros
- Sickle Cell Referral Center, Department of Internal Medicine, Henri-Mondor University Hospital, APHP, U-PEC, 94000 Créteil, France
| | - Giovanna Cannas
- Hospices Civils de Lyon, Edouard-Herriot Hospital, Internal Medicine, Reference Centre for Sickle-Cell Disease, Thalassemia and Other Red Blood Cell Disorders, 69003 Lyon, France
| | - Gylna Loko
- Martinique Hospital, 97212 Martinique, France
| | - Laure Joseph
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, 75610 Paris, France
| | - Pablo Bartolucci
- Sickle Cell Referral Center, Department of Internal Medicine, Henri-Mondor University Hospital, APHP, U-PEC, 94000 Créteil, France
| | - Justine Gellen-Dautremer
- Sickle Cell Referral Center, Department of Internal Medicine, Henri-Mondor University Hospital, APHP, U-PEC, 94000 Créteil, France
| | - Emmanuelle Bernit
- Antilles-Guyane Reference Centre for Sickle-Cell Disease, Thalassemia and Other Red Blood Cell Disorders, Pointe à Pitre, 97157 Guadeloupe, France
| | - Corine Charneau
- Antilles-Guyane Reference Centre for Sickle-Cell Disease, Thalassemia and Other Red Blood Cell Disorders, Pointe à Pitre, 97157 Guadeloupe, France
| | - Anoosha Habibi
- Sickle Cell Referral Center, Department of Internal Medicine, Henri-Mondor University Hospital, APHP, U-PEC, 94000 Créteil, France
| |
Collapse
|
8
|
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
|
9
|
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: 7.5] [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
|
10
|
Effects of Erythrocytapheresis Procedures on Delayed Bone Marrow Conversion in Sickle Cell Disease. Clin Hematol Int 2022; 4:144-147. [PMID: 36385464 PMCID: PMC9763515 DOI: 10.1007/s44228-022-00022-6] [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: 07/27/2022] [Accepted: 10/16/2022] [Indexed: 11/17/2022] Open
Abstract
The imaging appearances of the skeletal system have been well documented in sickle cell disease (SCD) but there is limited information about the impact of SCD treatments on skeletal abnormalities. We present two patients with SCD maintained on long-term erythrocytapheresis and the changes to their skeletal abnormalities on neuroimaging with this treatment. We observed a reversal of the bone marrow conversion process and the skull appearance was age appropriate without any radiographic findings of iron overload in the patients.
Collapse
|
11
|
Kassim AA, Leonard A. Debating the Future of Sickle Cell Disease Curative Therapy: Haploidentical Hematopoietic Stem Cell Transplantation vs. Gene Therapy. J Clin Med 2022; 11:jcm11164775. [PMID: 36013014 PMCID: PMC9409766 DOI: 10.3390/jcm11164775] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a well-established curative therapy for patients with sickle cell disease (SCD) when using a human leukocyte antigen (HLA)-matched sibling donor. Most patients with SCD do not have a matched sibling donor, thereby significantly limiting the accessibility of this curative option to most patients. HLA-haploidentical HSCT with post-transplant cyclophosphamide expands the donor pool, with current approaches now demonstrating high overall survival, reduced toxicity, and an effective reduction in acute and chronic graft-vs.-host disease (GvHD). Alternatively, autologous genetic therapies appear promising and have the potential to overcome significant barriers associated with allogeneic HSCT, such as donor availability and GvHD. Here the authors each take a viewpoint and discuss what will be the future of curative options for patients with SCD outside of a matched sibling transplantation, specifically haploidentical HSCT vs. gene therapy.
Collapse
Affiliation(s)
- Adetola A. Kassim
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt Meharry Sickle Cell Center of Excellence, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Correspondence: (A.A.K.); or (A.L.)
| | - Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20810, USA
- Division of Hematology, Children’s National Hospital, Washington, DC 20010, USA
- Correspondence: (A.A.K.); or (A.L.)
| |
Collapse
|
12
|
Hematopoietic Stem Cell Gene-Addition/Editing Therapy in Sickle Cell Disease. Cells 2022; 11:cells11111843. [PMID: 35681538 PMCID: PMC9180595 DOI: 10.3390/cells11111843] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/29/2022] [Accepted: 06/02/2022] [Indexed: 12/17/2022] Open
Abstract
Autologous hematopoietic stem cell (HSC)-targeted gene therapy provides a one-time cure for various genetic diseases including sickle cell disease (SCD) and β-thalassemia. SCD is caused by a point mutation (20A > T) in the β-globin gene. Since SCD is the most common single-gene disorder, curing SCD is a primary goal in HSC gene therapy. β-thalassemia results from either the absence or the reduction of β-globin expression, and it can be cured using similar strategies. In HSC gene-addition therapy, patient CD34+ HSCs are genetically modified by adding a therapeutic β-globin gene with lentiviral transduction, followed by autologous transplantation. Alternatively, novel gene-editing therapies allow for the correction of the mutated β-globin gene, instead of addition. Furthermore, these diseases can be cured by γ-globin induction based on gene addition/editing in HSCs. In this review, we discuss HSC-targeted gene therapy in SCD with gene addition as well as gene editing.
Collapse
|
13
|
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
|
14
|
Liggett LA, Cato LD, Weinstock JS, Zhang Y, Nouraie SM, Gladwin MT, Garrett ME, Ashley-Koch A, Telen M, Custer B, Kelly S, Dinardo C, Sabino EC, Loureiro P, Carneiro-Proietti A, Maximo C, Reiner AP, Abecasis GR, Williams DA, Natarajan P, Bick AG, Sankaran VG. Clonal hematopoiesis in sickle cell disease. J Clin Invest 2022; 132:156060. [PMID: 34990411 PMCID: PMC8843701 DOI: 10.1172/jci156060] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/04/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Curative gene therapies for sickle cell disease (SCD) are currently undergoing clinical evaluation. The occurrence of myeloid malignancies in these trials has prompted safety concerns. Individuals with SCD are predisposed to myeloid malignancies, but the underlying causes remain undefined. Clonal hematopoiesis (CH) is a pre-malignant condition that also confers significant predisposition to myeloid cancers. While it has been speculated that CH may play a role in SCD-associated cancer predisposition, limited data addressing this issue have been reported. METHODS Here, we leveraged 74,190 whole genome sequences to robustly study CH in SCD. Somatic mutation calling methods were used to assess CH in all samples and comparisons between individuals with and without SCD were performed. RESULTS While we had sufficient power to detect a greater than 2-fold increased rate of CH, we found no detectable variation in rate or clone properties between individuals affected by SCD and controls. The rate of CH in individuals with SCD was unaltered by hydroxyurea use. CONCLUSIONS We did not observe an increased risk for acquiring detectable CH in SCD, at least as measured by whole genome sequencing. These results should help guide ongoing efforts and further studies that seek to better define the risk factors underlying myeloid malignancy predisposition in SCD and help ensure that curative therapies can be more safely applied. FUNDING Funding was provided by the New York Stem Cell Foundation and National Institutes of Health. The funders had no role in study design or reporting.
Collapse
Affiliation(s)
- L Alexander Liggett
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, United States of America
| | - Liam D Cato
- Department of Human Genetics, Broad Institute of MIT and Harvard, Cambridge, United States of America
| | - Joshua S Weinstock
- Department of Biostatistics, University of Michigan, Ann Arbor, United States of America
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, United States of America
| | - S Mehdi Nouraie
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Mark T Gladwin
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Melanie E Garrett
- Department of Medicine, Duke University Medical Center, Durham, United States of America
| | - Allison Ashley-Koch
- Department of Medicine, Duke University Medical Center, Durham, United States of America
| | - Marilyn Telen
- Department of Medicine, Duke University Medical Center, Durham, United States of America
| | - Brian Custer
- Department of Epidemiology and Policy Science, Vitalant Research Institute, San Francisco, United States of America
| | - Shannon Kelly
- Division of Pediatric Hematology, UCSF Benioff Children's Hospital, Oakland, United States of America
| | - Carla Dinardo
- Department of Immunohematology, Fundação Pró-Sangue Hemocentro de São Paulo, Sao Paulo, Brazil
| | - Ester C Sabino
- Institute of Tropical Medicine, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil, Sao Paulo, Brazil
| | - Paula Loureiro
- Pernambuco State Center of Hematology and Hemotherapy, Fundação Hemope, Recife, Brazil
| | | | | | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, United States of America
| | - Gonçalo R Abecasis
- Department of Biostatistics, University of Michigan, Ann Arbor, United States of America
| | - David A Williams
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, United States of America
| | - Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, United States of America
| | - Alexander G Bick
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Vijay G Sankaran
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, United States of America
| |
Collapse
|
15
|
Green SE, Singh ZN, Baer MR. Primary myelofibrosis in a patient with sickle cell disease. Am J Hematol 2022; 97:160-161. [PMID: 34498303 DOI: 10.1002/ajh.26347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/18/2021] [Accepted: 08/30/2021] [Indexed: 01/07/2023]
Affiliation(s)
- Sarah E. Green
- Department of Medicine University of Maryland School of Medicine Baltimore Maryland USA
- Greenebaum Comprehensive Cancer Center University of Maryland School of Medicine Baltimore Maryland USA
| | - Zeba N. Singh
- Department of Pathology University of Maryland School of Medicine Baltimore Maryland USA
| | - Maria R. Baer
- Department of Medicine University of Maryland School of Medicine Baltimore Maryland USA
- Greenebaum Comprehensive Cancer Center University of Maryland School of Medicine Baltimore Maryland USA
| |
Collapse
|
16
|
Boulad F, Maggio A, Wang X, Moi P, Acuto S, Kogel F, Takpradit C, Prockop S, Mansilla-Soto J, Cabriolu A, Odak A, Qu J, Thummar K, Du F, Shen L, Raso S, Barone R, Di Maggio R, Pitrolo L, Giambona A, Mingoia M, Everett JK, Hokama P, Roche AM, Cantu VA, Adhikari H, Reddy S, Bouhassira E, Mohandas N, Bushman FD, Rivière I, Sadelain M. Lentiviral globin gene therapy with reduced-intensity conditioning in adults with β-thalassemia: a phase 1 trial. Nat Med 2022; 28:63-70. [PMID: 34980909 PMCID: PMC9380046 DOI: 10.1038/s41591-021-01554-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 09/23/2021] [Indexed: 01/05/2023]
Abstract
β-Thalassemias are inherited anemias that are caused by the absent or insufficient production of the β chain of hemoglobin. Here we report 6-8-year follow-up of four adult patients with transfusion-dependent β-thalassemia who were infused with autologous CD34+ cells transduced with the TNS9.3.55 lentiviral globin vector after reduced-intensity conditioning (RIC) in a phase 1 clinical trial ( NCT01639690) . Patients were monitored for insertional mutagenesis and the generation of a replication-competent lentivirus (safety and tolerability of the infusion product after RIC-primary endpoint) and engraftment of genetically modified autologous CD34+ cells, expression of the transduced β-globin gene and post-transplant transfusion requirements (efficacy-secondary endpoint). No unexpected safety issues occurred during conditioning and cell product infusion. Hematopoietic gene marking was very stable but low, reducing transfusion requirements in two patients, albeit not achieving transfusion independence. Our findings suggest that non-myeloablative conditioning can achieve durable stem cell engraftment but underscore a minimum CD34+ cell transduction requirement for effective therapy. Moderate clonal expansions were associated with integrations near cancer-related genes, suggestive of non-erythroid activity of globin vectors in stem/progenitor cells. These correlative findings highlight the necessity of cautiously monitoring patients harboring globin vectors.
Collapse
Affiliation(s)
- Farid Boulad
- Stem Cell Transplant and Cellular Therapy Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aurelio Maggio
- Department of Hematology and Rare Diseases, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Xiuyan Wang
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell Therapy and Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paolo Moi
- Ospedale Pediatrico Microcitemie 'A.Cao', A.O. 'G.Brotzu', Cagliari, Italy
| | - Santina Acuto
- Department of Hematology and Rare Diseases, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Friederike Kogel
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chayamon Takpradit
- Stem Cell Transplant and Cellular Therapy Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pediatrics, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Susan Prockop
- Stem Cell Transplant and Cellular Therapy Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge Mansilla-Soto
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Annalisa Cabriolu
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ashlesha Odak
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinrong Qu
- Cell Therapy and Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Keyur Thummar
- Cell Therapy and Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fang Du
- Cell Therapy and Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lingbo Shen
- Cell Therapy and Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simona Raso
- Department of Hematology and Rare Diseases, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Rita Barone
- Department of Hematology and Rare Diseases, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Rosario Di Maggio
- Department of Hematology and Rare Diseases, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Lorella Pitrolo
- Department of Hematology and Rare Diseases, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Antonino Giambona
- Department of Hematology and Rare Diseases, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Maura Mingoia
- Ospedale Pediatrico Microcitemie 'A.Cao', A.O. 'G.Brotzu', Cagliari, Italy
| | - John K Everett
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pascha Hokama
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aoife M Roche
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Vito Adrian Cantu
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Hriju Adhikari
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shantan Reddy
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eric Bouhassira
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Narla Mohandas
- Laboratory of Red Cell Physiology, New York Blood Center, New York, NY, USA
| | - Frederic D Bushman
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Isabelle Rivière
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell Therapy and Cell Engineering Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
17
|
Ribeil J. Primary myelofibrosis in untreated sickle cell disease: Are adult patients at higher risk for developing hematological myeloid neoplasms? Am J Hematol 2022; 97:4-6. [PMID: 34626435 DOI: 10.1002/ajh.26371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Jean‐Antoine Ribeil
- Section of Hematology/Oncology, Boston University and Boston Medical Center; Sickle Cell Center, Boston University School of Medicine Boston Massachusetts USA
| |
Collapse
|
18
|
Kanter J. Gene therapy for sickle cell disease: where we are now? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:174-180. [PMID: 34889358 PMCID: PMC8791177 DOI: 10.1182/hematology.2021000250] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The landscape of sickle cell disease (SCD) treatment continues to evolve rapidly, with new disease-modifying therapies in development and potentially curative options on the horizon. Until recently, allogeneic stem cell transplant has been the only proven cure for SCD. Gene therapy is rising to the forefront of the discussion as a potentially curative or highly disease- modifying option for abating the complications of the disease. Understanding the different types of gene therapy in use, the differences in their end points, and their potential risks and benefits will be key to optimizing the long-term use of this therapy.
Collapse
Affiliation(s)
- Julie Kanter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
19
|
Pincez T, Lee SSK, Ilboudo Y, Preuss M, Pham Hung d'Alexandry d'Orengiani AL, Bartolucci P, Galactéros F, Joly P, Bauer DE, Loos RJF, Lindsley RC, Lettre G. Clonal hematopoiesis in sickle cell disease. Blood 2021; 138:2148-2152. [PMID: 34521115 PMCID: PMC8617438 DOI: 10.1182/blood.2021011121] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/20/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Thomas Pincez
- Montreal Heart Institute, Montréal, QC, Canada
- Faculté de Médecine and
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Charles-Bruneau Cancer Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Simon S K Lee
- The Charles Bronfman Institute for Personalized Medicine, Icahn Mount Sinai School of Medicine, New York, NY
| | - Yann Ilboudo
- Montreal Heart Institute, Montréal, QC, Canada
- Faculté de Médecine and
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn Mount Sinai School of Medicine, New York, NY
| | - Anne-Laure Pham Hung d'Alexandry d'Orengiani
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est, Institut Mondor de Recherche Biomédicale (IMRB), Unité 955, Equipe 2, Créteil, France
| | - Pablo Bartolucci
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est, Institut Mondor de Recherche Biomédicale (IMRB), Unité 955, Equipe 2, Créteil, France
| | - Frédéric Galactéros
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est, Institut Mondor de Recherche Biomédicale (IMRB), Unité 955, Equipe 2, Créteil, France
| | - Philippe Joly
- Unité Fonctionnelle 34445 "Biochimie des Pathologies Érythrocytaires,' Laboratoire de Biochimie et Biologie Moléculaire Grand Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Equipe "Biologie Vasculaire et du Globule Rouge," Université Claude Bernard Lyon 1, Comité d'Universités et d'Etablissements, Lyon, France
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Department of Pediatrics, Harvard Medical School, Boston, MA; and
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn Mount Sinai School of Medicine, New York, NY
| | - R Coleman Lindsley
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Guillaume Lettre
- Montreal Heart Institute, Montréal, QC, Canada
- Faculté de Médecine and
| |
Collapse
|
20
|
Rosanwo TO, Bauer DE. Editing outside the body: Ex vivo gene-modification for β-hemoglobinopathy cellular therapy. Mol Ther 2021; 29:3163-3178. [PMID: 34628053 PMCID: PMC8571174 DOI: 10.1016/j.ymthe.2021.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/26/2022] Open
Abstract
Genome editing produces genetic modifications in somatic cells, offering novel curative possibilities for sickle cell disease and β-thalassemia. These opportunities leverage clinical knowledge of hematopoietic stem cell transplant and gene transfer. Advantages to this mode of ex vivo therapy include locus-specific alteration of patient hematopoietic stem cell genomes, lack of allogeneic immune response, and avoidance of insertional mutagenesis. Despite exciting progress, many aspects of this approach remain to be optimized for ideal clinical implementation, including the efficiency and specificity of gene modification, delivery to hematopoietic stem cells, and robust and nontoxic engraftment of gene-modified cells. This review highlights genome editing as compared to other genetic therapies, the differences between editing strategies, and the clinical prospects and challenges of implementing genome editing as a novel treatment. As the world's most common monogenic disorders, the β-hemoglobinopathies are at the forefront of bringing genome editing to the clinic and hold promise for molecular medicine to address human disease at its root.
Collapse
Affiliation(s)
- Tolulope O Rosanwo
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston MA, USA; Department of Pediatrics, Boston Medical Center, Boston, MA, USA
| | - Daniel E Bauer
- Department of Pediatrics, Harvard Medical School, Boston MA, USA; Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA; Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
21
|
Yannaki E, Psatha N, Papadopoulou A, Athanasopoulos T, Gravanis A, Roubelakis MG, Tsirigotis P, Anagnostopoulos A, Anagnou NP, Vassilopoulos G. Success Stories and Challenges Ahead in Hematopoietic Stem Cell Gene Therapy: Hemoglobinopathies as Disease Models. Hum Gene Ther 2021; 32:1120-1137. [PMID: 34662232 DOI: 10.1089/hum.2021.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Gene therapy is a relatively novel field that amounts to around four decades of continuous growth with its good and bad moments. Currently, the field has entered the clinical arena with the ambition to fulfil its promises for a permanent fix of incurable genetic disorders. Hemoglobinopathies as target diseases and hematopoietic stem cells (HSCs) as target cells of genetic interventions had a major share in the research effort toward efficiently implementing gene therapy. Dissection of HSC biology and improvements in gene transfer and gene expression technologies evolved in an almost synchronous manner to a point where the two fields seem to be functionally intercalated. In this review, we focus specifically on the development of gene therapy for hemoglobin disorders and look at both gene addition and gene correction strategies that may dominate the field of HSC-directed gene therapy in the near future and transform the therapeutic landscape for genetic diseases.
Collapse
Affiliation(s)
- Evangelia Yannaki
- Hematology Department-HCT Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Nikoletta Psatha
- Altius Institute for Biomedical Sciences, Seattle, Washington, USA
| | - Anastasia Papadopoulou
- Hematology Department-HCT Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | - Takis Athanasopoulos
- Cell and Gene Therapy (CGT), Medicinal Science and Technology (MST), GlaxoSmithKline (GSK), Medicines Research Centre, Stevenage, United Kingdom
| | - Achilleas Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Greece
| | - Maria G Roubelakis
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece and Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Panagiotis Tsirigotis
- 2nd Department of Internal Medicine, ATTIKO General University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Achilles Anagnostopoulos
- Hematology Department-HCT Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, Thessaloniki, Greece
| | | | - George Vassilopoulos
- BRFAA, Cell and Gene Therapy Lab, Athens, Greece.,Department of Hematology, UHL, University of Thessaly Medical School, Athens, Greece
| |
Collapse
|
22
|
American Society of Hematology 2021 guidelines for sickle cell disease: stem cell transplantation. Blood Adv 2021; 5:3668-3689. [PMID: 34581773 DOI: 10.1182/bloodadvances.2021004394c] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/23/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Sickle cell disease (SCD) is a life-limiting inherited hemoglobinopathy that results in significant complications and affects quality of life. Hematopoietic stem cell transplantation (HSCT) is currently the only curative intervention for SCD; however, guidelines are needed to inform how to apply HSCT in clinical practice. OBJECTIVE These evidence-based guidelines of the American Society of Hematology (ASH) are intended to support patients, clinicians, and health professionals in their decisions about HSCT for SCD. METHODS The multidisciplinary guideline panel formed by ASH included 2 patient representatives and was balanced to minimize potential bias from conflicts of interest. The Mayo Evidence-Based Practice Research Program supported the guideline development process, including performing systematic evidence reviews (through 2019). The panel prioritized clinical questions and outcomes according to their importance for clinicians and patients. The panel used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach, including GRADE Evidence-to-Decision frameworks, to assess evidence and make recommendations, which were subject to public comment. RESULTS The panel agreed on 8 recommendations to help patients and providers assess how individuals with SCD should consider the timing and type of HSCT. CONCLUSIONS The evidence review yielded no randomized controlled clinical trials for HSCT in SCD; therefore, all recommendations are based on very low certainty in the evidence. Key recommendations include considering HSCT for those with neurologic injury or recurrent acute chest syndrome at an early age and to improve nonmyeloablative regimens. Future research should include the development of a robust SCD registry to serve as a comparator for HSCT studies.
Collapse
|
23
|
Leonard A, Tisdale JF. A pause in gene therapy: Reflecting on the unique challenges of sickle cell disease. Mol Ther 2021; 29:1355-1356. [PMID: 33743192 DOI: 10.1016/j.ymthe.2021.03.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Alexis Leonard
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
24
|
Sezaki M, Hayashi Y, Wang Y, Johansson A, Umemoto T, Takizawa H. Immuno-Modulation of Hematopoietic Stem and Progenitor Cells in Inflammation. Front Immunol 2020; 11:585367. [PMID: 33329562 PMCID: PMC7732516 DOI: 10.3389/fimmu.2020.585367] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Lifelong blood production is maintained by bone marrow (BM)-residing hematopoietic stem cells (HSCs) that are defined by two special properties: multipotency and self-renewal. Since dysregulation of either may lead to a differentiation block or extensive proliferation causing dysplasia or neoplasia, the genomic integrity and cellular function of HSCs must be tightly controlled and preserved by cell-intrinsic programs and cell-extrinsic environmental factors of the BM. The BM had been long regarded an immune-privileged organ shielded from immune insults and inflammation, and was thereby assumed to provide HSCs and immune cells with a protective environment to ensure blood and immune homeostasis. Recently, accumulating evidence suggests that hemato-immune challenges such as autoimmunity, inflammation or infection elicit a broad spectrum of immunological reactions in the BM, and in turn, influence the function of HSCs and BM environmental cells. Moreover, in analogy with the emerging concept of “trained immunity”, certain infection-associated stimuli are able to train HSCs and progenitors to produce mature immune cells with enhanced responsiveness to subsequent challenges, and in some cases, form an inflammatory or infectious memory in HSCs themselves. In this review, we will introduce recent findings on HSC and hematopoietic regulation upon exposure to various hemato-immune stimuli and discuss how these challenges can elicit either beneficial or detrimental outcomes on HSCs and the hemato-immune system, as well as their relevance to aging and hematologic malignancies.
Collapse
Affiliation(s)
- Maiko Sezaki
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Yoshikazu Hayashi
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Yuxin Wang
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Alban Johansson
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Terumasa Umemoto
- Laboratory of Hematopoietic Stem Cell Engineering, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
25
|
Baseline TP53 mutations in adults with SCD developing myeloid malignancy following hematopoietic cell transplantation. Blood 2020; 135:1185-1188. [PMID: 32062672 DOI: 10.1182/blood.2019004001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
26
|
Aworanti OW, Fasola FA, Kotila TR, Olaniyi JA, Brown BJ. Acute leukemia in sickle cell disease patients in a tertiary health facility in Nigeria: a case series. Afr Health Sci 2020; 20:1304-1312. [PMID: 33402979 PMCID: PMC7751539 DOI: 10.4314/ahs.v20i3.36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Sickle cell disease(SCD) is a disorder of red cells resulting from the co-inheritance of haemoglobin S (HbS) with another abnormal haemoglobin. The diagnosis of acute leukaemia is uncommon in our patients with sickle cell disease more so the patients have high morbidity and mortality due to the sickling process. Acute leukemia is a malignant clonal disorder of haemopoietic precursor cells resulting in accumulation of immature blood cells in the bone marrow and blood. The objective of the case series was to highlight the challenges of diagnosis and management of SCD patients with acute leukaemia, the importance of peripheral blood film review and propound a possible risk factor. METHODS Records of 58 patients diagnosed and managed for acute leukaemia over a 7 year period at the University College Hospital, Ibadan were reviewed. The diagnosis of acute leukaemia was based on clinical features in addition to peripheral and bone marrow smears findings. Microsoft excel version 2013 was used for statistical analysis. RESULTS Five (8.6%) of the patients with acute leukaemia also had sickle cell disease: 3 males and 2 females were described. Recurrent fever and anaemia were the most consistent presenting features in the patients. All the patients were not on any routine medications meant for SCD patients and had poor history of clinic attendance prior to the diagnosis of acute leukaemia. The diagnosis of acute leukaemia was not made until the patients were seen by a haematologist. The principal tool of diagnosis in all the patients was peripheral blood film review. Two patients were discharged against medical advice. The treatment period ranged between one month and one year in the remaining three patients. CONCLUSION SCD patients are not exempted from developing acute leukaemias and the diagnoses of the two conditions overwhelms the social and economic support of patients and care givers. The study also underscores the relevance of high level of suspicion and prompt review of peripheral blood film of SCD patients particularly when patients present with unremitting symptoms associated with anaemia and fever.
Collapse
Affiliation(s)
- Oladapo W Aworanti
- Haematology Department, Central Laboratory, Synlab Nigeria Limited, Lagos
| | - Foluke A Fasola
- Department of Haematology, College of Medicine, University of Ibadan, Nigeria
| | - Taiwo R Kotila
- Department of Haematology, College of Medicine, University of Ibadan, Nigeria
| | - John A Olaniyi
- Department of Haematology, College of Medicine, University of Ibadan, Nigeria
| | - Biobele J Brown
- Department of Haematology, College of Medicine, University of Ibadan, Nigeria
| |
Collapse
|
27
|
Curative vs targeted therapy for SCD: does it make more sense to address the root cause than target downstream events? Blood Adv 2020; 4:3457-3465. [PMID: 32722787 DOI: 10.1182/bloodadvances.2020001469] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Sickle cell disease (SCD) places a heavy burden on a global and increasing population predominantly resident in resource-poor and developing countries. Progress continues to be made in preventing childhood mortality, and increasing numbers of chronically ill adults with disease are requiring care for disease sequelae. Curative therapies for SCD are therefore attractive to physicians and investigators focused on SCD. Gene therapies are being developed, and several are now in various stages of early-phase human clinical trials. However, we must also pursue avenues through which we can do the most good for the most people alive today. Such efforts include improving our understanding of disease mechanisms and which disease sequelae most strongly affect survival and interfere with quality of life. The pathways leading to disease sequelae are multiple, complex, and highly interactive. Four drugs have now been approved by the US Food and Drug Administration for SCD; however, each has a distinct mechanism and a measurable but limited effect on the many clinical sequelae of SCD. We therefore need to learn how to approach multi-agent therapy for SCD. The order of addition of each agent to treat a specific patient will need to be guided by response to previous therapy, risk factors identified for specific disease outcomes, and clinical studies to determine more comprehensively how the 4 currently approved drugs might interact and produce (or not) additive effects. Moreover, this will have to be accomplished with defined end points in mind, according to which pose the greatest threats to quality of life as well as survival.
Collapse
|
28
|
The double-edged sword of AlloHCT for SCD. Blood 2020; 135:1083-1085. [PMID: 32243517 DOI: 10.1182/blood.2020005118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
29
|
Chellapandian D, Nicholson CL. Haploidentical bone marrow transplantation in a patient with sickle cell disease and acute myeloid leukemia. Pediatr Transplant 2020; 24:e13641. [PMID: 31880407 DOI: 10.1111/petr.13641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/13/2019] [Accepted: 12/02/2019] [Indexed: 12/25/2022]
Abstract
Myeloid neoplasms in children with sickle cell disease have been rarely reported, and the exact underlying connection between these two conditions is not clearly understood. Whether the acute myeloid leukemia in hydroxyurea-treated sickle cell patients is co-incidental or related to therapy remains an unanswered question. Herein, we report a 14-year-old girl of Haitian descent with sickle beta zero thalassemia on chronic hydroxyurea therapy who developed FMS-like tyrosine kinase 3 (FLT3)-mutated acute myeloid leukemia and underwent a complete disease remission following a combination chemotherapy with sorafenib and was subsequently treated using a T cell replete unmanipulated haploidentical bone marrow transplantation followed by post-transplant cyclophosphamide.
Collapse
Affiliation(s)
- Deepak Chellapandian
- Blood and Marrow Transplant Program, Cancer and Blood Disorder Institute, Johns Hopkins All Children's Hospital, St Petersburg, Florida
| | - Cameron L Nicholson
- Pediatric Hematology/Oncology, Golisano Children's Hospital of Southwest Florida, Fort Myers, Florida
| |
Collapse
|
30
|
Zhao Y, Maule J, Li Y, Neff J, McCall CM, Hao T, Yang W, Rehder C, Yang LH, Wang E. Sequential development of human herpes virus 8-positive diffuse large B-cell lymphoma and chronic myelomonocytic leukemia in a 59 year old female patient with hemoglobin SC disease. Pathol Res Pract 2019; 215:152704. [PMID: 31699472 DOI: 10.1016/j.prp.2019.152704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/01/2022]
Abstract
Hematolymphoid neoplasms, including lymphoma and myeloid neoplasms, can occur in patients with sickle cell disease (SCD) or equivalent hemoglobinopathy, but an underlying connection between the two conditions has yet to be fully determined. Herein, we report a unique case of sequential development of two separate hematolymphoid neoplasms, human herpes virus 8 (HHV8)-positive diffuse large B-cell lymphoma (DLBCL) and chronic myelomonocytic leukemia, in a 59 year-old African American female with hemoglobin SC disease. While etiology of immunodeficiency is unknown, the potential causes include hydroxyurea therapy, disease related immunomodulation, chronic inflammation, and relatively old age. The leukemia cells demonstrated profound trilineage dysplasia and harbored complex cytogenetic abnormalities with loss of chromosome 5q and 7q, which are often observed in therapy-related myeloid neoplasms. Besides the potential causes listed above, we propose that myeloid leukemia in this setting may result from genomic changes due to excessive hematopoietic replication triggered by a hemolysis-induced cytokine storm. While myeloid neoplasms in the setting of SCD seems to herald a dismal clinical outcome per the literature, the HHV8-positive DLBCL in our case was apparently indolent, opposing the current perception of its clinical outcome.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Pathology, First affiliated Hospital and College of Basic Medical Science of China Medical University, Shenyang, 110122 PR China; Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710
| | - Jake Maule
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710
| | - Yang Li
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710; Division of Hematology/Oncology, Department of Medicine, Shengjing Hospital affiliated to China Medical University, Shenyang, 110004, PR China
| | - Jadee Neff
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710
| | - Chad M McCall
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710
| | - Tie Hao
- Department of Clinical Laboratory, Anshan Central Hospital, Anshan, Liaoning, 114001 PR China
| | - Weihong Yang
- Department of Clinical Laboratory, Anshan Central Hospital, Anshan, Liaoning, 114001 PR China
| | - Catherine Rehder
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710
| | - Lian-He Yang
- Department of Pathology, First affiliated Hospital and College of Basic Medical Science of China Medical University, Shenyang, 110122 PR China; Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710
| | - Endi Wang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA, 27710.
| |
Collapse
|