Therapeutic perspective for children and young adults living with thalassemia and sickle cell disease

Marta Ferraresi, Daniele Lello Panzieri, Simona Leoni, Maria Domenica Cappellini, Antonis Kattamis, Irene Motta, Marta Ferraresi, Daniele Lello Panzieri, Simona Leoni, Maria Domenica Cappellini, Antonis Kattamis, Irene Motta

Abstract

Hemoglobinopathies, including thalassemias and sickle cell disease, are the most common monogenic diseases worldwide, with estimated annual births of more than 330,000 affected infants. Hemoglobin disorders account for about 3.4% of deaths in children under 5 years of age. The distribution of these diseases is historically linked to current or previously malaria-endemic regions; however, immigration has led to a worldwide distribution of these diseases, making them a global health problem. During the last decade, new treatment approaches and novel therapies have been proposed, some of which have the potential to change the natural history of these disorders. Indeed, the first erythroid maturation agent, luspatercept, and gene therapy have been approved for beta-thalassemia adult patients. For sickle cell disease, molecules targeting vaso-occlusion and hemoglobin S polymerization include crizanlizumab, which has been approved for patients ≥ 16 years, voxelotor approved for patients ≥ 12 years, and L-glutamine for patients older than 5 years. Conclusion: We herein present the most recent advances and future perspectives in thalassemia and sickle cell disease treatment, including new drugs, gene therapy, and gene editing, and the current clinical trial status in the pediatric populations. What is Known: • Red blood cell transfusions, iron chelation therapy and hematopoietic stem cell transplantation have been the mainstay of treatment of thalassemia patients for decades. • For sickle cell disease, until 2005, treatment strategies were mostly the same as those for thalassemia, with the option of simple transfusion or exchange transfusion. In 2007, hydroxyurea was approved for patients ≥ 2 years old. What is New: • In 2019, gene therapy with betibeglogene autotemcel (LentiGlobin BB305) was approved for TDT patients ≥ 12 years old non β0/β0 without matched sibling donor. • Starting from 2017 several new drugs, such as L-glutamine (approved only by FDA), crizanlizumab (approved by FDA and EMA for patients ≥ 16 years), and lastly voxelotor (approved by FDA and EMA for patients ≥ 12 years old).

Keywords: Crizanlizumab; Gene editing; Gene therapy; Luspatercept; Sickle cell disease; Thalassemia.

Conflict of interest statement

MDC has been or is a current consultant for Sanofi-Genzyme, Novartis, Celgene Corp (Bristol Myers Squibb), Vifor Pharma, and Ionis Pharmaceuticals, and has received research funding from Sanofi-Genzyme, Novartis, Celgene Corp (Bristol Myers Squibb), La Jolla Pharmaceutical Company, Roche, Protagonist Therapeutics, and CRISPR Therapeutics. AK has received advisory board fees from Agios Pharmaceuticals, AMGEN, Celgene (Bristol Myers Squibb), Crisp/Vertex, Ionis, Novartis, Vifor Pharma; speaker fees from Celgene (Bristol Myers Squibb), Chiesi, Crisp/Vertex, Novartis, Vifor Pharma; and research funding from Celgene (Bristol Myers Squibb), Novartis. IM has received advisory board fees from Bristol Myers Squibb, Sanofi Genzyme, Amicus Therapeutics, and speaker fees from Bristol Myers Squibb, Sanofi Genzyme.

© 2023. The Author(s).

Figures

Fig. 1
Fig. 1
Milestones in the development of treatments for thalassemias and sickle cell disease. RBC, red blood cell; HSCT: hematopoietic stem cell transplantation

References

    1. Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ. 2008;86:480–487. doi: 10.2471/blt.06.036673.
    1. Makani J, Soka D, Rwezaula S, et al. Health policy for sickle cell disease in Africa: experience from Tanzania on interventions to reduce under-five mortality. Trop Med Int Health. 2015;20:184–187. doi: 10.1111/tmi.12428.
    1. Nnodu OE, Oron AP, Sopekan A, et al. Child mortality from sickle cell disease in Nigeria: a model-estimated, population-level analysis of data from the 2018 Demographic and Health Survey. Lancet Haematol. 2021;8:e723–e731. doi: 10.1016/S2352-3026(21)00216-7.
    1. Piel FB, Steinberg MH, Rees DC. Sickle cell disease. N Engl J Med. 2017;376:1561–1573. doi: 10.1056/NEJMra1510865.
    1. Piel FB, Weatherall DJ. The α-thalassemias. N Engl J Med. 2014;371:1908–1916. doi: 10.1056/NEJMra1404415.
    1. Inusa BPD, Colombatti R (2017) European migration crises: the role of national hemoglobinopathy registries in improving patient access to care. Pediatr Blood Cancer 64:e26515. 10.1002/pbc.26515
    1. Rund D, Rachmilewitz E. Beta-thalassemia. N Engl J Med. 2005;353:1135–1146. doi: 10.1056/NEJMra050436.
    1. Roberts I, de Montalembert M. Sickle cell disease as a paradigm of immigration hematology: new challenges for hematologists in Europe. Haematologica. 2007;92:865–871. doi: 10.3324/haematol.11474.
    1. Cappellini MD, Marcon A, Fattizzo B, Motta I (2021) Innovative treatments for rare anemias. Hemasphere 5:e576. 10.1097/HS9.0000000000000576
    1. Taher AT, Musallam KM, Cappellini MD. β-thalassemias. N Engl J Med. 2021;384:727–743. doi: 10.1056/NEJMra2021838.
    1. Kattamis A, Kwiatkowski JL, Aydinok Y. Thalassaemia Lancet. 2022;399:2310–2324. doi: 10.1016/S0140-6736(22)00536-0.
    1. Sinlapamongkolkul P, Surapolchai P (2020) Health-related quality of life in Thai children with thalassemia as evaluated by PedsQL and EQ-5D-Y: a single-center experience. Mediterr J Hematol Infect Dis 12:e2020036. 10.4084/MJHID.2020.036
    1. Chan WY, Leung AW, Luk CW et al (2018) Outcomes and morbidities of patients who survive haemoglobin Bart’s hydrops fetalis syndrome: 20-year retrospective review. Hong Kong Med J 24:107–118. 10.12809/hkmj176336
    1. Kreger EM, Singer ST, Witt RG, et al. Favorable outcomes after in utero transfusion in fetuses with alpha thalassemia major: a case series and review of the literature. Prenat Diagn. 2016;36:1242–1249. doi: 10.1002/pd.4966.
    1. Songdej D, Babbs C, Higgs DR, BHFS International Consortium An international registry of survivors with Hb Bart’s hydrops fetalis syndrome. Blood. 2017;129:1251–1259. doi: 10.1182/blood-2016-08-697110.
    1. Motta I, Bou-Fakhredin R, Taher AT, Cappellini MD. Beta thalassemia: new therapeutic options beyond transfusion and iron chelation. Drugs. 2020;80:1053–1063. doi: 10.1007/s40265-020-01341-9.
    1. Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia Lancet. 2018;391:155–167. doi: 10.1016/S0140-6736(17)31822-6.
    1. Guidelines for the management of non-transfusion dependent thalassaemia (NTDT) (2nd Edition – 2017). In: TIF. . Accessed 2 Feb 2023
    1. Forni GL, Gianesin B, Musallam KM, et al. Overall and complication-free survival in a large cohort of patients with β-thalassemia major followed over 50 years. Am J Hematol. 2023 doi: 10.1002/ajh.26798.
    1. Bou-Fakhredin R, Tabbikha R, Daadaa H, Taher AT. Emerging therapies in β-thalassemia: toward a new era in management. Expert Opin Emerg Drugs. 2020;25:113–122. doi: 10.1080/14728214.2020.1752180.
    1. Cappellini MD, Taher AT. The use of luspatercept for thalassemia in adults. Blood Adv. 2021;5:326–333. doi: 10.1182/bloodadvances.2020002725.
    1. Suragani RNVS, Cawley SM, Li R, et al. Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine β-thalassemia. Blood. 2014;123:3864–3872. doi: 10.1182/blood-2013-06-511238.
    1. Cappellini MD, Viprakasit V, Taher AT, et al. A phase 3 trial of luspatercept in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2020;382:1219–1231. doi: 10.1056/NEJMoa1910182.
    1. Food and Drug Administration (FDA). Reblozyl. . Accessed 02 Feb 2023
    1. European Medicines Agency (EMA). Reblozyl. . Accessed 29 Jan 2023
    1. Taher AT, Bou-Fakhredin R, Kattamis A et al (2021) Improving outcomes and quality of life for patients with transfusion-dependent β-thalassemia: recommendations for best clinical practice and the use of novel treatment strategies. Expert Rev Hematol 14:897–909. 10.1080/17474086.2021.1977116
    1. Taher AT, Cappellini MD, Kattamis A, et al. Luspatercept for the treatment of anaemia in non-transfusion-dependent β-thalassaemia (BEYOND): a phase 2, randomised, double-blind, multicentre, placebo-controlled trial. Lancet Haematol. 2022;S2352–3026(22):00208–213. doi: 10.1016/S2352-3026(22)00208-3.
    1. Al-Samkari H, van Beers EJ. Mitapivat, a novel pyruvate kinase activator, for the treatment of hereditary hemolytic anemias. Ther Adv Hematol. 2021;12:20406207211066070. doi: 10.1177/20406207211066070.
    1. Kattamis A. An energy booster for thalassaemic red blood cells. The Lancet. 2022;400:470–471. doi: 10.1016/S0140-6736(22)01431-3.
    1. Al-Samkari H, Galactéros F, Glenthøj A, et al. Mitapivat versus placebo for pyruvate kinase deficiency. N Engl J Med. 2022;386:1432–1442. doi: 10.1056/NEJMoa2116634.
    1. Mattè A, Kosinski PA, Federti E, et al. Mitapivat improves transfusion burden and reduces iron overload in thalassemic mice. Blood. 2021;138:2016. doi: 10.1182/blood-2021-153721.
    1. Kuo KHM, Layton DM, Lal A, et al. Long-term efficacy and safety of the oral pyruvate kinase activator mitapivat in adults with non-transfusion-dependent alpha- or beta-thalassemia. Blood. 2021;138:576. doi: 10.1182/blood-2021-150386.
    1. Agios Pharmaceuticals, Inc. (2022) A phase 3, double-blind, randomized, placebo-controlled, multicenter study evaluating the efficacy and safety of mitapivat in subjects with transfusion-dependent alpha- or beta-thalassemia (ENERGIZE-T).
    1. Agios Pharmaceuticals, Inc. (2022) A phase 3, double-blind, randomized, placebo-controlled, multicenter study evaluating the efficacy and safety of mitapivat in subjects with non-transfusion-dependent alpha- or beta-thalassemia (ENERGIZE).
    1. Imara, Inc. (2022) A phase 2 study to evaluate the safety and tolerability of IMR-687 in subjects with beta thalassemia.
    1. Porter J, Taher A, Viprakasit V, et al. Oral ferroportin inhibitor vamifeport for improving iron homeostasis and erythropoiesis in β-thalassemia: current evidence and future clinical development. Expert Rev Hematol. 2021;14:633–644. doi: 10.1080/17474086.2021.1935854.
    1. La Jolla Pharmaceutical Company (2021) A multi-center, randomized, open-label, parallel-group study with LJPC-401 for the treatment of myocardial iron overload in patients with transfusion-dependent beta thalassemia.
    1. Protagonist Therapeutics, Inc. (2021) A phase 2 study of PTG-300 in non-transfusion dependent (NTD) and transfusion-dependent (TD) β-thalassemia subjects with chronic anemia.
    1. Gelderman MP, Baek JH, Yalamanoglu A, et al. Reversal of hemochromatosis by apotransferrin in non-transfused and transfused Hbbth3/+ (heterozygous B1/B2 globin gene deletion) mice. Haematologica. 2015;100:611–622. doi: 10.3324/haematol.2014.117325.
    1. Langer AL, Esrick EB. β-Thalassemia: evolving treatment options beyond transfusion and iron chelation. Hematology Am Soc Hematol Educ Program. 2021;2021:600–606. doi: 10.1182/hematology.2021000313.
    1. Prothya Biosolutions (2022) Efficacy and safety of human apotransferrin in patients with β-thalassemia Intermedia.
    1. Musallam KM, Rivella S, Taher AT. Management of non-transfusion-dependent β-thalassemia (NTDT): the next 5 years. Am J Hematol. 2021;96:E57–E59. doi: 10.1002/ajh.26055.
    1. Nai A, Pagani A, Mandelli G, et al. Deletion of TMPRSS6 attenuates the phenotype in a mouse model of β-thalassemia. Blood. 2012;119:5021–5029. doi: 10.1182/blood-2012-01-401885.
    1. Schmidt PJ, Toudjarska I, Sendamarai AK, et al. An RNAi therapeutic targeting Tmprss6 decreases iron overload in Hfe(-/-) mice and ameliorates anemia and iron overload in murine β-thalassemia intermedia. Blood. 2013;121:1200–1208. doi: 10.1182/blood-2012-09-453977.
    1. Manwani D, Frenette PS. Vaso-occlusion in sickle cell disease: pathophysiology and novel targeted therapies. Blood. 2013;122:3892–3898. doi: 10.1182/blood-2013-05-498311.
    1. Sundd P, Gladwin MT, Novelli EM. Pathophysiology of sickle cell disease. Annu Rev Pathol. 2019;14:263–292. doi: 10.1146/annurev-pathmechdis-012418-012838.
    1. Payne AB, Mehal JM, Chapman C, et al. Trends in sickle cell disease-related mortality in the United States, 1979 to 2017. Ann Emerg Med. 2020;76:S28–S36. doi: 10.1016/j.annemergmed.2020.08.009.
    1. Bakshi N, Sinha CB, Ross D et al (2017) Proponent or collaborative: physician perspectives and approaches to disease modifying therapies in sickle cell disease. PLoS ONE 12:e0178413. 10.1371/journal.pone.0178413
    1. Meier ER. Treatment options for sickle cell disease. Pediatr Clin North Am. 2018;65:427–443. doi: 10.1016/j.pcl.2018.01.005.
    1. Food and Drug Administration (FDA). Droxia. . Accessed 02 Feb 2023
    1. European Medicines Agency (EMA). Xromi. . Accessed 02 Feb 2023
    1. Zhou AE, Travassos MA. Bringing sickle-cell treatments to children in sub-Saharan Africa. N Engl J Med. 2022;387:488–491. doi: 10.1056/NEJMp2201763.
    1. John CC, Opoka RO, Latham TS, et al. Hydroxyurea dose escalation for sickle cell anemia in sub-Saharan Africa. N Engl J Med. 2020;382:2524–2533. doi: 10.1056/NEJMoa2000146.
    1. Karkoska K, Pfeiffer A, Beebe DW, et al. Early hydroxyurea use is neuroprotective in children with sickle cell anemia. Am J Hematol. 2022 doi: 10.1002/ajh.26664.
    1. Wang WC, Ware RE, Miller ST, et al. Hydroxycarbamide in very young children with sickle-cell anaemia: a multicentre, randomised, controlled trial (BABY HUG) Lancet. 2011;377:1663–1672. doi: 10.1016/S0140-6736(11)60355-3.
    1. Charache S, Terrin ML, Moore RD, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med. 1995;332:1317–1322. doi: 10.1056/NEJM199505183322001.
    1. Shih S, Cohen LL. A systematic review of medication adherence interventions in pediatric sickle cell disease. J Pediatr Psychol. 2020;45:593–606. doi: 10.1093/jpepsy/jsaa031.
    1. de Montalembert M, Voskaridou E, Oevermann L, et al. Real-Life experience with hydroxyurea in patients with sickle cell disease: results from the prospective ESCORT-HU cohort study. Am J Hematol. 2021;96:1223–1231. doi: 10.1002/ajh.26286.
    1. Niihara Y, Miller ST, Kanter J, et al. A phase 3 trial of l-glutamine in sickle cell disease. N Engl J Med. 2018;379:226–235. doi: 10.1056/NEJMoa1715971.
    1. Food and Drug Administration (FDA). Endari. . Accessed 02 Feb 2023
    1. Vichinsky E, Hoppe CC, Ataga KI, et al. A phase 3 randomized trial of voxelotor in sickle cell disease. N Engl J Med. 2019;381:509–519. doi: 10.1056/NEJMoa1903212.
    1. Food and Drug Administration (FDA). Oxbryta. . Accessed 02 Feb 2023
    1. European Medicines Agency (EMA). Oxbryta. . Accessed 02 Feb 2023
    1. Food and Drug Administration (FDA). Adakveo. . Accessed 02 Feb 2023
    1. European Medicines Agency. Adakveo. . Accessed 02 Feb 2023
    1. Ataga KI, Kutlar A, Kanter J, et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. 2017;376:429–439. doi: 10.1056/NEJMoa1611770.
    1. Varelas C, Tampaki A, Sakellari I, et al. Complement in sickle cell disease: are we ready for prime time? J Blood Med. 2021;12:177–187. doi: 10.2147/JBM.S287301.
    1. Merle NS, Boudhabhay I, Leon J, et al. Complement activation during intravascular hemolysis: Implication for sickle cell disease and hemolytic transfusion reactions. Transfus Clin Biol. 2019;26:116–124. doi: 10.1016/j.tracli.2019.02.008.
    1. Rab MAE, Bos J, van Oirschot BA, et al. Decreased activity and stability of pyruvate kinase in sickle cell disease: a novel target for mitapivat therapy. Blood. 2021;137:2997–3001. doi: 10.1182/blood.2020008635.
    1. Schroeder P, Fulzele K, Forsyth S, et al. Etavopivat, a pyruvate kinase activator in red blood cells, for the treatment of sickle cell disease. J Pharmacol Exp Ther. 2022;380:210–219. doi: 10.1124/jpet.121.000743.
    1. Xu JZ, Conrey AK, Frey IC et al (2022) A phase 1 dose escalation study of the pyruvate kinase activator mitapivat (AG-348) in sickle cell disease. Blood 2022015403. 10.1182/blood.2022015403
    1. van Dijk MJ, Rab MAE, Rijneveld AW, et al. Safety and efficacy of mitapivat (AG-348), an oral activator of pyruvate kinase R, in subjects with sickle cell disease: a phase 2, open-label study (ESTIMATE) Blood. 2021;138:2047. doi: 10.1182/blood-2021-150234.
    1. Howard J, Kuo KHM, Oluyadi A, et al. A phase 2/3, randomized, double-blind, placebo-controlled study of mitapivat in patients with sickle cell disease. Blood. 2021;138:3109. doi: 10.1182/blood-2021-148370.
    1. Telen M, Brown R, Idowu M, et al. O-03: Etavopivat treatment for up to 12 weeks in patients with sickle cell disease is well tolerated and improves red blood cell health. Hemasphere. 2022;6:02–03. doi: 10.1097/01.HS9.0000872820.66998.56.
    1. Andemariam B, Mant T, Eleftheriou P, et al. Treatment with IMR-687, a highly selective PDE9 inhibitor, increases HbF and reduces VOCs in adults with sickle cell disease in a long-term, phase 2a, open-label extension study. Blood. 2021;138:2046. doi: 10.1182/blood-2021-149536.
    1. Imara, Inc. (2022) A phase 2b study to evaluate the safety and efficacy of IMR-687 in subjects with sickle cell disease.
    1. Heeney MM, Hoppe CC, Abboud MR, et al. A multinational trial of prasugrel for sickle cell vaso-occlusive events. N Engl J Med. 2016;374:625–635. doi: 10.1056/NEJMoa1512021.
    1. Heeney MM, Abboud MR, Githanga J, et al. Ticagrelor vs placebo for the reduction of vaso-occlusive crises in pediatric sickle cell disease: the HESTIA3 study. Blood. 2022;140:1470–1481. doi: 10.1182/blood.2021014095.
    1. Dampier CD, Telen MJ, Wun T, et al. A randomized clinical trial of the efficacy and safety of rivipansel for sickle cell vaso-occlusive crisis. Blood. 2023;141:168–179. doi: 10.1182/blood.2022015797.
    1. Casella JF, Barton BA, Kanter J, et al. Effect of poloxamer 188 vs placebo on painful vaso-occlusive episodes in children and adults with sickle cell disease: a randomized clinical trial. JAMA. 2021;325:1513–1523. doi: 10.1001/jama.2021.3414.
    1. Kanter J, Liem RI, Bernaudin F, et al. American Society of Hematology 2021 guidelines for sickle cell disease: stem cell transplantation. Blood Adv. 2021;5:3668–3689. doi: 10.1182/bloodadvances.2021004394C.
    1. Angelucci E, Matthes-Martin S, Baronciani D, et al. Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: indications and management recommendations from an international expert panel. Haematologica. 2014;99:811–820. doi: 10.3324/haematol.2013.099747.
    1. Angelucci E, Pilo F, Coates TD. Transplantation in thalassemia: revisiting the Pesaro risk factors 25 years later. Am J Hematol. 2017;92:411–413. doi: 10.1002/ajh.24674.
    1. EMA (2019) Zynteglo. In: European Medicines Agency. . Accessed 02 Feb 2023
    1. Food and Drug Administration (FDA). Zynteglo. . Accessed 02 Feb 2023
    1. Thompson AA, Walters MC, Kwiatkowski J, et al. Gene therapy in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2018;378:1479–1493. doi: 10.1056/NEJMoa1705342.
    1. Kwiatkowski J (2020) Long-term efficacy and safety of betibeglogene autotemcel gene therapy for the treatment of transfusion-dependent β-thalassemia: results in patients with up to 6 years of follow-up. ASH
    1. Ribeil J-A, Hacein-Bey-Abina S, Payen E, et al. Gene therapy in a patient with sickle cell disease. N Engl J Med. 2017;376:848–855. doi: 10.1056/NEJMoa1609677.
    1. bluebird bio. bluebird bio provides updated findings from reported case of acute myeloid leukemia (AML) in LentiGlobin for sickle cell disease (SCD) gene therapy program. . Accessed 02 Feb 2023
    1. Hsieh MM, Bonner M, Pierciey FJ, et al. Myelodysplastic syndrome unrelated to lentiviral vector in a patient treated with gene therapy for sickle cell disease. Blood Adv. 2020;4:2058–2063. doi: 10.1182/bloodadvances.2019001330.
    1. Bluebird bio. bluebird bio announces the lifting of FDA clinical hold for sickle cell disease and β-thalassemia studies - bluebird bio, Inc. . Accessed 02 Feb 2023
    1. Kanter J, Walters MC, Krishnamurti L, et al. Biologic and clinical efficacy of LentiGlobin for sickle cell disease. N Engl J Med. 2022;386:617–628. doi: 10.1056/NEJMoa2117175.
    1. Goyal S, Tisdale J, Schmidt M, et al. Acute myeloid leukemia case after gene therapy for sickle cell disease. N Engl J Med. 2022;386:138–147. doi: 10.1056/NEJMoa2109167.
    1. Bluebird Bio ends commercial operations in Europe (2021). Available from: . Accessed 10 Oct 2022
    1. Marktel S, Scaramuzza S, Cicalese MP, et al. Intrabone hematopoietic stem cell gene therapy for adult and pediatric patients affected by transfusion-dependent ß-thalassemia. Nat Med. 2019;25:234–241. doi: 10.1038/s41591-018-0301-6.
    1. Orchard Therapeutics (2022) A long-term safety and efficacy follow-on study in participants with transfusion dependent beta-thalassemia who have previously received OTL-300 (formerly know as GSK2696277) and completed the TIGET-BTHAL study.
    1. Assistance Publique - Hôpitaux de Paris (2021) A phase 1/2 open label study evaluating the safety and efficacy of gene therapy of the sickle cell disease by transplantation of an autologous CD34+ enriched Cell fraction that contains CD34+ cells transduced ex vivo with the GLOBE1 lentiviral vector expressing the βAS3 globin gene (GLOBE1 βAS3 modified autologous CD34+ Cells) in patients with sickle cell disease (SCD).
    1. Sankaran VG, Menne TF, Xu J, et al. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 2008;322:1839–1842. doi: 10.1126/science.1165409.
    1. Esrick EB, Lehmann LE, Biffi A, et al. Post-transcriptional genetic silencing of BCL11A to treat sickle cell disease. N Engl J Med. 2021;384:205–215. doi: 10.1056/NEJMoa2029392.
    1. Frangoul H (2020) Safety and efficacy of CTX001 in patients with transfusion-dependent β-thalassemia and sickle cell disease: early results from the Climb THAL-111 and Climb SCD-121 studies of Autologous CRISPR-CAS9–modified CD34+ Hematopoietic stem and progenitor cells. ASH
    1. Locatelli F (2022) Efficacy and safety of a single dose of exagamglogene autotemcel for transfusion-dependent β-thalassemia. ASH
    1. Motta I, Ghiaccio V, Cosentino A, Breda L (2019) Curing hemoglobinopathies: challenges and advances of conventional and new gene therapy approaches. Mediterr J Hematol Infect Dis 11:e2019067. 10.4084/MJHID.2019.067
    1. Mackenzie T (2022) A single-center, non-randomized study of the safety and efficacy of in utero hematopoietic stem cell transplantation for the treatment of fetuses with alpha thalassemia major.
    1. Horvei P, MacKenzie T, Kharbanda S. Advances in the management of α-thalassemia major: reasons to be optimistic. Hematology Am Soc Hematol Educ Program. 2021;2021:592–599. doi: 10.1182/hematology.2021000295.
    1. Witt R, MacKenzie TC, Peranteau WH. Fetal stem cell and gene therapy. Semin Fetal Neonatal Med. 2017;22:410–414. doi: 10.1016/j.siny.2017.05.003.
    1. Mensah C, Sheth S. Optimal strategies for carrier screening and prenatal diagnosis of α- and β-thalassemia. Hematology Am Soc Hematol Educ Program. 2021;2021:607–613. doi: 10.1182/hematology.2021000296.

Source: PubMed

3
Iratkozz fel