Defining curative endpoints for transfusion-dependent β-thalassemia in the era of gene therapy and gene editing.


Journal

American journal of hematology
ISSN: 1096-8652
Titre abrégé: Am J Hematol
Pays: United States
ID NLM: 7610369

Informations de publication

Date de publication:
15 Dec 2023
Historique:
revised: 16 10 2023
received: 23 06 2023
accepted: 06 11 2023
medline: 15 12 2023
pubmed: 15 12 2023
entrez: 15 12 2023
Statut: aheadofprint

Résumé

β-thalassemia is a monogenic disease that results in varying degrees of anemia. In the most severe form, known as transfusion-dependent β-thalassemia (TDT), the clinical hallmarks are ineffective erythropoiesis and a requirement of regular, life-long red blood cell transfusions, with the development of secondary clinical complications such as iron overload, end-organ damage, and a risk of early mortality. With the exception of allogeneic hematopoietic cell transplantation, current treatments for TDT address disease symptoms and not the underlying cause of disease. Recently, a growing number of gene addition and gene editing-based treatments for patients with TDT with the potential to provide a one-time functional cure have entered clinical trials. A key challenge in the design and evaluation of these trials is selecting endpoints to evaluate if these novel genetic therapies have a curative versus an ameliorative effect. Here, we present an overview of the pathophysiology of TDT, review emerging gene addition or gene editing therapeutic approaches for TDT currently in clinical trials, and identify a series of endpoints that can quantify therapeutic effects, including a curative outcome.

Identifiants

pubmed: 38100154
doi: 10.1002/ajh.27166
doi:

Types de publication

Journal Article Review

Langues

eng

Sous-ensembles de citation

IM

Subventions

Organisme : Vertex Pharmaceuticals

Informations de copyright

© 2023 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.

Références

Betts M, Flight PA, Paramore LC, Tian L, Milenkovic D, Sheth S. Systematic literature review of the burden of disease and treatment for transfusion-dependent beta-thalassemia. Clin Ther. 2020;42(2):322-337 e322.
Galanello R, Origa R. Beta-thalassemia. Orphanet J Rare Dis. 2010;5:11.
Origa R. Beta-thalassemia. Genet Med. 2017;19(6):609-619.
Thompson AA, Walters MC, Kwiatkowski J, et al. Gene therapy in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2018;378(16):1479-1493.
Jaing TH, Chang TY, Chen SH, Lin CW, Wen YC, Chiu CC. Molecular genetics of beta-thalassemia: a narrative review. Medicine (Baltimore). 2021;100(45):e27522.
Cappellini MD, Farmakis D, Porter J, Taher A. Guidelines for the Management of Transfusion Dependent Thalassemia (TDT). 2021. 2021 https://thalassaemia.org.cy/wp-content/uploads/2021/06/GUIDELINE-4th-SINGLE-PAGE.pdf
Cappellini MD, Porter JB, Viprakasit V, Taher AT. A paradigm shift on beta-thalassaemia treatment: how will we manage this old disease with new therapies? Blood Rev. 2018;32(4):300-311.
Taher AT, Musallam KM, Cappellini MD. Beta-thalassemias. N Engl J Med. 2021;384(8):727-743.
Hamilton JL, Kizhakkedathu JN. Polymeric nanocarriers for the treatment of systemic iron overload. Mol Cell Ther. 2015;3:3.
Brittenham GM. Iron-chelating therapy for transfusional iron overload. N Engl J Med. 2011;364(2):146-156.
Pennell DJ, Berdoukas V, Karagiorga M, et al. Randomized controlled trial of deferiprone or deferoxamine in beta-thalassemia major patients with asymptomatic myocardial siderosis. Blood. 2006;107(9):3738-3744.
Tanner MA, Galanello R, Dessi C, et al. A randomized, placebo-controlled, double-blind trial of the effect of combined therapy with deferoxamine and deferiprone on myocardial iron in thalassemia major using cardiovascular magnetic resonance. Circulation. 2007;115(14):1876-1884.
Cappellini MD, Bejaoui M, Agaoglu L, et al. Iron chelation with deferasirox in adult and pediatric patients with thalassemia major: efficacy and safety during 5 years’ follow-up. Blood. 2011;118(4):884-893.
Taher AT, Cappellini MD. Luspatercept for beta-thalassemia: beyond red blood cell transfusions. Expert Opin Biol Ther. 2021;21(11):1363-1371.
Shah FT, Sayani F, Trompeter S, Drasar E, Piga A. Challenges of blood transfusions in beta-thalassemia. Blood Rev. 2019;37:100588.
Shenoy S, Walters MC, Ngwube A, et al. Unrelated donor transplantation in children with thalassemia using reduced-intensity conditioning: the URTH trial. Biol Blood Marrow Transplant. 2018;24(6):1216-1222.
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(13):1219-1231.
Cappellini MD, Motta I. New therapeutic targets in transfusion-dependent and -independent thalassemia. Hematology Am Soc Hematol Educ Program. 2017;2017(1):278-283.
Makis A, Voskaridou E, Papassotiriou I, Hatzimichael E. Novel therapeutic advances in beta-thalassemia. Biology (Basel). 2021;10(6):546.
Taher AT, Cappellini MD. How I manage medical complications of beta-thalassemia in adults. Blood. 2018;132(17):1781-1791.
Meral A, Tuncel P, Surmen-Gur E, Ozbek R, Ozturk E, Gunay U. Lipid peroxidation and antioxidant status in beta-thalassemia. Pediatr Hematol Oncol. 2000;17(8):687-693.
Steinberg MH. Fetal hemoglobin in sickle cell anemia. Blood. 2020;136:2392-2400.
Steinberg MH, Sebastiani P. Genetic modifiers of sickle cell disease. Am J Hematol. 2012;87(8):795-803.
Thein SL, Menzel S. Discovering the genetics underlying foetal haemoglobin production in adults. Br J Haematol. 2009;145(4):455-467.
Steinberg MH. Treating sickle cell anemia: a new era dawns. Am J Hematol. 2020;95(4):338-342.
Uda M, Galanello R, Sanna S, et al. Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of β-thalassemia. Proc Natl Acad Sci U S A. 2008;105(5):1620-1625.
Musallam KM, Sankaran VG, Cappellini MD, Duca L, Nathan DG, Taher AT. Fetal hemoglobin levels and morbidity in untransfused patients with beta-thalassemia intermedia. Blood. 2012;119(2):364-367.
Forni GL, Gianesin B, Musallam KM, et al. Overall and complication-free survival in a large cohort of patients with beta-thalassemia major followed over 50 years. Am J Hematol. 2023;98(3):381-387.
Farmakis D, Giakoumis A, Angastiniotis M, Eleftheriou A. The changing epidemiology of the ageing thalassaemia populations: a position statement of the Thalassaemia International Federation. Eur J Haematol. 2020;105(1):16-23.
Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia. Lancet. 2018;391(10116):155-167.
Paramore C, Levine L, Bagshaw E, Ouyang C, Kudlac A, Larkin M. Patient- and caregiver-reported burden of transfusion-dependent beta-thalassemia measured using a digital application. Patient. 2021;14(2):197-208.
Shah F, Telfer P, Velangi M, et al. Routine management, healthcare resource use and patient and carer-reported outcomes of patients with transfusion-dependent beta-thalassaemia in the United Kingdom: a mixed methods observational study. EJHaem. 2021;2(4):738-749.
Sobota A, Yamashita R, Xu Y, et al. Quality of life in thalassemia: a comparison of SF-36 results from the thalassemia longitudinal cohort to reported literature and the US norms. Am J Hematol. 2011;86(1):92-95.
Tedone F, Lamendola P, Lopatriello S, Cafiero D, Piovani D, Forni GL. Quality of life and burden of disease in Italian patients with transfusion-dependent beta-thalassemia. J Clin Med. 2021;11(1):15.
Yengil E, Acipayam C, Kokacya MH, Kurhan F, Oktay G, Ozer C. Anxiety, depression and quality of life in patients with beta thalassemia major and their caregivers. Int J Clin Exp Med. 2014;7(8):2165-2172.
Li C, Mathews V, Kim S, et al. Related and unrelated donor transplantation for beta-thalassemia major: results of an international survey. Blood Adv. 2019;3(17):2562-2570.
Angelucci E, Pilo F, Coates TD. Transplantation in thalassemia: revisiting the Pesaro risk factors 25 years later. Am J Hematol. 2017;92(5):411-413.
Baronciani D, Angelucci E, Potschger U, et al. Hemopoietic stem cell transplantation in thalassemia: a report from the European Society for Blood and Bone Marrow Transplantation Hemoglobinopathy Registry, 2000-2010. Bone Marrow Transplant. 2016;51(4):536-541.
Kanate AS, Majhail NS, Savani BN, et al. Indications for hematopoietic cell transplantation and immune effector cell therapy: guidelines from the American Society for Transplantation and Cellular Therapy. Biol Blood Marrow Transplant. 2020;26(7):1247-1256.
Galgano L, Hutt D. HSCT: how does it work? In: Kenyon M, Babic A, eds. The European Blood and Marrow Transplantation Textbook for Nurses: Under the Auspices of EBMT. Springer; 2018:23-36.
Strocchio L, Locatelli F. Hematopoietic stem cell transplantation in thalassemia. Hematol Oncol Clin North Am. 2018;32(2):317-328.
Anurathapan U, Pakakasama S, Songdej D, et al. Haploidentical hematopoietic stem cell transplantation in thalassemia. Hemoglobin. 2022;46(1):2-6.
Gluckman E, Cappelli B, Bernaudin F, et al. Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood. 2017;129(11):1548-1556.
Kenyon M, Murray J, Quinn B, Greenfield D, Trigoso E. Late effects and long-term follow-up. In: Kenyon M, Babic A, eds. The European Blood and Marrow Transplantation Textbook for Nurses: Under the Auspices of EBMT. Springer; 2018:271-299.
Locatelli F, Thompson AA, Kwiatkowski JL, et al. Betibeglogene autotemcel gene therapy for non-β(0)/β(0) genotype β-thalassemia. N Engl J Med. 2022;386(5):415-427.
Mulas O, Mola B, Caocci G, La Nasa G. Conditioning regimens in patients with beta-thalassemia who underwent hematopoietic stem cell transplantation: a scoping review. J Clin Med. 2022;11(4):907.
Wingard JR, Hsu J, Hiemenz JW. Hematopoietic stem cell transplantation: an overview of infection risks and epidemiology. Infect Dis Clin North Am. 2010;24(2):257-272.
Marktel S, Scaramuzza S, Cicalese MP, et al. Intrabone hematopoietic stem cell gene therapy for adult and pediatric patients affected by transfusion-dependent beta-thalassemia. Nat Med. 2019;25(2):234-241.
ClinicalTrials.gov. Long-term Follow-up of Subjects Treated With OTL-300 for Transfusion Dependent Beta-thalassemia Study (TIGET-BTHAL). 2022. https://clinicaltrials.gov/ct2/show/NCT03275051. Accessed February 17, 2023
Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 gene editing for sickle cell disease and beta-thalassemia. N Engl J Med. 2021;384(3):252-260.
Locatelli F, Lang, P, Corbacioglu, S et al. Transfusion independence and elimination of vaso-occulsive crises after exagamglogene autotemcel in transfusion-independent beta thalassemia and severe sickle cell disease. In: European Hematology Association Annual Meeting, 2023.
Alavi A, Krishnamurti L, Abedi M, et al. Preliminary safety and efficacy results from Precizn-1: an ongoing phase 1/2 study on zinc finger nuclease-modified autologous CD34+HSPCs for sickle cell disease (SCD). Blood. 2021;138(1):2930.
Dube K, Willenberg L, Dee L, et al. Re-examining the HIV ‘functional cure’ oxymoron: time for precise terminology? J Virus Erad. 2020;6(4):100017.
Choi HSJ, Tonthat A, Janssen HLA, Terrault NA. Aiming for functional cure with established and novel therapies for chronic hepatitis B. Hepatol Commun. 2022;6(5):935-949.
Cornberg M, Lok AS, Terrault NA, Zoulim F, Faculty E-AHTEC. Guidance for design and endpoints of clinical trials in chronic hepatitis B-report from the 2019 EASL-AASLD HBV treatment endpoints conference (double dagger). J Hepatol. 2020;72(3):539-557.
Terpos ERM, Ntanasis-Stathopoulos I, Kanellias N, et al. Functional cure, defined as PFS of more than 7 years, is achieved in 9% of myeloma patients in the era of conventional chemotherapy and of first-generation novel anti-myeloma agents; a single-center experience over 20-year period. Blood. 2018;132(1):1968.
Bourgeois S, Sawatani T, Van Mulders A, et al. Towards a functional cure for diabetes using stem cell-derived Beta cells: are we there yet? Cell. 2021;10(1):191.
Baronciani D, Boumendil A, Dalissier A, et al. Hematopoietic cell transplantation in thalassemia and sickle cell disease: report from the European Society for Blood and Bone Marrow Transplantation Hemoglobinopathy Registry: 2000-2017. Blood. 2018;132(1):168.
Kattamis A, Forni GL, Aydinok Y, Viprakasit V. Changing patterns in the epidemiology of beta-thalassemia. Eur J Haematol. 2020;105(6):692-703.
Caocci G, Orofino MG, Vacca A, et al. Long-term survival of beta thalassemia major patients treated with hematopoietic stem cell transplantation compared with survival with conventional treatment. Am J Hematol. 2017;92(12):1303-1310.
Yesilipek MA, Karasu G, Kaya Z, et al. A phase II, multicenter, single-arm study to evaluate the safety and efficacy of deferasirox after hematopoietic stem cell transplantation in children with beta-thalassemia major. Biol Blood Marrow Transplant. 2018;24(3):613-618.
Inati A, El Hachem Y, Tawk S, Kassm SA, Abbas HA. Pediatric immune thrombocytopenia in Lebanon: treatment and predictors of outcome. Pediatr Blood Cancer. 2016;63(2):375-376.
Varni JW, Zebracki K, Hwang M, Mulcahey MJ, Vogel LC. Pain, pain interference, social and school/work functioning in youth with spinal cord injury: a mediation analysis. J Spinal Cord Med. 2022;1-7:1-7.
El Hoss S, Cochet S, Godard A, et al. Fetal hemoglobin rescues ineffective erythropoiesis in sickle cell disease. Haematologica. 2021;106(10):2707-2719.
Steinberg MH. Targeting fetal hemoglobin expression to treat beta hemoglobinopathies. Expert Opin Ther Targets. 2022;26(4):347-359.
Ribeil JA, Arlet JB, Dussiot M, Moura IC, Courtois G, Hermine O. Ineffective erythropoiesis in beta-thalassemia. ScientificWorldJournal. 2013;2013:394295.
Origa R. Beta-thalassemia. In: Adam MP, Mirzaa GM, Pagon RA, et al., eds. GeneReviews (R). University of Washington; 1993.
Haworth KG, Kiem HP. Next-generation conditioning for bone marrow transplantation: paving the way for CAR-T cell-based conditioning. Mol Ther. 2018;26(5):1167-1168.

Auteurs

Selim Corbacioglu (S)

University of Regensburg, Regensburg, Germany.

Haydar Frangoul (H)

Sarah Cannon Research Institute and the Children's Hospital at TriStar Centennial, Nashville, Tennessee, USA.

Franco Locatelli (F)

IRCCS, Ospedale Pediatrico Bambino, Gesù Rome, Catholic University of the Sacred Heart, Rome, Italy.

William Hobbs (W)

Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA.

Mark Walters (M)

Department of Pediatrics, UCSF Benioff Children's Hospital Oakland, Oakland, California, USA.

Classifications MeSH