Skip to main content

MINI REVIEW article

Front. Hematol., 04 January 2023
Sec. Hematopoiesis and Stem Cells
This article is part of the Research Topic Editors' Showcase: Hematopoiesis and Stem Cells View all 6 articles

Innovative and Needs-led research on β-thalassemia treatment methods

  • Department of Genetics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania

Beta-thalassemia is a well-known blood genetic disorder inherited in an autosomal recessive manner. Beta-thalassemia is found everywhere in the world as a rare, relatively rare, or common disease depending on the ethnic population. Affected individuals have chronic anemia associated with delayed growth, pale skin, weakness, fatigue, and more serious complications resulting in early death. Those with the severe form need frequent lifelong transfusions and depend on blood donations to survive. This literature mini-review highlights the healthcare needs that are not optimally met by people living with beta-thalassemia. The needs-led research can help to improve clinical outcomes through more appropriate management of the disease, increase provider satisfaction, and reduce the cost of care.

1 Introduction

β-thalassemia (BT) is a quantitative disorder of β -globin synthesis characterized by the absence (β 0) or reduced (β +) synthesis of the normal β -globin chains of hemoglobin A. Even though β-thalassemia turned out to be an ancient disease (1), and the life expectancy of patients with severe thalassemia has increased alongside therapeutic progresses over time (2), it still imposes an economic burden on the communities, and healthcare systems worldwide. Moreover, the therapeutic protocols can physically and psychologically impact patients and caregivers. Patients with β-thalassemia have a lower health-related quality of life than the general population, especially for those who require lifelong regular blood cell transfusions and have more disease complications and chelation-related side effects (3). Hematopoietic stem cell transplantation from healthy donors is a cure for β-thalassemia, however it is not done very often due to the significant risk involved and its high cost (usually not covered by the medical insurance). Moreover, BT is included as a disability in the Rights of Persons with Disabilities Act, 2016 that promotes “the access to the health services they need, when and where they need them, without suffering financial hardship” (4). More treatment options would help people with thalassemia to make the appropriate choice and develop the desired clinical improvements.

2 Information about BT in different populations

BT is found everywhere in the world being a rare, relatively rare, or common disease, depending on the ethnic population. It originates in endemic populations from malaria regions, such as the Mediterranean basin, the Middle East, South-East Africa, and Sub-Saharan Africa (5). The selective geographic distribution of BT is thought to be because of red blood cell morphological abnormalities that may play a protective role against malaria disease (6). At that point, the number of cases of BT occurring in a certain population was low, such as in North-West Europe, and high in people of Mediterranean, African, Middle Eastern, Asian Indian, Chinese, and Southeast Asian countries. The descents of people from endemic areas carry a greater risk of developing the disease. There are three main forms of BT in the population based on the degrees of phenotypic severity and blood transfusion requirement: β-thalassemia major (severe microcytic and hypochromic anaemia and clinical course requiring regular, lifelong transfusions), β-thalassemia intermedia (moderate symptoms of anaemia, occasional transfusions required) and β-thalassemia minor or β-thalassemia carrier (asymptomatic or mild symptoms of anaemia) (7).

Nowadays, BT becomes a global health problem through the migration of people from endemic areas throughout Europe, the Americas, and Australia, leading to the global distribution of the disease (8). In the last 50 years, the pattern of BT epidemiology has changed. In addition to migration, implementation of β-thalassemia prevention and screening programs, or survival rate due to current therapeutic approaches, decreased the prevalence in endemic populations (1, 8, 9). So, there is a significant variation of prevalence/incidence among different countries, but according to Orphanet, the annual incidence, at birth, of symptomatic BT is approximately one in every 100,000 individuals in the general population (10). The estimates based on the group of people with different types of BT (depending on the severity of symptoms) show that around 1.5% of the global population are carriers, approximately 60000 children are born with β-thalassemia annually, and about 63% have a longer life expectancy surviving through the age of 50 (11, 12).

3 What are the functional changes that accompany β-thalassemia?

The β-thalassemia is caused by more than 350 pathogenic variants of b-gene involved in the defective β-globin chains synthesis (13). The result is either reduced or absent synthesis of β-globin chains but the unaffected chains, such as a-globin chains, continue to be synthesized at relatively normal levels. As a result, the HbA (a2b2) formation does not work properly. Excess free a-chains precipitate in the cytoplasm of erythroid precursors. The molecular aggregates are toxic and highly insoluble, and form inclusions in nucleated erythroid precursors in the bone marrow. These inclusion bodies cause accelerated red blood cell destruction by apoptosis and intramedullary haemolysis leading to ineffective erythropoiesis responsible for anaemia. Along with haematological features, abnormal iron metabolism and bone abnormalities are present. Both patients with dependent and non-dependent transfusion experience an iron overload because of an inappropriate increase in intestinal iron absorption. Excess iron deposited in the heart, pancreas, liver, and other organs damages tissues and disfunctions. Symptomatic patients exhibit erythroid hyperplasia, bone marrow expansion, and extramedullary haematopoiesis. Under these conditions, the bones show marked decrease in mineral density. Skull and face bone deformities, cortical thinning, and pathological fractures of long bones are noted (1420).

Considering all the actual burdens, there is no other option than seeking new available, accesible, and affordable therapies to improve the BT patients’ quality of life. Therefore, what treatments bring hope in the patients’ community? Is any new therapy that can treat Beta-thalassemia without side effects?

4 Novel therapeutic methods

4.1 Pharmacological approach

4.1.1 Luspatercept

A promising therapy recently approved by the FDA (in 2019) and EMA (in 2020) for the treatment of β-thalassemia is luspatercept (ACE-536) (21). It acts by inhibiting the Smad2/3 signaling pathway, which promotes the attenuation of ineffective erythropoiesis. It shows as well an improvement of iron balance parameters (2225). It is currently approved only for the treatment of transfusion-dependent BT, but there is hope it could also be used for non-transfusion-dependent BT, according to ongoing clinical trials (26). However, an important disadvantage of using luspatercept today is the very high cost of the drug, as it is estimated that the total annual amount that a patient could pay for luspatercept reaches up to $170,000 (27).

4.1.2 Hydroxyurea

Of all the drug therapies tested or used in β thalassemia, hydroxyurea certainly remains a mystery, but equally a challenge for the medical world to unravel the mysteries behind this substance. In addition to its antineoplastic effect that recommends it for the treatment of many types of cancer, it is currently approved for the SCD patients, but is also used in some cases of β thalassemia. Its importance has increased since the start of the COVID-19 pandemic as a result of the difficulties encountered by medical systems related to the adequate implementation of transfusion therapies (28). The main mechanism of action justifies its cytotoxic activity, as it blocks ribonucleoside diphosphate reductase (rNDP), an important enzyme involved in DNA synthesis. Therefore, it does not allow cells to go beyond the S phase of the cell cycle. It is also an HbF-inducer by interfering with various transcription factors (represses BCL11A and GATA1, stimulates GATA2), but also by modulating some epigenetic processes (21, 28). Recently, the results of the first placebo-controlled, double-blind, randomized clinical trial which aimed to study the effectiveness and safety of oral administration of hydroxyurea, for 6 months, in patients with TDT, were published (29). The primary outcome (a significant improvement in blood transfusion volume) was not met. However, 89% of participants who received the drug experienced increases in HbF during treatment, and 79% experienced decreases in serum sTfR (soluble transferrin receptor) levels, which is associated with improvements in hematopoiesis. All these data regarding the mentioned drugs support the idea that they can play the role of adjunctive therapies that bring an additional therapeutic benefit in β thalassemia. Therefore, more trials using therapeutic combinations should be organized.

4.2 Gene therapy

One of the researchers’ priorities when treating β-thalassemia is to increase the γ chain production, which binds the α chains, leading to the production of HbF levels. This results in reduced precipitation of free α chains and mitigation of iron overload and cellular oxidative stress (30). Thus, it combates both the hemolytic syndrome and the dyserythropoiesis. Gene editing came in as a solution to increase the levels of γ-globin, by manipulating the genome of the hematopoietic stem and progenitor cells (HSPCs) from patients (31). In this regard, the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) system is continuously tested.

Nonetheless, the last decade has witnessed the development of treatments based on lentiviral vectors, which rely on the gene transfer of healthy HBB genes (32, 33).

4.2.1 Genome editing-based treatments

Researchers turned their attention to the genetic manipulation of the BCL11A gene enhancer in red blood cells. BCL11A is considered to be the most important regulator of HBG expression. By deleting its enhancer through CRISPR/Cas9, the transcription of BCL11A is suppressed, which allows the resumption of HBG activity and HbF synthesis (34, 35).

A phase 1/2/3 study (CLIMB-111), supported by Vertex and CRISPR Therapeutics, developed a cell therapy product called CTX001 using CRISPR/Cas9 mechanisms in CD34+ HSPCs (NCT03655678). Researchers deleted the BCL11A gene enhancer, thus increasing the levels of γ-globin and restoring the production of fetal hemoglobin, when transplanted in the patients’ blood (36). This study offers promising preliminary data: remarkable and sustained increases in total hemoglobin (new values ​​being between 8.9 and 16.9 g/dL) and HbF (between 67.3% and 99.6%) for all 15 participants at 4-26 months after infusion. This progress allowed patients to be categorized as transfusion-independent (37).

Analogous to CLIMB-111, a recently initiated Phase 1/2 study (NCT04211480) also uses the CRISPR/Cas9 editing tool in HSPCs, targeting the BCL11A enhancer loci. It is dedicated exclusively to pediatric patients aged between 5 and 15 years and the partial results are encouraging as well (38).

Gene editing techniques can be used to modulate the α chain level, too. There are 2 ways to apply these tools in order to prevent the excess of free α chains. The first of them damages directly the α gene. Using CRISPR/Cas9 technology, its deletion can be induced [640], genetically “simulating” the appearance of α thalassemia. The main reason why this “association” (α and β thalassemia) is favoured is represented by the clinical and molecular observations made during the last decades that show the patients with co-inheritance of α and β thalassemia manifest a milder form of the disease (39), precisely because of an α/β ratio less unbalanced and with lower levels of cellular oxidative stress compared to β thalassemia patients. As a consequence of these findings, an in vitro study applying the theory was conducted and revealed remarkable results due to a significant reduction in the precipitation of free α chains (40).

The second way to control the α gene expression through gene editing techniques involves affecting gene enhancers. It is considered that gene activity is mediated by 4 enhancers (MCS-R1 to R4), but among them MCS-R2 is the key element as researches show (39, 41, 42). According to an in vitro study, the deletion of MCS-R2 with CRISPR/Cas9 led to significant reductions in the α chains’ levels: 60% in the case of monoallelic mutation and 90% if the deletion targeted both alleles of the enhancer (41, 42). These results demonstrate the huge potential that gene therapy techniques have in the context of treating β thalassemia by modifying HBA activity and suggest the need for further investigations to study the concrete efficacy of these techniques.

4.2.2 Gene transfer-based treatments

August 2022 saw the first FDA approval of a genetic treatment method for transfusion-dependent β-thalassemia, Zynteglo (betibeglogene autotemcel) (43). Such treatment represents an innovation for patients suffering from BT, as it is a one-time therapy with an efficacy rate of over 86%, including pediatric patients. It uses the patient’s own CD34+ bone marrow cells, modifying them genetically, via the lentiviral vector BB305, in order to encode the β-globin (βA-T87Q) gene, thus increasing the production of β-globin (44). Moreover, Zynteglo proves itself as a more favorable treatment method than the standard of care, from an economical perspective, as compared to lifelong treatment (45). BB305 is a lentiviral vector constructed after the structure of Human Immunodeficiency Virus-1 (HIV-1), but lacking in HIV-1 protein-coding genes, which are replaced by the β-globin ones. It does, however, maintain the structures needed in the processes of viral genome packaging and cellular transductions (46). The principle behind this treatment has been under investigation for the last 12 years, thus it is expected that more treatment options of this kind will emerge, such as the DEST LVV and others (4749).

5 Conclusion

Considering the current needs of the patients, the new therapies should increase the quality of life by reducing the transfusion burden, disease complications and chelation-related side effects. Moreover, the outcomes of these newly-approved treatments should encourage the patients’ community to support further research.

Author contributions

These authors contributed equally to this work. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

BT, β-thalassemia; FDA, Food and Drug Administration; EMA, European Medicines Agency; HbF, Hemoglobin F; HBB, Hemoglobin Subunit Beta; HBG, Hemoglobin Subunit Gamma; CRISPR, clustered regularly interspaced short palindromic repeats; Cas9, CRISPR associated protein 9; HSPC, hematopoietic stem and progenitor cells; HIV-1, Human Immunodeficiency Virus-1; LVV, lentiviral vector.

References

1. De Sanctis V, Kattamis C, Canatan D, Soliman AT, Elsedfy H, Karimi M, et al. β-thalassemia distribution in the old world: An ancient disease seen from a historical standpoint. Mediterr J Hematol Infect Dis (2017) 9(1):e2017018. doi: 10.4084/MJHID.2017.018

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Esmaeilzadeh F, Azarkeivan A, Emamgholipour S, Akbari Sari A, Yaseri M, Ahmadi B, et al. Economic burden of thalassemia major in Iran, 2015. J. Res. Health Sci. (2016) 16(3):111–5. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7191027/.

PubMed Abstract | Google Scholar

3. Sobota A, Yamashita R, Xu Y, Trachtenberg F, Kohlbry P, Kleinert DA, 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):92–5. doi: 10.1002/ajh.21896

PubMed Abstract | CrossRef Full Text | Google Scholar

4. The Rights of Persons with Disabilities (RPwD) Act. (2016). Available at: https://disabilityaffairs.gov.in/content/page/acts.php (Accessed September 30, 2022).

Google Scholar

5. Kountouris P, Kousiappa I, Papasavva T, Christopoulos G, Pavlou E, Petrou M, et al. The molecular spectrum and distribution of haemoglobinopathies in Cyprus: A 20-year retrospective study. Sc. Rep (2016) 6:26371. doi: 10.1038/srep26371

CrossRef Full Text | Google Scholar

6. Highlights of ASH in the Mediterranean basin. Available at: https://www.ashclinicalnews.org/features/blood-beyond-borders-malaria-thalassemia-mediterranean-basin (Accessed August 23, 2022).

Google Scholar

7. Cao A, Galanello R. Beta-thalassemia. Genet Med (2010) 12:61–76. doi: 10.1097/GIM.0b013e3181cd68ed

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Modell B. EC Concerted action on developing patient registers as a tool for improving service delivery for haemoglobin disorders. In: Fracchia GN, Theophilatou M, editors. Health services research. Amsterdam: IOS Press (1993).

Google Scholar

9. Kattamis A, Forni GL, Aydinok Y, Viprakasit V. Changing patterns in the epidemiology of β-thalassemia. Eur JHaematol (2020) 105:692–703. doi: 10.1111/ejh.13512

CrossRef Full Text | Google Scholar

10. Orphanet β -thalassemia. Available at: https://www.orpha.net/consor/cgi-bin (Accessed August 25, 2022).

Google Scholar

11. Galanello R, Origa R. β -thalassemia. Orphanet J Rare Dis (2010) 5:11. doi: 10.1186/1750-1172-5-11

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Vogiatzi MG, Macklin EA, Trachtenberg FL, Fung EB, Cheung AM, Vichinsky E, et al. Differences in the prevalence of growth, endocrine and vitamin d abnormalities among the various thalassaemia syndromes in north America. Br J Haematol (2009) 146(5):546–56. doi: 10.1111/j.1365-2141.2009.07793.x

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kountouris P, Lederer CW, Fanis P, Feleki X, Old J, Kleanthous M. IthaGenes: An interactive database for haemoglobin variations and epidemiology. PloS One (2014) 9:e103020. doi: 10.1371/journal.pone.0103020

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Kautz L, Jung G, Valore EV, Rivella S, Nemeth E, Ganz T. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat Genet (2014) 46:678–84. doi: 10.1038/ng.2996

PubMed Abstract | CrossRef Full Text | Google Scholar

15. 2021 Guidelines for the management of transfusion dependent thalassaemia (TDT). Available at: https://www.tasca.org.au/wp-content/uploads/2021/07/GUIDELINE-4th-SINGLE-PAGE.pdf (Accessed November 17, 2022).

Google Scholar

16. Schrier SL. Pathophysiology of thalassemia. Curr Opin Hematol (2002) 9:123–6. doi: 10.1097/00062752-200203000-00007

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Cappellini MD, Cohen A, Porter J, Taher A, Viprakasit V. Guidelines for the management of transfusion dependent thalassaemia (TDT). Nicosia. Thalassaemia Int Fed (2014):22–25.

Google Scholar

18. Taher A, Vichinsky E, Musallam K, Cappellini MD, Viprakasit V. Guidelines for the management of non transfusion dependent thalassaemia (NTDT). Nicosia: Thalassaemia Int Fed (2013):35–50. doi: 10.1182/blood.2021011045

CrossRef Full Text | Google Scholar

19. Cazzola M. Ineffective erythropoiesis and its treatment. Blood (2022) 139(16):2460–70. doi: 10.1182/blood.2021011045

CrossRef Full Text | Google Scholar

20. Sanchez-Villalobos M, Blanquer M, Moraleda JM, Salido EJ, Perez-Oliva AB. New insights into pathophysiology of β-thalassemia. Front Med (2022) 9:880752. doi: 10.3389/fmed.2022.880752

CrossRef Full Text | Google Scholar

21. Longo F, Piolatto A, Ferrero GB, Piga A. Ineffective erythropoiesis in β-thalassaemia: Key steps and therapeutic options by drugs. Int J Mol Sci (2021) 22(13):7229. doi: 10.3390/ijms22137229

PubMed Abstract | CrossRef Full Text | Google Scholar

22. 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(2):113–22. doi: 10.1080/14728214.2020.1752180

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Cappellini MD, Viprakasit V, Taher AT, Georgiev P, Kuo KHM, Coates T, et al. A phase 3 trial of luspatercept in patients with transfusion-dependent β-thalassemia. N Engl J Med (2020) 382(13):1219–31. doi: 10.1056/NEJMoa1910182

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Cappellini MD. Taher AT.(2021) the use of luspatercept for thalassemia in adults. Blood Adv (2021) 5(1):326–33. doi: 10.1182/bloodadvances.2020002725

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Taher AT, Bou-Fakhredin R, Kattamis A, Viprakasit V. Cappellini MD.(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 (2021) 14(10):897–909. doi: 10.1080/17474086.2021.1977116

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Taher AT, Cappellini MD, Kattamis A, Voskaridou E, Perrotta S, Piga AG, 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.Aug (2022) 22:S2352–3026(22)00208-3. doi: 10.1016/S2352-3026(22)00208-3

CrossRef Full Text | Google Scholar

27. FDA approves first drug for anemia tied to rare blood disorder. Available at: https://www.biopharmadive.com/news/celgene-win-fda-approval-luspatercept-reblozyl-beta-thalassemia/566968/ (Accessed October 23, 2022).

Google Scholar

28. Yasara N, Premawardhena A, Mettananda S. A comprehensive review of hydroxyurea for β-haemoglobinopathies: the role revisited during COVID-19 pandemic. Orphanet J Rare Dis (2021) 16:114. doi: 10.1186/s13023-021-01757-w

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Yasara N, Wickramarathne N, Mettananda C, Silva I, Hameed N, Attanayaka K, et al. A randomised double-blind placebo-controlled clinical trial of oral hydroxyurea for transfusion-dependent β-thalassaemia. Sci Rep (2022) 12:2752. doi: 10.1038/s41598-022-06774-8

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Fibach E, Dana M. Oxidative stress in β-thalassemia. Mol Diagn Ther (2019) 23(2):245–61. doi: 10.1007/s40291-018-0373-5

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Magrin E, Miccio A, Cavazzana M. Lentiviral and genome editing strategies for the treatment of β-hemoglobinopathies. Blood (2019) 134:1203–13. doi: 10.1182/blood.2019000949

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Ernst MPT, Broeders M, Herrero-Hernandez P, Oussoren E, van der Ploeg AT, Pijnappel W. Ready for repair? gene editing enters the clinic for the treatment of human disease. Molec Ther Methods Clin Dev (2020) 18:532–57. doi: 10.1016/j.omtm.2020.06.02

CrossRef Full Text | Google Scholar

33. Cavazzana M, Bushman FD, Miccio A, André-Schmutz I, Six E. Gene therapy targeting haematopoietic stem cells for inherited diseases: Progress and challenges. Nat Rev Drug Discov (2019) 18:447–62. doi: 10.1038/s41573-019-0020-9

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Adelvand P, Hamid M, Sardari S. The intrinsic genetic and epigenetic regulator factors as therapeutic targets, and the effect on fetal globin gene expression. Expert Rev Hematol (2018) 11(1):71–81. doi: 10.1080/17474086.2018.1406795

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Lohani N, Bhargava N, Munshi A, Ramalingam S. Pharmacological and molecular approaches for the treatment of β-hemoglobin disorders. J Cell Physiol (2018) 233(6):4563–77. doi: 10.1002/jcp.26292

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Frangoul H, Altshuler D, Cappellini MD, Chen Y-S, Domm J, Eustace BK, et al. CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. New Engl J Med. (2020) 384(3):252–60. doi: 10.1056/NEJMoa2031054

CrossRef Full Text | Google Scholar

37. Vertex and CRISPR Therapeutics Present New Data in 22 Patients With Greater Than 3 Months Follow-Up Post-Treatment With Investigational CRISPR/Cas9 Gene-Editing Therapy, CTX001TM at European Hematology Association Annual Meeting. Available at: http://ir.crisprtx.com/news-releases/news-release-details/vertex-and-crispr-therapeutics-present-new-data-22-patients (Accessed October 3, 2022).

Google Scholar

38. Fu B, Liao J, Chen S, Li W, Wang Q, Hu J, et al. CRISPR–Cas9-mediated gene editing of the BCL11A enhancer for pediatric β00 transfusion-dependent β-thalassemia. Nat Med (2022) 28:1573–80. doi: 10.1038/s41591-022-01906-z

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Mettananda S, Gibbons RJ, Higgs DR. α-globin as a molecular target in the treatment of β-thalassemia. Blood (2015) 125(24):3694–701. doi: 10.1182/blood-2015-03-633594

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Pavani G, Fabiano A, Laurent M, Amor F, Cantelli E, Chalumeau A, et al. Correction of β-thalassemia by CRISPR/Cas9 editing of the α-globin locus in human hematopoietic stem cells. Blood Adv (2021) 5(5):1137–53. doi: 10.1182/bloodadvances.2020001996

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Mettananda S. Genetic and epigenetic therapies for β-thalassaemia by altering the expression of α-globin gene. Front Genome Ed (2021) 3:752278. doi: 10.3389/fgeed.2021.752278

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Mettananda S, Fisher CA, Hay D, Badat M, Quek L, Clark K, et al. Editing an α-globin enhancer in primary human hematopoietic stem cells as a treatment for β-thalassemia. Nat Commun (2017) 8(1):424. doi: 10.1038/s41467-017-00479-7

PubMed Abstract | CrossRef Full Text | Google Scholar

43. FDA Approves first cell-based gene therapy to treat adult and pediatric patients with β -thalassemia who require regular blood transfusions (Accessed October 1, 2022).

Google Scholar

44. Locatelli F, Thompson AA, Kwiatkowski JL, Porter JB, Thrasher AJ, Hongeng S, et al. Betibeglogene autotemcel gene therapy for non-β0/β0 genotype β-thalassemia. N Engl J Med (2022) 386(5):415–27. doi: 10.1056/NEJMoa2113206

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Kansal AR, Reifsnider OS, Brand SB, Hawkins N, Coughlan A, Li S, et al. Economic evaluation of betibeglogene autotemcel (Beti-cel) gene addition therapy in transfusion-dependent β-thalassemia. J Mark Access Health Policy (2021) 9(1):1922028. doi: 10.1080/20016689.2021.1922028

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Hongeng S, Anurathapan U, Songdej D, Phuphuakrat A, Jongrak K, Parsons G, et al. Wild-type HIV infection after treatment with lentiviral gene therapy for β-thalassemia. Blood Adv (2021) 5(13):2701–6. doi: 10.1182/bloodadvances.2020003680

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Breda L, Kleinert DA, Casu C, Casula L, Cartegni L, Fibach E, et al. A preclinical approach for gene therapy of β -thalassemia. Ann N Y Acad Sci (2010) 1202:134–40. doi: 10.1111/j.1749-6632.2010.05594.x

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Abbasalipour M, Khosravi MA, Zeinali S, Khanahmad H, Azadmanesh K, Karimipoor M. Lentiviral vector containing β -globin gene for β thalassemia gene therapy. Gene Rep (2022) 27:101615. doi: 10.1016/j.genrep.2022.101615

CrossRef Full Text | Google Scholar

49. Nualkaew T, Sii-Felice K, Giorgi M, McColl B, Gouzil J, Glaser A, et al. Coordinated β-globin expression and α2-globin reduction in a multiplex lentiviral gene therapy vector for β-thalassemia. Mol Ther (2021) 29(9):2841–53. doi: 10.1016/j.ymthe.2021.04.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: beta-thalassemia, unmet needs, needs-led research, innovative therapy, pattern of epidemiology

Citation: Dan M-O, Gutu B-I, Severin E and Tanase V-G (2023) Innovative and Needs-led research on β-thalassemia treatment methods. Front. Hematol. 1:1085952. doi: 10.3389/frhem.2022.1085952

Received: 31 October 2022; Accepted: 29 November 2022;
Published: 04 January 2023.

Edited by:

Maegan Capitano, Indiana University Bloomington, United States

Reviewed by:

Sachith Mettananda, University of Kelaniya, Sri Lanka

Copyright © 2023 Dan, Gutu, Severin and Tanase. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Emilia Severin, emiliaseverin11@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.