Skip to main content
Blood Transfusion logoLink to Blood Transfusion
. 2021 Oct 21;20(1):78–88. doi: 10.2450/2021.0265-21

Advancing the care of β-thalassaemia patients with novel therapies

Rayan Bou-Fakhredin 1, Irene Motta 2, Maria Domenica Cappellini 1,2,
PMCID: PMC8796844  PMID: 34694225

Abstract

The β-thalassaemias are a group of inherited disorders of haemoglobin synthesis characterised by chronic anaemia of varying severity. Currently available conventional therapies in thalassaemia have many challenges and limitations. A better understanding of the pathology of β-thalassaemia has led to the development of new treatment options, most of which are currently in clinical trials. These could have the potential of reducing red blood cell transfusion burden, raising haemoglobin levels, and improving patients’ overall quality of life. In this review, we will provide an overview of the novel therapeutic approaches that are currently under development to advance the care of β-thalassaemia patients.

Keywords: β-thalassaemia, clinical trial, novel therapies, quality of life, transfusion burden

INTRODUCTION

The thalassaemias constitute one of the most common inherited monogenic diseases in the world. They are characterised by autosomal recessive inherited defects in the production of haemoglobin (Hb). Today, thalassaemia patients are commonly categorised according to their need for blood transfusion. Transfusion-dependent thalassaemia (TDT) patients frequently present with severe anaemia in early childhood and their survival depends on lifelong treatment with transfusions. Non-transfusion-dependent thalassaemia (NTDT) patients commonly present with mild to moderate anaemia in a later stage of childhood, or even in adulthood, and may require occasional or short-course transfusions for the management or prevention of some disease manifestations1,2. Over the last decade, the improved management of symptoms and associated comorbidities for β-thalassaemia patients via regular red blood cell (RBC) transfusions, iron chelation therapy (ICT), and supporting therapies has allowed more patients to remain healthy and move into adulthood3,4. However, unmet needs in terms of safety, efficacy and adherence to conventional therapies have posed many challenges, limitations and an additional burden not only to the patients themselves, but also to their families5. Moreover, advances in the understanding of the disease itself has enabled clinicians and researchers to move forward towards the development of novel therapeutic modalities6,7. These can be classified into three major categories based on their efforts to address different features of the underlying pathophysiology: correction of the α/β-globin chain imbalance, targeting ineffective erythropoiesis, and targeting iron dysregulation7 (Figure 1). In this review, we will provide an overview of the novel therapeutic approaches that are currently in development to advance the care of β-thalassaemia patients.

Figure 1.

Figure 1

Summary of novel targets and agents in β-thalassaemia

CORRECTION OF THE α/β-GLOBIN CHAIN IMBALANCE

Allogeneic haematopoietic stem cell transplantation

Allogeneic haematopoietic stem cell transplantation (HSCT) from a human leukocyte antigen (HLA)-matched sibling donor (MSD), performed in childhood, has been the gold standard for TDT patients for decades8,9. Unfortunately, only a minority of patients have siblings as potential donors. Therefore, fully matched unrelated donors (MUD) have been the second choice for cure, with similar results in terms of overall survival (OS). Many key studies on HSCT for TDT patients using MUDs have been conducted, with efforts focused on better engraftment, prevention of graft rejection and toxicity, and targeting conditioning regimens1015. Results from a recent international retrospective study of 1,110 TDT patients, aged ≤25 years, who received grafts from HLA-matched related (61%), HLA-mismatched related (7%), HLA-matched unrelated (23%), and HLA-mismatched unrelated (9%) donors between 2000 and 2016 showed that the highest OS was seen in patients aged ≤6 years. There was no significant difference in OS rates between the groups transplanted from MSDs or MUDs (89 vs 87% and 86 vs 82%, respectively)16.

On the other hand, haploidentical transplantation has been performed more frequently during the last decade, with promising results1722. Current practice mainly includes a preconditioning phase and then either selective depletion of T-cell receptor aβ+/CD19+ graft lymphocytes or the use of post-transplantation cyclophosphamide (post-Cy). The basic advantage of haploidentical donor HSCT is immediate donor accessibility, with no ethnic or racial restrictions. This offers a viable solution for thalassemia patients belonging to ethnic minorities8.

However, several aspects limit allogeneic HSCT as a curative therapy. First, MSDs are available to only 20–30% of patients, depending on nationality and ethnic groups. Second, HSCT using alternative donors, mainly MUDs, haploidentical donors and cord blood transplants, have not yielded results that can equal those of allogeneic MSDs2325. Third, the probability of real-life, long-term complications such as hypogonadism and thyroid dysfunction after HSCT needs to be considered2628. Further data concerning secondary cancer are required29. Moreover several new conditioning regimens are now being evaluated in clinical trials as an effort to improve HSCT outcomes8,9.

Gene therapy

Gene therapy is a novel and potentially curative treatment strategy for TDT patients that has been designed to correct the underlying α/β-globin chain ratio, thus improving the production of functional Hb, the erythropoiesis, and the chronic anaemia. After isolating haematopoietic stem and progenitor cells (HSPCs), exogenous β-globin genes are incorporated into the host-cell genome using a self-inactivating lentiviral vector. After full or partial myeloablative busulfan conditioning, these genetically modified autologous HSPCs are returned to the patient where they replicate and repopulate in the blood compartment and facilitate normal Hb synthesis30,31. In order for gene therapy to be successful in β-thalassaemia, certain requirements have to be met: high-efficiency haematopoietic stem cell (HSC) engraftment and gene transfer, high expression of the β/γ-globin gene and appropriate expression, with minimal to no risk of insertional mutagenesis. The use of gene therapy technology has been proven to be effective and curative in many animal models of β-thalassaemia3235 and in proof-of-principle studies in an adult patient with TDT36,37.

The outcomes of two phase I/II studies (HGB-204 [NCT01745120] and HGB-205 [NCT02151526])38 conducted on 22 TDT patients (12–35 years of age) who were reinfused with cells transduced ex vivo with the LentiGlobin BB305 vector led to the conditional approval in Europe of the gene therapy product betibeglogene autotemcel (Zynteglo, bluebird bio Inc., Cambridge, MA, USA) in June 2019. In these studies, and at a median of 26 months after receiving gene therapy, 12 of the 13 patients who had a non-β00 genotype had achieved transfusion independence, and biological markers indicated that ineffective erythropoiesis had been corrected. In 9 patients with β0β0 genotype the median annualised transfusion volume was decreased by 73%, and 3 of these patients discontinued RBC transfusions. The adverse events (AEs) attributed to the LentiGlobin BB305 vector were reported and were found to be similar to those associated with autologous HSCT, and no clonal dominance related to lentiviral vector integration was observed38.

The HGB-207 study (Northstar-2, NCT02906202) and the HGB-212 study (Northstar-3, NCT03207009) are currently ongoing phase III studies using the LentiGlobin BB305 vector for TDT subjects with non-β0β0 and β0β0 genotypes. Interim data from these studies projected after a median follow-up of 24.3-months showed that 30/34 (88.2%) patients became transfusion independent (6/7 [85.7%] β00 and 24/27 [88.9%] non-β00) maintained for a median of 20.6 months)39. AEs were similar to those seen following myeloablative conditioning with busulfan. There were no deaths and no evidence of clonal dominance or insertional oncogenesis in these studies39. Following these initial positive results, the phase III studies were expanded to also include paediatric patients. Interim results in the paediatric population showed that 27 paediatric patients achieved a similar rate of transfusion independence and showed a similar safety profile to that of adults40.

Most recently, preliminary data from 44 patients enrolled in the ongoing LTF-303 study (NCT02633943) with a median follow-up of 45.6 months (range: 22.9–76.4) showed that transfusion independence was achieved and sustained in 15/22 (68.2%) patients from the phase I/II studies and in 20/22 (90.9%) of patients treated in the phase III studies. Weighted average Hb during transfusion independence was 10.3 g/dL and 11.8 g/dL in patients in phase I/II and phase III trials, respectively. This was maintained for over three years of follow-up. Moreover, most patients who achieved transfusion independence had a decrease in liver iron concentration (LIC) approaching normal levels. While some serious AEs during LTF-303 study were observed, no deaths, replication-competent lentivirus, or insertional oncogenesis were reported41.

A decision by bluebird bio to temporarily suspend all clinical trials and marketing of betibeglogene autotemcel was made on 16 February 2021, following two reports of acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS) in sickle cell patients recruited on the HGB-206 trial42. The MDS diagnosis was later revised to transfusion-dependent anaemia and the AML case was reported to be unlikely related to treatment43. While the conditional approval of betibeglogene autotemcel was given by the European Medicines Agency (EMA) in 2019, bluebird bio announced on August 9, 2021 that they will discontinue their operations in Europe due to payor challenges44.

Other gene insertion approaches and vectors have also been evaluated in animal models and validated for clinical investigation. For example, 3 adult and 6 paediatric TDT patients with β0 or severe β+ mutations were enrolled and treated in a phase I/II trial (TIGET-BTHAL, NCT02453477) with an intrabone administration of HSCs transduced with the lentiviral vector GLOBE, which removes the HS4 locus control region element. Transfusion reduction was seen in 3 adults and complete independence was achieved in 3/4 evaluated children45. Data from the phase I TNS9 clinical trial (NCT01639690), which was conducted on 4 adult TDT patients with β0+ and β 00 genotypes treated with autologous CD34+ HSPCs transduced with the TNS9.3.55 lentiviral vector, showed durable and stable gene marking, no evidence of clonal dominance, and no toxicity profile higher than grade 3 in all patients. One patient experienced a significant decrease in transfusion requirements that lasted for more than five years46,47. Ongoing and future studies in gene therapy will focus on improving transduction efficiency by optimising protocols preserving biological HSC features and unravelling biological key factors for prediction of favourable outcomes.

Gene editing

Newer approaches have been under development to correct genetic mutations at the DNA level of the cell or to disrupt specific DNA sequences in the genome. BCL11A, which is located on chromosome 2, has been shown to be a potential and promising target for genome editing30. It acts as a key regulator of the foetal-to-adult Hb switch and HbF silencing48,49. Genetic variation in the expression of BCL11A and persistence of HbF production was shown to reduce clinical severity in β-thalassaemia50,51. Many in vitro studies have shown that knock-out gene-editing technology could permanently produce foetal Hb (HbF) in adults with thalassaemia5254. Knock-down studies of BCL11A using RNA interference (RNAi) showed it to be an effective method to silence the γ-globin gene49,55,56. A new approach to induce HbF by silencing BCL11A involves the use of an erythroid-specific promoter to drive a microRNA-adapted short hairpin (shRNA)53,57. Other gene-editing approaches to inhibit BCL11A have been described and are currently in pre-clinical and clinical development; these include clustered, regularly interspaced, palindromic repeats (CRISPR) in association with Cas9 (CRISPR-Cas9), transcription activator-like effector nucleases (TALENS), and zinc finger nucleases (ZFNs)5861.

The first major product under study is CTX011 (CRISPR-Cas9), which is produced using ex vivo editing of the erythroid enhancer region of the BCL11A in CD34+ HSPCs and reduces erythroid-specific expression of BCL11A. Infusion of CTX0011 was shown to increase HbF levels in erythroid cells in vivo. CLIMB THAL-111 (NCT03655678) is a phase I/II trial evaluating the safety and efficacy of CTX001 in 45 patients with TDT (age 12–35 years). Interim results from 10 TDT patients who have been followed up for 3.8–21.5 months, and who received CTX001 infusion, showed that engraftment of neutrophils and platelets was achieved at medians of 30 and 38.5 days, respectively; clinically meaningful levels of total Hb, HbF, and F-cell pancellularity were observed early and were maintained over time across all 10 patients (mean total Hb at baseline 10.6 g/dL including 0.2 g/dL HbF, mean total Hb at 21 months 13.3 g/dL including 12.5 g/dL HbF). Patients also stopped transfusions within two months of CTX001 infusion and were transfusion-free up to 23.8 months of follow-up. The safety profile of CTX0011 was also generally consistent with myeloablative conditioning and autologous HSCT62,63.

The use of ZFN technology is also currently being studied in a clinical trial. THALES (NCT03432364) is a phase I/II clinical trial evaluating the safety, tolerability and efficacy of ST-400 (ZFN) in reducing transfusion requirement (frequency and volume) in 6 adults (18–40 years) with TDT. Preliminary results from 2 patients showed rapid haematopoietic reconstitution following myeloablative conditioning, and elevated HbF levels64. These data are preliminary, and additional patients and longer follow-up will be required to better understand the safety and efficacy of this therapy.

Foetal-haemoglobin-inducing agents

Considerable efforts are also being made to stimulate γ-globin and HbF production by using pharmacological agents. Most notable of them all is IMR-687, a potent, specific, and highly selective small molecule inhibitor of phosphodiesterase (PDE) 9, that increases intracellular cGMP levels and reactivation of HbF65. A phase II, randomised, double-blind, placebo-controlled study is currently underway to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of IMR-687 given once daily for 36 weeks in a total of 120 β-thalassemia patients (TDT and NTDT) (NCT04411082), aged 18–65 years of age. The primary objective of this study is to assess the safety and tolerability of IMR-687 in adult subjects with TDT and NTDT. Secondary objectives in the TDT patients include reduction in RBC transfusion burden (TB), iron load rate, mean number of transfusion events, and mean change in ICT dose and serum ferritin levels. Secondary objectives in NTDT patients include change in Hb, mean change in HbF levels, and mean change in ICT dose and serum ferritin levels.

TARGETING INEFFECTIVE ERYTHROPOIESIS

Erythroid maturation agents

Sotatercept or ACE-011 has been shown to correct ineffective erythropoiesis by acting as a ligand trap to inhibit negative regulators of late-stage erythropoiesis in the TGF-β superfamily. Sotatercept was evaluated up to a phase II trial (NCT01571635) on 16 TDT patients and 30 NTDT patients66. Most patients with NTDT treated with higher doses of sotatercept achieved sustained increases in Hb level. On the other hand, TDT patients treated with higher doses of sotatercept achieved notable reductions in transfusion requirements. Although sotatercept was efficacious, exhibited an overall good safety profile, and was tolerated by most patients, a decision was made not to take sotatercept forward to phase III trials.

Luspatercept67,68, an erythroid maturation agent, is a recombinant fusion protein that binds to the transforming growth factor beta (TGF-β) superfamily ligands to inhibit aberrant SMAD2/SMAD3 signalling and promote late-stage erythropoiesis69,70. Luspatercept was approved for the treatment of anaemia in adult patients with TD β-thalassemia by the US Food and Drug Administration (FDA) in November 2019 and by the EMA in June 202071,72. The development of this drug was suggested when RAP-536 fusion proteins were used in mouse models with thalassaemia and showed beneficial effects on the reduction of ineffective erythropoiesis, splenomegaly, and iron overload70,73. Following on from this, a multicentre, open-label, randomised dose-finding phase II study (NCT01749540, NCT02268409) demonstrated that luspatercept at 0.2–1.25 mg/kg administered subcutaneously every three weeks for at least 5 cycles was effective and well-tolerated in 64 adults with β-thalassaemia74. This paved the way for the pivotal phase III BELIEVE trial (NCT02604433) which involved 336 adult patients with TDT or Hb E/β-thalassaemia with no transfusion-free period of >35 days within the 24 weeks before randomisation. Patients were randomized 2:1 to receive luspatercept (1.0 mg/kg, titration up to 1.25 mg/kg) or placebo every three weeks for ≥48 weeks75. A significantly greater percentage of patients receiving luspatercept achieved the primary endpoint of a ≥33% reduction in TB from baseline during weeks 13–24, with a reduction of ≥2 RBC units compared with placebo. The percentage of patients achieving a ≥50% reduction in TB versus baseline was greater in the luspatercept group than the placebo group for all time points examined. The least squares mean difference in the key secondary end point of mean change from baseline in TB from weeks 13–24 was also in favour of luspatercept75. All patient subgroups showed a benefit from luspatercept treatment, including those with the more severe β00 genotype75. In addition to reductions in TB, patients treated with luspatercept were more likely to achieve clinically meaningful improvements in health-related quality of life as measured by the Transfusion-Dependent Quality of Life (TranQoL) and the 36-item Short Form Health Survey (SF-36) questionnaires, compared with placebo76. Moreover, a higher proportion of luspatercept responders achieved clinically meaningful improvements in physical functioning and overall physical component summary compared with placebo. Initial data from 5-year open-label extension phase of the BELIEVE trial, which is currently ongoing, showed that luspatercept treatment was associated with a lower mean cumulative RBC transfusion units and visits through week 48 across levels of baseline RBC TB. Luspatercept responders also showed meaningful reductions in RBC transfusion units and visits across levels of baseline RBC TB through week 12077. In addition, compared with placebo, luspatercept treatment was associated with a significant reduction in serum iron levels, LIC, and myocardial iron levels during the first 96 weeks of treatment78. Recruitment is expected to begin soon for a phase IIa study (NCT04143724) to evaluate the safety of luspatercept in paediatric patients who require regular RBC transfusions.

BEYOND (NCT03342404) is a phase II, double-blind, randomised (2:1), placebo-controlled, multicentre study evaluating the efficacy and safety of luspatercept in 145 adult patients with NTDT patients and a Hb level ≤10 g/dL79. The BEYOND study met its primary end point of mean increase in Hb of ≥1.0 g/dL from baseline over a continuous 12-week interval during weeks 13–24 in the absence of RBC transfusions with a statistically significant difference in favour of luspatercept. Treatment with luspatercept resulted in clinically significant and sustained improvements in anaemia in adults with NTDT, as measured by Hb levels, with >50% of patients receiving luspatercept achieving and maintaining a mean Hb increase of ≥1.5 g/dL79. Improvement in quality of life, as measured by the NTDT-patient-reported outcome Tiredness/Weakness (NTDT-PRO-T/W) domain score, favoured luspatercept and was correlated with increases in Hb levels. Luspatercept was well-tolerated over a prolonged period of time79.

Pyruvate kinase activators

The enzyme pyruvate kinase (PK) plays a significant role in the last stage of glycolysis in the RBC, and its presence is, therefore, essential for energy production in the RBC. Based on preclinical studies conducted on PK-deficient mice, metabolic disturbance in PK deficiency alters not only the survival of RBCs, but also the maturation of erythroid progenitors, resulting in ineffective erythropoiesis80. The use of an oral PK activator known as AG-348 (mitapivat) in healthy subjects has been shown to increase ATP and has been proven to be efficient and safe in PK deficiency patients81,82. This same agent showed increased ATP levels, reduced markers of ineffective erythropoiesis, and improved anaemia, RBC survival, and indices of iron overload in mouse models of β-thalassaemia83. Interim results from a phase II open-label, multicentre study (NCT03692052) of mitapivat in 15 NTDT adult patients showed an increase in Hb of ≥1.0 g/dL in 11 of 15 patients at 12 weeks with favourable changes in markers of erythropoiesis and haemolysis. No serious AEs were reported84. Based on this, two phase III double-blind, randomised, placebo-controlled, multicentre studies evaluating the efficacy and safety of mitapivat (100 mg orally, twice daily) in 240 adult patients with TDT (ENERGIZE-T, NCT04770779) and 171 adult patients with NTDT (ENERGIZE, NCT04770753) are scheduled to begin soon85.

Janus kinase 2 inhibitors

Preclinical studies have shown that Janus kinase 2 (JAK2) inhibition not only improved ineffective erythropoiesis but also reversed splenomegaly in an NTDT mouse model86. More data from other pre-clinical studies on TDT and NTDT mouse models also showed that JAK2 inhibitors are able to reduce splenomegaly87. These findings have suggested that the use of ruxolitinib, a JAK1/JAK2 inhibitor, could benefit β-thalassaemia patients. A single-arm, phase IIa study to evaluate the efficacy and safety of the JAK2 inhibitor ruxolitinib (INCB018424; INC424) administered orally at a starting dose of 10 mg twice daily among 30 adults with TDT and splenomegaly has been conducted (NCT02049450)88. A decrease in spleen size from baseline was observed in ruxolitinib-treated patients. There was also a mean change in spleen volume from baseline to week 12 (n=26) and week 30 (n=25)88. At week 30, one patient who initially had had a 15% decrease in spleen volume at week 12 demonstrated an increase in spleen volume88. However, there was no clinically significant improvement in pre-transfusion Hb, thus no related reduction in transfusion requirement. Moreover, although hepcidin levels increased in the ruxolitinib treatment group, no significant changes in iron parameters were observed over time88. For all the above-mentioned reasons, the study did not proceed to phase III88.

Glycine transporter 1 inhibitor

The oral administration of bitopertin (RO-4917838), a small molecule selective inhibitor of glycine transporter 1 (GlyT1), resulted in reduced anaemia and haemolysis, enhanced in vivo survival of erythrocytes, and diminished ineffective erythropoiesis in β-thalassaemia mice89. Markers of cellular damage induced by reactive oxygen species were also substantially improved. A phase II study of oral bitopertin in 12 adults with NTDT (NCT03271541) was conducted. The first 8 patients assessed at an 8-week preliminary efficacy analysis showed a reduction in mean total Hb. However, the study failed to show clinically significant improvements in haematologic and chemical biomarkers of disease activity and was, therefore, stopped early90.

TARGETING IRON DYSREGULATION

Minihepcidins

Minihepcidins are known to restrict iron absorption. The use of minihepcidins on young Hbbth3/+ mice led to amelioration of ineffective erythropoiesis, anaemia, splenomegaly, and iron overload91. In old Hbbth3/+ mice, a combined administration of minihepcidin with the iron chelator deferiprone also improved ineffective erythropoiesis, and anaemia and reversed splenomegaly91. A new mouse model (Hbbth1/th2), which closely resembles the human TDT phenotype, was generated to assess the efficacy of minihepcidins. In non-transfused Hbbth1/th2 mice, administration of high-dose minihepcidin also improved RBC lifespan, and ameliorated ineffective erythropoiesis, anaemia, splenomegaly and iron overload92. With these encouraging data, the efficacy and safety of minihepcidins was assessed in several clinical trials. LJPC-401, a synthetic human hepcidin given as a subcutaneous injection, was evaluated in a phase II, multicentre, randomised, open-label study (NCT03381833) in 84 adult patients with TDT. This trial was stopped early for lack of efficacy as determined by interim end point analyses93. PTG-300, another injectable hepcidin mimetic, was also evaluated in the phase II TRANSCEND (NCT03802201) trial in 63 adult β-thalassaemia patients. Due to efficacy issues with the drug, however, this study has also recently been stopped.

Transmembrane protease serine 6

Other novel therapeutic approaches to target iron dysregulation include increasing the hepatic synthesis of hepcidin. This can be achieved by suppressing the gene transmembrane protease serine 6 (TMPRSS6). In mouse models with NTDT, the inhibition of TMPRSS6 with small interfering RNA (siRNA) or antisense oligonucleotides caused a rise of hepcidin and an amelioration of anaemia and iron deposition94,95. Two major currently ongoing clinical trials are looking at stimulators of hepcidin production. The first is a phase I, randomised, single-blind, placebo-controlled, single-ascending and multiple-dose study (NCT04718844) to investigate the safety, tolerability, pharmacokinetic, and pharmacodynamic response of SLN124 [a GalNAc conjugated double-stranded fully modified siRNA that targets TMPRSS6 messenger RNA (mRNA)] in 112 adults with NTDT (α- and β-thalassaemia) and MDS. The second is phase IIa, randomised, open-label trial (NCT04059406) in 36 adults with NTDT who will be administered with TMPRSS6-LRx (a generation 2+ ligand-conjugated ASO targeting TMPRSS6). Interim data from both of these clinical trials are awaited.

Ferroportin inhibitors

Direct inhibition of the ferroportin receptor represents a novel approach to targeting the hepcidin-ferroportin axis96. VIT-2763 is an oral ferroportin inhibitor that has been specifically designed to target the hepcidin-ferroportin axis and ameliorate ineffective erythropoiesis. Preclinical data from NTDT mouse models showed that VIT-2763 decreased organ iron levels and improved haematologic parameters, including Hb level and RBC count, demonstrating amelioration of anaemia and improved erythropoiesis97. In addition, findings from a study of combination treatment with VIT-2763 and the iron-chelating agent deferasirox in the same preclinical model of NTDT revealed no negative impact of vamifeport on the efficacy of deferasirox or vice versa98. Based on this, a phase I, randomised, double-blind, placebo-controlled study was conducted on 72 healthy adult volunteers and showed that VIT-2763 administered at single oral doses up to 240 mg or multiple oral doses up to 120 mg twice daily was well-tolerated compared with placebo. There were no serious or severe AEs or discontinuations due to AEs99. A phase IIa trial (VIT-2763-THAL-201) to evaluate the efficacy of multiple doses of VIT-2763 in NTDT patients is currently ongoing. This study will assess improvements in haematologic parameters, including Hb and RBC indices, iron-related markers, and biomarkers for haemopoietic/erythropoietic activity. A phase IIb, multiple-dose, double-blind, randomised, placebo-controlled, parallel-group, multicentre trial in adult TDT patients is also being planned comprising a 24-week treatment phase with VIT-2763. This planned clinical trial will also assess and observe changes in Hb and RBC indices, iron metabolism parameters, and markers of erythropoietic activity96.

CONCLUSIONS

A new era of novel therapies is emerging to advance care and improve outcomes for thalassaemia patients. It is now a reality that gene therapy can modify HSCs to cure diseases like β-thalassemia. However, major limitations to this approach remain. Manipulation and expansion of HSCs ex vivo is challenging and HSCs cannot be targeted in vivo. Moreover, current myeloablative regimens are toxic. Until gene therapeutic strategies are proven to be safe in terms of long-term complications, and based on the available scientific data, HSCT using alternative donors (first MUDs and then haploidentical donors), remains the only curative approach for β-thalassaemia patients with no sibling donor. Ultimately, in order for gene therapy to be useful it is important to establish the profile of the patient for whom it is a priority. This is particularly important as it becomes more widely used outside experimental trials conducted in highly selected and equipped centres. Moreover, current costs, limited availability and resources represent a further indication for a rational and progressive decision-making process regarding this innovative treatment. Identifying and selecting patients for gene therapy must be carried out through a consensus decision with a precise set of criteria100. Gene disruption via genome editing approaches can be very efficient. Specifically, the base editing approach can correct mutant genes through the achievement of a physiological and tissue specific expression of endogenous genes. However, chromothripsis, a potential phenomenon that is subsequent to the introduction of the double break in the DNA has been described101 and should be kept in mind as a potential negative outcome. Such damage to the DNA is associated with a very high and dense region of mutations. Future genome editing approaches should aim to improve the targeting of quiescent cells and reducing potential genome toxicity.

The approval of luspatercept has led to the availability of another new treatment modality for TDT patients and may provide long-term sustained reductions in transfusion requirements and the need for ICT. Data from the long-term phase III BELIEVE trial will provide further valuable insights. Moreover, the clinical benefit of luspatercept treatment has now been observed in patients with NTDT from the phase II BEYOND trial, as measured by a meaningful improvement in anaemia. This could implicate the potential use of this agent in NTDT patients in the near future. In conclusion, we currently have several novel therapeutic agents in the field that are currently under development. Once the efficacy and safety of all these novel therapies has been established, long-term, head-to-head, and comparison trials are needed to guide the integration of these novel therapies into the standard care of β-thalassaemia. Moreover, cost-effectiveness and health-economic studies are warranted to select the best therapy for the individual patient in addition to demonstrating the cost/benefit of a particular therapy in prolonging a patient’s life, while taking into consideration the resources available.

Footnotes

CONFLICTS OF INTEREST

RB reports no conflicts of interest. IM reports receiving honoraria from Sanofi-Genzyme and Amicus Therapeutics. MDC has been or is a current consultant for Novartis, Celgene Corp (Bristol Myers Squibb), Vifor Pharma and Ionis Pharmaceuticals, and has received research funding from Novartis, Celgene Corp (Bristol Myers Squibb), La Jolla Pharmaceutical Company, Roche, Protagonist Therapeutics, and CRISPR Therapeutics.

REFERENCES

  • 1.Taher AT, Musallam KM, Cappellini MD. beta-Thalassemias. N Engl J Med. 2021;384:727–43. doi: 10.1056/NEJMra2021838. [DOI] [PubMed] [Google Scholar]
  • 2.Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia. Lancet. 2018;391:155–67. doi: 10.1016/S0140-6736(17)31822-6. [DOI] [PubMed] [Google Scholar]
  • 3.Modell B, Khan M, Darlison M, et al. Improved survival of thalassaemia major in the UK and relation to T2* cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2008;10:42. doi: 10.1186/1532-429X-10-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Taher AT, Cappellini MD. How I manage medical complications of beta-thalassemia in adults. Blood. 2018;132:1781–91. doi: 10.1182/blood-2018-06-818187. [DOI] [PubMed] [Google Scholar]
  • 5.Grech L, Borg K, Borg J. Novel therapies in beta-thalassaemia. Br J Clin Pharmacol. 2021 doi: 10.1111/bcp.14918. [Online ahead of print.] [DOI] [PubMed] [Google Scholar]
  • 6.Taher AT, Bou-Fakhredin R, Kattamis A, et al. Improving outcomes and quality of life for patients with transfusion-dependent beta-thalassemia: recommendations for best clinical practice and the use of novel treatment strategies. Expert Rev Hematol. 2021;14:897–908. doi: 10.1080/17474086.2021.1977116. [DOI] [PubMed] [Google Scholar]
  • 7.Musallam KM, Bou-Fakhredin R, Cappellini MD, Taher AT. 2021 update on clinical trials in beta-thalassemia. Am J Hematol. 2021;96:1518–31. doi: 10.1002/ajh.26316. [DOI] [PubMed] [Google Scholar]
  • 8.Oikonomopoulou C, Goussetis E. HSCT remains the only cure for patients with transfusion-dependent thalassemia until gene therapy strategies are proven to be safe. Bone Marrow Transplant. 2021;56:2882–8. doi: 10.1038/s41409-021-01461-0. [DOI] [PubMed] [Google Scholar]
  • 9.Makis A, Voskaridou E, Papassotiriou I, Hatzimichael E. Novel therapeutic advances in β-thalassemia. Biology (Basel) 2021;10:546. doi: 10.3390/biology10060546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Shenoy S, Thompson AA. Unrelated donor stem cell transplantation for transfusion-dependent thalassemia. Ann N Y Acad Sci. 2016;1368:122–6. doi: 10.1111/nyas.13019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hussein AA, Al-Zaben A, Ghatasheh L, et al. Risk adopted allogeneic hematopoietic stem cell transplantation using a reduced intensity regimen for children with thalassemia major. Pediatr Blood Cancer. 2013;60:1345–9. doi: 10.1002/pbc.24493. [DOI] [PubMed] [Google Scholar]
  • 12.Anurathapan U, Pakakasama S, Rujkijyanont P, et al. Pretransplant immunosuppression followed by reduced-toxicity conditioning and stem cell transplantation in high-risk thalassemia: a safe approach to disease control. Biol Blood Marrow Transplant. 2013;19:1259–62. doi: 10.1016/j.bbmt.2013.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gaziev J, Isgro A, Sodani P, et al. Optimal outcomes in young class 3 patients with thalassemia undergoing HLA-identical sibling bone marrow transplantation. Transplantation. 2016;100:925–32. doi: 10.1097/TP.0000000000000928. [DOI] [PubMed] [Google Scholar]
  • 14.Mathews V, George B, Viswabandya A, et al. Improved clinical outcomes of high risk beta thalassemia major patients undergoing a HLA matched related allogeneic stem cell transplant with a treosulfan based conditioning regimen and peripheral blood stem cell grafts. PLoS One. 2013;8:e61637. doi: 10.1371/journal.pone.0061637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.John MJ, Mathew A, Philip CC, et al. Unrelated and related donor transplantation for beta-thalassemia major: a single-center experience from India. Pediatr Transplant. 2018;22:e13209. doi: 10.1111/petr.13209. [DOI] [PubMed] [Google Scholar]
  • 16.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;3:2562–70. doi: 10.1182/bloodadvances.2019000291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gaziev J, Isgro A, Sodani P, et al. Haploidentical HSCT for hemoglobinopathies: improved outcomes with TCRalphabeta(+)/CD19(+)-depleted grafts. Blood Adv. 2018;2:263–70. doi: 10.1182/bloodadvances.2017012005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Anurathapan U, Hongeng S, Pakakasama S, et al. Hematopoietic stem cell transplantation for severe thalassemia patients from haploidentical donors using a novel conditioning regimen. Biol Blood Marrow Transplant. 2020;26:1106–12. doi: 10.1016/j.bbmt.2020.01.002. [DOI] [PubMed] [Google Scholar]
  • 19.Sun Q, Wu B, Lan H, et al. Haploidentical haematopoietic stem cell transplantation for thalassaemia major based on an FBCA conditioning regimen. Br J Haematol. 2018;182:554–8. doi: 10.1111/bjh.15438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bolanos-Meade J, Cooke KR, Gamper CJ, et al. Effect of increased dose of total body irradiation on graft failure associated with HLA-haploidentical transplantation in patients with severe haemoglobinopathies: a prospective clinical trial. Lancet Haematol. 2019;6:e183–e93. doi: 10.1016/S2352-3026(19)30031-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Vellaichamy Swaminathan V, Ravichandran N, Ramanan KM, et al. Augmented immunosuppression and PTCY-based haploidentical hematopoietic stem cell transplantation for thalassemia major. Pediatr Transplant. 2021;25:e13893. doi: 10.1111/petr.13893. [DOI] [PubMed] [Google Scholar]
  • 22.Anurathapan U, Hongeng S, Pakakasama S, et al. Hematopoietic stem cell transplantation for homozygous beta-thalassemia and beta-thalassemia/hemoglobin E patients from haploidentical donors. Bone Marrow Transplant. 2016;51:813–8. doi: 10.1038/bmt.2016.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Swaminathan VV, Uppuluri R, Patel S, et al. Matched family versus alternative donor hematopoietic stem cell transplantation for patients with thalassemia major: experience from a Tertiary Referral Center in South India. Biol Blood Marrow Transplant. 2020;26:1326–31. doi: 10.1016/j.bbmt.2020.03.016. [DOI] [PubMed] [Google Scholar]
  • 24.Oikonomopoulou C, Paisiou A, Komitopoulou A, et al. Increased incidence of autoimmune cytopenias after allogeneic haematopoietic stem cell transplantation using a matched unrelated donor in children with beta-thalassaemia. Br J Haematol. 2021;192:e127–e9. doi: 10.1111/bjh.17335. [DOI] [PubMed] [Google Scholar]
  • 25.Strocchio L, Romano M, Cefalo MG, et al. Cord blood transplantation in children with hemoglobinopathies. Expert Opinion on Orphan Drugs. 2015;3:1125–36. [Google Scholar]
  • 26.Rahal I, Galambrun C, Bertrand Y, et al. Late effects after hematopoietic stem cell transplantation for beta-thalassemia major: the French national experience. Haematologica. 2018;103:1143–9. doi: 10.3324/haematol.2017.183467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Angelucci E. Complication free survival long-term after hemopoietic cell transplantation in thalassemia. Haematologica. 2018;103:1094–6. doi: 10.3324/haematol.2018.196071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Vlachopapadopoulou E, Kitra V, Peristeri J, et al. Gonadal function of young patients with beta-thalassemia following bone marrow transplantation. J Pediatr Endocrinol Metab. 2005;18:477–83. doi: 10.1515/jpem.2005.18.5.477. [DOI] [PubMed] [Google Scholar]
  • 29.Santarone S, Pepe A, Meloni A, et al. Secondary solid cancer following hematopoietic cell transplantation in patients with thalassemia major. Bone Marrow Transplant. 2018;53:39–43. doi: 10.1038/bmt.2017.214. [DOI] [PubMed] [Google Scholar]
  • 30.Goodman MA, Malik P. The potential of gene therapy approaches for the treatment of hemoglobinopathies: achievements and challenges. Ther Adv Hematol. 2016;7:302–15. doi: 10.1177/2040620716653729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rivella S. beta-thalassemias: paradigmatic diseases for scientific discoveries and development of innovative therapies. Haematologica. 2015;100:418–30. doi: 10.3324/haematol.2014.114827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Breda L, Casu C, Gardenghi S, et al. Therapeutic hemoglobin levels after gene transfer in beta-thalassemia mice and in hematopoietic cells of beta-thalassemia and sickle cells disease patients. PLoS One. 2012;7:e32345. doi: 10.1371/journal.pone.0032345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.May C, Rivella S, Callegari J, et al. Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin. Nature. 2000;406:82–6. doi: 10.1038/35017565. [DOI] [PubMed] [Google Scholar]
  • 34.May C, Rivella S, Chadburn A, Sadelain M. Successful treatment of murine beta-thalassemia intermedia by transfer of the human beta-globin gene. Blood. 2002;99:1902–8. doi: 10.1182/blood.v99.6.1902. [DOI] [PubMed] [Google Scholar]
  • 35.Rivella S, May C, Chadburn A, et al. A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta-globin gene transfer. Blood. 2003;101:2932–9. doi: 10.1182/blood-2002-10-3305. [DOI] [PubMed] [Google Scholar]
  • 36.Cavazzana-Calvo M, Payen E, Negre O, et al. Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature. 2010;467:318–22. doi: 10.1038/nature09328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Negre O, Bartholomae C, Beuzard Y, et al. Preclinical evaluation of efficacy and safety of an improved lentiviral vector for the treatment of beta-thalassemia and sickle cell disease. Current gene therapy. 2015;15:64–81. doi: 10.2174/1566523214666141127095336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Thompson AA, Walters MC, Kwiatkowski J, et al. Gene therapy in patients with transfusion-dependent beta-thalassemia. N Engl J Med. 2018;378:1479–93. doi: 10.1056/NEJMoa1705342. [DOI] [PubMed] [Google Scholar]
  • 39.Locatelli F, Kwiatkowski JL, Walters MC, et al. Betibeglogene autotemcel in patients with transfusion-dependent β-thalassemia: updated results from HGB-207 (Northstar-2) and HGB-212 (Northstar-3) [abstract] Hemasphere. 2021;5(S2):91. [Google Scholar]
  • 40.Kulozik AE, Thuret I, Kwiatkowski JL, et al. Interim results of betibeglogene autotemcel gene therapy in pediatric patients with transfusion-dependent β thalassemia (TDT) treated In the phase 3 Northstar-2 and Northstar-3 studies [abstract] Hemasphere. 2021;5(S2):628. [Google Scholar]
  • 41.Yannaki E, Locatelli F, Kwiatkowski JL, et al. Betibeglogene autotemcel gene therapy for the Treatment of transfusion-dependent β-thalassemia: Updated long-term efficacy and safety results [abstract] Hemasphere. 2021;5(S2):86–7. [Google Scholar]
  • 42.Bluebird bio Announces Temporary Suspension on Phase 1/2 and Phase 3 Studies of LentiGlobin Gene Therapy for Sickle Cell Disease (bb1111). Press Release. Feb 16, 2021. [Accessed on 28/09/2021]. Available at: https://investor.bluebirdbio.com/news-releases/news-release-details/bluebird-bio-announces-temporary-suspension-phase-12-and-phase-3.
  • 43.Bluebird bio announces the lifting of FDA clinical hold for sickle cell disease and β-thalassemia studies. bluebird bio press release. Jun 7, 2021. [Accessed on 28/09/2021]. Available at: https://www.businesswire.com/news/home/20210607005267/en/bluebird-bio-Announces-the-Lifting-of-FDA-Clinical-Hold-for-Sickle-Cell-Disease-and-%CE%B2-Thalassemia-Studies.
  • 44.Bluebird bio reports second quarter financial results and provides operational update. bluebird bio press release. Aug 9, 2021. [Accessed on 28/09/2021.]. Available at: https://www.businesswire.com/news/home/20210809005334/en/bluebird-bio-Reports-Second-Quarter-Financial-Results-and-Provides-Operational-Update.
  • 45.Marktel S, Scaramuzza S, Cicalese MP, et al. Intrabone hematopoietic stem cell gene therapy for adult and pediatric patients affected by transfusion-dependent β-thalassemia. Nat Med. 2019;25:234–41. doi: 10.1038/s41591-018-0301-6. [DOI] [PubMed] [Google Scholar]
  • 46.Boulad F, Mansilla-Soto J, Cabriolu A, et al. Gene Therapy and Genome Editing. Hematol Oncol Clin North Am. 2018;32:329–42. doi: 10.1016/j.hoc.2017.11.007. [DOI] [PubMed] [Google Scholar]
  • 47.Boulad F, Wang X, Qu J, et al. Safe mobilization of CD34+ cells in adults with beta-thalassemia and validation of effective globin gene transfer for clinical investigation. Blood. 2014;123:1483–6. doi: 10.1182/blood-2013-06-507178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Menzel S, Garner C, Gut I, et al. A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15. Nat Genet. 2007;39:1197–9. doi: 10.1038/ng2108. [DOI] [PubMed] [Google Scholar]
  • 49.Sankaran VG, Menne TF, Xu J, et al. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 2008;322:1839–42. doi: 10.1126/science.1165409. [DOI] [PubMed] [Google Scholar]
  • 50.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 beta-thalassemia. Proc Natl Acad Sci U S A. 2008;105:1620–5. doi: 10.1073/pnas.0711566105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Galanello R, Sanna S, Perseu L, et al. Amelioration of Sardinian beta0 thalassemia by genetic modifiers. Blood. 2009;114:3935–7. doi: 10.1182/blood-2009-04-217901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Costa FC, Fedosyuk H, Neades R, et al. Induction of Fetal Hemoglobin In Vivo Mediated by a Synthetic gamma-Globin Zinc Finger Activator. Anemia. 2012;2012:507894. doi: 10.1155/2012/507894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Guda S, Brendel C, Renella R, et al. miRNA-embedded shRNAs for Lineage-specific BCL11A Knockdown and Hemoglobin F Induction. Mol Ther. 2015;23:1465–74. doi: 10.1038/mt.2015.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mishra B, Chou S, Lin MI, et al. Crispr/Cas9-mediated genome editing of human CD34+ cells upregulate fetal hemoglobin to clinically relevant levels in single cell-derived Erythroid Colonies. Blood. 2016;128:3623. [Google Scholar]
  • 55.Basak A, Hancarova M, Ulirsch JC, et al. BCL11A deletions result in fetal hemoglobin persistence and neurodevelopmental alterations. J Clin Invest. 2015;125:2363–8. doi: 10.1172/JCI81163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Giani FC, Fiorini C, Wakabayashi A, et al. Targeted application of human genetic variation can improve red blood cell production from stem cells. Cell Stem Cell. 2016;18:73–8. doi: 10.1016/j.stem.2015.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Brendel C, Guda S, Renella R, et al. Lineage-specific BCL11A knockdown circumvents toxicities and reverses sickle phenotype. J Clin Invest. 2016;126:3868–78. doi: 10.1172/JCI87885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bjurstrom CF, Mojadidi M, Phillips J, et al. Reactivating fetal hemoglobin expression in human adult erythroblasts through BCL11A knockdown using targeted endonucleases. Mol Ther Nucleic Acids. 2016;5:e351. doi: 10.1038/mtna.2016.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cavazzana M, Antoniani C, Miccio A. Gene therapy for beta-hemoglobinopathies. Mol Ther. 2017;25:1142–54. doi: 10.1016/j.ymthe.2017.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Makis A, Hatzimichael E, Papassotiriou I, Voskaridou E. 2017 Clinical trials update in new treatments of beta-thalassemia. Am J Hematol. 2016;91:1135–45. doi: 10.1002/ajh.24530. [DOI] [PubMed] [Google Scholar]
  • 61.Genovese P, Schiroli G, Escobar G, et al. Targeted genome editing in human repopulating haematopoietic stem cells. Nature. 2014;510:235–40. doi: 10.1038/nature13420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 Gene Editing for Sickle Cell Disease and beta-Thalassemia. N Engl J Med. 2021;384:252–60. doi: 10.1056/NEJMoa2031054. [DOI] [PubMed] [Google Scholar]
  • 63.Locatelli F, Ailinca-Luchian S, Bobruff Y, et al. CTX001 for transfusion-dependent β-thalassemia: safety and efficacy results from the ongoing CLIMB THAL-111 study of autologous CRISPR-cas9-modified CD34+ hematopoietic stem and progenitor cells [abstract] Hemasphere. 2021;5(S2):335–6. [Google Scholar]
  • 64.Smith AR, Schiller GJ, Vercellotti GM, et al. Preliminary results of a phase 1/2 clinical study of zinc finger nuclease-mediated editing of BCL11A in autologous hematopoietic stem cells for transfusion-dependent beta thalassemia [abstract] Blood. 2019;134(Suppl 1):3455. [Google Scholar]
  • 65.McArthur JG, Svenstrup N, Chen C, et al. A novel, highly potent and selective phosphodiesterase-9 inhibitor for the treatment of sickle cell disease. Haematologica. 2020;105:623–31. doi: 10.3324/haematol.2018.213462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cappellini MD, Porter J, Origa R, et al. Sotatercept, a novel transforming growth factor beta ligand trap, improves anemia in beta-thalassemia: a phase II, open-label, dose-finding study. Haematologica. 2019;104:477–84. doi: 10.3324/haematol.2018.198887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cappellini MD, Taher AT. The use of luspatercept for thalassemia in adults. Blood Adv. 2021;5:326–33. doi: 10.1182/bloodadvances.2020002725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Taher AT, Cappellini MD. Luspatercept for beta-thalassemia: beyond red blood cell transfusions. Expert Opin Biol Ther. 2021;21:1363–71. doi: 10.1080/14712598.2021.1968825. [DOI] [PubMed] [Google Scholar]
  • 69.Suragani RN, Cadena SM, Cawley SM, Sako D, Mitchell D, Li R, et al. Transforming growth factor-beta superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med. 2014;20:408–14. doi: 10.1038/nm.3512. [DOI] [PubMed] [Google Scholar]
  • 70.Suragani RN, Cawley SM, Li R, Wallner S, Alexander MJ, Mulivor AW, et al. Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine β-thalassemia. Blood. 2014;123:3864–72. doi: 10.1182/blood-2013-06-511238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.FDA [internet] REBLOZYL label. Silver Spring MD: FDA; 2019. [Accessed on 28/09/2021]. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/761136lbl.pdf. [Google Scholar]
  • 72.European Medicines Agency (EMA) Meeting highlights from the Committee for Medicinal Products for Human Use (CHMP); 28–30 April 2020; Amsterdam: EMA; 2020. [Accessed on 28/09/2021.]. Available at: https://www.ema.europa.eu/en/news/meeting-highlights-committee-medicinal-products-human-use-chmp-28-30-april-2020. [Google Scholar]
  • 73.Martinez PA, Li R, Ramanathan HN, et al. Smad2/3-pathway ligand trap luspatercept enhances erythroid differentiation in murine beta-thalassaemia by increasing GATA-1 availability. J Cell Mol Med. 2020;24:6162–77. doi: 10.1111/jcmm.15243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Piga A, Perrotta S, Gamberini MR, et al. Luspatercept improves hemoglobin levels and blood transfusion requirements in a study of patients with β-thalassemia. Blood. 2019;133:1279–89. doi: 10.1182/blood-2018-10-879247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cappellini MD, Viprakasit V, Taher AT, et al. A Phase 3 Trial of Luspatercept in Patients with Transfusion-Dependent beta-Thalassemia. N Engl J Med. 2020;382:1219–31. doi: 10.1056/NEJMoa1910182. [DOI] [PubMed] [Google Scholar]
  • 76.Cappellini MD, Taher AT, Piga A, et al. Health-related quality of life outcomes for patients with transfusion-dependent beta-thalassemia treated with luspatercept in the believe trial [abstract] Blood. 2020;136(Suppl 1):8–9. [Google Scholar]
  • 77.Taher AT, Viprakasit V, Hermine O, et al. Sustained reductions in red blood cell (RBC) transfusion burden and events in β-Thalassemia with luspatercept: longitudinal results of the believe trial [abstract] Blood. 2020;136(Suppl 1):45–6. [Google Scholar]
  • 78.Hermine O, Cappellini MD, Taher AT, et al. Longitudinal effect of luspatercept treatment on iron overload and iron chelation therapy (ICT) in adult patients (pts) with β-thalassemia in the BELIEVE trial [abstract] Blood. 2020;136(Suppl 1):47–8. [Google Scholar]
  • 79.Taher AT, Cappellini MD, Kattamis A, et al. The BEYOND study: results of a phase 2, double-blind, randomized, placebo-controlled multicenter study of luspatercept in adult patients with non-transfusion dependent beta-thalassemia [abstract] Hemasphere. 2021;5(S2):1–2. [Google Scholar]
  • 80.Aizawa S, Harada T, Kanbe E, et al. Ineffective erythropoiesis in mutant mice with deficient pyruvate kinase activity. Exp Hematol. 2005;33:1292–8. doi: 10.1016/j.exphem.2005.07.008. [DOI] [PubMed] [Google Scholar]
  • 81.Yang H, Merica E, Chen Y, et al. Phase 1 single- and multiple-ascending-dose randomized studies of the safety, pharmacokinetics, and pharmacodynamics of AG-348, a first-in-class allosteric activator of pyruvate kinase r, in healthy volunteers. Clin Pharmacol Drug Dev. 2019;8:246–59. doi: 10.1002/cpdd.604. [DOI] [PubMed] [Google Scholar]
  • 82.Grace RF, Rose C, Layton DM, et al. Safety and efficacy of mitapivat in pyruvate kinase deficiency. N Engl J Med. 2019;381:933–44. doi: 10.1056/NEJMoa1902678. [DOI] [PubMed] [Google Scholar]
  • 83.Matte A, Beneduce E, Siciliano A, et al. The pyruvate kinase activator AG-348 improves murine β-thalassemic anemia and corrects ineffective erythropoiesis [abstract] Haematologica. 2016;101(S1):18. (S135) [Google Scholar]
  • 84.Kuo KH, Layton DM, Lal A, et al. Results from a phase 2, open-label, multicenter study of the oral pyruvate kinase activator mitapivat in adults with non-transfusion dependent alpha- or beta-thalassemia [abstract] Hemasphere. 2021;5(S2):92. [Google Scholar]
  • 85.Kuo KH, Layton DM, Al-Samkari H, et al. ENERGIZE and ENERGIZE-T: two phase 3, randomized, double-blind, placebo-controlled studies of mitapivat in adults with non-transfusion dependent or transfusion-dependent alpha- or beta-thalassemia [abstract] Hemasphere. 2021;5:848–9. [Google Scholar]
  • 86.Libani IV, Guy EC, Melchiori L, et al. Decreased differentiation of erythroid cells exacerbates ineffective erythropoiesis in beta-thalassemia. Blood. 2008;112:875–85. doi: 10.1182/blood-2007-12-126938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Casu C, Presti VL, Oikonomidou PR, et al. Short-term administration of JAK2 inhibitors reduces splenomegaly in mouse models of beta-thalassemia intermedia and major. Haematologica. 2018;103:e46–e9. doi: 10.3324/haematol.2017.181511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Taher AT, Karakas Z, Cassinerio E, et al. Efficacy and safety of ruxolitinib in regularly transfused patients with thalassemia: results from a phase 2a study. Blood. 2018;131:263–5. doi: 10.1182/blood-2017-06-790121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Matte A, Federti E, Winter M, et al. Bitopertin, a selective oral GLYT1 inhibitor, improves anemia in a mouse model of β-thalassemia. JCI Insight. 2019;4:e130111. doi: 10.1172/jci.insight.130111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Taher AT, Viprakasit V, Cappellini MD, et al. Haematological effects of oral administration of bitopertin, a glycine transport inhibitor, in patients with non-transfusion-dependent beta-thalassaemia. Br J Haematol. 2021;194:474–7. doi: 10.1111/bjh.17479. [DOI] [PubMed] [Google Scholar]
  • 91.Casu C, Oikonomidou PR, Chen H, et al. Minihepcidin peptides as disease modifiers in mice affected by beta-thalassemia and polycythemia vera. Blood. 2016;128:265–76. doi: 10.1182/blood-2015-10-676742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Casu C, Chessa R, Liu A, et al. Minihepcidins improve ineffective erythropoiesis and splenomegaly in a new mouse model of adult β-thalassemia major. Haematologica. 2020;105:1835. doi: 10.3324/haematol.2018.212589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Motta I, Bou-Fakhredin R, Taher AT, Cappellini MD. Beta thalassemia: new therapeutic options beyond transfusion and iron chelation. Drugs. 2020;80:1053–63. doi: 10.1007/s40265-020-01341-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Guo S, Casu C, Gardenghi S, et al. Reducing TMPRSS6 ameliorates hemochromatosis and beta-thalassemia in mice. J Clin Invest. 2013;123:1531–41. doi: 10.1172/JCI66969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.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 beta-thalassemia intermedia. Blood. 2013;121:1200–8. doi: 10.1182/blood-2012-09-453977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Porter J, Taher A, Viprakasit V, et al. Oral ferroportin inhibitor vamifeport for improving iron homeostasis and erythropoiesis in beta-thalassemia: current evidence and future clinical development. Expert Rev Hematol. 2021;14:633–44. doi: 10.1080/17474086.2021.1935854. [DOI] [PubMed] [Google Scholar]
  • 97.Manolova V, Nyffenegger N, Flace A, et al. Oral ferroportin inhibitor ameliorates ineffective erythropoiesis in a model of beta-thalassemia. J Clin Invest. 2019;130:491–506. doi: 10.1172/JCI129382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Nyffenegger N, Flace A, Doucerain C, et al. The oral ferroportin inhibitor VIT-2763 improves erythropoiesis without interfering with iron chelation therapy in a mouse model of beta-thalassemia. Int J Mol Sci. 2021;22:873. doi: 10.3390/ijms22020873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Richard F, van Lier JJ, Roubert B, et al. Oral ferroportin inhibitor VIT-2763: first-in-human, phase 1 study in healthy volunteers. Am J Hematol. 2020;95:68–77. doi: 10.1002/ajh.25670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Baronciani D, Casale M, De Franceschi L, et al. Selecting beta-thalassemia patients for gene therapy: a decision-making algorithm. Hemasphere. 2021;5:e555. doi: 10.1097/HS9.0000000000000555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Leibowitz ML, Papathanasiou S, Doerfler PA, et al. Chromothripsis as an on-target consequence of CRISPR-Cas9 genome editing. Nat Genet. 2021;53:895–905. doi: 10.1038/s41588-021-00838-7. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Blood Transfusion are provided here courtesy of SIMTI Servizi

RESOURCES