Alpha-Thalassemia: A Practical Overview (PDF)

Document Details

BelovedJackalope

Uploaded by BelovedJackalope

Khaled M. Musallam, M. Domenica Cappellini, Thomas D. Coates, Kevin H.M. Kuo, Hanny Al-Samkari, Sujit Sheth, Vip Viprakasit, Ali T. Taher

Tags

alpha-thalassemia blood disorders hemoglobinopathies medical review

Summary

This article provides a practical overview of alpha-thalassemia, a genetic blood disorder. It covers the molecular genetics, genotype-phenotype correlations, clinical presentations (from silent carrier to severe forms), and management strategies. The review is intended for clinicians practicing in regions where alpha-thalassemia might not be as common.

Full Transcript

Blood Reviews xxx (xxxx) xxx Contents lists available at ScienceDirect Blood Reviews journal homepage: www.elsevier.com/locate/issn/0268960X Review Аlpha-thalassemia: A practical overview Khaled M. Musallam a, M. Domenica Cappellini b, Thomas D. Coates c, Kevin H.M. Kuo d, Hanny Al-Samkari e, Sujit...

Blood Reviews xxx (xxxx) xxx Contents lists available at ScienceDirect Blood Reviews journal homepage: www.elsevier.com/locate/issn/0268960X Review Аlpha-thalassemia: A practical overview Khaled M. Musallam a, M. Domenica Cappellini b, Thomas D. Coates c, Kevin H.M. Kuo d, Hanny Al-Samkari e, Sujit Sheth f, Vip Viprakasit g, Ali T. Taher h, * a Center for Research on Rare Blood Disorders (CR-RBD), Burjeel Medical City, Abu Dhabi, United Arab Emirates Department of Clinical Sciences and Community, University of Milan, Ca’ Granda Foundation IRCCS Maggiore Policlinico Hospital, Milan, Italy c Hematology Section, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA d Division of Hematology, Department of Medicine, University of Toronto, Toronto, ON, Canada e Center for Hematology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA f Division of Pediatric Hematology and Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA g Department of Pediatrics & Thalassemia Center, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand h Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon b A R T I C L E I N F O A B S T R A C T Keywords: alpha-thalassemia Hemolysis anemia Diagnosis Management Transfusion α-Thalassemia is an inherited blood disorder characterized by decreased synthesis of α-globin chains that results in an imbalance of α and β globin and thus varying degrees of ineffective erythropoiesis, decreased red blood cell (RBC) survival, chronic hemolytic anemia, and subsequent comorbidities. Clinical presentation varies depending on the genotype, ranging from a silent or mild carrier state to severe, transfusion-dependent or lethal disease. Management of patients with α-thalassemia is primarily supportive, addressing either symptoms (eg, RBC transfusions for anemia), complications of the disease, or its transfusion-dependence (eg, chelation therapy for iron overload). Several novel therapies are also in development, including curative gene manipulation techniques and disease modifying agents that target ineffective erythropoiesis and chronic hemolytic anemia. This review of α-thalassemia and its various manifestations provides practical information for clinicians who practice beyond those regions where it is found with high frequency. 1. Introduction The thalassemias are inherited, autosomal recessive disorders of hemoglobin (Hb) synthesis characterized by a variety of molecular de­ fects and are among the most common genetic diseases worldwide [1–3]. The principal forms, α- and β-thalassemia, result from mutations in globin genes that lead to a deficit or qualitative change in the pro­ duction of the α-globin and β-globin chains of adult Hb, respectively. The global disease burden of thalassemia is substantial, with 5% to 20% of the world population carrying one or more α-thalassemia mu­ tations, and ≈1.5% carrying one or more β-thalassemia mutations [5–7]. Prevalence rates vary among regions but are highest in tropical and subtropical countries, particularly in Southeast Asia and the Mediter­ ranean. α-Thalassemia has almost reached fixation (frequency has reached 100% in the population) in some parts of southern Asia, with 80% to 90% of the population being carriers [9–11]. The thalassemias are more common in areas where falciparum malaria has been wide­ spread and endemic, possibly conferring a protective advantage in ma­ larial environments [12–16]. The total burden of thalassemia on economic and healthcare systems is not currently known but is under­ stood to be increasing, not only in countries with high prevalence where more patients are surviving and living longer lives , but also where prevalence is increasing due to immigration and demographic transi­ tions [3,18]. Although there are significant gaps in our knowledge of the prevalence and health burden of α-thalassemia, the increasing speed and decreasing cost of genetic testing and other screening methods may aid in the goal of treatment and future prevention. Of the hemoglobinopathies, α-thalassemia, with its complex muta­ tions, is particularly challenging to clinicians. Clinically diverse forms of the disease present across a wide spectrum of phenotypes, ranging from asymptomatic or mild carrier states to severe, transfusion-dependent or lethal types, with a broad range of clinical manifestations in between. * Corresponding author at: Medicine, Hematology & Oncology, Naef K. Basile Cancer Institute, Research, Department of Internal Medicine, American University of Beirut Medical Center, P.O. Box 11-0236, Beirut 11072020, Lebanon. E-mail address: [email protected] (A.T. Taher). https://doi.org/10.1016/j.blre.2023.101165 Available online 3 January 2024 0268-960X/© 2024 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/bync-nd/4.0/). Please cite this article as: Khaled M. Musallam et al., Blood Reviews, https://doi.org/10.1016/j.blre.2023.101165 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx This overview aims to describe the disease and its various manifestations to clinicians who practice in regions where it is relatively rare. We re­ view the molecular genetics and genotype/phenotype correlations to establish a firm knowledge base and promote the recognition, differ­ ential diagnosis, counseling, and clinical management of α-thalassemia. New information regarding emerging therapies and agents in develop­ ment is also provided. It is worth noting that overall, evidence-based observations in α-thalassemia are relatively limited, with several as­ pects of diagnosis and management extrapolated from studies of β-thalassemia. In this review, we use our experience in treating the disease over the past few decades to fill such evidence gaps. 2. Molecular understanding 2.1. Molecular genetics of normal hemoglobin Human Hb synthesis is directed by an α-globin gene cluster (con­ taining 1 embryonic ζ gene and a linked pair of fetal/adult α genes) located on chromosome 16 and a β-globin gene cluster containing 1 embryonic ε gene, 2 embryonic/fetal γ genes, and the adult δ and β genes (1 each) located on chromosome 11 (Fig. 1A) [19–22]. 2.2. Normal hemoglobin synthesis Hemoglobin is a tetrameric protein consisting of 2 homodimers of globin subunits, each comprising a globin chain conjugated with a heme moiety having a liganded iron atom at the center that binds one oxygen A B C Fig. 1. Human globin synthesis. (A) Chromosomal location and organization of the α- and β-globin gene clusters. The α- and β-globin gene clusters are located on chromosomes 16p13.3 and 11p15.4, respectively. The ζ-globin, α2-globin, and α1-globin genes are driven by 4 conserved enhancer elements (MCS-R1 through MCSR4), and the ε-globin, γ1-globin, γ2-globin, δ-globin, and β-globin genes are driven by an enhancer cluster, the locus control region (βLCR). Adapted from Farashi S, et al. Blood Cells Mol Dis 2018;70:43–53 with kind permission from Elsevier. (B) Hemoglobin switching at the α- and β-globin loci. During primitive erythropoiesis in the yolk sac, the embryonic ζ-globin (from the α-globin locus) and ε-globin (from the β-globin locus) genes are expressed until approximately 8 weeks’ gestation, when these genes are silenced and there is a maturational switch to α- and γ-globin expression during fetal life. A second switch from γ- to β-globin occurs at birth. Embryonic Hb (ζ2γ2 [Hb Portland], ζ2ε2 [Hb Gower 1], and α2ε2 [Hb Gower 2]) is produced through the yolk sac stage of development, when ζ- and ε-globin chain production stop, resulting in the formation of fetal Hb (α2γ2 [HbF]). As the fetus approaches birth, γ-globin production slowly decreases as β-globin production increases. Shortly after birth, β- and δ-globin production have replaced γ-globin production, resulting in the production of adult Hb (α2β2 [HbA1] and α2δ2 [HbA2]). This figure was first published in Bunn HF, et al. Human Hemoglobins. Philadelphia, PA: WB Saunders; 1977; adapted with kind permission from Elsevier. (C) Deletion or inactivation of one of the α-globin genes in a linked pair is designated α+-thalassemia. α+-Thalassemia is “heterozygous” when on only one of the two chromosome 16 and “homozygous” when on both. Deletion or inactivation of both α-globin genes in a linked pair on the same chromosome 16 is referred to as α0thalassemia. Abbreviations: βLCR, β-locus control region; Hb, hemoglobin; α1, fetal/adult α-gene 1; α2, fetal/adult α-gene 2; β, postnatal β-gene; δ, postnatal δ-gene; ε, embryonic ε-gene; γ1, embryonic/fetal γ-gene 1; γ2, embryonic/fetal γ-gene 2; ζ, embryonic ζ-gene; MCS, multispecies conserved sequence. 2 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx molecule [19,22,23]. Expression of human globin genes switches during transition from embryo to fetus to adult (Fig. 1B) [24,25]. Thus, the composition of Hb tetramers also changes from embryonic hemoglobins ζ2γ2 (Hb Portland), ζ2ε2 (Hb Gower 1), and α2ε2 (Hb Gower 2) embryonic Hb; to α2γ2 (HbF) fetal Hb; and then to α2β2 (HbA1) and α2δ2 (HbA2) adult Hb. The usual proportion of Hb types in normal adults is 95% to 98% HbA1 (α2β2), 2% to 3% HbA2 (α2δ2), and < 2% HbF (α2γ2) [17,26,27]. reduced red cell survival and a varying degree of hemolytic anemia [33,34]. The amount of α-globin produced impacts the amount of β-globin or γ-globin available to form HbH or Hb Barts. Newborns who inherit HbH disease (one functional α-globin gene) have 20% to 40% of Hb Barts at birth [32,35]. 3.2. Clinical forms and definitions α-Thalassemia presents in 3 carrier states (heterozygous or homo­ zygous α+- and α0-thalassemia) and 2 clinically relevant forms (HbH disease and Hb Barts hydrops fetalis syndrome; Fig. 2). Persons with heterozygous α+-thalassemia (silent carrier or α-thalassemia minima; 3 functional genes) are either clinically silent (asymptomatic, with normal red cell parameters, and normal HbA and HbF) or show normal HbA and HbF with mild hematological changes (eg, mild anemia with low mean corpuscular volume [MCV]) [27,36,37]. Persons with α0-thalassemia or homozygous α+-thalassemia (α-thalassemia trait or α-thalassemia minor; 2 functional genes) are usually asymptomatic but have a mild microcytic anemia with normal proportions of HbA and HbF, depending on the amount of α-globin produced [27,37]. Individuals with HbH disease (α-thalassemia intermedia; 1 func­ tional gene) have considerable variability in clinical severity that cor­ relates with the type of mutation and extent of α-globin deficit [36,38,39]. When the disease stems from α-globin gene deletions (termed deletional HbH), anemia is mild to moderate and may require intermittent blood transfusions during periods of stress-induced hemo­ lysis [38,40]. Studies in China, Canada, and California have shown that >50% of patients with HbH disease have deletional mutations [40–42]. When the genotype involves a nondeletional mutation, (termed non­ deletional HbH), the disease is often associated with an earlier and more serious phenotype, with profound microcytic anemia with hypochromia (often in utero); low absolute amounts of HbA and HbF; and transfusion dependence and the need for regular iron chelation [38,40–43]. The most common nondeletional HbH genotype is Hb Constant Spring (HbCS; − − / αCS α) due to a mutation in the termination codon of the α2globin gene that creates an unstable mRNA transcript. Any globin translated from this mRNA is unstable because of translation read­ through [44–46]. Recently, scoring systems have been proposed that may help eval­ uate HbH disease severity and support making treatment decisions in clinical practice [47,48]. In the first, the final validated scoring system comprised age at diagnosis 800 μg/L (and corresponding LIC >5 mg/g) have been associated with increased he­ patic, endocrine, and vascular morbidity and with increased mortality in NTD β-thalassemia and are used to flag the need for iron chelation therapy [106–109]. Similar associations between elevated serum ferritin and LIC and hepatic disease have also been observed in patients with HbH disease [97,110], though the prevalence of iron overload in NTD α-thalassemia is likely much lower than in NTD β-thalassemia because of the lower degree of ineffective erythropoiesis. Evaluating serial serum ferritin levels is the easiest and most widely available way to assess iron overload; however, results may be affected by factors beyond body iron stores, such as hemolysis, ascorbate defi­ ciency, inflammation, and liver disease. When available, assess­ ment of LIC and cardiac T* by noninvasive MRI techniques can provide valuable information on hepatic and cardiac iron overload and are strongly recommended to allow tailoring of iron chelation therapy. Recommendations for the monitoring (and management, see later sec­ tion [Iron Chelation]) of iron overload in patients with HbH disease are summarized in Fig. 4. 7.4. Specific morbidities Several complications linked to chronic anemia and ineffective erythropoiesis include hyperhemolysis, endocrinopathies (especially diabetes mellitus) and bone disease, cardiovascular disease, chronic leg ulcers, liver complications, gallstones, and EMH. 7.4.1. Hyperhemolysis Although most patients with HbH disease remain clinically healthy without regular transfusions, sporadic episodes of hyperhemolysis requiring RBC transfusions may occur. Hyperhemolysis leading to se­ vere anemia may be triggered by acute infection and high fever, or other stressors such as surgery. Hb levels may rapidly drop to as low as ≤3 g/dL , accompanied by hemoglobinemia and hemoglobinuria (intravascular hemolysis) and resulting in renal damage and insuffi­ ciency, and ultimately acute renal failure. Patients must therefore be continually monitored, especially during acute infections and py­ rexia, oxidative stress, hypersplenism, or pregnancy. 7.4.2. Endocrinopathies and bone disease The medullary expansion that follows from ineffective erythropoiesis leads to many of the bone complications observed in children and adults with thalassemia, including bone deformities and osteoporosis [75,76,113]. In addition, an association between iron overload and growth retardation, delayed sexual development, and endocrinopathies has been well established in patients with TD β-thalassemia [113–116]. A study of 361 TCRN patients with any thalassemia syndrome showed Fig. 4. Monitoring and management of anemia and iron overload in hemoglobin H disease. Common adverse events for iron chelators include: DFO, ocular and auditory symptoms, bone-growth retardation, local reactions, and allergy; DFP, gastrointestinal symptoms, arthralgia, agranulocytosis, and neutropenia; DFX, gastrointestinal symptoms, increased creatinine levels, and increased hepatic enzyme levels. Adherence should be regularly monitored in all patients on iron che­ lation. Abbreviations: AEs, adverse events; DFO, deferoxamine; DFP, deferiprone; DFX, deferasirox; Hb, hemoglobin; LIC, liver iron concentration in mg/g; mo, months; SF, serum ferritin in μg/L; Q, every. 9 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx that 60% to 75% of adult patients showed reduced bone mass for their age. In addition, many patients aged 20 to 30 years who may not yet have attained peak bone mass, had early onset of osteopenia and osteoporosis, especially those dependent on transfusions. Estimated mean (95% confidence interval) spine bone mineral density (BMD) Zscores of a 20 year old with deletional and nondeletional HbH disease were − 1.64 (− 2.16,− 1.11) and − 2.00 (− 2.52,− 1.49), respectively, indicating suboptimal peak bone mass. High osteoporosis rates were also observed in a single-center, prospective study of patients with NTDT in Thailand (26.3%). Once again, most of the data are from patients with β-thalassemia and data specific to α-thalassemia are relatively sparse. An analysis in Guangxi Province, China of 200 children with HbH disease (deletional and nondeletional) aged 3 to 12 years showed that 68.5% of patients had growth retardation, 84.0% hypogonadism, 14.5% hypoparathyroidism, 13% hypothyroidism, and 7% diabetes mellitus, all significantly more prevalent compared with age-matched normal controls (p ≤ 0.001). A prospective study in Thailand of 57 adult patients with NTDT aged 18 to 74 years (59.6% β-thalassemia and 40.4% HbH disease) showed that in patients with HbH disease (n = 23), 13.0% had osteo­ porosis, 4.3% hypogonadism, and 4.3% diabetes mellitus. In Thailand, 40 children, adolescents, and young adults aged 10 to 25 years with HbH disease (nondeletional n = 17; deletional n = 23) were fol­ lowed for 2 years. There was a high prevalence of patients (52.5%) with an abnormal insulinogenic index, which indicated abnormal early-phase insulin secretion from β-cells, with no significant difference between nondeletional and deletional HbH disease. with thalassemia [52,53,97,110]. Moreover, hepatitis B or C are inde­ pendent risk factors, which together with iron overload can hasten progression, even leading to the development of hepatocellular carci­ noma. Liver disease is recently regarded as a major cause of mor­ tality in both NTDT and TDT patients, especially those not receiving adequate iron chelation therapy [122–124]. 7.4.5. Gallstones Approximately 38% to 52% of patients with HbH disease have “silent gallstones” [45,110]. These patients may develop acute and/or chronic cholecystitis (with or without known gallstones), which is very difficult to diagnose based on clinical symptoms alone. Gallstone formation is a factor of the degree of anemia and hemolysis. Patients with thalassemia (including those with HbH disease) who present with “peptic ulcer–like” symptoms should be referred for ultrasonography if symptoms do not dissipate with treatment [52,135]. Patients with HbH disease who are homozygotes or double heterozygotes for Gilbert alleles have a significantly increased risk of gallstones and jaundice. 7.4.6. Leg ulcers Leg ulcers are a painful and indolent complication of NTDT , which are attributed to chronic hypoxia, local iron overload deposits, and hypercoagulability-related ischemia; they may increase disability and disrupt quality of life. 7.4.7. Extramedullary hematopoiesis Extramedullary hematopoiesis is common in patients with thalas­ semia not requiring regular transfusions (NTDT]) [76,138]. It is thought that >80% of EMH cases may remain asymptomatic, with pseudotumors discovered only incidentally by radiologic techniques. A 3-year case-control study showed that of 17 patients with HbH disease enrolled and screened by computed tomography or MRI, 47% had EMH pseudotumors. In other observational studies, prevalence rates were lower than those reported in β-thalassemia and ranged between 0 and 8% [110,140,141]. Although different anatomical locations may be involved, paraspinal involvement is most concerning due to poten­ tially debilitating permanent clinical consequences secondary to spinal cord compression. EMH in the spleen and liver can lead to hep­ atosplenomegaly , which is more common in nondeletional HbH disease. 7.4.3. Cardiovascular disease Cardiomyopathy and arrhythmias are the main consequences of cardiac iron overload in patients with TDT, especially those reaching serum ferritin values >2500 μg/L and cardiac T2* values 5 mg/g considering iron-related morbidity and mortality above these thresholds (starting age of 10 years, or higher in HbH disease). In a small study of 17 patients with HbH disease who were not transfusion dependent, treatment with DFP significantly decreased serum ferritin levels after 6 months and 18 months of treat­ ment, and this decrease was maintained until 24 months. The first study to investigate a chelating agent in a large cohort of patients with NTDT was the THALASSA (Assessment of Exjade in NonTransfusionDependent Thalassemia) trial (2008–2011; reported in 2012). In this phase 2, prospective, randomized, double-blind, placebo-controlled trial, the efficacy and safety of DFX were evaluated in 166 patients ≥10 years of age with NTDT and iron overload, including 22 patients with α-thalassemia. At 1 year, both LIC and serum ferritin levels decreased significantly with DFX treatment compared with placebo, with re­ ductions notably seen in α-thalassemia patients receiving dispersible tablet doses as low 5 mg/kg/day. The frequency of adverse events (AEs) in patients receiving DFX was similar to placebo. The most com­ mon drug-related AEs were nausea (6.6%), rash (4.8%), and diarrhea (3.6%). The study also included a preplanned, 1-year, open-label extension phase. Here, LIC continued to decrease, and almost 40% of patients achieved LIC 60% of women carrying a fetus with Hb Barts hydrops fetalis syndrome develop hypertension during the pregnancy, 30% develop severe preeclampsia, and 11% develop antepartum hemorrhage [204,206]. Other serious complications may include worsening anemia, premature labor, placental abruption, oligohydramnios, potential miscarriage, renal failure, and congestive heart failure [205–208]. Postpartum complications can include retained placenta, hemorrhage, life-threatening hypertension, puerperal pyrexia, and anemia [209–211]. Thus, patients should be carefully monitored for these issues throughout the pregnancy and after delivery. 10.2. Intrauterine allogeneic stem cell transplantation Intrauterine allogeneic stem cell transplantation is currently an experimental approach that attempts to exploit the bidirectional maternal-fetal tolerance during pregnancy to correct the anemia before the baby is born. A single-center, non-randomized study (NCT02986698) enrolled fetuses with Hb Barts hydrops fetalis syn­ drome between 18 and 26 weeks’ gestation diagnosed by chorionic villus sampling, amniocentesis, cordocentesis, or by identification of parents as genetic carriers, and identification of fetal anemia or signs of impending hydrops without a second major anatomic anomaly (not related to thalassemia). Fetuses underwent a single intrauterine 9.4. Pregnancy in patients with HbH disease Key aspects of monitoring and management of pregnancy in patients with HbH disease are summarized in Table 1. 13 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx infusion of CD34+ cells from 300 mL of maternal marrow, followed by intrauterine red cell transfusion every 3 weeks. Two patients have un­ dergone this procedure thus far. Both had low levels of engraftment. One patient showed reactivity against maternal antigens, while the other was offered but declined booster transplant. Practice points In patients with α-thalassemia, globin chain imbalance leads to ineffective erythropoiesis and hemolysis, resulting in chronic ane­ mia, iron overload, and associated short- and long-term complications. There is a direct correlation between a patient’s genotype (number of α-globin genes affected, with deletional or nondeletional mutations) and severity of clinical phenotype. Blood transfusion is the only available intervention for patients with symptomatic anemia, while iron chelation therapy is indicated in patients with evidence of iron overload (due to increased intestinal absorption or regular transfusion therapy). 10.3. Gene therapy Early clinical studies are evaluating the safety and efficacy of autologous HSCT using autologous CD34+ HSCs transduced with a lentiviral vector encoding the human α-globin gene for treating transfusion-dependent patients with α-thalassemia (HGI-002 [NCT05851105]; GMCN-508 A [NCT05757245]). Research agenda 10.3.1. Disease modifying agents Mitapivat is an oral, small-molecule allosteric activator of the RBCspecific form of pyruvate kinase (PK) that leads to increased adenosine triphosphate (ATP) production in RBCs and improves their survival in thalassemia mouse models [213–215]. Mitapivat is approved in the US for the treatment of hemolytic anemia in adults with PK deficiency and in the EU for the treatment of PK deficiency in adult patients. In an openlabel, multicenter phase 2 study, 20 patients with NTDT (α-thalassemia, n = 5; β-thalassemia, n = 15) and Hb levels ≤10 g/dL received mitapivat 50 mg twice daily for 6 weeks and then 100 mg twice daily for 18 weeks ; 80% (p < 0.0001) showed a Hb response, defined as Hb levels increasing ≥1.0 g/dL from baseline at ≥1 assessments between weeks 4 and 12, inclusive. An Hb response was observed in all 5 patients with α-thalassemia. Improvements in markers of hemolysis and inef­ fective erythropoiesis were also seen. Mitapivat was well tolerated in this study, and global phase 3 trials of mitapivat for treating adults with TD α-thalassemia and β-thalassemia (ENERGIZE-T; NCT04770779) and NTD α-thalassemia and β-thalassemia (ENERGIZE; NCT04770753) are ongoing. The efficacy and safety of another PK activator, etavopivat, is currently being studied in patients with α-thalassemia, β-thalassemia, or sickle cell disease in a phase 2 trial (NCT04987489). As a result of ineffective erythropoiesis, hepcidin expression is sup­ pressed through various erythroid factors, resulting in increased iron absorption and primary iron overload [216,217]. Hepcidin is under the negative control of transmembrane serine protease 6 (TMPRSS6). Theoretically, inhibition of TMPRSS6 expression could increase hepci­ din production and reduce anemia and iron overload. A phase 1 single-dose study of a small interfering RNA (siRNA) conjugate (SLN124) optimized for hepatic targeting of TMPRSS6 is being con­ ducted in adults with NTD α-thalassemia and β-thalassemia (NCT04718844). Luspatercept, a recombinant fusion protein comprising a modified extracellular domain of activin receptor type IIB fused to the FC domain of human IgG1, is an erythroid maturation agent (ie, affects late-stage erythropoiesis) approved to treat anemia secondary to β-thalassemia in adult patients requiring regular RBC transfusions (US and EU) as well as those with NTD β-thalassemia (EU) [220–222]. The efficacy and safety of luspatercept in adult patients with HbH disease is presently being studied in a multinational phase 2 clinical trial (NCT05664737). Development of local disease registries to inform epidemiology. Establishment of collaborative longitudinal cohorts to evaluate dis­ ease outcomes, treatment patterns, and risk factors for morbidity and mortality. Development of novel therapies targeting the underlying genetic anomaly or the ineffective erythropoiesis. Author contributions All authors contributed to manuscript conceptualization, critical review of all drafts, and provision of suggestions on the scientific con­ tent. The authors retained full editorial control and provided final approval on all content. Role of funding source Medical writing and editorial support were provided by Symbiotix, LLC, and funded by Agios Pharmaceuticals. Data sharing and data availability Data sharing is not applicable to this article as no new data were created or analyzed in this review. Declaration of competing interest K.M.M. reports consultancy fees from Novartis, Celgene Corp (Bristol Myers Squibb), Agios Pharmaceuticals, CRISPR Therapeutics, Vifor Pharma, and Pharmacosmos; and research funding from Agios Phar­ maceuticals and Pharmacosmos. M.D.C. reports consultancy fees from Novartis, Celgene/Bristol Myers Squibb, Vifor Pharma, and Ionis Phar­ maceuticals; and research funding from Novartis, Celgene/Bristol Myers Squibb, La Jolla Pharmaceutical Company, Roche, Protagonist Thera­ peutics, and CRISPR Therapeutics. T.D.C. provides advisory support to Agios Pharma, Bristol Meyers Squibb, and Chiesi. K.H.M.K. reports consultancy fees from Agios Pharmaceuticals, Alexion Pharmaceuticals, Bristol Myers Squibb, Forma Therapeutics, Pfizer, NovoNordisk, and Vertex Pharmaceuticals; honoraria from Agios Pharmaceuticals and Bristol Myers Squibb; membership on an advisory committee for Bio­ verativ/Sanofi/Sangamo; and research funding from Agios Pharma­ ceuticals and Pfizer. H.A-S. reports consultancy fees from Novartis, Forma Therapeutics, Agios Pharmaceuticals, argenx, Moderna, Phar­ macosmos, and Sobi; and research funding from Agios Pharmaceuticals, Amgen, Sobi, Novartis, and Vaderis Therapeutics. S.S. reports consul­ tancy fees from Agios Pharmaceuticals, bluebird bio, Fulcrum Thera­ peutics, Chiesi, Celgene Corp (Bristol Myers Squibb), and Vertex; honoraria for CME activities from Plexus, CCO, and Physicians’ Educa­ tion Resource; advisory board travel from Agios Pharmaceuticals, Cel­ gene Corp (Bristol Myers Squibb), and bluebird bio; research funding from Celgene Corp (Bristol Myers Squibb), Agios Pharmaceuticals, and 11. Conclusion and future considerations α-Thalassemia is a genetic disorder with complex pathophysiology and multi-organ involvement. There is a need for heightened awareness of α-thalassemia in the medical community so that accurate diagnosis, early intervention to treat complications, and successful management of the condition is achieved. Beyond the treating hematologist or PCP, a multi-disciplinary team of experts is needed for monitoring and to deal with emerging complications in vital organs. Collaborative research efforts are also needed to establish the evidence-base on disease epide­ miology and unmet needs, to support the continued development of novel therapies and management approaches. 14 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx Forma Therapeutics; and serving on a clinical trial steering committee for CRISPR/Vertex CTX001 for thalassemia. V.V. reports consultancy fees from Celgene Corp (Bristol Myers Squibb), Agios Pharmaceuticals, Novartis, Vifor Pharma, Pharmacosmos, IONIS Pharmaceuticals, Inc., and DisperSol Technologies, LLC; and research funding from Celgene Corp (Bristol Myers Squibb), Agios Pharmaceuticals, Novartis, Vifor Pharma, Pharmacosmos, IONIS Pharmaceuticals, Inc., DisperSol Tech­ nologies, LLC, and The Government Pharmaceutical Organization (GPO). A.T.T. reports consultancy fees from Novartis, Celgene Corp (Bristol Myers Squibb), Agios Pharmaceuticals, Vifor Pharma, and Pharmacosmos; and research funding from Novartis, Celgene Corp (Bristol Myers Squibb), Agios Pharmaceuticals, Vifor Pharma, and Pharmacosmos. Goh SH, Lee YT, Bhanu NV, Cam MC, Desper R, Martin BM, et al. A newly discovered human alpha-globin gene. Blood 2005;106(4):1466–72. Turilli-Ghisolfi ES, Lualdi M, Fasano M. Ligand-based regulation of dynamics and reactivity of hemoproteins. Biomolecules 2023;13(4):683. Stamatoyannopoulos G. Control of globin gene expression during development and erythroid differentiation. Exp Hematol 2005;33(3):259–71. Wilber A, Nienhuis AW, Persons DA. Transcriptional regulation of fetal to adult hemoglobin switching: new therapeutic opportunities. Blood 2011;117(15): 3945–53. Motiani A, Sonagra AD. Laboratory evaluation of alpha thalassemia. In: StatPearls [internet]. Treasure Islan,. FL: StatPearls Publishing; 2023. Tesio N, Bauer DE. Molecular basis and genetic modifiers of thalassemia. Hematol Oncol Clin North Am 2023;37(2):273–99. Harteveld CL, Higgs DR. Alpha-thalassaemia. Orphanet J Rare Dis 2010;5:13. Galanello R, Cao A. Alpha-thalassemia. Genet Med 2011;13(2):83–8. Liebhaber SA, Cash FE, Ballas SK. Human alpha-globin gene expression. The dominant role of the alpha 2-locus in mRNA and protein synthesis. J Biol Chem 1986;261(32):15327–33. Liebhaber SA, Kan YW. Differentiation of the mRNA transcripts originating from the alpha 1- and alpha 2-globin loci in normals and alpha-thalassemics. J Clin Invest 1981;68(2):439–46. Chui DH, Fucharoen S, Chan V. Hemoglobin H disease: not necessarily a benign disorder. Blood 2003;101(3):791–800. Yuan J, Bunyaratvej A, Fucharoen S, Fung C, Shinar E, Schrier SL. The instability of the membrane skeleton in thalassemic red blood cells. Blood 1995;86(10): 3945–50. Chinprasertsuk S, Wanachiwanawin W, Piankijagum A. Effect of pyrexia in the formation of intraerythrocytic inclusion bodies and vacuoles in haemolytic crisis of haemoglobin H disease. Eur J Haematol 1994;52(2):87–91. Lorey F, Cunningham G, Vichinsky EP, Lubin BH, Witkowska HE, Matsunaga A, et al. Universal newborn screening for Hb H disease in California. Genet Test 2001;5(2):93–100. Galanello R, Cao A. Gene test review. Alpha-thalassemia Genet Med 2011;13(2): 83–8. Kalle Kwaifa I, Lai MI, Md Noor S. Non-deletional alpha thalassaemia: a review. Orphanet J Rare Dis 2020;15(1):166. Viprakasit V. Alpha-thalassemia syndromes: from clinical and molecular diagnosis to bedside management. Hematology Education 2013;7:329–38. Origa R, Sollaino MC, Giagu N, Barella S, Campus S, Mandas C, et al. Clinical and molecular analysis of haemoglobin H disease in Sardinia: haematological, obstetric and cardiac aspects in patients with different genotypes. Br J Haematol 2007;136(2):326–32. Lal A, Goldrich ML, Haines DA, Azimi M, Singer ST, Vichinsky EP. Heterogeneity of hemoglobin H disease in childhood. N Engl J Med 2011;364(8):710–8. Chen FE, Ooi C, Ha SY, Cheung BM, Todd D, Liang R, et al. Genetic and clinical features of hemoglobin H disease in Chinese patients. N Engl J Med 2000;343(8): 544–50. Waye JS, Eng B, Patterson M, Walker L, Carcao MD, Olivieri NF, et al. Hemoglobin H (Hb H) disease in Canada: molecular diagnosis and review of 116 cases. Am J Hematol 2001;68(1):11–5. Kanavakis E, Papassotiriou I, Karagiorga M, Vrettou C, MetaxotouMavrommati A, Stamoulakatou A, et al. Phenotypic and molecular diversity of haemoglobin H disease: a Greek experience. Br J Haematol 2000;111(3):915–23. Vichinsky EP. Changing patterns of thalassemia worldwide. Ann N Y Acad Sci 2005;1054:18–24. Fucharoen S, Viprakasit V. Hb H disease: clinical course and disease modifiers. Hematology Am Soc Hematol Educ Program 2009:26–34. Traivaree C, Boonyawat B, Monsereenusorn C, Rujkijyanont P, Photia A. Clinical and molecular genetic features of Hb H and AE Bart’s diseases in central Thai children. Appl Clin Genet 2018;11:23–30. Songdej D, Tandhansakul M, Wongwerawattanakoon P, Sirachainan N, Charoenkwan P, Chuansumrit A. Severity scoring system to guide transfusion management in pediatric non-deletional HbH. Pediatr Int 2023;65(1):e15568. Liu Y, Zhuang Y, Chen J, Zhong Z, Fang J, Li X, et al. Quantitative evaluation of the clinical severity of hemoglobin H disease in a cohort of 591 patients using a scoring system based on regression analysis. Haematologica 2023. https://doi. org/10.3324/haematol.2023.283211 [Online ahead of print]. Vanaparthy R, Mahdy H. Hydrops Fetalis. In: StatPearls [internet]. Treasure Island (FL): StatPearls Publishing; 2022. Tamary H, Dgany O. Alpha-thalassemia. 2005 Nov 1 [updated 2020 Oct 1]. In: Adam MP, Mirzaa GM, Pagon RA, Wallace SE, bean LJH, Gripp KW, et al., editors. GeneReviews® [internet]. Seattle (WA): University of Washington, Seattle; 19932023. Musallam KM, Cappellini MD, Viprakasit V, Kattamis A, Rivella S, Taher AT. Revisiting the non-transfusion-dependent (NTDT) vs. transfusion-dependent (TDT) thalassemia classification 10 years later. Am J Hematol 2021;96(2) [E54E6]. Cappellini MD, Farmakis D, Porter J, Taher A, editors. 2021 Guidelines for the Management of Transfusion-Dependent Thalassemia (TDT). 4th ed. Nicosia, Cyprus: Thalassaemia International Federation; 2021. Taher A, Musallam K, Cappellini MD, editors. Guidelines for the Management of non-Transfusion–Dependent Thalassaemia (NTDT). 2nd ed. Nicosia, Cyprus: Thalassaemia International Federation; 2017. Taher AT, Musallam KM, Cappellini MD, editors. Guidelines for the Management of non-Transfusion–Dependent β-Thalassaemia. 3rd ed. Nicosia, Cyprus: Thalassaemia International Federation; 2023. Acknowledgments Medical writing and editorial support were provided by Elizabeth Smith, MS, and Linda M Ritter, PhD, of Symbiotix, LLC, and were funded by Agios Pharmaceuticals. The authors received no honorarium/fee or other form of financial support related to the development of this article. References 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. Williams TN, Weatherall DJ. World distribution, population genetics, and health burden of the hemoglobinopathies. Cold Spring Harb Perspect Med 2012;2(9): a011692. Weatherall D, Akinyanju O, Fucharoen S, Olivieri N, Musgrove P. Inherited disorders of hemoglobin. In: Jamison DT, Breman JG, Measham AR, Alleyne G, Claeson M, Evans DB, et al., editors. Disease control priorities in developing countries. 2nd ed. Washington (DC): The International Bank for Reconstruction and Development/The World Bank; 2006. Kattamis A, Kwiatkowski JL, Aydinok Y. Thalassaemia. Lancet 2022;399(10343): 2310–24. Vichinsky EP. Clinical manifestations of alpha-thalassemia. Cold Spring Harb Perspect Med 2013;3(5):a011742. Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ 2008;86(6):480–7. Galanello R, Origa R. Beta-thalassemia. Orphanet J Rare Dis 2010;5:11. Kotila TR. Thalassaemia is a tropical disease. Ann Ib Postgrad Med 2012;10(2): 11–5. Modiano G, Morpurgo G, Terrenato L, Novelletto A, Di Rienzo A, Colombo B, et al. Protection against malaria morbidity: near-fixation of the alpha-thalassemia gene in a Nepalese population. Am J Hum Genet 1991;48(2):390–7. Yenchitsomanus P, Summers KM, Board PG, Bhatia KK, Jones GL, Johnston K, et al. Alpha-thalassemia in Papua New Guinea. Hum Genet 1986;74(4):432–7. Hockham C, Ekwattanakit S, Bhatt S, Penman BS, Gupta S, Viprakasit V, et al. Estimating the burden of alpha-thalassaemia in Thailand using a comprehensive prevalence database for Southeast Asia. Elife 2019:8. Williams TN, Mwangi TW, Wambua S, Peto TE, Weatherall DJ, Gupta S, et al. Negative epistasis between the malaria-protective effects of alpha+− thalassemia and the sickle cell trait. Nat Genet 2005;37(11):1253–7. Weatherall DJ. Genetic variation and susceptibility to infection: the red cell and malaria. Br J Haematol 2008;141(3):276–86. Wambua S, Mwangi TW, Kortok M, Uyoga SM, Macharia AW, Mwacharo JK, et al. The effect of alpha+− thalassaemia on the incidence of malaria and other diseases in children living on the coast of Kenya. PLoS Med 2006;3(5):e158. Ndila CM, Uyoga S, Macharia AW, Nyutu G, Peshu N, Ojal J, et al. Human candidate gene polymorphisms and risk of severe malaria in children in Kilifi, Kenya: a case-control association study. Lancet Haematol 2018;5(8) [e333-e45]. Williams TN, Wambua S, Uyoga S, Macharia A, Mwacharo JK, Newton CR, et al. Both heterozygous and homozygous alpha+ thalassemias protect against severe and fatal plasmodium falciparum malaria on the coast of Kenya. Blood 2005;106 (1):368–71. Weatherall DJ, Clegg JB. Inherited haemoglobin disorders: an increasing global health problem. Bull World Health Organ 2001;79(8):704–12. Angastiniotis M, Vives Corrons JL, Soteriades ES, Eleftheriou A. The impact of migrations on the health services for rare diseases in Europe: the example of haemoglobin disorders. ScientificWorldJournal 2013;2013:727905. Farashi S, Harteveld CL. Molecular basis of alpha-thalassemia. Blood Cells Mol Dis 2018;70:43–53. Higgs DR, Vickers MA, Wilkie AO, Pretorius IM, Jarman AP, Weatherall DJ. A review of the molecular genetics of the human alpha-globin gene cluster. Blood 1989;73(5):1081–104. Antonarakis SE, Boehm CD, Giardina PJ, Kazazian Jr HH. Nonrandom association of polymorphic restriction sites in the beta-globin gene cluster. Proc Natl Acad Sci U S A 1982;79(1):137–41. 15 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx Vitrano A, Meloni A, Addario Pollina W, Karimi M, El-Beshlawy A, Hajipour M, et al. A complication risk score to evaluate clinical severity of thalassaemia syndromes. Br J Haematol 2021;192(3):626–33. Vitrano A, Musallam KM, Meloni A, Karimi M, Daar S, Ricchi P, et al. Development of a thalassemia international prognostic scoring system (TIPSS). Blood Cells Mol Dis 2023;99:102710. El Goundali K, Chebabe M, Zahra Laamiri F, Hilali A. The determinants ofconsanguineous marriages among the Arab population: a systematic review. Iran J Public Health 2022;51(2):253–65. Hamamy H, Antonarakis SE, Cavalli-Sforza LL, Temtamy S, Romeo G, Kate LP, et al. Consanguineous marriages, pearls and perils: Geneva international consanguinity workshop report. Genet Med 2011;13(9):841–7. Hamamy H. Consanguineous marriages: preconception consultation in primary health care settings. J Community Genet 2012;3(3):185–92. Bittles AH. Global consanguinity map. Available from: https://consang.net/map s2/worldmap2022.pdf; 2022. Goh LPW, Chong ETJ, Lee PC. Prevalence of alpha(alpha)-thalassemia in Southeast Asia (2010− 2020): a meta-analysis involving 83,674 subjects. Int J Environ Res Public Health 2020;17(20):7354. Musallam KM, Lombard L, Kistler KD, Arregui M, Gilroy KS, Chamberlain C, et al. Epidemiology of clinically significant forms of alpha- and beta-thalassemia: a global map of evidence and gaps. Am J Hematol 2023;98(9):1436–51. Weatherall DJ. The evolving spectrum of the epidemiology of thalassemia. Hematol Oncol Clin North Am 2018;32(2):165–75. Vichinsky EP, MacKlin EA, Waye JS, Lorey F, Olivieri NF. Changes in the epidemiology of thalassemia in North America: a new minority disease. Pediatrics 2005;116(6):e818–25. Michlitsch J, Azimi M, Hoppe C, Walters MC, Lubin B, Lorey F, et al. Newborn screening for hemoglobinopathies in California. Pediatr Blood Cancer 2009;52 (4):486–90. He J, Song W, Yang J, Lu S, Yuan Y, Guo J, et al. Next-generation sequencing improves thalassemia carrier screening among premarital adults in a high prevalence population: the Dai nationality. China Genet Med 2017;19(9): 1022–31. He J, Zeng H, Zhu L, Li H, Shi L, Hu L. Prevalence and spectrum of thalassaemia in Changsha, Hunan province, China: discussion of an innovative screening strategy. J Genet 2017;96(2):327–32. Bozkurt G. Results from the North Cyprus thalassemia prevention program. Hemoglobin 2007;31(2):257–64. Weatherall DJ. The inherited diseases of hemoglobin are an emerging global health burden. Blood 2010;115(22):4331–6. Halim-Fikri BH, Lederer CW, Baig AA, Mat-Ghani SNA, Syed-Hassan S-NR-K, Yusof W, et al. Global globin network consensus paper: classification and stratified roadmaps for improved thalassaemia care and prevention in 32 countries. J Pers Med 2022;12(4):552. Giordano P. Newborn screening for haemoglobinopathies. In: Old J, editor. Prevention of Thalassaemias and other Haemoglobin disorders: Volume 1: Principles [internet]. 2nd ed. Nicosia, Cyprus: Thalassaemia International Federation; 2013. Giordano PC. Strategies for basic laboratory diagnostics of the hemoglobinopathies in multi-ethnic societies: interpretation of results and pitfalls. Int J Lab Hematol 2013;35(5):465–79. Giordano PC. Newborn screening for hemoglobinopathies using capillary electrophoresis. Methods Mol Biol 2013;919:131–45. Mills A. Health care systems in low- and middle-income countries. N Engl J Med 2014;370(6):552–7. Rivella S. Ineffective erythropoiesis and thalassemias. Curr Opin Hematol 2009; 16(3):187–94. Rivella S. The role of ineffective erythropoiesis in non-transfusion-dependent thalassemia. Blood Rev 2012;26(Suppl. 1):S12–5. Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia. Lancet 2018;391(10116): 155–67. Chansai S, Yamsri S, Fucharoen S, Fucharoen G, Teawtrakul N. Phosphatidylserine-exposed red blood cells and ineffective erythropoiesis biomarkers in patients with thalassemia. Am J Transl Res 2022;14(7):4743–56. Sabath DE. Molecular diagnosis of thalassemias and hemoglobinopathies: an ACLPS critical review. Am J Clin Pathol 2017;148(1):6–15. Tubman VN, Fung EB, Vogiatzi M, Thompson AA, Rogers ZR, Neufeld EJ, et al. Guidelines for the standard monitoring of patients with thalassemia: report of the thalassemia longitudinal cohort. J Pediatr Hematol Oncol 2015;37(3):e162–9. Taher AT, Cappellini MD. How I manage medical complications of betathalassemia in adults. Blood 2018;132(17):1781–91. Taher AT, Musallam KM, Cappellini MD. beta-Thalassemias. N Engl J Med 2021; 384(8):727–43. Galanello R, Pirastu M, Melis MA, Paglietti E, Moi P, Cao A. Phenotype-genotype correlation in haemoglobin H disease in childhood. J Med Genet 1983;20(6): 425–9. Galanello R, Aru B, Dessì C, Addis M, Paglietti E, Melis MA, et al. HbH disease in Sardinia: molecular, hematological and clinical aspects. Acta Haematol 1992;88 (1):1–6. Langer AL, Lombard L, Gilroy K, Li J, Zhao J, Lew CR, et al. Clinical burden of alpha- and beta-thalassemia compared to matched controls in therealworldsetting. Blood 2022;140(Suppl. 1):5362–4. Songdej D, Fucharoen S. Alpha-thalassemia: diversity of clinical phenotypes and update on the treatment. Thalassemia Reports 2022;12(4):157–72. Singer ST, Kim HY, Olivieri NF, Kwiatkowski JL, Coates TD, Carson S, et al. Hemoglobin H-constant spring in North America: an alpha thalassemia with frequent complications. Am J Hematol 2009;84(11):759–61. Musallam KM, Cappellini MD, Taher AT. Variations in hemoglobin level and morbidity burden in non-transfusion-dependent beta-thalassemia. Ann Hematol 2021;100(7):1903–5. Musallam KM, Taher AT, Cappellini MD, Hermine O, Kuo KHM, Sheth S, et al. Untreated anemia in nontransfusion-dependent beta-thalassemia: time to sound the alarm. Hemasphere 2022;6(12):e806. Kuo KHM, Layton DM, Lal A, Al-Samkari H, Bhatia J, Kosinski PA, et al. Safety and efficacy of mitapivat, an oral pyruvate kinase activator, in adults with nontransfusion dependent α-thalassaemia or β-thalassaemia: an open-label, multicentre, phase 2 study. Lancet 2022;400(10351):493–501. Dhanya R, Sedai A, Ankita K, Parmar L, Agarwal RK, Hegde S, et al. Life expectancy and risk factors for early death in patients with severe thalassemia syndromes in South India. Blood Adv 2020;4(7):1448–57. Wallace DF. The regulation of iron absorption and homeostasis. Clin Biochem Rev 2016;37(2):51–62. Musallam KM, Cappellini MD, Wood JC, Taher AT. Iron overload in nontransfusion-dependent thalassemia: a clinical perspective. Blood Rev 2012;26 (Suppl. 1):S16–9. Taher AT, Saliba AN. Iron overload in thalassemia: different organs at different rates. Hematology Am Soc Hematol Educ Program 2017:265–71. Tso SC, Loh TT, Todd D. Iron overload in patients with haemoglobin H disease. Scand J Haematol 1984;32(4):391–4. Hsu HC, Lin CK, Tsay SH, Tse E, Ho CH, Chow MP, et al. Iron overload in Chinese patients with hemoglobin H disease. Am J Hematol 1990;34(4):287–90. Chan LKL, Mak VWM, Chan SCH, Yu ELM, Chan NCN, Leung KFS, et al. Liver complications of haemoglobin H disease in adults. Br J Haematol 2021;192(1): 171–8. Huang Y, Yang G, Wang M, Wei X, Pan L, Liu J, et al. Iron overload status in patients with non-transfusion-dependent thalassemia in China. Ther Adv Hematol 2022;13 (20406207221084639). Au WY, Lam WW, Chu WW, Tam S, Wong WK, Lau J, et al. Organ-specific hemosiderosis and functional correlation in Chinese patients with thalassemia intermedia and hemoglobin H disease. Ann Hematol 2009;88(10):947–50. Taher A, El Rassi F, Isma’eel H, Koussa S, Inati A, Cappellini MD. Correlation of liver iron concentration determined by R2 magnetic resonance imaging with serum ferritin in patients with thalassemia intermedia. Haematologica 2008;93 (10):1584–6. Musallam KM, Cappellini MD, Taher AT. Iron overload in beta-thalassemia intermedia: an emerging concern. Curr Opin Hematol 2013;20(3):187–92. Borgna-Pignatti C, Rugolotto S, De Stefano P, Zhao H, Cappellini MD, Del Vecchio GC, et al. Survival and complications in patients with thalassemia major treated with transfusion and deferoxamine. Haematologica 2004;89(10): 1187–93. Kirk P, Roughton M, Porter JB, Walker JM, Tanner MA, Patel J, et al. Cardiac T2* magnetic resonance for prediction of cardiac complications in thalassemia major. Circulation 2009;120(20):1961–8. Angelucci E, Brittenham GM, McLaren CE, Ripalti M, Baronciani D, Giardini C, et al. Hepatic iron concentration and total body iron stores in thalassemia major. N Engl J Med 2000;343(5):327–31. Jensen PD, Jensen FT, Christensen T, Eiskjaer H, Baandrup U, Nielsen JL. Evaluation of myocardial iron by magnetic resonance imaging during iron chelation therapy with deferrioxamine: indication of close relation between myocardial iron content and chelatable iron pool. Blood 2003;101(11):4632–9. Musallam KM, Cappellini MD, Daar S, Karimi M, El-Beshlawy A, Graziadei G, et al. Serum ferritin level and morbidity risk in transfusion-independent patients with beta-thalassemia intermedia: the ORIENT study. Haematologica 2014;99 (11):e218–21. Musallam KM, Cappellini MD, Wood JC, Motta I, Graziadei G, Tamim H, et al. Elevated liver iron concentration is a marker of increased morbidity in patients with beta thalassemia intermedia. Haematologica 2011;96(11):1605–12. Musallam KM, Cappellini MD, Taher AT. Evaluation of the 5mg/g liver iron concentration threshold and its association with morbidity in patients with betathalassemia intermedia. Blood Cells Mol Dis 2013;51(1):35–8. Musallam KM, Vitrano A, Meloni A, Pollina SA, Karimi M, El-Beshlawy A, et al. Risk of mortality from anemia and iron overload in nontransfusion-dependent beta-thalassemia. Am J Hematol 2022;97(2):E78–80. Ekwattanakit S, Siritanaratkul N, Viprakasit V. A prospective analysis for prevalence of complications in Thai nontransfusion-dependent Hb E/betathalassemia and alpha-thalassemia (Hb H disease). Am J Hematol 2018;93(5): 623–9. Shah FT, Porter JB, Sadasivam N, Kaya B, Moon JC, Velangi M, et al. Guidelines for the monitoring and management of iron overload in patients with haemoglobinopathies and rare anaemias. Br J Haematol 2022;196(2):336–50. Jetsrisuparb A, Sanchaisuriya K, Fucharoen G, Fucharoen S, Wiangnon S, Jetsrisuparb C, et al. Development of severe anemia during fever episodes in patients with hemoglobin E trait and hemoglobin H disease combinations. J Pediatr Hematol Oncol 2006;28(4):249–53. Wong P, Fuller PJ, Gillespie MT, Milat F. Bone disease in thalassemia: a molecular and clinical overview. Endocr Rev 2016;37(4):320–46. Wood JC. Impact of iron assessment by MRI. Hematology Am Soc Hematol Educ Program 2011:443–50. 16 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx Belhoul KM, Bakir ML, Saned MS, Kadhim AM, Musallam KM, Taher AT. Serum ferritin levels and endocrinopathy in medically treated patients with beta thalassemia major. Ann Hematol 2012;91(7):1107–14. Taher AT, Musallam KM, Inati A. Iron overload: consequences, assessment, and monitoring. Hemoglobin 2009;33(Suppl. 1). S46-57m. Vogiatzi MG, Macklin EA, Fung EB, Cheung AM, Vichinsky E, Olivieri N, et al. Bone disease in thalassemia: a frequent and still unresolved problem. J Bone Miner Res 2009;24(3):543–57. Luo HC, Luo QS, Huang FG, Wang CF, Wei YS. Impact of genotype on endocrinal complications of children with alpha-thalassemia in China. Sci Rep 2017;7(1): 2948. Nakavachara P, Kajchamaporn W, Pooliam J, Viprakasit V. Early development of decreased beta-cell insulin secretion in children and adolescents with hemoglobin H disease and its relationship with levels of anemia. Pediatr Blood Cancer 2020; 67(4):e28109. Olivieri NF, Nathan DG, MacMillan JH, Wayne AS, Liu PP, McGee A, et al. Survival in medically treated patients with homozygous beta-thalassemia. N Engl J Med 1994;331(9):574–8. Modell B, Khan M, Darlison M, Westwood MA, Ingram D, Pennell DJ. Improved survival of thalassaemia major in the UK and relation to T2* cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2008;10(1):42. Forni GL, Gianesin B, Musallam KM, Longo F, Rosso R, Lisi R, 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):381–7. Voskaridou E, Kattamis A, Fragodimitri C, Kourakli A, Chalkia P, Diamantidis M, et al. National registry of hemoglobinopathies in Greece: updated demographics, current trends in affected births, and causes of mortality. Ann Hematol 2019;98 (1):55–66. Musallam KM, Vitrano A, Meloni A, Pollina SA, Karimi M, El-Beshlawy A, et al. Survival and causes of death in 2,033 patients with non-transfusion-dependent beta-thalassemia. Haematologica 2021;106(9):2489–92. Chansai S, Fucharoen S, Fucharoen G, Jetsrisuparb A, Chumpia W. Elevations of thrombotic biomarkers in hemoglobin H disease. Acta Haematol 2018;139(1): 47–51. Taher AT, Musallam KM, Karimi M, El-Beshlawy A, Belhoul K, Daar S, et al. Overview on practices in thalassemia intermedia management aiming for lowering complication rates across a region of endemicity: the OPTIMAL CARE study. Blood 2010;115(10):1886–92. Taher AT, Musallam KM, Karimi M, El-Beshlawy A, Belhoul K, Daar S, et al. Splenectomy and thrombosis: the case of thalassemia intermedia. J Thromb Haemost 2010;8(10):2152–8. Taher AT, Cappellini MD, Bou-Fakhredin R, Coriu D, Musallam KM. Hypercoagulability and vascular disease. Hematol Oncol Clin North Am 2018;32 (2):237–45. Cappellini MD, Robbiolo L, Bottasso BM, Coppola R, Fiorelli G, Mannucci AP. Venous thromboembolism and hypercoagulability in splenectomized patients with thalassaemia intermedia. Br J Haematol 2000;111(2):467–73. Taher AT, Cappellini MD, Musallam KM. Development of a thalassemia-related thrombosis risk scoring system. Am J Hematol 2019;94(8) [E207-E9]. Pinto VM, Musallam KM, Derchi G, Graziadei G, Giuditta M, Origa R, et al. Mortality in beta-thalassemia patients with confirmed pulmonary arterial hypertension on right heart catheterization. Blood 2022;139(13):2080–3. Derchi G, Galanello R, Bina P, Cappellini MD, Piga A, Lai ME, et al. Prevalence and risk factors for pulmonary arterial hypertension in a large group of betathalassemia patients using right heart catheterization: a Webthal study. Circulation 2014;129(3):338–45. Tantiworawit A, Tapanya S, Phrommintikul A, Saekho S, Rattarittamrong E, Norasetthada L, et al. Prevalence and risk factors for cardiac iron overload and cardiovascular complications among patients with thalassemia in northern Thailand. Southeast Asian J Trop Med Public Health 2016;47(6):1335–42. Yin XL, Zhang XH, Wu ZK, Zhao DH, Zhou YL, Yu YH, et al. Pulmonary hypertension risk in patients with hemoglobin H disease: low incidence and absence of correlation with splenectomy. Acta Haematol 2013;130(3):153–9. Chandrcharoensin-Wilde C, Chairoongruang S, Jitnuson P, Fucharoen S, Vathanopas V. Gallstones in thalassemia. Birth Defects Orig Artic Ser 1988;23 (5B):263–7. Au WY, Cheung WC, Hu WH, Chan GC, Ha SY, Khong PL, et al. Hyperbilirubinemia and cholelithiasis in Chinese patients with hemoglobin H disease. Ann Hematol 2005;84(10):671–4. Daneshmend TK, Peachey RD. Leg ulcers in alpha-thalassaemia (haemoglobin H disease). Br J Dermatol 1978;98(2):233–5. Wu JH, Shih LY, Kuo TT, Lan RS. Intrathoracic extramedullary hematopoietic tumor in hemoglobin H disease. Am J Hematol 1992;41(4):285–8. Huang Y, Liu R, Wei X, Liu J, Pan L, Yang G, et al. Erythropoiesis and iron homeostasis in non-transfusion-dependent thalassemia patients with extramedullary hematopoiesis. Biomed Res Int 2019;2019:4504302. Winichakoon P, Tantiworawit A, Rattanathammethee T, Hantrakool S, ChaiAdisaksopha C, Rattarittamrong E, et al. Prevalence and risk factors for complications in patients with nontransfusion dependent alpha- and betathalassemia. Anemia 2015;2015:793025. Ricchi P, Ammirabile M, Costantini S, Spasiano A, Di Matola T, Verna R, et al. Soluble form of transferrin receptor as a biomarker of overall morbidity in patients with non-transfusion-dependent thalassaemia: a cross-sectional study. Blood Transfus 2016;14(6):538–40. Chao YH, Wu KH, Wu HP, Liu SC, Peng CT, Lee MS. Clinical features and molecular analysis of Hb H disease in Taiwan. Biomed Res Int 2014;2014:271070. Muncie Jr HL, Campbell J. Alpha and beta thalassemia. Am Fam Physician 2009; 80(4):339–44. Zurlo MG, De Stefano P, Borgna-Pignatti C, Di Palma A, Piga A, Melevendi C, et al. Survival and causes of death in thalassaemia major. Lancet 1989;2(8653):27–30. Borgna-Pignatti C, Cappellini MD, De Stefano P, Del Vecchio GC, Forni GL, Gamberini MR, et al. Survival and complications in thalassemia. Ann N Y Acad Sci 2005;1054:40–7. Angastiniotis M, Lobitz S. Thalassemias: an overview. Int J Neonatal Screen 2019; 5(1):16. Chan WY, Leung AW, Luk CW, Li RC, Ling AS, Ha SY. Outcomes and morbidities of patients who survive haemoglobin Bart’s hydrops fetalis syndrome: 20-year retrospective review. Hong Kong Med J 2018;24(2):107–18. Beaudry MA, Ferguson DJ, Pearse K, Yanofsky RA, Rubin EM, Kan YW. Survival of a hydropic infant with homozygous alpha-thalassemia-1. J Pediatr 1986;108(5 Pt 1):713–6. Bianchi DW, Beyer EC, Stark AR, Saffan D, Sachs BP, Wolfe L. Normal long-term survival with alpha-thalassemia. J Pediatr 1986;108(5 Pt 1):716–8. Lee SY, Chow CB, Li CK, Chiu MC. Outcome of intensive care of homozygous alpha-thalassaemia without prior intra-uterine therapy. J Paediatr Child Health 2007;43(7–8):546–50. Yi JS, Moertel CL, Baker KS. Homozygous alpha-thalassemia treated with intrauterine transfusions and unrelated donor hematopoietic cell transplantation. J Pediatr 2009;154(5):766–8. Fung TY, Lau TK, Tam WH, Li CK. In utero exchange transfusion in homozygous alpha-thalassaemia: a case report. Prenat Diagn 1998;18(8):838–41. Torcharus K, Pankaew T. Health-related quality of life in Thai thalassemic children treated with iron chelation. Southeast Asian J Trop Med Public Health 2011;42(4):951–9. Taher AT. Thalassemia. Hematol Oncol Clin North Am 2018;32(2):xv–xvi. Alshamsi S, Hamidi S, Narci HO. Healthcare resource utilization and direct costs of transfusion-dependent thalassemia patients in Dubai, United Arab Emirates: a retrospective cost-of-illness study. BMC Health Serv Res 2022;22(1):304. Musallam KM, Lombard L, Gilroy K, Vinals L, Tam C, Rizzo M. Characterizing the clinical, health-related quality of life and economic burden of alpha-thalassemia: a systematic literature review and evidence gaps assessment. Blood 2022;140 (Suppl. 1):2491–3. Vichinsky E. Advances in the treatment of alpha-thalassemia. Blood Rev 2012;26 (Suppl. 1):S31–4. Tang CH, Furnback W, Wang BCM, Tang J, Tang D, Lu MY, et al. Relationship between transfusion burden, healthcare resource utilization, and complications in patients with beta-thalassemia in Taiwan: a real-world analysis. Transfusion 2021;61(10):2906–17. Betts M, Flight PA, Paramore LC, Tian L, Milenkovic D, Sheth S. Systematic literature review of the burden of disease and treatment for transfusiondependent beta-thalassemia. Clin Ther 2020;42(2):322–37 e2. Hirsh J, Dacie JV. Persistent post-splenectomy thrombocytosis and thromboembolism: a consequence of continuing anaemia. Br J Haematol 1966;12(1): 44–53. Tso SC, Chan TK, Todd D. Venous thrombosis in haemoglobin H disease after splenectomy. Aust N Z J Med 1982;12(6):635–8. Fortenko OM, Schaef Johns GJ, Kudva GC. Erythropoietin for hemoglobin H disease. Ann Hematol 2009;88(2):179–80. Singer ST, Vichinsky EP, Sweeters N, Rachmilewitz E. Darbepoetin alfa for the treatment of anaemia in alpha- or beta- thalassaemia intermedia syndromes. Br J Haematol 2011;154(2):281–4. Cappellini MD, Bejaoui M, Agaoglu L, Canatan D, Capra M, Cohen A, 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):884–93. Davis BA, Porter JB. Long-term outcome of continuous 24-hour deferoxamine infusion via indwelling intravenous catheters in high-risk beta-thalassemia. Blood 2000;95(4):1229–36. Taher AT, Origa R, Perrotta S, Kourakli A, Ruffo GB, Kattamis A, et al. New filmcoated tablet formulation of deferasirox is well tolerated in patients with thalassemia or lower-risk MDS: results of the randomized, phase II ECLIPSE study. Am J Hematol 2017;92(5):420–8. Pennell DJ, Porter JB, Piga A, Lai YR, El-Beshlawy A, Elalfy M, et al. Sustained improvements in myocardial T2* over 2 years in severely iron-overloaded patients with beta thalassemia major treated with deferasirox or deferoxamine. Am J Hematol 2015;90(2):91–6. Pennell DJ, Porter JB, Cappellini MD, Chan LL, El-Beshlawy A, Aydinok Y, et al. Deferasirox for up to 3 years leads to continued improvement of myocardial T2* in patients with beta-thalassemia major. Haematologica 2012;97(6):842–8. Tanner MA, Galanello R, Dessi C, Smith GC, Westwood MA, Agus A, 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): 1876–84. Maggio A, Kattamis A, Felisi M, Reggiardo G, El-Beshlawy A, Bejaoui M, et al. Evaluation of the efficacy and safety of deferiprone compared with deferasirox in paediatric patients with transfusion-dependent haemoglobinopathies (DEEP-2): a multicentre, randomised, open-label, non-inferiority, phase 3 trial. Lancet Haematol 2020;7(6) [e469-e78]. Pennell DJ, Berdoukas V, Karagiorga M, Ladis V, Piga A, Aessopos A, et al. Randomized controlled trial of deferiprone or deferoxamine in beta-thalassemia major patients with asymptomatic myocardial siderosis. Blood 2006;107(9): 3738–44. 17 K.M. Musallam et al. Blood Reviews xxx (xxxx) xxx Chan JC, Chim CS, Ooi CG, Cheung B, Liang R, Chan TK, et al. Use of the oral chelator deferiprone in the treatment of iron overload in patients with Hb H disease. Br J Haematol 2006;133(2):198–205. Taher AT, Porter J, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox reduces iron overload significantly in nontransfusion-dependent thalassemia: 1-year results from a prospective, randomized, double-blind, placebo-controlled study. Blood 2012;120(5):970–7. Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox effectively reduces iron overload in non-transfusion-dependent thalassemia (NTDT) patients: 1-year extension results from the THALASSA study. Ann Hematol 2013;92(11):1485–93. Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox demonstrates a dose-dependent reduction in liver iron concentration and consistent efficacy across subgroups of non-transfusiondependent thalassemia patients. Am J Hematol 2013;88(6):503–6. Lai YR, Cappellini MD, Aydinok Y, Porter J, Karakas Z, Viprakasit V, et al. An open-label, multicenter, efficacy, and safety study of deferasirox in ironoverloaded patients with non-transfusion-dependent thalassemia (THETIS): 5year results. Am J Hematol 2022;97(8) [E281-E4]. JADENU® (deferasirox). package insert. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 2020. Angelucci E. Hematopoietic stem cell transplantation in thalassemia. Hematology Am Soc Hematol Educ Program 2010:456–62. Algeri M, Lodi M, Locatelli F. Hematopoietic stem cell transplantation in thalassemia. Hematol Oncol Clin North Am 2023;37(2):413–32. Jaing TH. Is the benefit-risk ratio for patients with transfusion-dependent thalassemia treated by unrelated cord blood transplantation favorable? Int J Mol Sci 2017;18(11):2472. Ruggeri A, Eapen M, Scaravadou A, Cairo MS, Bhatia M, Kurtzberg J, et al. Umbilical cord blood transplantation for children with thalassemia and sickle cell disease. Biol Blood Marrow Transplant 2011;17(9):1375–82. La Nasa G, Argiolu F, Giardini C, Pession A, Fagioli F, Caocci G, et al. Unrelated bone marrow transplantation for beta-thalassemia patients: the experience of the Italian bone marrow transplant group. Ann N Y Acad Sci 2005;1054:186–95. Li C, Mathews V, Kim S, George B, Hebert K, Jiang H, et al. Related and unrelated donor transplantation for β-thalassemia major: results of an international survey. Blood Adv 2019;3(17):2562–70. Horvei P, MacKenzie T, Kharbanda S. Advances in the management of α-thalassemia major: reasons to be optimistic. Hematology Am Soc Hematol Educ Program 2021:592–9. Chik KW, Shing MM, Li CK, Leung TF, Tsang KS, Yuen HL, et al. Treatment of hemoglobin Bart’s hydrops with bone marrow transplantation. J Pediatr 1998; 132(6):1039–42. Zhou X, Ha SY, Chan GC, Luk CW, Chan V, Hawkins B, et al. Successful mismatched sibling cord blood transplant in Hb Bart’s disease. Bone Marrow Transplant 2001;28(1):105–7. Thornley I, Lehmann L, Ferguson WS, Davis I, Forman EN, Guinan EC. Homozygous alpha-thalassemia treated with intrauterine transfusions and postnatal hematopoietic stem cell transplantation. Bone Marrow Transplant 2003; 32(3):341–2. Joshi DD, Nickerson HJ, McManus MJ. Hydrops fetalis caused by homozygous alpha-thalassemia and Rh antigen alloimmunization: report of a survivor and literature review. Clin Med Res 2004;2(4):228–32. Gumuscu B, Thompson EI, Grovas AC, Zach TL, Warkentin PI, Coccia PF. Successful unrelated cord blood transplantation for homozygous alphathalassemia. J Pediatr Hematol Oncol 2013;35(7):570–2. Pongtanakul B, Sanpakit K, Chongkolwatana V, Viprakasit V. Normal cognitive functioning in a patient with Hb Bart’s hydrops successfully cured by hematopoietic SCT. Bone Marrow Transplant 2014;49(1):155–6. Elsaid MY, Capitini CM, Diamond CA, Porte M, Otto M, DeSantes KB. Successful matched unrelated donor stem cell transplant in hemoglobin Bart’s disease. Bone Marrow Transplant 2016;51(11):1522–3. Pecker LH, Guerrera MF, Loechelt B, Massaro A, Abraham AA, Fasano RM, et al. Homozygous alpha-thalassemia: challenges surrounding early identification, treatment, and cure. Pediatr Blood Cancer 2017;64(1):151–5. Kreger EM, Singer ST, Witt RG, Sweeters N, Lianoglou B, Lal A, et al. Favorable outcomes after in utero transfusion in fetuses with alpha thalassemia major: a case series and review of the literature. Prenat Diagn 2016;36(13):1242–9. Chan WYK, Lee PPW, Lee V, Chan GCF, Leung W, Ha SY, et al. Outcomes of allogeneic transplantation for hemoglobin Bart’s hydrops fetalis syndrome in Hong Kong. Pediatr Transplant 2021;25(6):e14037. Surapolchai P, Sirachainan N, So CC, Hongeng S, Pakakasama S, Anurathapan U, et al. Curative stem cell transplantation for severe Hb H disease manifesting from early infancy: phenotypic and genotypic analyses. Hemoglobin 2016;40(1):70–3. Musallam K, Cappellini MD, Taher A. Challenges associated with prolonged survival of patients with thalassemia: transitioning from childhood to adulthood. Pediatrics 2008;121(5):e1426–9. Forni GL, Puntoni M, Boeri E, Terenzani L, Balocco M. The influence of treatment in specialized centers on survival of patients with thalassemia major. Am J Hematol 2009;84(5):317–8. Anie KA, Massaglia P. Psychological therapies for thalassaemia. Cochrane Database Syst Rev 2014;2014(3):CD002890. Keshvar Y, Sabeghi S, Sharifi Z, Fatemi KS, Fouladi P, Younesi Khah S, et al. A decade of molecular preimplantation genetic diagnosis of 350 blastomeres for beta-thalassemia combined with HLA typing, aneuploidy screening and sex selection in Iran. BMC Pregnancy Childbirth 2022;22(1):330. Piel FB, Weatherall DJ. The alpha-thalassemias. N Engl J Med 2014;371(20): 1908–16. Vichinsky EP. Alpha thalassemia major–new mutations, intrauterine management, and outcomes. Hematology Am Soc Hematol Educ Program 2009: 35–41. Lee HH, Mak AS, Poon CF, Leung KY. Prenatal ultrasound monitoring of homozygous α(0)-thalassemia-induced fetal anemia. Best Pract Res Clin Obstet Gynaecol 2017;39:53–62. Scotchman E, Shaw J, Paternoster B, Chandler N, Chitty LS. Non-invasive prenatal diagnosis and screening for monogenic disorders. Eur J Obstet Gynecol Reprod Biol 2020;253:320–7. Dame C, Albers N, Hasan C, Bode U, Eigel A, Hansmann M, et al. Homozygous alpha-thalassaemia and hypospadias—common aetiology or incidental association? Long-term survival of Hb Bart’s hydrops syndrome leads to new aspects for counselling of alpha-thalassaemic traits. Eur J Pediatr 1999;158(3): 217–20. Chui DHK, Waye JS. Hydrops fetalis caused by α-thalassemia: an emerging health care problem. Blood 1998;91(7):2213–22. Liang ST, Wong VC, So WW, Ma HK, Chan V, Todd D. Homozygous alphathalassaemia: clinical presentation, diagnosis and management. A review of 46 cases. Br J Obstet Gynaecol 1985;92(7):680–4. Taher AT, Iolascon A, Matar CF, Bou-Fakhredin R, de Franceschi L, Cappellini MD, et al. Recommendations for pregnancy in rare inherited anemias. Hemasphere 2020;4(4):e446. Leung WC, Leung KY, Lau ET, Tang MH, Chan V. Alpha-thalassaemia. Semin Fetal Neonatal Med 2008;13(4):215–22. Tan SL, Tseng AM, Thong PW. Bart’s hydrops fetalis—clinical presentation and management–an analysis of 25 cases. Aust N Z J Obstet Gynaecol 1989;29(3 Pt 1): 233–7. Guy G, Coady DJ, Jansen V, Snyder J, Zinberg S. alpha-thalassemia hydrops fetalis: clinical and ultrasonographic considerations. Am J Obstet Gynecol 1985; 153(5):500–4. Bowman E, Watts J, Burrows R, Chui DH. Hemoglobin barts hydrops fetalis syndrome. Haematologia (Budap) 1987;20(3):125–30. MacKenzie TC, Frascoli M, Sper R, Lianoglou BR, Gonzalez Velez J, Dvorak CC, et al. In utero stem cell transplantation in patients with alpha thalassemia major: interim results of a phase 1 clinical trial. Blood 2020;136(Suppl. 1):1. Kung C, Hixon J, Kosinski PA, Cianchetta G, Histen G, Chen Y, et al. AG-348 enhances pyruvate kinase activity in red blood cells from patients with pyruvate kinase deficiency. Blood 2017;130(11):1347–56. Matte A, Kosinski PA, Federti E, Dang L, Recchiuti A, Russo R, et al. Mitapivat, a pyruvate kinase activator, improves transfusion burden and reduces iron overload in beta-thalassemic mice. Haematologica 2023;108(9):2535–41. Matte A, Federti E, Kung C, Kosinski PA, Narayanaswamy R, Russo R, et al. The pyruvate kinase activator mitapivat reduces hemolysis and improves anemia in a beta-thalassemia mouse model. J Clin Invest 2021;131(10). Papanikolaou G, Tzilianos M, Christakis JI, Bogdanos D, Tsimirika K, MacFarlane J, et al. Hepcidin in iron overload disorders. Blood 2005;105(10): 4103–5. Rivella S. Iron metabolism under conditions of ineffective erythropoiesis in betathalassemia. Blood 2019;133(1):51–8. Camaschella C, Nai A, Silvestri L. Iron metabolism and iron disorders revisited in the hepcidin era. Haematologica 2020;105(2):260–72. Vadolas J, Ng GZ, Kysenius K, Crouch PJ, Dames S, Eisermann M, et al. SLN124, a GalNac-siRNA targeting transmembrane serine protease 6, in combination with deferiprone therapy reduces ineffective erythropoiesis and hepatic iron-overload in a mouse model of β-thalassaemia. Br J Haematol 2021;194(1):200–10. 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(4):408–14. 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 betathalassemia. N Engl J Med 2020;382(13):1219–31. Taher AT, Cappellini MD, Kattamis A, Voskaridou E, Perrotta S, Piga AG, et al. Luspatercept for the treatment of anaemia in non-transfusion-dependent betathalassaemia (BEYOND): a phase 2, randomised, double-blind, multicentre, placebo-controlled trial. Lancet Haematol 2022;9(10) [e733-e44]. 18

Use Quizgecko on...
Browser
Browser