Fact-checked by Grok 2 weeks ago

Beta thalassemia

Beta thalassemia is an inherited blood disorder caused by mutations in the HBB gene on , which encodes the beta-globin subunit of , resulting in reduced or absent beta-globin production and leading to microcytic . This impairment disrupts normal hemoglobin synthesis, causing ineffective , , and a shortage of functional red blood cells, which carry oxygen throughout the body. The condition manifests in various severities depending on the number and type of mutations, with over 300 variants identified, including β⁰ (no production) and β⁺ (reduced production) alleles. Clinically, beta thalassemia is classified into three main forms based on and : thalassemia major (homozygous or compound heterozygous for severe , presenting as transfusion-dependent in infancy), thalassemia intermedia (milder leading to variable that may or may not require regular transfusions), and thalassemia minor (heterozygous carriers who are typically but exhibit mild microcytosis). Symptoms in severe cases include , , , poor growth, , and skeletal abnormalities due to , often appearing between 6 and 24 months of age. Complications from chronic and from transfusions can involve cardiac issues, endocrine disorders, and increased risk. Beta thalassemia follows an autosomal recessive inheritance pattern, meaning individuals must inherit two mutated HBB alleles—one from each parent—to develop the full disorder, while carriers (trait) have one mutated and a 25% risk of passing the condition to offspring if both parents are carriers. It has the highest global prevalence in regions with historical endemicity, such as the (e.g., up to 14% carrier rate in ), , , and , affecting thousands of infants annually worldwide. Diagnosis involves showing microcytosis, revealing elevated HbA₂ and HbF levels, and confirmatory of the HBB gene. Management strategies are tailored to severity and include regular blood transfusions for major to maintain levels, disease-modifying treatments such as luspatercept to reduce transfusion requirements, (e.g., ) to prevent overload, and supportive care like folic acid supplementation and monitoring for complications. Curative options have advanced with offering potential cure in pediatric patients with matched donors, and recent therapies such as betibeglogene autotemcel (beti-cel, approved in 2022) and exagamglogene autotemcel (Casgevy, approved in 2024), which insert a functional beta-globin or edit the to restore production. and prenatal screening are recommended for at-risk populations to inform .

Pathophysiology

Hemoglobin structure and function

is a composed of two α-globin and two β-globin subunits (α₂β₂), forming a quaternary structure that enables cooperative oxygen binding. Each subunit contains a , consisting of a ring with a central iron (Fe²⁺) atom, which reversibly binds one oxygen in the distal pocket of the heme. This binding occurs via a covalent interaction that completes the octahedral coordination of the iron, allowing to transport up to four oxygen molecules per tetramer. The α and β subunits are structurally similar, each comprising about 140-150 folded into eight α-helices, with the heme nestled in a hydrophobic pocket stabilized by a proximal residue. The β-globin chain is synthesized primarily in erythroid precursor cells within the , where it is produced in a coordinated manner with α-globin to assemble functional . This synthesis is regulated by the HBB gene, located on the short arm of (11p15.4), which encodes the 146-amino-acid β-globin polypeptide. Transcription of HBB is controlled by enhancers and promoters within the β-globin , ensuring high-level expression during terminal erythroid differentiation. In normal oxygen transport, deoxygenated in the lungs binds oxygen cooperatively, resulting in a sigmoidal oxygen dissociation curve that facilitates efficient loading at high s of oxygen (pO₂). Upon reaching tissues, oxygen is unloaded due to allosteric modulation: the reduces hemoglobin's oxygen affinity in response to decreased and increased of (pCO₂), promoting release in metabolically active sites. Additionally, 2,3-bisphosphoglycerate (2,3-BPG), an erythrocyte , binds specifically to the central cavity of deoxyhemoglobin, stabilizing the low-affinity T-state conformation and further enhancing oxygen delivery, with typical physiological levels shifting the P₅₀ (oxygen at 50% saturation) to approximately 26-27 mmHg. A precise quantitative balance between α- and β-globin chains, typically at a 1:1 molar ratio, is crucial for stable tetramer formation, as imbalances can lead to precipitation of unpaired chains and impaired assembly. This stoichiometric equilibrium is maintained through coordinated and during .

Genetic mutations and inheritance

Beta thalassemia is inherited in an autosomal recessive pattern, requiring two mutated alleles in the HBB gene for disease manifestation, while heterozygotes serve as asymptomatic carriers with beta thalassemia minor. In this inheritance mode, each parent contributes one mutated allele, resulting in a 25% chance of an affected child per pregnancy for carrier couples. Rarely, dominant forms occur due to specific HBB mutations causing inclusion body thalassemia, but the recessive pattern predominates. The , located on chromosome 11p15.4, encodes the beta-globin subunit of , and over 350 disrupt its function, primarily through point affecting transcription, splicing, or . These include nonsense like codon 39 C>T (c.118C>T), which introduce premature stop codons leading to no beta-globin production (β⁰-thalassemia), and splice-site variants such as IVS1-5 G>C (c.92+5G>C), which abolish splicing and also result in β⁰. Other common types are frameshift deletions, like the 4-bp deletion at codons 41/42 (-TCTT), causing β⁰, and promoter such as -28 A>G, which reduce transcription and produce β⁺-thalassemia with partial beta-globin output. Larger deletions spanning up to 67 kb are less frequent but can eliminate the gene entirely. Clinical severity correlates with the genotype: beta thalassemia major arises from homozygous β⁰/β⁰ or severe β⁺/β⁺ combinations, leading to transfusion-dependent , while intermedia results from compound heterozygous states with at least one milder β⁺ , causing non-transfusion-dependent . Heterozygous carriers exhibit beta thalassemia minor with minimal or no symptoms. Genotype-phenotype variability is modulated by factors like co-inheritance of deletions, which reduce alpha-globin excess and ameliorate severity, or increased alpha gene copy number (up to four in normal individuals), which exacerbates imbalance in β-thalassemia patients. Additionally, polymorphisms in regulators, such as BCL11A, influence residual gamma-globin compensation.

Disease mechanisms

In beta thalassemia, mutations in the HBB gene result in reduced or absent beta-globin chain synthesis, leading to an imbalance in globin chain production. This imbalance causes excess unpaired alpha-globin chains to accumulate in erythroid precursors. The excess alpha chains precipitate as insoluble inclusions within erythroid cells, forming aggregates that damage cell membranes and the cytoskeleton. These precipitates generate reactive oxygen species (ROS), free heme, and iron, which promote oxidative damage to lipids, proteins, and other cellular components. The resulting oxidative stress triggers apoptosis, particularly in polychromatophilic erythroblasts, contributing to ineffective erythropoiesis where most erythroid precursors fail to mature into red blood cells. In the periphery, the surviving mature red blood cells (RBCs) are unstable due to residual alpha-chain precipitates and ongoing oxidative injury, leading to membrane rigidity and reduced deformability. This instability causes extravascular hemolysis in the spleen and liver, as well as some intravascular hemolysis, resulting in chronic . The persistent anemia stimulates elevated production, which drives compensatory erythroid in the . Bone marrow expansion can increase the marrow cavity volume up to 25-30 times normal, leading to skeletal deformities such as maxillary hyperplasia and thinning of long bones. When the marrow reaches its capacity, extramedullary hematopoiesis occurs in sites like the spleen, liver, and paraspinal regions, causing organ enlargement and potential complications such as spinal cord compression or pulmonary masses. This hyperplasia also imposes stress on multiple organs through chronic inflammation and cytokine dysregulation. Secondary iron overload arises from two main sources: increased intestinal iron absorption due to hepcidin dysregulation and repeated blood transfusions in severe cases. Ineffective erythropoiesis suppresses hepcidin expression via factors like growth differentiation factor 15 (GDF15) from erythroid precursors, leading to inappropriately low hepcidin levels that fail to inhibit ferroportin-mediated iron export from enterocytes and macrophages. This hyperabsorption, combined with transfused iron that accumulates in tissues, results in systemic iron overload, particularly in the liver, heart, and endocrine organs, exacerbating oxidative damage and organ dysfunction.

Clinical features

Signs and symptoms

Beta thalassemia manifests with a range of clinical primarily driven by chronic , with severity varying from carriers to severe presentations in transfusion-dependent forms. Common anemia-related symptoms include , , and dyspnea, which arise from reduced oxygen-carrying capacity of the blood. In children, growth retardation is a frequent observation, reflecting the impact of persistent anemia on . Enlargement of the spleen () and liver () often occurs, resulting from and as the body compensates for ineffective . In transfusion-dependent cases, such as beta major, expansion of the leads to characteristic facial bone changes known as chipmunk facies, featuring prominent cheekbones and maxillary overgrowth. In beta thalassemia intermedia, symptoms are more variable and typically milder than in , including intermittent , from , and leg ulcers due to poor tissue oxygenation. Beta thalassemia carriers (trait) are often , though some may experience mild fatigue or from subtle .

Classification and severity

Beta thalassemia is classified into several clinical subtypes based on the degree of reduced beta-globin synthesis, ranging from asymptomatic carriers to severe, transfusion-dependent forms. The primary subtypes include beta thalassemia major, intermedia, trait (minor), and silent carrier, with severity determined by the underlying genotype and modifying factors. Beta major, also known as transfusion-dependent or Cooley's , represents the most severe form, characterized by absent or markedly reduced beta-globin production from both alleles (typically β⁰/β⁰ ). Affected individuals develop profound with levels below 7 g/dL, leading to transfusion dependence starting in infancy, usually between 6 and 24 months of age. Without regular transfusions every 2-4 weeks, symptoms such as and organ enlargement rapidly progress. Beta thalassemia intermedia encompasses a heterogeneous group with moderate severity, often resulting from compound heterozygous mutations (e.g., β⁺/β⁰ or β⁺/β⁺), leading to partial beta-globin production. Patients typically experience hemoglobin levels of 7-10 g/dL and are non-transfusion dependent, though occasional transfusions may be needed during stress such as or ; onset is variable, often after 2 years of age. This subtype's milder course is frequently influenced by genetic modifiers, such as coinheritance of (HPFH), which elevates gamma-globin expression and compensates for beta-globin deficiency. Beta thalassemia trait, or minor, occurs in heterozygous carriers with one affected HBB , resulting in mild ( 9.5-12.5 g/dL) that is usually and detected incidentally through routine tests. No transfusions are required, and individuals lead normal lives without significant clinical impact. The silent carrier state involves a very mild (e.g., mild β⁺ variant) with normal levels and no hematologic abnormalities, identifiable only by . Disease severity and progression in beta thalassemia are shaped by prognostic factors including the specific , which dictates the extent of beta-globin reduction, and co-modifiers such as elevated gamma-globin levels from variants in loci like BCL11A or HBG2 that promote fetal hemoglobin production. Coinheritance of can further ameliorate severity by balancing globin chain ratios, while environmental influences like nutritional status—particularly adequate and iron management—play a role in mitigating complications and supporting .

Associated complications

Beta thalassemia is associated with several long-term complications primarily stemming from chronic , ineffective , and . , resulting from repeated blood transfusions and increased intestinal absorption, leads to deposition in various organs. Myocardial iron accumulation causes , which manifests as and arrhythmias, accounting for a significant portion of mortality in affected patients. Endocrinopathies arise from iron-mediated damage to the pituitary, , , and gonads, resulting in conditions such as (prevalence up to 24% in intermedia forms), diabetes mellitus (2-7%), and (2-3%). Hepatic iron deposition promotes and , elevating the risk of liver complications like when combined with . Bone complications are multifactorial, involving expanded bone marrow activity, hormonal deficiencies from iron overload, and direct iron toxicity to osteoblasts. affects up to 81% of patients with beta thalassemia intermedia, characterized by reduced bone mineral density in the , , and . This leads to an increased incidence of pathologic fractures (prevalence around 12%), often exacerbated by and . In severe cases, marrow expansion contributes to skeletal deformities, though these are more pronounced in transfusion-dependent forms. Pulmonary hypertension is another important complication, particularly in non-transfusion-dependent such as , where chronic , , and hypercoagulability contribute to its development. Echocardiographic screening estimates prevalence at 10-75%, while confirmed cases by right heart catheterization range from 2% to 24%, and it is associated with increased mortality. Other notable complications include heightened infection risk following , which impairs clearance of encapsulated bacteria like , leading to in vulnerable patients. Chronic promotes overload, resulting in pigment gallstones that are more prevalent in thalassemia intermedia than major (up to 40-50% in some cohorts). Thromboembolic events, such as deep vein thrombosis, , and , occur at higher rates in intermedia variants due to chronic hypercoagulability from platelet activation and red cell abnormalities, with further elevating risk fivefold. Neurological issues are rare but include ischemic strokes from hypercoagulability or cerebral iron deposition, with silent brain infarcts detected in 27-60% of splenectomized intermedia patients via .

Diagnosis

Screening programs

Screening programs for beta thalassemia focus on identifying at the population level to enable early intervention and reduce the incidence of severe through targeted reproductive planning. These initiatives are particularly vital in regions with high carrier frequencies, where they combine initial hematological assessments with confirmatory molecular testing to detect individuals. Newborn screening is a cornerstone in high-prevalence areas, such as the and , where beta thalassemia carrier rates can exceed 10%. This approach employs (HPLC) or on dried blood spots collected shortly after birth to identify abnormal , such as elevated (HbF) or reduced HbA, allowing for early and management of affected infants. In these programs, cutoffs for abnormal patterns are established based on local , with follow-up testing recommended for presumptive positives to distinguish thalassemia from other hemoglobinopathies. Premarital and antenatal carrier screening typically initiates with a (CBC) to detect microcytosis, characterized by (MCV) below 80 fL, which prompts further evaluation. Subsequent quantification of HbA2 via HPLC or is performed, with levels exceeding 3.5% serving as a key indicator of beta thalassemia trait. These strategies are integrated into routine healthcare in many at-risk communities, often as voluntary or mandatory components of premarital counseling or , to identify at-risk couples before conception. Positive initial screens are referred for confirmatory to verify carrier status. Exemplary programs in endemic regions demonstrate the impact of systematic screening. In , a mandatory premarital testing initiative launched in the , combined with voluntary prenatal , has reduced the birth of beta thalassemia major by over 95%, from approximately 1 in 158 live births in 1970 to near elimination by the 2000s, through widespread carrier detection and informed . Similar models in countries like and have achieved substantial declines in affected births by adapting premarital screening to cultural and legal contexts. Despite successes, screening programs face significant challenges, including substantial costs for laboratory infrastructure and personnel, which limit implementation in low-resource settings where the disease burden is highest. Access barriers, such as geographic remoteness and inadequate healthcare networks, further hinder coverage in rural or underserved areas. Additionally, false positives can arise during initial screening due to overlapping microcytosis in , necessitating careful differentiation through iron studies or repeat testing to avoid unnecessary anxiety and follow-up expenses. Coexisting may also lower HbA2 levels, potentially leading to under-detection of carriers if not addressed.

Confirmatory diagnostic tests

Confirmatory diagnostic tests for beta thalassemia are essential to verify the following initial clinical suspicion or screening, utilizing hematological, biochemical, and genetic approaches to characterize the and identify underlying molecular defects. Hematological evaluations provide the initial confirmation of the characteristic . A (CBC) typically demonstrates microcytic , with reduced (MCV <80 fL) and mean corpuscular hemoglobin (MCH <27 pg), alongside a normal or elevated red blood cell count and hemoglobin levels that vary by severity (e.g., normal or mildly reduced by 1-2 g/dL in trait carriers). Reticulocyte count is often normal or mildly elevated in carriers (1-3%) and higher in severe forms (up to 15%), reflecting compensatory bone marrow hyperactivity and ineffective erythropoiesis. Peripheral blood smear examination reveals distinctive morphological changes, including target cells (codocytes), basophilic stippling, teardrop cells (dacrocytes), and poikilocytosis, which are more pronounced in intermedia and major forms. Hemoglobin analysis via high-performance liquid chromatography (HPLC) or capillary electrophoresis quantifies variant hemoglobins to confirm the beta chain defect. In beta thalassemia trait, HbA2 is elevated (3.5-7%), while HbF may be mildly increased; in homozygous beta thalassemia major, HbA is absent or severely reduced (<10%), with HbF comprising 92-95% of total hemoglobin and HbA2 at 3-6%. These methods distinguish beta thalassemia from other microcytic anemias, such as iron deficiency, by the absence of proportional hypochromia relative to microcytosis. Molecular diagnostics definitively identify the genetic basis through targeted analysis of the HBB gene on chromosome 11. Polymerase chain reaction (PCR)-based techniques, including gap-PCR for large deletions and direct sequencing or next-generation sequencing for point mutations, splice site variants, and promoter alterations, detect over 200 known pathogenic variants (e.g., β0 null alleles like codon 39 C>T or β+ mild alleles like IVS1-5 G>C). correlates with phenotypic severity and is recommended for all suspected cases, particularly in at-risk populations or for family studies. Prenatal confirmatory testing is offered to at-risk pregnancies for early detection of fetal genotypes. (CVS) at 10-12 weeks gestation or at 15-18 weeks provides fetal DNA for HBB gene analysis via and sequencing, enabling of homozygous or compound heterozygous mutations with >99% accuracy. These invasive procedures carry a small risk of (0.5-1%) but are critical for informed reproductive decisions. Emerging non-invasive (NIPT) using maternal and targeted sequencing can detect fetal HBB mutations with high accuracy (>99%) from 10 weeks gestation, avoiding risks associated with invasive procedures, though availability is limited to specialized centers as of 2025.

Prevention

Genetic counseling

Genetic counseling for beta thalassemia involves providing at-risk individuals and families with comprehensive, non-directive information about the autosomal recessive inheritance pattern of the disorder, enabling informed reproductive decisions. Counselors educate clients on the fundamentals of status and disease transmission, emphasizing that if both parents are , each carries a 25% risk of an affected child, a 50% risk of a offspring, and a 25% chance of an unaffected non-carrier child, often illustrated using Punnett squares to clarify these probabilities. This education is tailored to the client's understanding, incorporating visual aids and addressing misconceptions about the condition's severity. A core component of counseling is risk assessment through pedigree analysis, where geneticists construct family trees to identify patterns and calculate recurrence risks based on confirmed genotypes. Psychosocial support is integral, offering emotional guidance to mitigate anxiety, guilt, or family conflicts arising from status revelations, with counselors trained to recognize and address these emotional impacts. For instance, brief reference to screening results may be used to contextualize personal risks without delving into testing procedures. Preconception counseling advises prospective parents on partner testing to ascertain mutual carrier status, facilitating proactive family planning. Options discussed include limiting family size after having unaffected children, pursuing adoption, or considering gamete donation to avoid transmission risks, all presented without bias to respect individual values. In endemic regions, such as parts of the Mediterranean and South Asia, counselors highlight alternatives that align with cultural norms around family expansion. Ethical considerations underscore the importance of , ensuring clients fully comprehend risks, benefits, and alternatives before any decisions. Cultural sensitivities are paramount, particularly in communities where stigma may lead to or marriage challenges; for example, in some Muslim or South Asian groups, carrier status can evoke , necessitating sensitive discussions that integrate religious perspectives, such as permissibility of certain reproductive choices. Geneticists play a pivotal role by providing specialized expertise in and phenotype prediction, collaborating with multidisciplinary teams to deliver holistic, culturally attuned support.

Prenatal and preconception screening

Prenatal and preconception screening for beta thalassemia focuses on identifying at-risk pregnancies early and enabling informed reproductive choices for carrier couples, primarily through of fetal or embryonic material. Non-invasive prenatal testing (NIPT) represents a first-line approach, utilizing (cffDNA) circulating in maternal blood to detect HBB gene mutations associated with beta thalassemia. This , often employing techniques like droplet digital (ddPCR) or population-based haplotyping (PBH), targets common mutations such as CD41/42, IVS1-1, and others, and can be performed as early as 7-10 weeks of with reported of 94-100% and specificity of 92-99% depending on the and , as demonstrated in validated cohorts. By avoiding invasive procedures, NIPT reduces risks to the pregnancy while identifying affected fetuses, allowing progression to confirmatory testing only for high-risk results. For definitive diagnosis during pregnancy, invasive procedures such as (CVS) or are employed following initial screening. CVS involves sampling placental tissue via a needle through the or , typically between 11 and 14 weeks of , to extract DNA for HBB via PCR-based analysis. , performed later from 15 weeks onward, collects containing fetal cells for similar . Both procedures carry a miscarriage risk of approximately 0.5-1%, making them suitable for couples confirmed as carriers through preconception testing. Positive results for affected fetuses often lead to options for termination, with termination rates reaching 98% in large-scale programs among diagnosed cases. Preconception screening incorporates (PGD), integrated with in vitro fertilization (IVF), to select unaffected embryos before implantation and thereby prevent the birth of affected children. In PGD, oocytes undergo or blastomere , followed by multiplex and digestion or short analysis to HBB mutations and linked markers, ensuring exclusion of dropout errors. Unaffected or carrier embryos are then transferred, achieving pregnancy success rates of up to 30.8% in fertile couples across multiple cycles, with 100% avoidance of affected births when viable unaffected embryos are available. This approach, particularly beneficial for at-risk couples, has resulted in healthy live births confirmed free of beta thalassemia through postnatal verification. Overall, these screening strategies, supported by , have significantly reduced the incidence of beta thalassemia major in high-prevalence populations by empowering reproductive autonomy.

Management

Supportive therapies

Supportive therapies for beta thalassemia primarily aim to alleviate symptoms of , prevent complications from , and support overall health in patients with transfusion-dependent forms such as beta thalassemia major, while providing targeted interventions for non-transfusion-dependent cases like . These measures do not alter the underlying genetic defect but are essential for improving and longevity. Management should align with the 2025 Thalassaemia International Federation (TIF) Guidelines for the Management of Transfusion-Dependent β-Thalassaemia (5th edition). Regular (RBC) transfusions form the cornerstone of management for severe beta thalassemia, administered every 3-4 weeks to maintain pre-transfusion levels at 9.5-10.5 g/dL, thereby reducing , growth retardation, and organ stress associated with chronic . Each transfusion typically involves 8-15 mL/kg of leukocyte-depleted, packed RBCs infused over 1-2 hours, with careful phenotyping for ABO and Rh antigens at the outset to minimize alloimmunization risks. For beta thalassemia intermedia, transfusions may be episodic, triggered by stressors such as or , rather than routine. Iron chelation therapy is critical to counteract the iron overload that accumulates from repeated transfusions, which can lead to cardiac, hepatic, and endocrine damage if unmanaged. Chelation is tailored to the individual's profile and initiated in young children after approximately 10-20 lifetime transfusions or when serum exceeds 1,000 μg/L, with ongoing via MRI T2* annually from age 8-10 to assess myocardial and hepatic iron concentrations and guide adjustments. Common agents include , administered subcutaneously via over 8-12 hours, 5-6 nights per week; , an oral tablet taken once daily; and , an oral option used thrice daily or in combination with for refractory cases. Folic acid supplementation supports enhanced in beta thalassemia patients, who exhibit increased turnover and potential demands. A daily dose of 1-5 mg is recommended, particularly for those on transfusions or with dietary deficiencies, to help sustain production without altering transfusion requirements. Splenectomy may be considered in beta thalassemia intermedia or major patients experiencing hypersplenism, where an enlarged sequesters RBCs and escalates transfusion needs beyond 200-220 mL/kg annually. The procedure is generally deferred until age 6-7 years to allow immune maturation, preceded by vaccinations against encapsulated such as , , and type b, along with prophylactic penicillin to mitigate postoperative infection risks.

Disease-modifying treatments

Luspatercept, an erythropoiesis-stimulating that acts as a ligand trap for transforming growth factor-β superfamily members, is a key for beta thalassemia. It promotes late-stage erythroid maturation and production, thereby addressing ineffective . The U.S. approved luspatercept-aamt (Reblozyl) on November 8, 2019, for the treatment of in adult patients with beta thalassemia who require regular transfusions. In the phase 3 BELIEVE , a multicenter, randomized, double-blind, -controlled study involving 336 adults with transfusion-dependent beta thalassemia, luspatercept (administered subcutaneously at 1.0 mg/kg every 3 weeks, up to 1.25 mg/kg) significantly reduced transfusion burden compared to . The primary endpoint of at least a 33% reduction in transfusion volume with a decrease of at least 2 units over weeks 13 through 24 was met by 21.4% of luspatercept-treated patients versus 4.5% of recipients (p < 0.001). Over any 12-week interval, 70.5% achieved at least a 33% reduction, and 40.2% achieved at least a 50% reduction, corresponding to mean transfusion reductions of approximately 30-50% in responders. Long-term follow-up confirmed durable efficacy, with sustained transfusion reductions in up to 80% of patients over 3 years. Common side effects include bone pain (28%), arthralgia (21%), headache (20%), and fatigue (15%), generally mild to moderate; thromboembolic events occurred in 3.7% of patients. Efficacy is monitored via serial assessments of transfusion requirements, hemoglobin levels, and erythropoietic markers, with dose adjustments based on response and tolerability. Hydroxyurea, a ribonucleotide reductase inhibitor, induces fetal hemoglobin (HbF) production through stress erythropoiesis and nitric oxide-mediated pathways, offering a disease-modifying option particularly for non-transfusion-dependent or intermedia forms of beta thalassemia. It is typically administered orally at 10-20 mg/kg daily, starting at 10 mg/kg and titrated upward by 5 mg/kg every 4-6 weeks to maximize HbF induction while monitoring for myelosuppression. In a meta-analysis of 16 studies involving over 500 patients with beta thalassemia major or intermedia, hydroxyurea achieved a pooled response rate of 65% in transfusion-dependent cases, with 37% becoming transfusion-independent and significant HbF elevations (mean increase of 5-10 g/L). For non-transfusion-dependent patients, 53% experienced at least a 1 g/dL hemoglobin increase. Side effects are primarily hematologic, including neutropenia (12%) and leukopenia (24%), which are reversible upon dose reduction; long-term use shows good tolerability with low rates of severe toxicity. Monitoring involves monthly complete blood counts, HbF levels, and transfusion records to assess sustained hemoglobin improvements. Emerging agents include mitapivat, an oral activator of pyruvate kinase that enhances red blood cell glycolysis and ATP production to improve anemia in beta thalassemia. In the phase 3 ENERGIZE-T trial (NCT04770779), a double-blind, placebo-controlled study of 258 adults with transfusion-dependent alpha- or beta-thalassemia, mitapivat (100 mg twice daily) met its primary endpoint, with 30.4% of patients achieving at least a 50% reduction in transfusion units (with ≥2 units decrease) over 48 weeks versus 12.6% on placebo (p = 0.0003); 9.9% achieved transfusion independence for ≥8 weeks. In the parallel ENERGIZE trial for non-transfusion-dependent disease, 42% of mitapivat-treated patients had a ≥1.0 g/dL hemoglobin increase from baseline (weeks 12-24) versus 2% on placebo (p < 0.0001), with improvements in fatigue scores. Adverse events were similar to placebo, including headache (22%) and nausea (12%), with no new safety signals. As of November 2025, mitapivat remains under FDA review with a PDUFA target date of December 7, 2025; it received a positive CHMP opinion in the EU on October 17, 2025, and was approved for adults with alpha- or beta-thalassemia in Saudi Arabia on August 5, 2025. Efficacy is tracked through hemoglobin response rates, transfusion metrics, and patient-reported outcomes. Sotatercept, an earlier ligand trap in the same class as luspatercept, showed promise in a phase 2 trial where 60% of non-transfusion-dependent patients achieved ≥1.0 g/dL hemoglobin increases, but development for thalassemia was discontinued in favor of pulmonary hypertension indications.

Curative interventions

Hematopoietic stem cell transplantation (HSCT) remains the established curative intervention for beta thalassemia, particularly in patients with transfusion-dependent disease. Allogeneic HSCT from an HLA-matched sibling donor achieves cure rates exceeding 90% overall survival and thalassemia-free survival in young patients, with optimal outcomes in those under 14 years of age where survival rates reach 90-96%. However, the procedure carries significant risks, including graft-versus-host disease (GVHD), which occurs in up to 20-30% of cases, as well as infections and organ toxicity, necessitating careful donor matching and conditioning regimens. Gene therapy has emerged as a promising autologous curative approach, utilizing ex vivo modification of the patient's hematopoietic stem cells to restore functional beta-globin production. Betibeglogene autotemcel (Zynteglo), a lentiviral vector-based therapy that inserts a functional into autologous stem cells, was approved by the FDA in August 2022 for adult and pediatric patients (aged 4 years and older) with transfusion-dependent . Clinical trials demonstrated transfusion independence in approximately 90% of treated patients, with sustained hemoglobin levels above 9 g/dL for at least one year in phase 3 studies. Similarly, exagamglogene autotemcel (Casgevy), a CRISPR-Cas9 edited therapy that reactivates fetal hemoglobin by disrupting the , received FDA approval in January 2024 for patients 12 years and older with transfusion-dependent , achieving transfusion independence in 91% of trial participants. These therapies involve myeloablative conditioning followed by reinfusion of modified cells, offering a one-time treatment but at high costs, with Zynteglo priced at $2.8 million per patient in the US. Patient selection for curative interventions prioritizes younger individuals with low-risk disease features, such as no hepatomegaly or portal fibrosis, to maximize success and minimize complications; HSCT is ideal for children under 14 with matched donors, while gene therapies suit those without suitable donors but face barriers like accessibility and cost exceeding $2 million. Emerging strategies include in vivo gene editing approaches, such as fetal therapies targeting hematopoietic stem cells during gestation to prevent disease onset, which remain in preclinical stages but show promise in animal models for early intervention. Ongoing phase 1/2 trials for additional CRISPR-based edits of the HBB locus continue to evaluate safety and efficacy, building on approved therapies to expand curative options.

Epidemiology

Global incidence and prevalence

Beta thalassemia exhibits a heterogeneous global distribution, with the highest carrier frequencies observed in regions historically associated with malaria endemicity, including the Mediterranean basin, the Middle East, , and . In the Mediterranean region, carrier rates range from 5% to 15%, particularly in Italy where prevalence reaches 10-12% in Sardinia and up to 8% in southern mainland areas, and in Greece where the mean frequency is approximately 7.4%. In the Middle East, carrier frequencies vary from 1% to over 10% in populations such as those in Iran and Saudi Arabia, while in , India reports a mean prevalence of 3-4% among diverse ethnic groups. Southeast Asian countries like Malaysia and Thailand show carrier rates of 4-5%, often compounded by interactions with hemoglobin E variants. Worldwide, approximately 1.5% of the population—equating to 80-90 million individuals—are carriers of beta thalassemia mutations, with the majority residing in low- and middle-income countries where access to care is limited. As of 2021, the global prevalence of thalassemia (including beta forms) is estimated at 1.31 million cases. Recent estimates indicate approximately 23,000 infants are born annually worldwide with transfusion-dependent beta thalassemia major (as of 2020s data), with broader symptomatic forms affecting up to around 40,000 historically but declining due to prevention efforts, imposing a significant health burden primarily in resource-constrained settings. Screening and prevention programs have led to substantial declines in incidence in high-prevalence areas; for instance, in , voluntary carrier screening and prenatal diagnosis implemented since 1975 reduced the birth rate of affected infants from 1 in 250 to 1 in 4,000, achieving over 90% prevention. Demographic shifts due to migration have conversely increased cases in low-prevalence regions, such as Northern Europe and the United States; in the , approximately 20 new cases of beta thalassemia major are born annually in England alone, reflecting a rising trend linked to immigration from endemic areas. In the US, the prevalence has grown by about 7.5% over the past 50 years, with current estimates of 1,000-1,500 transfusion-dependent patients.

Evolutionary and population genetics

Beta thalassemia persists at high frequencies in certain human populations due to heterozygote advantage conferred by resistance to severe malaria, a phenomenon first hypothesized by in 1949. Heterozygous carriers exhibit altered erythrocyte properties that inhibit parasite growth and reduce the severity of infection, with studies estimating that they are 50-80% less likely to develop severe malaria compared to non-carriers. This protective effect arises from mechanisms such as increased oxidative stress in red blood cells and impaired cytoadherence of infected erythrocytes to vascular endothelium. The condition exemplifies balanced polymorphism, where the survival benefit against malaria in heterozygotes outweighs the fitness cost of severe disease in homozygotes, leading to elevated carrier rates in historically malaria-endemic regions such as the Mediterranean, Middle East, Southeast Asia, and parts of Africa. In these zones, the mild anemia experienced by carriers is offset by enhanced survival during malaria epidemics, maintaining allele frequencies through natural selection. Population genetic models indicate that this selective pressure has stabilized beta thalassemia alleles at polymorphic levels, preventing their elimination despite the homozygous disadvantage. Genetic drift and founder effects have further amplified the prevalence of specific beta thalassemia mutations in isolated populations, such as through historical bottlenecks in Mediterranean islands and Southeast Asian communities. For instance, limited gene flow in founder populations has concentrated rare variants, contributing to localized high carrier rates independent of ongoing malaria selection. In modern contexts, the intensification of malaria control efforts and eradication programs has diminished the selective advantage of beta thalassemia alleles, leading to observed declines in carrier prevalence in some regions, as evidenced by longitudinal studies in the Maldives. However, global migration continues to sustain genetic diversity by dispersing carriers to non-endemic areas, where the alleles persist without historical selective pressures.

Combined disorders

Coinheritance with alpha thalassemia

Coinheritance of with mitigates the severity of the disease by addressing the underlying alpha/beta globin chain imbalance characteristic of . In , reduced beta globin production leads to an excess of , which precipitate and cause ineffective erythropoiesis, hemolysis, and organ damage. , resulting from deletions or mutations in the or genes, decreases synthesis, thereby reducing this excess and improving red blood cell survival and overall phenotype. This interaction is particularly relevant in populations with high carrier frequencies for both disorders, where co-occurrence can shift a severe presentation toward a milder form. Genotypic examples illustrate this modifying effect through gene dosage alterations. For instance, individuals homozygous for severe beta thalassemia mutations (e.g., β0/β0 genotypes) who also carry two alpha globin gene deletions, such as the --SEA deletion (removing both alpha genes on one chromosome) or compound -α3.7/-α4.2, often exhibit a thalassemia intermedia phenotype rather than transfusion-dependent major disease. This occurs because the twofold reduction in alpha globin output balances the chain ratio more effectively, leading to less severe anemia. In Southeast Asia, where alpha thalassemia prevalence reaches 22.6% overall and co-inheritance with beta thalassemia affects up to 17.8% of beta carriers in regions like Guangdong Province, China, such genotypes are common and contribute significantly to phenotypic variability. The diagnostic importance of alpha genotyping cannot be overstated, as it enables precise prediction of disease severity in beta thalassemia carriers and affected individuals. Incorporating alpha globin gene analysis alongside beta mutation testing refines prognostic assessments, identifying those at risk for milder courses and guiding counseling on transfusion requirements or complications. Clinically, this coinheritance results in reduced transfusion needs, with affected patients often classified as non-transfusion-dependent (e.g., <8 units/year) and showing higher hemoglobin levels (>7 g/dL in many cases) compared to those without alpha deletions. However, persistent microcytosis remains a hallmark, with (MCV) values elevated but still below normal (e.g., 70.1 fL versus 64.8 fL in isolated beta thalassemia), reflecting ongoing mild imbalance.

Interactions with hemoglobin variants

Beta thalassemia often occurs in compound heterozygous states with structural , resulting in hemoglobinopathies with clinical phenotypes that differ from homozygous beta thalassemia or the variant alone. These interactions arise when one carries a beta thalassemia (reducing or abolishing beta-globin production) and the other carries a structural variant in the beta-globin gene, leading to imbalanced globin chain synthesis, , and variable severity influenced by factors like the specific type and co-inherited modifiers such as . A prominent example is sickle beta thalassemia (HbS/β-thal), where the HbS variant (β6 Glu→Val) coexists with a beta thalassemia . In HbS/β⁰-thalassemia, no normal beta-globin is produced from the thalassemic , resulting in a severe akin to homozygous sickle cell disease, characterized by chronic (hemoglobin levels typically 7-9 g/dL), high HbS proportions (70-90%), elevated (HbF, often 5-15%), vaso-occlusive crises, , and increased risk of or . In contrast, HbS/β⁺-thalassemia allows partial beta-globin production (HbA 5-30%), yielding a milder form with higher hemoglobin levels (8-11 g/dL), reduced vaso-occlusive events, but heightened susceptibility to compared to HbS/β⁰ cases. This condition is prevalent in regions with overlapping sickle cell and thalassemia frequencies, such as the Mediterranean, , and , affecting clinical management with hydroxyurea often more effective in β⁺ cases due to residual HbA. Another significant interaction is with (HbE/β-thalassemia), where the HbE variant (β26 Glu→Lys) acts as a mild beta⁺-thalassemic mutation, reducing beta-globin output by about 80% while producing an unstable . This compound state accounts for roughly half of severe beta thalassemia cases worldwide, predominantly in (e.g., , , and , with carrier rates up to 50% in some populations). Clinical severity varies widely—from mild ( >10 g/dL) to transfusion-dependent thalassemia intermedia or major—due to the interacting beta thalassemia 's impact on globin balance, often featuring , growth retardation, and , though many patients require only occasional transfusions. Genetic modifiers, including coinheritance or XmnI polymorphisms boosting HbF, can ameliorate outcomes, emphasizing the need for personalized monitoring of growth and organ function. Less common but notable interactions include those with HbC (β6 Glu→Lys) and HbD-Punjab (β121 Glu→Gln), which typically produce mild to moderate hemolytic anemias in heterozygotes with beta thalassemia. HbC/β-thalassemia, seen mainly in West African descent populations, results in target cell formation, mild , and hemoglobin levels of 9-12 g/dL, rarely requiring transfusions. Similarly, HbD/β-thalassemia, prevalent in parts of and the , mimics thalassemia intermedia with chronic but lower complication rates than HbS/β-thal. Rare variants, such as Hb Vancleave or Hb , can exacerbate or modify beta thalassemia phenotypes, sometimes leading to unexpected erythrocytosis or altered electrophoresis patterns, highlighting the importance of in atypical presentations. Overall, these interactions underscore the clinical heterogeneity of beta thalassemia, where and are essential for accurate diagnosis and tailored therapy.

References

  1. [1]
    Beta-Thalassemia - GeneReviews® - NCBI Bookshelf
    Feb 8, 2024 · Beta-thalassemia (β-thalassemia) has two clinically significant forms, β-thalassemia major and β-thalassemia intermedia, caused by absent or reduced synthesis ...
  2. [2]
  3. [3]
    Thalassemia - Symptoms & causes - Mayo Clinic
    May 6, 2025 · Thalassemia is caused by gene changes in cells that make hemoglobin. Hemoglobin is the protein in red blood cells that carries oxygen throughout the body.
  4. [4]
    Beta thalassemia: MedlinePlus Genetics
    **Summary of Beta Thalassemia (Last updated: February 8, 2024)**
  5. [5]
    Entry - #613985 - BETA-THALASSEMIA - OMIM - (OMIM.ORG)
    A number sign (#) is used with this entry because beta-thalassemia can be caused by homozygous or compound heterozygous mutation in the beta-globin gene ...
  6. [6]
    Molecular genetics of β-thalassemia: A narrative review - PMC
    Nov 12, 2021 · The HBB maps clustered with other β-like genes on chromosome 11 (11p 15.15). The cluster also contains 4 other functional genes, ε-globin gene ( ...
  7. [7]
    Entry - *141900 - HEMOGLOBIN--BETA LOCUS; HBB - OMIM
    They found 7 different beta-thalassemia mutations. The nonsense codon 39 accounted for 64%, whereas the IVS1 position 110 mutation (141900.0364), the most ...
  8. [8]
    Genetic Manipulation Strategies for β-Thalassemia: A Review
    Jun 14, 2022 · HBB mutations that result in no β-globin production leads to β0-thalassemia. Other mutations that impair the β-globin synthesis at a variable ...Missing: inheritance | Show results with:inheritance
  9. [9]
    Pathophysiology and treatment of patients with beta-thalassemia - NIH
    Dec 20, 2017 · The unpaired globin chains are unstable; they precipitate intracellularly, resulting in hemolysis, premature destruction of red blood cell [RBC] ...Missing: expansion | Show results with:expansion
  10. [10]
    Oxidative stress in β-thalassaemia and sickle cell disease - PMC - NIH
    Insufficient β-globin production results in the accumulation and precipitation of unpaired α-globin chains, leading to haemolysis and ineffective erythropoiesis ...
  11. [11]
    Could Anti-Erythropoietin Receptor Antibodies Be the Future ...
    These highly unstable chains precipitate within erythroid precursors in the bone marrow causing direct membrane damage and premature cell death by apoptosis.
  12. [12]
    Hepcidin in β-thalassemia - PMC - NIH
    Hepcidin deficiency is the main or contributing factor of iron overload in iron-loading anemias such as β-thalassemia.Missing: secondary | Show results with:secondary
  13. [13]
    Beta Thalassemia: Symptoms, Causes & Treatment - Cleveland Clinic
    Symptoms of beta thalassemia · Fatigue · Dizziness or weakness · Frequent headaches · Skin paleness · Shortness of breath · Heart palpitations · Yellow skin or ...
  14. [14]
    Beta Thalassemia | Johns Hopkins Medicine
    What are the symptoms of beta thalassemia? · Extreme tiredness (fatigue) · Pale skin · Slow or delayed growth · Weak bones · Enlarged spleen ...
  15. [15]
    Beta Thalassemia Clinical Presentation - Medscape Reference
    Jan 26, 2024 · Thalassemia can result in maxillary enlargement, leading to an appearance known as chipmunk face, along with increased spaces between teeth, ...
  16. [16]
    Beta Thalassemia - Symptoms, Causes, Treatment | NORD
    May 23, 2023 · Affected individuals may experience classic symptoms of anemia including fatigue, weakness, shortness of breath, dizziness or headaches. Severe ...
  17. [17]
    Thalassemia - Knowledge @ AMBOSS
    Sep 11, 2025 · High forehead; Prominent zygomatic bones and maxilla (referred to as “chipmunk facies”); Hair-on-end (also known as “crew cut”) sign.
  18. [18]
    Thalassemia Intermedia Clinical Presentation - Medscape Reference
    Mar 7, 2024 · Skin ulcers. Older thalassemia intermedia patients can develop leg ulcers, as oxygen reduction causes subcutaneous tissue to thin. Iron overload.
  19. [19]
    Thalassaemia carriers - NHS
    This can cause symptoms such as tiredness and pale skin, but may only be picked up with a blood test. This type of anaemia is different from iron deficiency ...
  20. [20]
    Molecular Basis and Genetic Modifiers of Thalassemia - PMC - NIH
    THE α AND β GLOBIN GENE LOCI​​ Human hemoglobin is a tetramer assembled of two homodimers of α and β globular subunits, each composed of a globin chain tightly ...The α And β Globin Gene... · α-Thalassemia · β-Thalassemia
  21. [21]
    Beta-thalassemia | Orphanet Journal of Rare Diseases | Full Text
    May 21, 2010 · Cardiac disease caused by myocardial siderosis is the most important life-limiting complication of iron overload in beta-thalassemia. In fact, ...
  22. [22]
    Endocrine and Bone Complications in β-Thalassemia Intermedia - NIH
    Thalassemia intermedia, with iron overload, cardiac failure, diabetes mellitus, hypopituitarism and porphyrinuria. The American Journal of Medicine. 1967;42 ...
  23. [23]
    Impact of bone disease and pain in thalassemia - ASH Publications
    Dec 8, 2017 · Bone disease in thalassemia. Untreated thalassemia is associated with anemia, erythroid marrow hyperplasia, and skeletal deformities.Missing: erythropoietin- | Show results with:erythropoietin-
  24. [24]
    Thalassaemia Intermedia and HbE - NCBI
    Gallstones are much more common in thalassaemia intermedia than in thalassaemia major as a result of ineffective erythropoiesis and peripheral haemolysis.Missing: hemolysis | Show results with:hemolysis
  25. [25]
    β-Thalassemia Intermedia: A Clinical Perspective - PMC - NIH
    β-thalassemia intermedia seriously impacts almost every organ system. Complications are often due to primary iron overload and hypercoagulability.Missing: post- | Show results with:post-
  26. [26]
    Neurological complications of beta-thalassemia - PubMed
    Chronic hypoxia, iron overload, desferrioxamine neurotoxicity, and bone ... This review summarizes current knowledge of the neurological complications.
  27. [27]
    Carrier screening for Beta-thalassaemia: a review of international ...
    Jun 23, 2010 · This review compares the key characteristics of β-thalassaemia carrier screening programmes implemented in countries across the world.
  28. [28]
    NEWBORN SCREENING FOR HAEMOGLOBINOPATHIES - NCBI
    Those most commonly used in neonatal screening are high-performance liquid chromatography (HPLC), capillary electrophoresis (CE) and isoelectric focusing (IEF).
  29. [29]
    Newborn Screening Practices for Beta-Thalassemia in the United ...
    Dec 13, 2021 · A comprehensive survey of all US NBS programs to assess beta-thalassemia testing methodologies, the cutoffs for defining beta-thalassemia major, and the ...Missing: premarital antenatal
  30. [30]
    HbA2 levels in children with β-thalassemia trait & iron deficiency
    Jul 23, 2024 · A critical factor in β-thalassemia trait screening is a hemoglobin A2 (HbA2) level of 3.5% or higher. In children with iron deficiency, HbA2 ...
  31. [31]
    Update in Laboratory Diagnosis of Thalassemia - Frontiers
    Alpha-thalassemia carriers had Hb A2 = 3.5% (Figures 1B,E), but β-thalassemia carrier had Hb A2 > 3.5% (Figures 1C,F). Under the HPLC system, Hb A2 and Hb E ...
  32. [32]
    [PDF] A systematic review on thalassaemia screening and birth reduction ...
    May 3, 2024 · The success rate of the prevention programme in Cyprus has been estimated to be 96%.48. To hasten the reduction initiative, the government ...
  33. [33]
    Significant haemoglobinopathies: A guideline for screening and ...
    Apr 19, 2023 · HPLC usually provides accurate quantification of Hb A2 and is, therefore, suitable for the diagnosis of β thalassaemia trait. Automated HPLC ...
  34. [34]
    Challenges in Thalassemia Carrier Detection in a Low Resource ...
    Jan 1, 2021 · The aim of this study was to assess the possibility of thalassemia carrier screening in the eastern part of Indonesia, where laboratory facility is limited by ...
  35. [35]
    Point-of-Care Diagnostic Test for Beta-Thalassemia - MDPI
    ... β-thalassemia is most prevalent, the diagnosis and screening for β-thalassemia are still challenging due to the cost and logistical burden of laboratory ...<|control11|><|separator|>
  36. [36]
    Thalassemias | Choose the Right Test - ARUP Consult
    Apr 16, 2025 · Laboratory testing for thalassemias includes screening and diagnosis via routine blood tests, structural Hb testing (eg, high-performance liquid chromatography ...
  37. [37]
    Beta Thalassemia Workup - Medscape Reference
    Jan 26, 2024 · Elevation of the Hb A2 level, demonstrated by electrophoresis or column chromatography, confirms the diagnosis of beta thalassemia trait. The Hb ...Missing: confirmatory | Show results with:confirmatory
  38. [38]
    Thalassemia - Diagnosis & treatment - Mayo Clinic
    Blood tests can reveal the number of red blood cells and irregular changes in their size, shape or color. Blood tests also can be used to look for gene changes ...
  39. [39]
    Molecular prenatal diagnosis of alpha and beta thalassemia in ... - NIH
    The term invasive prenatal diagnosis refers to obtaining fetal material by CVS, amniocentesis, or cordocentesis. Mutations in the α‐ and β‐globin genes can ...
  40. [40]
    GENETIC COUNSELLING - Prevention of Thalassaemias ... - NCBI
    Genetic counselling should be non-directive and the counsellors main role is to provide information in a non-biased manner and support decisions that are ...COMPLEXITIES IN GENETIC... · ETHICS AND RELIGION IN...
  41. [41]
    Social and cultural influences on genetic screening programme ...
    This study outlines the experiences and attitudes of adults with thalassemia, their family members, and screen‐identified thalassemia carriers toward ...
  42. [42]
    Non-invasive prenatal screening & diagnosis of β-thalassaemia in ...
    Jun 20, 2023 · The results of this study suggest that NIPT utilizing the ddPCR assay can be effectively used for the screening and diagnosis of foetal β-thalassaemia in at ...
  43. [43]
    Noninvasive prenatal testing of α-thalassemia and β-thalassemia ...
    Feb 5, 2021 · This study demonstrated the feasibility of PBH-NIPT for thalassemia. PBH-NIPT can be used after carrier screening for thalassemia. For high-risk ...
  44. [44]
    Screening for sickle cell and thalassaemia - NHS
    Screening is offered to find out if you're a carrier of a gene for sickle cell or thalassaemia and therefore likely to pass it on to your baby.Missing: premarital | Show results with:premarital
  45. [45]
    From Prenatal to Preimplantation Genetic Diagnosis of β ... - NIH
    Feb 20, 2018 · The success rate of pregnancies increased from 11.1% to 30.8% when, crucial law changes were enacted, and PGD was offered to fertile women as ...
  46. [46]
    Preimplantation Diagnosis of Thalassemias - PMC - NIH
    Purpose:Preimplantation genetic diagnosis (PGD) is an important option for couples at risk of having children with β-globin mutations to avoid selective ...
  47. [47]
    Thalassemia - Treatment | NHLBI, NIH
    Jun 1, 2022 · Blood transfusions are the main treatment for thalassemia. Learn more about other treatments like iron chelation therapy and blood and bone ...
  48. [48]
    Beta Thalassemia Treatment & Management - Medscape Reference
    Jan 26, 2024 · Supportive measures include folic acid replacement and monitoring for the development of complications such as pulmonary hypertension, ...
  49. [49]
    Thalassaemia - Treatment - NHS
    Read about treatments for thalassaemia, including blood transfusions, medicine to remove excess iron from the body, and stem cell transplants.
  50. [50]
    How I treat thalassemia | Blood | American Society of Hematology
    Sep 29, 2011 · Folic acid (1-5 mg/day) can be given when the diet is deficient in folate and/or in the presence of infection, malabsorption, and where the ...
  51. [51]
    FDA approves luspatercept-aamt for anemia in patients with beta ...
    Nov 8, 2019 · The Food and Drug Administration approved luspatercept-aamt (REBLOZYL, Celgene Corp.) for treatment of anemia in adult patients with beta thalassemia.
  52. [52]
    Phase 3 Trial of Luspatercept in β-Thalassemia
    Mar 25, 2020 · Here, we report the results of the phase 3 BELIEVE trial, which evaluated the efficacy and safety of luspatercept in adults with transfusion- ...
  53. [53]
    Efficacy and Safety of Luspatercept in the Treatment of β-Thalassemia
    Nov 16, 2022 · Luspatercept was approved via the Food and Drug Administration (FDA) on November 8, 2019 [10]. It was also authorized in 2020 via the ...
  54. [54]
    Efficacy and safety of hydroxyurea therapy on patients with β ...
    Jan 14, 2025 · Although start dose of HU was 10 mg/kg per day and increased by 5 mg/kg per day every 4–6 weeks until toxicity or according to clinical response ...Introduction · Methods · Results · Discussion
  55. [55]
    Agios Presents Positive Results from Phase 3 ENERGIZE-T Study of ...
    Dec 8, 2024 · ENERGIZE-T is first Phase 3 study to demonstrate efficacy of an oral, disease-modifying treatment for transfusion-dependent alpha- and beta-thalassemia.
  56. [56]
  57. [57]
    Sotatercept, a novel transforming growth factor β ligand trap ... - NIH
    This study demonstrated the safety and efficacy of sotatercept in patients with β-thalassemia. An improvement in hemoglobin levels and reduction in transfusion ...
  58. [58]
    Stem Cell Transplantation Still the Main Treatment Option for Beta ...
    Dec 27, 2024 · In addition, the study showed that the threshold age for optimal transplant outcomes was around 14 years, with OS rates ranging from 90% to 96% ...
  59. [59]
    Cure for thalassemia major – from allogeneic hematopoietic stem ...
    The limited data available suggests that the long-term outcome is also excellent, with a >90% survival rate, but for the best results, hematopoietic stem cell ...
  60. [60]
    ZYNTEGLO | FDA
    Sep 19, 2022 · August 17, 2022 Approval Letter - ZYNTEGLO · August 17, 2022 Summary Basis for Regulatory Action - ZYNTEGLO · Approval History, Letters, Reviews ...
  61. [61]
    Long-Term Data Presented at ASH Support beti-cel as a Potentially ...
    Among patients in the Phase 3 studies, 90.2% (37/41) achieved transfusion independence (TI). As of the data cutoff date, TI was maintained through last follow- ...Missing: rate | Show results with:rate
  62. [62]
    FDA Approves First Gene Therapies to Treat Patients with Sickle ...
    Dec 8, 2023 · The FDA approved the first cell-based gene therapies, Casgevy and Lyfgenia, for the treatment of sickle cell disease in patients 12 years ...
  63. [63]
    With $2.8M gene therapy, Bluebird sets new bar for US drug pricing
    Aug 18, 2022 · Approved for severe beta thalassemia, Zynteglo will test insurers' willingness to pay for expensive one-time treatments.
  64. [64]
    Recent advancements in gene therapy for sickle cell disease and β ...
    In this review article, we discuss the current genetic approaches and innovative strategies to ensure safe and effective gene therapy for SCD and β-thalassemia.
  65. [65]
    Advances in β-Thalassemia Gene Therapy: CRISPR/Cas Systems ...
    Oct 14, 2025 · The clinical translation of CRISPR-based genome editing in β-thalassemia has advanced from proof-of-concept studies to pivotal trials (Table 3), ...
  66. [66]
    Antenatal Screening and Diagnosis of B-thalassemia in High-Risk ...
    Nov 2, 2022 · Sardinia, an Italian island in the Mediterranean sea with a population of 1,7 millions is at high risk incidence of β-thalassemia, with 10-12% ...
  67. [67]
    β-Thalassemia Distribution in the Old World: an Ancient Disease ...
    Greece is a country of approximately 11 million people with a mean frequency of β-thalassaemia carriers of 7.4%. Extensive studies have disclosed that the ...
  68. [68]
    Prevalence of β-thalassemia carriers in the countries on the...
    Beta-thalassemia exhibits higher occurrence rates in nations situated around the Mediterranean, Southeast Asia, and Eastern Europe.
  69. [69]
    Global distribution of β-thalassemia mutations: An update
    Feb 20, 2024 · Only 20 β thalassemia mutations account for more than 80% of the β thalassemia mutation across the globe due to phenomenon of geographical clustering.
  70. [70]
    Epidemiology of clinically significant forms of alpha‐ and beta ...
    Jun 26, 2023 · In two studies from the Middle East, the birth prevalence of beta-thalassemia major was reported as 23.9/100 000 births in 2006 in Iran and 40/ ...
  71. [71]
    Sur - Journal of Hematology
    Earlier studies have shown that the overall prevalence of β-thalassemia is 3-4% with an estimate of around 8,000 - 10,000 new births with major disease each ...<|control11|><|separator|>
  72. [72]
    Molecular spectrum and carrier frequency of deletional hereditary ...
    Sep 29, 2025 · In Malaysia, it is estimated that approximately 4.5% of the population are beta (β)-thalassemia carriers, with certain ethnic groups, such as ...
  73. [73]
    Beta-Thalassemia Major and Intermedia - DynaMed
    Nov 27, 2024 · Incidence/Prevalence · Thailand: 556 in 100,000 live births (beta-thalassemia major and beta-thalassemia/hemoglobin E) · Iraq: 34.6 per 100,000 ...Definitions · Epidemiology · Etiology And Pathogenesis
  74. [74]
    Beta-Thalassemia Prevalence, Pathophysiology and Inheritance
    About 1.5% of the global population (80-90 million people) are carriers of beta-thalassemia. 60,000 symptomatic individuals are born annually.3,5. TDT greatly ...
  75. [75]
    Beta-thalassemia - ThinkGenetic Foundation
    It has been estimated that about 1.5% of the global population (80 to 90 million people) are carriers of beta-thalassemia. How is beta-thalassemia diagnosed?
  76. [76]
    Changing patterns in the epidemiology of β‐thalassemia - PMC
    Worldwide, an estimated 25 500 infants with TDT are born every year; overall, only about 12% of these patients receive blood transfusions, with 2.7% of patients ...3. Prevalence Of... · 4. Factors Affecting... · 4.2. Prevention Programs
  77. [77]
    Carrier screening for Beta-thalassaemia: a review of international ...
    Jun 23, 2010 · This review compares the key characteristics of β-thalassaemia carrier screening programmes implemented in countries across the world.
  78. [78]
    Information and choices for women and couples at risk of having a ...
    Feb 5, 2025 · About 20 babies are born with thalassaemia major every year in England. People with thalassaemia major cannot make enough haemoglobin. This is a ...
  79. [79]
    [PDF] REPORT AT A GLANCE: BETA THALASSEMIA - ICER
    There are likely between 1,000 – 1,500 people in the US living with TDT, but there are estimated to be about 1.25 million carriers of the genetic defect that is ...
  80. [80]
    The Thalassemias: The Role of Molecular Genetics in an Evolving ...
    Although J. B. S. Haldane originally suggested in 1949 that the high frequency of the thalassemias might reflect heterozygote advantage against severe malaria ( ...
  81. [81]
    Human genetic variations conferring resistance to malaria - PMC
    In malaria-endemic regions, HbS exhibits a heterozygote advantage. HbAA individuals are free from SCD but face a higher risk of death from malaria [22], while ...
  82. [82]
    Population genetics of malaria resistance in humans | Heredity
    Mar 23, 2011 · Overall, the original 'malaria hypothesis' of Haldane that diseases such as thalassemia are polymorphisms with an advantage to heterozygotes ...
  83. [83]
    Resistance to malaria in humans: the impact of strong, recent selection
    Oct 22, 2012 · ... β-thalassaemia heterozygotes had an increased fitness in the presence of malaria. Therefore, citation of [3] seems more correct for the ...
  84. [84]
    Beta Thalassemia - StatPearls - NCBI Bookshelf
    Beta-thalassemia refers to an inherited mutation of the beta-globin gene, causing a reduced beta-globin chain of hemoglobin. The highest prevalence of beta- ...
  85. [85]
    β-Thalassemia | Genetics in Medicine - Nature
    Nov 3, 2016 · Gene drift and founder effects are other reasons that thalassemias are most frequent in southeastern and southern Asia, the Middle East, the ...
  86. [86]
    Falling prevalence of beta-thalassaemia and eradication of malaria ...
    It is concluded that the observed fall in thalassaemia carrier prevalence in the Maldives is consistent with the predicted effect of malaria eradication and ...
  87. [87]
    Beta‐thalassemia and the advent of new interventions beyond ...
    Apr 3, 2022 · Because carriers have migrated there owing to demographic migration, β-thalassaemia can now be detected in areas other than malaria-endemic ...
  88. [88]
    Alpha globin gene alterations modifying the phenotype of ...
    May 27, 2024 · Co-inheritance of alpha thalassemia mutations is known to ameliorate the severity of HbE-β thalassemia. However, the role of alpha globin gene ...
  89. [89]
    Co-inheritance of α-thalassaemia and β-thalassaemia in a prenatal ...
    Aug 12, 2014 · Similarly, the coexistence of β-thalassaemia is a background modifier for ameliorating disease severity in patients with haemoglobin H disease ...<|control11|><|separator|>
  90. [90]
    Prevalence of Alpha(α)-Thalassemia in Southeast Asia (2010–2020)
    Oct 9, 2020 · Overall, the prevalence of α-thalassemia is 22.6%. The highest α-thalassemia prevalence was observed in Vietnam (51.5%) followed by Cambodia ( ...
  91. [91]
    Molecular genetics of β-thalassemia: A narrative review - Medicine
    Nov 12, 2021 · β-thalassemia is a hereditary hematological disease caused by over 350 mutations in the β-globin gene (HBB).
  92. [92]
    Full article: Sickle cell/β-thalassemia: Comparison of Sβ0 and Sβ+ ...
    May 28, 2016 · The objective of this study was to provide an overview of the clinical and laboratory characteristics of patients with the syndrome, and of the ...
  93. [93]
    Hb E/beta-thalassaemia: a common & clinically diverse disorder - NIH
    Haemoglobin E-beta thalassaemia (Hb E/β-thalassaemia) is the genotype responsible for approximately one-half of all severe beta-thalassaemia worldwide.
  94. [94]
    Clinicohematological and molecular analysis of hemoglobin D ...
    β-thalassemia is particularly common in Pakistan, but data on other hemoglobin variants remain limited. This study aimed to investigate HbD syndrome, identify ...
  95. [95]
    Rare coinheritance of hemoglobin vancleave with severe beta ...
    Apr 23, 2024 · Here, we report the first case of Hb Vancleave combined with a severe beta-thalassemia mutation associated with secondary erythrocytosis in a ...Missing: modification | Show results with:modification
  96. [96]
    Co-inheritance of Hb Ottawa and HbS: a rare interaction leading to ...
    Sep 14, 2022 · Interaction of HbS with other α and β globin chain variants is known to occur. We report a case showing interaction of HbS with a rare Hb ...