Deferasirox is an orally bioavailable iron chelator medication primarily used to treat chronic iron overload in patients receiving long-term blood transfusions, such as those with beta-thalassemia major, sickle cell disease, or myelodysplastic syndromes.[1] It binds selectively to ferric iron (Fe³⁺) in a 2:1 complex, facilitating its excretion mainly through the feces and thereby preventing toxic accumulation in organs like the liver, heart, and endocrine glands.[2] Approved by the U.S. Food and Drug Administration (FDA) in 2005 as the first oral agent for this purpose, deferasirox is indicated for patients aged 2 years and older with transfusion-related iron overload, as well as for those aged 10 years and older with non-transfusion-dependent thalassemia syndromes who have elevated liver iron concentrations.[1][3]Available under brand names such as Exjade (tablets for oral suspension in 125 mg, 250 mg, and 500 mg strengths) and Jadenu (sprinkle granules and tablets), deferasirox is typically dosed at 20–40 mg/kg body weight once daily for Exjade or an equivalent of 14–28 mg/kg for Jadenu, adjusted based on serum ferritin levels and iron intake from transfusions.[4][5] Its absolute bioavailability is approximately 70%, with a half-life of 7–16 hours, and it exhibits high affinity for iron while having minimal binding to essential metals like zinc or copper.[2] The drug has been a significant advancement in managing transfusional hemosiderosis, reducing the need for parenteral chelators like deferoxamine.[1]Despite its efficacy, deferasirox carries serious risks, including renal toxicity, hepatic failure, and potentially fatal gastrointestinal hemorrhage, particularly in elderly patients, those with pre-existing kidney or liver impairment, or individuals on concomitant medications like NSAIDs or anticoagulants.[3] Regular monitoring of renal function (via serum creatinine and proteinuria), hepatic enzymes, and complete blood counts is essential during treatment, with dose interruptions or reductions recommended if abnormalities occur.[1] Common side effects include nausea, diarrhea, rash, and abdominal pain, while hypersensitivity reactions and auditory or ocular disturbances have also been reported.[3]
Medical Uses
Indications
Deferasirox is primarily indicated for the treatment of chronic iron overload resulting from blood transfusions (transfusional hemosiderosis) in patients aged 2 years and older with conditions such as beta-thalassemia major, sickle cell disease, and other transfusion-dependent anemias.[6][7] This approval is based on clinical trials demonstrating its ability to reduce liver iron concentration and serum ferritin levels in these populations, thereby addressing the iron accumulation from repeated transfusions.[6][8]Secondary indications include the management of chronic iron overload in patients with non-transfusion-dependent thalassemia (NTDT) syndromes, approved for patients aged 10 years and older with liver iron concentration (LIC) of at least 5 mg Fe/g dry weight and serum ferritin greater than 300 mcg/L.[6][8] In Europe, it is also indicated for chronic iron overload due to blood transfusions in beta-thalassemia major patients aged 2 to 5 years with frequent transfusions (≥7 mL/kg/month of packed red blood cells) when deferoxamine is contraindicated or inadequate, as well as for infrequent transfusions (<7 mL/kg/month) in patients aged 2 years and older when deferoxamine is contraindicated or inadequate.[8] Efficacy in these secondary uses is supported by studies such as EPIC (Study A2409), which showed significant reductions in serum ferritin over 52 weeks compared to baseline.[8]For patients with myelodysplastic syndromes (MDS) requiring frequent blood transfusions, deferasirox is used to treat chronic iron overload, though controlled clinical trials specifically in this population have not been conducted.[6] Pediatric use is approved starting from age 2 years for transfusional iron overload in beta-thalassemia and other anemias in the US, and from age 2 years in the EU when deferoxamine is contraindicated or inadequate (general use from age 6 years in the EU for beta-thalassemia major), with safety and efficacy profiles similar to adults in supportive studies.[6][8] Off-label applications include limited evidence for iron overload management in sickle cell disease beyond standard transfusional settings and other rare anemias, though these are not formally approved and rely on case reports rather than large trials.[6]
Dosage and Administration
Deferasirox is administered orally once daily, with dosing tailored to the patient's iron overload status, body weight, and renal function. For patients with chronic iron overload due to frequent blood transfusions (transfusion-dependent), the recommended initial dose is 20 mg/kg body weight per day for those aged 2 years and older with an estimated glomerular filtration rate (eGFR) greater than 60 mL/min/1.73 m² (for Exjade; 14 mg/kg/day for Jadenu).[6][7] This dose is adjusted every 3 to 6 months in increments of 5 to 10 mg/kg based on serumferritin trends and liver iron concentration (LIC), aiming to maintain ferritin below 1,000 mcg/L while avoiding levels below 500 mcg/L, which may warrant interruption of therapy; the maximum dose is 40 mg/kg per day (28 mg/kg for Jadenu).[6][7]For pediatric patients aged 2 years and older with transfusion-dependent iron overload, the same weight-based dosing applies as in adults, with careful monitoring during acute illnesses involving volume depletion, where temporary interruption may be necessary.[6] In cases of non-transfusion-dependent thalassemia syndromes (for patients aged 10 years and older), the initial dose is lower at 10 mg/kg per day (7 mg/kg for Jadenu), which may be increased to 20 mg/kg per day (14 mg/kg for Jadenu) after 4 weeks if baseline LIC exceeds 15 mg Fe/g dry weight; subsequent adjustments occur every 6 months based on LIC, with interruption recommended if LIC falls below 3 mg Fe/g dry weight or ferritin below 300 mcg/L, and a maximum of 20 mg/kg per day (14 mg/kg for Jadenu).[6][7]The medication should be taken on an empty stomach at least 30 minutes before food, preferably at the same time each day, to optimize absorption (Exjade).[6] Tablets are not to be chewed or swallowed whole but dispersed in water, orange juice, or apple juice (using 3.5 ounces for doses under 1 g and 7 ounces for 1 g or more); the resulting suspension is swallowed immediately, with any residue rinsed and ingested.[6] Administration should avoid proximity to dairy products, calcium-fortified juices, or aluminum-containing antacids, which can impair absorption.[6] For the Jadenu formulation (tablets or sprinkles), it may be taken with a light meal, with equivalent doses approximately 30% lower due to higher bioavailability.[7]Dose adjustments are required for hepatic or renal impairment: reduce by 50% in moderate hepatic impairment (Child-Pugh B) or eGFR 40-60 mL/min/1.73 m², and avoid entirely in severe cases (Child-Pugh C or eGFR below 40 mL/min/1.73 m²).[6][7] For concurrent use with potent UGT inducers like rifampin, the initial dose may be increased by 50%, with close monitoring.[6] If serum creatinine rises more than 33% above baseline on two consecutive monthly measurements, reduce the dose by 10 mg/kg in transfusion-dependent patients or interrupt/reduce in non-transfusion-dependent cases.[6]Ongoing monitoring is essential to guide therapy and mitigate risks, including monthly assessments of serum ferritin, renal function (serum creatinine, eGFR, urine protein), liver enzymes (transaminases, bilirubin), and complete blood counts.[6] For non-transfusion-dependent patients, LIC should be evaluated every 6 months via MRI or biopsy.[6] Baseline and periodic auditory and ophthalmic examinations are also recommended, with therapy interruption during episodes of dehydration or gastrointestinal disturbances until resolution.[6]
Adverse Effects
Common Side Effects
The most common adverse reactions associated with deferasirox therapy are gastrointestinal disturbances, which occur frequently but are generally mild to moderate in severity and often appear early in treatment. In pivotal clinical trials, such as the EPIC study in patients with transfusion-dependent anemias and the ESCALATOR study in those with β-thalassemia major, nausea was reported in up to 23% of patients, vomiting in up to 21%, diarrhea in up to 20%, and abdominal pain in up to 28%.[9] These effects typically resolve with continued use or dose adjustment, and symptomatic management, such as antiemetics or antidiarrheal agents, is recommended if they persist.[9]Other frequent reactions include dermatologic issues and mild renal function changes. Skin rash occurred in 8-11% of patients across these trials, often resolving spontaneously, while pruritus was noted less commonly but managed similarly with topical treatments or dose reduction if needed.[9][10]Fatigue was reported infrequently, affecting fewer than 5% of participants, and usually did not require intervention beyond monitoring.[9]Elevated serum creatinine, indicating potential renal effects, was observed as mild increases in 30-40% of patients in the EPIC and ESCALATOR studies, with rises greater than 33% above baseline in approximately 36-38%.[9] These elevations are typically non-progressive and reversible upon dose reduction (e.g., by 10 mg/kg/day) or temporary interruption, with regular monitoring of renal function advised during therapy.[9] Overall, most common side effects in these trials led to discontinuation in less than 5% of cases, emphasizing their manageability with appropriate adjustments.[10]
Serious Risks
Deferasirox is associated with serious renal toxicity, including acute kidney injury and renal failure, which prompted a boxed warning from the U.S. Food and Drug Administration (FDA). In clinical trials, dose-dependent elevations in serum creatinine occurred in approximately 38% of patients receiving deferasirox compared to 14% on deferoxamine, with renal tubulopathy reported primarily in pediatric patients with β-thalassemia major. Acute renal failure has been reported in post-marketing surveillance, with rare incidence (<1%) in clinical use, though rates may be higher in elderly patients, those with dehydration, or individuals with baseline renal impairment. Risk factors for renal toxicity include pre-existing renal dysfunction (e.g., estimated glomerular filtration rate 40-60 mL/min/1.73 m²), concomitant use of other nephrotoxic agents, and conditions causing volume depletion such as vomiting or diarrhea.[1][11][12]Hepatic effects represent another major concern, with deferasirox carrying an FDA boxed warning for hepatic impairment and failure, including fatal outcomes. Elevations in transaminases greater than five times the upper limit of normal were observed in 5.7% to 8.4% of patients in clinical studies, leading to discontinuation in some cases due to drug-induced hepatitis. These events are more frequent in patients over 55 years with comorbidities such as myelodysplastic syndrome. Gastrointestinal hemorrhage, also subject to a boxed warning, has resulted in fatal cases, particularly in elderly patients with advanced hematologic malignancies or thrombocytopenia (platelet count <50 × 10⁹/L); caution is advised with concurrent use of ulcerogenic drugs.[1][1]Hematologic adverse events include severe cytopenias such as agranulocytosis and bone marrow suppression, reported in post-marketing surveillance. The FDA issued an early communication in 2007 alerting healthcare providers to cases of cytopenias, including fatal outcomes, in patients treated with deferasirox, emphasizing the need for regular monitoring of blood counts. These effects may occur independently or exacerbate other toxicities.[13][1]Overall mortality linked to deferasirox includes suspected drug-related deaths reported in pharmacovigilance databases, with over 1,300 cases noted by 2009 and additional reports through 2012 totaling more than 4,100, often attributed to multi-organ failure involving renal, hepatic, or gastrointestinal complications. As of 2024, FDA post-marketing reviews have not identified new safety signals in pediatric patients, though ongoing monitoring is recommended. Common gastrointestinal side effects like nausea may occasionally precede severe hemorrhage.[14][15][16]
Pharmacology
Mechanism of Action
Deferasirox functions as a tridentate iron chelator that selectively binds ferric iron (Fe³⁺) ions with high affinity, forming a stable neutral complex in a 2:1 molar ratio (two deferasirox molecules per iron atom).[2] This binding occurs primarily with the labile iron pool, the biologically reactive fraction of iron available for chelation, and the overall stability constant for the Fe(DFX)₂ complex is log β₂ = 36.9 at 25°C in 0.1 M KCl aqueous solution.[17] The selectivity for Fe³⁺ over other metals, such as zinc and copper, is high, minimizing interference with essential metal homeostasis.[2]Due to its lipophilic properties, deferasirox readily penetrates cell membranes to access intracellular iron stores, particularly the cytosolic labile iron pool in hepatocytes and myocytes.[18] This intracellular action enables effective removal of iron accumulated in tissues prone to overload, such as the liver and heart, contrasting with deferoxamine, which primarily acts extracellularly by binding non-transferrin-bound iron (NTBI) in plasma and requires parenteral administration.[18]The iron-deferasirox complex is lipid-soluble and undergoes biliary elimination, leading to fecal excretion as the primary route (approximately 84% of the dose), with minimal renal clearance (about 8%).[2] Compared to deferiprone, a hydrophilic bidentate chelator that also provides oral efficacy but forms 3:1 complexes with iron, deferasirox's greater lipophilicity enhances its tissue penetration and sustained action, contributing to reductions in serum NTBI levels over 24 hours.[18][19][20]
Pharmacokinetics
Deferasirox is administered orally and exhibits approximately 70% absolute bioavailability compared to intravenous administration.[9] The time to reach peak plasma concentration (T_max) occurs at a median of 1.5 to 4 hours post-dose, with both maximum plasma concentration (C_max) and area under the curve (AUC) increasing linearly with dose.[9] Ingestion with food can increase bioavailability by up to twofold, depending on meal fat and caloric content, though the drug is recommended to be taken on an empty stomach at least 30 minutes before meals to ensure consistent absorption and minimize variability. [9]Following absorption, deferasirox is highly bound to plasma proteins, approximately 99% to serum albumin, with only about 5% confined to blood cells.[9] The steady-state volume of distribution is 14.37 ± 2.69 L in adults, indicating moderate distribution into extracellular fluid.[9] As a lipophilic compound, deferasirox and its metabolites penetrate various tissues, including the liver, spleen, and other organs relevant to iron overload.[19]Deferasirox undergoes primarily hepatic metabolism through glucuronidation by uridine 5'-diphospho-glucuronosyltransferase (UGT) enzymes, mainly UGT1A1 and UGT1A3, forming an inactive acyl glucuronidemetabolite that accounts for the majority of circulating drug products.[9] Minor metabolism occurs via cytochrome P450 enzymes, particularly CYP3A4, contributing to about 8% of the total metabolic pathways, with potential for enterohepatic recirculation following deconjugation of the glucuronide.[9]Elimination of deferasirox is predominantly fecal, with 84% of the dose recovered in feces primarily as the glucuronidemetabolite, and only 8% excreted renally.[9] The terminal half-life ranges from 8 to 16 hours, supporting once-daily dosing.[9] Steady-state plasma concentrations are achieved after 3 to 4 days of daily administration, with an accumulation factor of 1.3 to 2.3.[21]
Chemistry
Physical and Chemical Properties
Deferasirox is chemically designated as 4-[3,5-bis(2-hydroxyphenyl)-1H-1,2,4-triazol-1-yl]benzoic acid, with the molecular formula C_{21}H_{15}N_3O_4. It functions as a tridentate ligand incorporating salicylic acid and hydrazone moieties that enable iron chelation. The compound has a molar mass of 373.36 g/mol.[22][23]Physically, deferasirox exists as a white to slightly yellow, non-hygroscopic crystalline powder. It exhibits a melting point of 264–265 °C and a logP value of 3.6, conferring lipophilic properties that influence its membrane permeability.[22][2]
Deferasirox demonstrates poor aqueous solubility, with values below 0.1 mg/mL in water at neutralpH, limiting its dissolution in physiological conditions. It is readily soluble in dimethyl sulfoxide (DMSO, up to 75 mg/mL) and in alkaline media, where deprotonation enhances solubility. The compound possesses pKa values of 4.57 for the carboxylic acid group and 8.71 for the phenolic hydroxyl group, reflecting its amphoteric nature.[2][24][25]Under standard conditions, deferasirox is chemically stable and photostable, maintaining integrity for extended periods such as 60 months at 25 °C and 60% relative humidity. Commercially, it is presented in film-coated tablet formulations at strengths of 125 mg, 250 mg, and 500 mg to facilitate oral administration.[23]
Synthesis
The synthesis of deferasirox employs a two-step process utilizing commercially available starting materials: salicylic acid, salicylamide, and 4-hydrazinobenzoic acid hydrochloride.[26][27]In the first step, salicylic acid is converted to salicyloyl chloride, typically using thionyl chloride, followed by condensation with salicylamide under heating at approximately 170°C to form the key intermediate 2-(2-hydroxyphenyl)-1,3-benzoxazin-4-one.[26][27] This dehydration reaction proceeds via nucleophilic attack and cyclization, yielding the benzoxazinone intermediate as slightly yellow crystals after cooling and crystallization from ethanol.[26]The second step involves the reaction of the benzoxazinone intermediate with 4-hydrazinobenzoic acid hydrochloride in ethanol under reflux conditions for several hours, promoting ring opening and triazole formation.[26][28] The reaction mixture is then cooled, and the pH is adjusted to 4.0-4.5 using dilute hydrochloric acid to precipitate the product, followed by filtration, washing with water, and drying to obtain deferasirox. Purification is achieved through recrystallization, ensuring high purity.The industrial process, conducted under good manufacturing practice (GMP) conditions, achieves an overall yield greater than 80%, with process controls ensuring no major impurities exceed qualification thresholds as per ICH guidelines.[29][23] This scalable route supports commercial production while maintaining product quality.[23]
History
Development
Deferasirox, known during development as ICL670, was discovered by Novartis in the late 1990s through extensive structure-activity relationship studies focused on tridentate iron chelators. Building on the earlier identification of desferrithiocin in 1980—a tridentate ligand with oral activity but significant toxicity—researchers synthesized and evaluated over 700 molecular entities, including more than 40 bis-hydroxyphenyltriazole derivatives, to optimize iron-binding affinity, oral bioavailability, and safety. This rational design process at Novartis identified ICL670 as a promising candidate due to its high selectivity for ferric iron and favorable pharmacokinetic profile compared to existing chelators like deferoxamine.[30]Preclinical studies in animal models, including rats, gerbils, and marmosets, demonstrated deferasirox's superior iron mobilization capabilities, particularly in the liver and heart, relative to deferiprone. In iron-overloaded gerbils, deferasirox reduced hepatic iron more effectively than deferiprone while achieving comparable cardiac iron removal, highlighting its potential for addressing transfusional iron overload. Acute and chronic toxicity assessments revealed low overall toxicity, with no significant adverse effects on major organs at doses achieving effective chelation, supporting progression to human trials.[31][32][33]Key milestones in early development included the initiation of Phase I trials in 2001, which evaluated single oral doses up to 80 mg/kg in healthy volunteers and confirmed good tolerability with no serious adverse events. These studies established a safe dosing range and bioavailability suitable for once-daily administration. The multinational EPIC trial, launched in 2005, further advanced development by enrolling over 1,700 patients with transfusion-dependent anemias to assess long-term efficacy in reducing liver iron concentration, providing pivotal data on dose-dependent iron removal.[34][35]The primary rationale for deferasirox's development was to provide a convenient oral therapy as an alternative to the subcutaneous infusions required for deferoxamine, thereby improving patient compliance and quality of life in transfusion-dependent conditions like thalassemia major. This addressed the longstanding challenge of chronic iron overload management, where injectable chelators often led to poor adherence due to pain and inconvenience.[36][37]
Regulatory Approvals and Commercial Status
Deferasirox received initial approval from the U.S. Food and Drug Administration (FDA) on November 2, 2005, for the treatment of chronic iron overload due to blood transfusions (transfusional hemosiderosis) in patients aged 2 years and older.[38] The European Medicines Agency (EMA) granted marketing authorization on August 28, 2006, for similar indications in patients with chronic iron overload from blood transfusions associated with conditions such as beta-thalassemia and other anemias.[8] This approval extended to pediatric patients aged 2 years and older from the outset, with subsequent label updates in 2011 and 2013 supporting use in younger children through formulations like dispersible tablets and sprinkles for easier administration.[1] In 2015, the FDA approved Jadenu, a film-coated tablet formulation of deferasirox with improved bioavailability.[39] Deferasirox was included in the World Health Organization (WHO) Model List of Essential Medicines starting from the 22nd list (2021) and retained in the 23rd list (2023) as an oral alternative to deferoxamine for managing chronic iron overload due to transfusions.[40]Marketed primarily under the brand name Exjade by Novartis Pharmaceuticals, deferasirox became widely available following the introduction of generic versions. In the United States, the first generic approvals occurred in 2019, with Teva Pharmaceuticals launching deferasirox tablets for oral suspension in March of that year.[41] However, Teva discontinued its generic formulations in July 2020 due to manufacturing and supply chain challenges, though other generics from manufacturers like Aurobindo and Cipla continued to fill the market.[42] Exjade remains available in many regions, but generics have significantly expanded access post-2019. As of 2025, multiple generic manufacturers supply the U.S. market.Safety concerns prompted several regulatory updates. In May 2007, the FDA issued an early warning about potential renal and hepatic risks, including kidney failure observed in post-marketing reports, leading to enhanced monitoring recommendations.[43] This evolved into a boxed warning added to the U.S. label in February 2010, highlighting risks of renal failure, hepatic failure, and gastrointestinal (GI) hemorrhage, particularly in patients with comorbidities.[44] A 2013 label revision further emphasized GI bleeding risks, noting increased potential when combined with ulcerogenic drugs and requiring discontinuation in cases of severe hemorrhage.[9] In 2019, FDA analysis of post-marketing data reinforced these warnings, linking deferasirox to fatal outcomes including deaths from renal failure, hepatic injury, and GI hemorrhage, especially in elderly patients or those with advanced hematologic malignancies.[45]Deferasirox is available in over 100 countries worldwide, supporting global management of transfusional iron overload. Prior to generic entry, annual treatment costs exceeded $30,000 per patient in the U.S., driven by branded pricing; as of 2024, generics have reduced this to approximately $3,000–$8,000 annually, improving affordability in low- and middle-income settings.[46][47]