MedPath

Deferoxamine Advanced Drug Monograph

Published:Aug 12, 2025

Generic Name

Deferoxamine

Brand Names

Desferal

Drug Type

Small Molecule

Chemical Formula

C25H48N6O8

CAS Number

70-51-9

Associated Conditions

Aluminum overload, Chronic Iron Overload, Chronic aluminum overload, Iron Overload

Deferoxamine (DB00746): A Comprehensive Pharmacological and Clinical Monograph

1.0 Executive Summary & Introduction

1.1 Overview

Deferoxamine, also known by its chemical name desferrioxamine and the brand name Desferal, is a hexadentate chelating agent with a long and established history in clinical medicine.[1] It is classified as a small molecule drug with the DrugBank identification number DB00746 and CAS Number 70-51-9.[2] The primary therapeutic application of Deferoxamine is the management of metal toxicity, specifically as a first-line parenteral antidote for acute iron intoxication and as a cornerstone therapy for chronic iron overload resulting from conditions that necessitate frequent blood transfusions, such as thalassemia and sickle cell disease.[1] Furthermore, it is employed in an off-label capacity for the treatment of aluminum toxicity, particularly in patients with renal failure undergoing dialysis.[1] The molecule itself is a natural siderophore, a high-affinity iron-chelating compound, isolated from the actinomycete

Streptomyces pilosus.[2] This biological origin is fundamental to its potent and specific therapeutic action.

1.2 Historical and Clinical Context

The journey of Deferoxamine from a microbial metabolite to a life-saving medication is a compelling narrative of scientific serendipity and methodical re-evaluation. Its development began not as a quest for a chelator, but during research into iron-containing antibiotics.[8] This initial line of inquiry led to the isolation of ferrioxamines, which were initially conceived as potential iron supplements. However, when these iron-laden molecules proved to be so stable that the iron was not bioavailable and was simply excreted, a crucial insight emerged: the iron-free precursor must be an exceptionally powerful iron binder.[8] This pivot led to the production of Deferoxamine, which received FDA approval in 1968 and has since been recognized as an essential medicine by the World Health Organization.[1]

The natural origin of Deferoxamine is not merely a historical curiosity; it is the very foundation of its pharmacological efficacy. Siderophores are molecules that bacteria and other microorganisms have evolved over millennia for the express purpose of scavenging ferric iron (Fe3+), a vital but scarce nutrient, from their environment.[7] This evolutionary pressure has resulted in a molecule that is exquisitely optimized for binding iron with extremely high affinity and specificity. The clinical application of Deferoxamine, therefore, represents a brilliant repurposing of a highly specialized natural product. Rather than designing a chelator from first principles, researchers harnessed a molecule already perfected by nature for the task. This inherent biological purpose explains its remarkable potency and its ability to effectively remove toxic iron from human tissues without disrupting essential iron-containing proteins like hemoglobin.[5]

For decades, Deferoxamine was the sole agent available for managing chronic iron overload, dramatically improving survival in patients with transfusion-dependent anemias.[1] However, its clinical use is constrained by a challenging pharmacokinetic profile that necessitates prolonged, painful parenteral infusions.[1] This significant burden on patients, particularly children and adolescents, has led to issues with compliance and, consequently, suboptimal treatment outcomes. This major unmet clinical need was the primary catalyst for the development and eventual adoption of a new generation of oral iron chelators, namely Deferasirox and Deferiprone.[11] Today, Deferoxamine is no longer the only option but holds a vital and nuanced position within a more sophisticated, multi-agent approach to chelation therapy.

2.0 Chemical Identity and Physicochemical Properties

2.1 Identifiers and Nomenclature

To ensure unambiguous identification in scientific literature, regulatory documents, and clinical practice, Deferoxamine is cataloged under a comprehensive set of identifiers.

  • Drug Name: Deferoxamine [1]
  • Synonyms and Brand Names: Desferrioxamine, Desferrioxamine B, DFOA, Desferal, Desferrin, Desferan [1]
  • DrugBank ID: DB00746 [1]
  • CAS Number: 70-51-9 (for the free base) [2]
  • IUPAC Name: 1-Amino-6,17-dihydroxy-7,10,18,21-tetraoxo-27-(N-acetyl hydroxylamino)-6,11,17,22-tetraazaheptaeicosane [13]
  • Other Key Identifiers:
  • UNII: J06Y7MXW4D [2]
  • ChEBI ID: CHEBI:4356 [2]
  • ChEMBL ID: CHEMBL556 [2]
  • European Community (EC) Number: 200-738-5 [2]

2.2 Chemical and Physical Characteristics

The physicochemical properties of Deferoxamine are critical determinants of its formulation, pharmacokinetic behavior, and mechanism of action. It is typically supplied for clinical use as Deferoxamine mesylate, a salt form that enhances its stability and handling properties.[2]

  • Molecular Formula: C25​H48​N6​O8​ [13]
  • Molecular Weight: Approximately 560.68 g/mol for the free base; 656.79 g/mol for the mesylate salt [14]
  • Physical Description: A sterile, white to off-white lyophilized powder.[2]
  • Solubility: Deferoxamine mesylate is freely soluble in water, with a reported solubility of 12000 mg/L at 20°C, and is slightly soluble in methanol.[2] This high water solubility is essential for its parenteral formulation and for the renal excretion of its metal complexes.
  • Melting Point: The melting point of the free base is approximately 140°C. Different salt forms have distinct melting points; for instance, the methanesulfonate (mesylate) salt melts at 148-149°C.[2]
  • Partition Coefficient (LogP): The LogP value is reported as -2.2, indicating that the molecule is highly hydrophilic (water-loving) and has low lipid solubility.[2] This property explains its poor absorption from the gastrointestinal tract and its limited ability to cross cell membranes without active transport mechanisms.
  • Stability: When reconstituted, solutions of Deferoxamine mesylate are stable for up to two weeks when stored at room temperature, which provides some flexibility in clinical settings.[2]

The consolidation of these fundamental identifiers and properties into a single reference table provides a clear and efficient data sheet for researchers, clinicians, and regulatory scientists, preventing ambiguity and facilitating cross-referencing across diverse information sources.

Table 2.1: Summary of Deferoxamine Identifiers and Physicochemical Properties

PropertyValueSource(s)
Identifiers
DrugBank IDDB007462
CAS Number (Free Base)70-51-92
CAS Number (Mesylate Salt)138-14-713
UNIIJ06Y7MXW4D2
IUPAC Name1-Amino-6,17-dihydroxy-7,10,18,21-tetraoxo-27-(N-acetyl hydroxylamino)-6,11,17,22-tetraazaheptaeicosane13
Physicochemical Properties
Molecular FormulaC25​H48​N6​O8​13
Molecular Weight (Free Base)~560.68 g/mol14
Physical DescriptionWhite to off-white lyophilized powder2
Water Solubility (20°C)12000 mg/L (freely soluble)2
LogP-2.22
Melting Point (Free Base)~140°C2

3.0 Historical Context and Development

3.1 Serendipitous Discovery

The discovery of Deferoxamine is a classic example of scientific progress arising from unexpected observations and the willingness to pivot research direction. In the late 1950s and early 1960s, a collaboration between scientists at Ciba (now Novartis), the Swiss Federal Institute of Technology in Zurich, and the University Hospital in Freiburg was investigating soil microorganisms for novel antibiotics.[1] Their primary goal was to isolate iron-containing antibiotic compounds known as ferrimycines from the actinomycete

Streptomyces pilosus.[8] During this process, they also identified and isolated iron-containing impurities that acted as antagonists to these antibiotics; these were named ferrioxamines.[8] The initial project focused on the antibiotic properties was soon abandoned due to the rapid development of bacterial resistance. However, the research team astutely shifted its focus to the ferrioxamine impurities, exploring their potential therapeutic applications.[8]

3.2 From Failed Supplement to Successful Chelator

The first hypothesis for the therapeutic use of ferrioxamines was logical but ultimately incorrect. Given that these molecules contained iron, the investigators posited that they could serve as effective iron supplements for patients with iron-deficiency anemia.[8] To their surprise, animal and human studies revealed that when administered, the ferrioxamine complex was so stable that very little iron was released or absorbed by the body; the compound was largely excreted intact.[8] This "failure" proved to be the critical turning point. The researchers reasoned that if the iron-containing form of the molecule was so exceptionally stable and had such a high affinity for iron, then the

iron-free version of the molecule must be an incredibly powerful agent for binding and removing excess iron from the body.[8] This deductive leap led directly to the creation of the iron-free preparation, Deferoxamine (DFO), first produced in December 1960.[8]

3.3 Path to Clinical Use

With this new hypothesis, research proceeded rapidly. Animal experiments in rabbits and dogs confirmed that Deferoxamine administration led to a significant increase in urinary iron excretion and demonstrated a favorable safety profile.[8] Following successful tolerability tests in human volunteers, the first patient with hemochromatosis was treated with Deferoxamine in 1961, with promising results that validated the new therapeutic concept.[9] Further clinical trials were conducted, and the drug's path to market was remarkably swift for its time. Deferoxamine was officially registered in Switzerland in 1963, only two years after its first therapeutic use.[8] The United States Food and Drug Administration (FDA) granted its approval for use as an iron chelator in 1968.[1]

3.4 Global Recognition

The profound impact of Deferoxamine on the management of iron overload disorders was quickly recognized worldwide. In 1979, it was added to the World Health Organization's List of Essential Medicines, a designation reserved for medications considered to be the most effective and safe to meet the most important needs in a health system.[1] This cemented its status as a critical, life-saving medication on a global scale, a status it retains to this day.

4.0 Pharmacodynamics: Molecular Mechanisms of Action

The therapeutic effects of Deferoxamine stem from its potent and highly specific ability to chelate metal ions, primarily iron and aluminum, as well as from more recently discovered pleiotropic effects on cellular pathways such as ferroptosis and hypoxia signaling.

4.1 Primary Mechanism: Iron Chelation

The principal mechanism of action of Deferoxamine is the chelation of trivalent (ferric) iron, Fe3+.[5]

  • Binding Stoichiometry and Complex Formation: Deferoxamine is a linear molecule containing three bidentate hydroxamic acid groups ((−C(=O)N(−OH)−)).[7] These three groups act as hexadentate ligands, meaning they provide six binding sites that wrap around a single ferric iron ion in a precise 1:1 molar ratio, forming an exceptionally stable, octahedral complex known as ferrioxamine.[5] This complex is highly water-soluble, which facilitates its excretion from the body.[6] The binding affinity of Deferoxamine for Fe3+ is extremely high, which allows it to effectively compete for and sequester iron from physiological storage sites. Quantitatively, 100 mg of Deferoxamine is theoretically capable of binding approximately 8.5 mg of ferric iron.[18]
  • Source of Chelated Iron: Deferoxamine acts on specific, pathologically relevant iron pools. It readily chelates iron from storage proteins, namely ferritin and hemosiderin, which are the primary sites of iron accumulation in overload states.[5] It also effectively binds free iron in the plasma, including non-transferrin-bound iron (NTBI) and labile plasma iron (LPI), which are highly redox-active and responsible for catalyzing the formation of damaging free radicals via the Fenton reaction.[5]
  • Selectivity and Safety Profile: A critical feature of Deferoxamine's pharmacodynamic profile is its selectivity. It does not readily remove iron that is already incorporated into functionally essential hemoproteins like hemoglobin and myoglobin, or from iron-containing enzymes such as the cytochromes.[5] It also does not easily chelate iron from transferrin, the body's primary iron transport protein. This selectivity is paramount to its clinical safety, as it allows the drug to target toxic, excess iron while sparing the iron required for normal physiological processes like oxygen transport and cellular respiration.

4.2 Aluminum Chelation

In addition to iron, Deferoxamine is an effective chelator of aluminum, which forms the basis for its off-label use in patients with aluminum toxicity, a condition most often seen in individuals with chronic kidney disease on long-term dialysis.[1] The mechanism involves Deferoxamine binding to aluminum that is deposited in tissues (such as bone) and in the plasma, forming a stable, water-soluble complex called aluminoxane.[21]

This process demonstrates a powerful synergistic interplay between pharmacology and a physical medical procedure. The chelation by Deferoxamine mobilizes aluminum from tissues into the bloodstream, which acutely increases the serum aluminum concentration.[5] This elevated plasma level creates a much steeper concentration gradient between the patient's blood and the dialysis fluid (dialysate) across the semipermeable dialysis membrane.[21] According to the principles of diffusion, this enhanced gradient drives a more efficient removal of the aluminoxane complex from the blood during the hemodialysis session.[5] Thus, Deferoxamine's efficacy in this setting is not merely chemical; it relies on the subsequent physical removal by dialysis to complete the detoxification process. This dual mechanism also explains a key safety concern: administering Deferoxamine to patients with very high baseline serum aluminum levels (e.g., >200 mcg/L) can be dangerous. The initial, sharp increase in circulating aluminum, before it can be cleared by dialysis, can precipitate or worsen neurological dysfunction, including seizures and dialysis dementia.[5]

4.3 Emerging and Pleiotropic Mechanisms

Beyond its established role as a metal chelator, ongoing research has revealed that Deferoxamine exerts several other biologically significant effects, opening up new avenues for its therapeutic application.

  • Ferroptosis Inhibition: Deferoxamine is a potent inhibitor of ferroptosis, a recently identified form of regulated cell death that is dependent on iron and characterized by the accumulation of lipid peroxides.[2] The core mechanism of ferroptosis involves the iron-catalyzed Fenton reaction, which generates highly destructive hydroxyl radicals that attack polyunsaturated fatty acids in cell membranes.[5] By tightly binding and sequestering free, redox-active iron, Deferoxamine effectively shuts down the Fenton reaction, thereby preventing the lipid peroxidation that drives ferroptotic cell death.[20] This mechanism is under active investigation for its potential neuroprotective effects in conditions associated with iron-mediated oxidative stress, such as intracerebral hemorrhage and spinal cord injury, and for its potential role in cancer therapy.[1]
  • HIF-1α Induction: Deferoxamine has been shown to induce the production of Hypoxia-Inducible Factor-1 alpha (HIF-1α).[15] HIF-1α is a master transcription factor that regulates the cellular response to low oxygen levels (hypoxia). By chelating iron, which is a necessary cofactor for the prolyl hydroxylase enzymes that mark HIF-1α for degradation, Deferoxamine stabilizes HIF-1α and allows it to accumulate, even under normal oxygen conditions, thereby mimicking a hypoxic state.[1] This leads to the upregulation of genes involved in angiogenesis (new blood vessel formation), erythropoiesis, and cell survival. This mechanism is the rationale behind its promising investigational use in improving blood flow and healing in chronic wounds and radiation-induced soft tissue injury.[8]
  • Anti-proliferative and Pro-apoptotic Effects: By depriving rapidly dividing cells of essential iron, Deferoxamine can exert anti-proliferative and anti-tumor effects.[15] Some studies have also shown that it can induce apoptosis (programmed cell death) and autophagy in certain cancer cell lines, further highlighting its potential as a repurposed oncologic agent.[15]

The discovery of these pleiotropic effects represents a significant evolution in the understanding of this half-century-old drug. What was once considered a simple "metal sponge" is now recognized as a modulator of fundamental cellular processes. This "re-branding" of Deferoxamine is driving a wave of research into its repurposing for a wide range of modern therapeutic challenges, including neurodegenerative diseases, ischemic injuries, and regenerative medicine, far beyond its original indication.

5.0 Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

The pharmacokinetic profile of Deferoxamine is the single most critical factor influencing its clinical use, administration regimens, and the historical drive for therapeutic alternatives. Its properties dictate that it must be administered parenterally via prolonged infusion, a reality that has shaped patient experience and clinical practice for decades.

5.1 Absorption

Deferoxamine is characterized by very poor absorption from the gastrointestinal tract when the mucosa is intact.[21] This low oral bioavailability necessitates its administration via parenteral routes. Following intramuscular (IM) or subcutaneous (SC) injection, the drug is rapidly absorbed into the systemic circulation.[21] Studies utilizing SC infusion have shown it follows a zero-order absorption model, achieving a steady plasma concentration over the duration of the infusion.[23]

5.2 Distribution

Once in the bloodstream, Deferoxamine exhibits the following distribution characteristics:

  • Plasma Protein Binding: It has very low affinity for plasma proteins, with less than 10% being bound in vitro.[21] This means the vast majority of the drug in circulation is free and available to chelate iron.
  • Distribution Model and Volume: Pharmacokinetic studies have shown that Deferoxamine distribution is best described by a bicompartmental model.[23] A key study in thalassemic patients receiving a continuous intravenous infusion reported a volume of distribution at steady state ( Vss​) of 1.35 ± 0.65 L/kg and a volume of distribution at the terminal phase (Vz​) of 1.88 ± 1.0 L/kg.[24] These values indicate that the drug distributes beyond the plasma volume into the extracellular fluid.
  • Cellular Penetration: Due to its high molecular weight and hydrophilic nature (low lipid solubility), Deferoxamine does not readily diffuse across cell membranes. It is thought to enter the intracellular compartment slowly via endocytic processes to access intracellular iron stores like ferritin.[23]

5.3 Metabolism

The metabolism of Deferoxamine occurs primarily in the plasma, mediated by plasma enzymes; hepatic metabolism is considered minimal.[18] The specific enzymatic pathways responsible for its biotransformation have not been fully elucidated, but several metabolites have been isolated.[21] An important pharmacodynamic consideration is that some of these metabolites, most notably the product of oxidative deamination, retain iron-chelating activity.[21] This suggests that metabolism does not necessarily terminate the drug's therapeutic effect, and these active metabolites contribute to the overall iron excretion. Studies have shown that in patients with low iron stores relative to their Deferoxamine dose, the proportion of plasma metabolites is higher, suggesting that metabolite levels could potentially be used to identify patients at risk of excessive dosing.[25]

5.4 Excretion

The elimination of Deferoxamine and its iron chelate, ferrioxamine, is biphasic and occurs through both renal and biliary routes.

  • Elimination Half-Life: Studies in healthy volunteers have described a biphasic elimination pattern, with a rapid initial phase half-life (t1/2​) of approximately 1 hour, followed by a slower terminal phase half-life of about 6 hours.[21] In iron-overloaded thalassemic patients, a study reported a terminal elimination half-life of 3.05 ± 1.30 hours following IV infusion.[24] This relatively short half-life is the primary reason why continuous, prolonged infusion is required to maintain therapeutic plasma concentrations.
  • Excretion Pathway: The ferrioxamine complex is highly water-soluble and is readily filtered by the kidneys, leading to its excretion in the urine.[5] This process imparts a characteristic reddish or pinkish "vin rosé" color to the urine, which can serve as a visible, albeit qualitative, indicator of effective chelation.[1] A portion of the chelate is also excreted in the feces via the bile.[5] The same renal excretion pathway applies to the aluminoxane complex formed during the treatment of aluminum toxicity.[5]

The pharmacokinetic profile of Deferoxamine is, in essence, its greatest strength and its most significant weakness. Its hydrophilic nature ensures that its metal complexes are readily excreted by the kidneys, but this same property contributes to its poor oral absorption. The short elimination half-life is a direct consequence of its efficient clearance, but it mandates the burdensome and often painful daily infusion regimens that can last 8-12 hours.[1] This administration challenge has been a major driver of patient non-compliance, which in turn leads to inadequate chelation, progressive organ damage from iron accumulation, and increased morbidity and mortality. This entire causal chain, rooted in the drug's fundamental ADME properties, created the profound unmet clinical need that directly spurred the development of oral iron chelators and continues to fuel research into novel, long-acting formulations of Deferoxamine, such as deferoxamine-conjugated nanoparticles (DFO-NPs).[23]

Table 5.1: Key Pharmacokinetic Parameters of Deferoxamine

ParameterValue / DescriptionSource(s)
Absorption
Oral BioavailabilityPoorly absorbed from the GI tract21
Parenteral AbsorptionRapidly absorbed after IM or SC administration21
Distribution
Plasma Protein Binding< 10%21
Volume of Distribution (Vss​)1.35 ± 0.65 L/kg (in thalassemic patients)24
Cellular EntrySlow, via endocytosis23
Metabolism
Site of MetabolismPrimarily plasma enzymes; minimal hepatic metabolism18
MetabolitesSeveral isolated, some retain chelating activity21
Excretion
Elimination PatternBiphasic21
Elimination Half-Life (t1/2​)Rapid phase: ~1 hour; Slow phase: ~6 hours (healthy volunteers) Terminal phase: ~3.05 hours (thalassemic patients)21
Route of ExcretionPrimarily renal (urine); some fecal (bile)5

6.0 Clinical Applications and Therapeutic Efficacy

Deferoxamine is a cornerstone therapy for managing toxic accumulation of iron and aluminum. Its clinical applications are well-defined, with two primary FDA-approved indications and several important off-label uses.

6.1 FDA-Approved Indications

The U.S. Food and Drug Administration has approved Deferoxamine for the treatment of both acute and chronic iron overload states.[3]

6.1.1 Acute Iron Intoxication

In the setting of acute iron poisoning, which is a medical emergency particularly common in small children, Deferoxamine serves as a critical antidote.[1] It is important to note that it is indicated as an

adjunct to, and not a substitute for, standard supportive measures. These primary interventions include gastrointestinal decontamination (e.g., gastric lavage, whole-bowel irrigation), maintenance of a clear airway, correction of metabolic acidosis, and hemodynamic support to control shock with intravenous fluids and vasopressors.[4]

Chelation therapy with Deferoxamine is specifically recommended for patients who present with severe manifestations of iron toxicity, such as coma, seizures, persistent vomiting, metabolic acidosis, or signs of shock, or for those with a peak serum iron concentration exceeding 500 mcg/dL.[10] For patients who are hemodynamically stable, the preferred route of administration is intramuscular (IM). For patients in a state of cardiovascular collapse or shock, slow intravenous (IV) infusion is required.[4]

6.1.2 Chronic Transfusional Iron Overload

Deferoxamine is indicated for the treatment of chronic iron overload that results from multiple blood transfusions in patients with chronic anemias.[3] Since the human body has no physiological mechanism for excreting excess iron, patients who receive regular transfusions—such as those with beta-thalassemia, sickle cell disease, myelodysplastic syndromes (MDS), or other transfusion-dependent anemias—inevitably accumulate toxic levels of iron in their organs.[1]

Long-term chelation therapy with Deferoxamine has been proven to be life-saving. It promotes the excretion of excess iron, thereby reducing the total body iron burden. This has profound beneficial effects, including slowing the accumulation of iron in the liver, which retards or even reverses the progression of hepatic fibrosis, and improving cardiac function by preventing or delaying the onset of iron-associated cardiomyopathy.[1] Treatment with Deferoxamine has been shown to reduce mortality in transfusion-dependent patients with beta-thalassemia and sickle cell disease.[1]

Clinical guidelines recommend initiating chelation therapy once there is clear evidence of significant iron overload, which is typically defined by a serum ferritin level that consistently exceeds 1,000 mcg/L or after a patient has received approximately 100 mL/kg of packed red blood cells (equivalent to about 20 units for a 40 kg person).[4] Its efficacy is supported by numerous clinical trials, including studies specifically in patients with beta-thalassemia (NCT00061750), sickle cell disease (NCT00067080), and MDS (NCT00110266).[31]

6.2 Off-Label and Investigational Uses

Beyond its approved indications, Deferoxamine is utilized in several other clinical contexts and is the subject of ongoing research for new applications.

  • Aluminum Toxicity: Deferoxamine is widely accepted as the standard of care for the diagnosis and treatment of aluminum-associated pathology, such as osteomalacia (bone disease) and encephalopathy, in patients with chronic renal failure on dialysis.[1] It is important to note that while this is a common and guideline-supported practice, it remains an off-label use in the United States.[1]
  • Aceruloplasminemia: It is used in the management of aceruloplasminemia, a rare autosomal recessive genetic disorder characterized by impaired iron metabolism, leading to iron accumulation in the brain and visceral organs.[1]
  • Neuroprotection: There is growing interest in Deferoxamine's potential neuroprotective effects. It has been studied in the context of intracerebral hemorrhage, where the breakdown of blood releases iron that causes significant secondary oxidative injury to brain tissue.[1] By chelating this free iron, Deferoxamine may mitigate this damage, although the clinical evidence for its efficacy and safety in this indication is still considered weak.[1]
  • Wound Healing and Radiation-Induced Fibrosis: A particularly promising area of investigation involves the use of Deferoxamine to treat chronic, non-healing wounds and radiation-induced fibrosis. This application leverages its ability to induce HIF-1α, which promotes angiogenesis and improves blood flow to ischemic, damaged tissues.[8]
  • Primary Hemochromatosis: Deferoxamine is not a first-line therapy for primary (hereditary) hemochromatosis. The standard of care for this condition is therapeutic phlebotomy (blood removal).[4] However, Deferoxamine may be considered a beneficial alternative for patients in whom phlebotomy is contraindicated or not tolerated.[10]

7.0 Comparative Analysis: Deferoxamine in the Modern Chelation Landscape

The introduction of oral iron chelators, Deferasirox (Exjade®, Jadenu®) and Deferiprone (Ferriprox®), has transformed the management of chronic iron overload, shifting the paradigm from a single-drug approach to a more nuanced, multi-agent strategy. A comparative analysis of Deferoxamine against these newer agents is essential for contemporary clinical decision-making.

7.1 Deferoxamine vs. Deferasirox (Oral)

  • Efficacy: The comparative efficacy of Deferoxamine and Deferasirox has yielded somewhat mixed results across different studies, likely due to variations in patient populations, study design, and endpoints. A study involving patients with beta-thalassemia concluded that Deferasirox was more effective than Deferoxamine in reducing serum ferritin and serum iron levels.[11] Conversely, a highly controlled metabolic iron balance study in six thalassemia patients found that Deferoxamine was more effective than Deferasirox in all participants, achieving negative iron balance in all six, whereas Deferasirox did so in only two.[35] This highlights that while Deferasirox is an effective oral agent, its efficacy can be variable, and Deferoxamine remains a highly potent standard.
  • Administration and Compliance: The most significant advantage of Deferasirox is its oral, once-daily administration, which offers a dramatic improvement in convenience and quality of life compared to the burdensome 8-12 hour daily subcutaneous infusions of Deferoxamine. This is expected to lead to better long-term patient compliance, a critical factor in preventing the consequences of chronic iron overload.[12]
  • Safety Profile: The safety profiles of the two drugs differ. Deferasirox is associated with a notable risk of renal toxicity (increased serum creatinine), hepatic toxicity (elevated transaminases), and gastrointestinal adverse events (abdominal pain, nausea, diarrhea).[12] While Deferoxamine has its own distinct safety concerns (ocular, otic, and injection site reactions), it is generally not associated with the same degree of renal or primary hepatic risk as Deferasirox.

7.2 Deferoxamine vs. Deferiprone (Oral)

  • Efficacy and Organ Specificity: Deferiprone has demonstrated non-inferiority to Deferoxamine in terms of overall iron reduction, as measured by changes in liver iron concentration (LIC) and serum ferritin.[38] The most critical distinction between these two agents lies in their organ-specific effects. Multiple studies have established that Deferiprone is superior to Deferoxamine in removing iron from the heart.[36] This is evidenced by greater improvements in cardiac T2* MRI values (a measure of myocardial iron) and better preservation or improvement of ventricular function in patients treated with Deferiprone. In contrast, the same studies often show that Deferoxamine is more effective at reducing liver iron concentration.[36] This differential organ targeting is a key concept in modern chelation therapy.
  • Safety Profile: Deferiprone's major safety concern is the risk of agranulocytosis (a severe drop in white blood cells), which occurs in a small percentage of patients and requires regular blood count monitoring. This is a serious adverse event not associated with Deferoxamine therapy.[11]

7.3 Combination Therapy

The complementary profiles of the available chelators—particularly the superior cardiac efficacy of Deferiprone and the potent hepatic efficacy of Deferoxamine—have led to the widespread clinical use of combination therapies. Combining Deferoxamine with an oral agent (either Deferiprone or Deferasirox) is a strategy employed for patients with severe iron overload or for those who do not respond adequately to monotherapy.[39] This approach aims to achieve more comprehensive and rapid iron removal from all affected organs. For instance, combining Deferoxamine and Deferasirox has been shown to produce a synergistic effect, significantly increasing urinary iron excretion and enabling patients to achieve negative iron balance when they could not with either drug alone.[35]

The evolution of iron chelation therapy has moved beyond a simple "one-size-fits-all" model. There is no longer a single "best" chelator, but rather a best therapeutic strategy tailored to the individual patient's specific clinical situation, organ involvement, and lifestyle considerations. A patient with significant liver iron but minimal cardiac involvement might be managed well with Deferoxamine or Deferasirox. However, a patient with evidence of developing iron-induced cardiomyopathy would be a prime candidate for Deferiprone, either as monotherapy or in combination with Deferoxamine, to specifically target the heart. The rise of these personalized, organ-targeted, and often multi-agent regimens reflects a sophisticated understanding of the distinct pharmacodynamic properties of each available drug. Deferoxamine, the oldest agent, remains an indispensable tool in this modern therapeutic toolkit.

Table 7.1: Comparative Profile of Deferoxamine, Deferasirox, and Deferiprone

FeatureDeferoxamineDeferasiroxDeferiprone
Route of AdministrationParenteral (SC, IV, IM)Oral (once daily)Oral (three times daily)
Dosing FrequencyDaily (8-12 hour infusion)Once dailyThree times daily
Primary Excretion RouteRenal (urine), Biliary (feces)Primarily Biliary (feces)Primarily Renal (urine)
Efficacy (Liver Iron)HighHigh (variable)Moderate
Efficacy (Cardiac Iron)ModerateLow to ModerateHigh (Superior)
Key ToxicitiesOcular/Otic Toxicity, Injection Site Reactions, Growth Retardation (peds)Renal Toxicity, Hepatic Toxicity, GI UpsetAgranulocytosis/Neutropenia, Arthralgia, GI Upset
MonitoringEyes, Ears, Renal function, Growth (peds)Renal function, Liver function, Auditory/OcularWeekly Blood Counts, Liver function
Sources11

8.0 Comprehensive Safety, Tolerability, and Risk Management

Deferoxamine has a well-characterized safety profile established over more than five decades of clinical use. While generally well-tolerated when used appropriately, it is associated with a range of adverse reactions, from common local effects to rare but serious systemic toxicities that require careful monitoring.

8.1 Adverse Reactions (by System Organ Class)

  • Local/Injection Site Reactions (Very Common): Given its parenteral route, reactions at the injection site are the most frequently reported adverse events. These include pain, swelling, erythema (redness), induration (hardening of tissue), pruritus (itching), and occasionally eschar or crust formation.[18] These reactions may be associated with systemic allergic features.
  • Musculoskeletal System (Common): Arthralgia (joint pain) and myalgia (muscle pain) are common systemic effects.[41] In pediatric patients, long-term therapy, particularly with high doses (e.g., >60 mg/kg/day), is associated with a significant risk of growth retardation and bone disorders, such as metaphyseal dysplasia.[1]
  • Special Senses - Ocular Toxicity (Rare but Serious): Prolonged use of Deferoxamine, especially at high doses or in patients with low iron stores, can lead to various forms of ocular toxicity. Reported effects include blurred vision, decreased visual acuity, visual field defects (scotoma), impaired color vision (dyschromatopsia) and night vision, optic neuritis, and the formation of cataracts.[1] Retinal pigmentary abnormalities have also been observed. In most cases, these disturbances are reversible if detected early and treatment is promptly discontinued.[18]
  • Special Senses - Otic Toxicity (Rare but Serious): Auditory abnormalities are another well-documented risk. These can manifest as tinnitus (ringing in the ears) and high-frequency sensorineural hearing loss.[1] As with ocular toxicity, the risk is higher with excessive dosing, and the effects are often reversible upon cessation of the drug.[18]
  • Renal System (Rare): Although the drug and its chelate are excreted by the kidneys, renal toxicity is uncommon but can be serious. Postmarketing reports include cases of increased serum creatinine, renal tubular disorders, and acute renal failure.[18] Patients with pre-existing renal impairment require careful monitoring.
  • Respiratory System (Rare but Potentially Fatal): A severe and potentially fatal complication is the development of Acute Respiratory Distress Syndrome (ARDS). This has been reported in patients receiving excessively high doses of Deferoxamine, particularly via continuous intravenous infusion lasting longer than 24 hours.[10] Symptoms include acute dyspnea, cyanosis, and interstitial infiltrates on chest imaging.
  • Hypersensitivity Reactions: Systemic allergic reactions can occur, ranging from generalized rash and urticaria (hives) to severe anaphylactic reactions with or without shock and angioedema.[18] Rapid intravenous infusion is known to cause a non-allergic reaction characterized by flushing, urticaria, hypotension, and shock, which is why slow infusion rates are mandated.[18]
  • Infections: Deferoxamine therapy is associated with an increased susceptibility to certain opportunistic infections. This is because Deferoxamine itself is a siderophore, and some bacteria, notably Yersinia enterocolitica and Yersinia pseudotuberculosis, can utilize the ferrioxamine complex to acquire iron, which enhances their growth.[18] If a Yersinia infection develops, Deferoxamine treatment should be interrupted until the infection is resolved. Rare cases of fatal mucormycosis (a fungal infection) have also been reported.[10]

8.2 Contraindications and Boxed Warnings

  • Contraindications: Deferoxamine is contraindicated in patients with a known hypersensitivity to the active substance. It is also contraindicated in patients with severe renal disease or anuria (inability to produce urine), because the drug and its iron chelate are primarily excreted by the kidneys.[4]
  • Boxed Warnings: The product labeling for Deferoxamine does not contain a boxed warning.[4]

8.3 Drug Interactions

The potential for drug interactions with Deferoxamine requires clinical vigilance. There are 37 known drug interactions, with 22 classified as major and 15 as moderate.[44]

  • Vitamin C (Ascorbic Acid): This is the most clinically significant and complex interaction. Concomitant administration of low-dose Vitamin C (e.g., up to 200 mg/day in adults) can be beneficial, as it mobilizes iron from intracellular stores, thereby increasing the amount of chelatable iron and potentiating the effect of Deferoxamine.[5] However, this same mobilization of iron can be dangerous. In patients with severe chronic iron overload and pre-existing cardiac dysfunction, the sudden increase in labile iron can potentiate iron's cardiac toxicity, leading to impaired cardiac function.[5] Therefore, high doses of Vitamin C should be avoided, and it should not be given at all to patients with known heart failure undergoing Deferoxamine therapy. If used, Vitamin C supplementation should only be started after at least one month of regular Deferoxamine therapy, and cardiac function should be monitored closely.[29]
  • Prochlorperazine: Concurrent use of Deferoxamine and prochlorperazine, a phenothiazine antiemetic and antipsychotic, may lead to a temporary loss of consciousness. The mechanism is believed to be a pharmacodynamic synergism leading to enhanced neurological effects.[29]
  • Gallium-67: Deferoxamine can interfere with diagnostic imaging. As it chelates Gallium-67, it can cause rapid urinary excretion of the radioisotope, leading to distorted or uninterpretable scan images. It is recommended to discontinue Deferoxamine 48 hours prior to undergoing a gallium scan.

Table 8.1: Clinically Significant Drug Interactions with Deferoxamine

Interacting AgentInteraction TypeClinical ConsequenceRecommended ManagementSource(s)
Vitamin C (Ascorbic Acid)PharmacodynamicLow Dose: Potentiates iron chelation. High Dose / Heart Failure: Potentiates cardiac iron toxicity, may impair cardiac function.Avoid in patients with heart failure. If used, start after 1 month of DFO therapy. Limit dose (e.g., ≤200 mg/day). Monitor cardiac function.5
ProchlorperazinePharmacodynamic SynergismMay cause temporary loss of consciousness and other neurological effects.Avoid concurrent use if possible. Monitor patient closely if combination is necessary.29
Oral Iron/Mineral SupplementsPharmacokinetic (Absorption)Deferoxamine chelates iron and other metals, preventing their absorption.Applies only to oral forms. DFO is not given orally. Interaction is primarily relevant if DFO were used to treat toxicity from oral supplements.29
Betibeglogene autotemcel / Exagamglogene autotemcel (Gene Therapies)Pharmacodynamic (Myelosuppression)Increased risk of infection due to myelosuppressive effects.Discontinue iron chelators at least 7 days prior to conditioning for gene therapy. Avoid use for 6 months post-infusion.29
Gallium-67Diagnostic InterferenceBinds Gallium-67, leading to rapid urinary excretion and distorted scan results.Discontinue Deferoxamine 48 hours prior to Gallium-67 scintigraphy.10

9.0 Dosing, Administration, and Clinical Monitoring Protocols

The appropriate dosing, administration, and monitoring of Deferoxamine are crucial for maximizing its efficacy while minimizing the risk of its significant toxicities. Protocols differ substantially between the treatment of acute intoxication and chronic overload.

9.1 Dosing and Administration

Deferoxamine is supplied as a lyophilized powder in vials (e.g., 500 mg and 2 g) that must be reconstituted for parenteral administration.[18]

9.1.1 Acute Iron Intoxication

  • Intramuscular (IM) Route: This is the preferred route for all patients who are not in shock. The typical initial dose is 1000 mg. This may be followed by 500 mg every 4 hours for two doses, and then subsequent doses of 500 mg every 4 to 12 hours, depending on the clinical response.[4]
  • Intravenous (IV) Route: This route should be reserved for patients who are in a state of cardiovascular collapse or shock. The initial dose is 1000 mg administered as a slow IV infusion. The infusion rate must be strictly controlled and should not exceed 15 mg/kg/hour to avoid hypotension.[28] Subsequent doses can be given at a rate not exceeding 125 mg/hour.
  • Maximum Dose: The total amount administered should not exceed 6,000 mg in a 24-hour period.[4]

9.1.2 Chronic Iron Overload

  • Subcutaneous (SC) Infusion: This is the standard method for long-term administration. The usual daily dose is between 1000 mg and 2000 mg, which corresponds to an average daily dose of 20 to 60 mg/kg. This dose is infused slowly over a period of 8 to 24 hours using a small, portable infusion pump. Therapy is typically administered 5 to 7 days per week.[1]
  • Intravenous (IV) Infusion: For patients who already have long-term IV access (e.g., a central venous catheter), IV infusion is an alternative. The dose is typically 40-50 mg/kg/day for adults and 20-40 mg/kg/day for children, infused over 8 to 12 hours, 5 to 7 days per week. The maximum recommended daily dose for children is 40 mg/kg/day until growth has ceased, and for adults, the average dose should not exceed 60 mg/kg/day. The infusion rate should not exceed 15 mg/kg/hour.[28]
  • Intramuscular (IM) Administration: A daily dose of 500-1000 mg may be administered IM, but this route is less effective than slow infusion for chronic chelation and is generally reserved for situations where infusion is not feasible. The total daily IM dose should not exceed 1000 mg.[28]

9.2 Clinical Monitoring Protocols

Regular and comprehensive monitoring is essential to guide therapy and ensure patient safety.

9.2.1 Efficacy Monitoring

  • Serum Ferritin: Serum ferritin levels are the most common tool for monitoring total body iron stores. In chronic overload, therapy is often initiated when levels exceed 1,000 mcg/L. The goal of therapy is to reduce and maintain ferritin levels below this threshold, often targeting a level above 500 mcg/L to avoid over-chelation.[4]
  • Liver and Cardiac MRI: Magnetic Resonance Imaging (MRI) techniques are the gold standard for non-invasively quantifying organ-specific iron deposition. Liver Iron Concentration (LIC) is measured to assess hepatic iron stores, while cardiac T2* (or R2*) MRI is used to measure myocardial iron, which is a key predictor of cardiac complications.[5] These assessments are crucial for tailoring therapy, especially when considering combination chelation.
  • Urine Color: The characteristic reddish "vin rosé" color of the urine indicates that the drug is actively chelating and excreting iron. While not a quantitative measure, its absence may suggest that the chelatable iron pool has been depleted.[1]

9.2.2 Safety Monitoring

  • Ophthalmologic Monitoring: Due to the risk of ocular toxicity, a baseline ophthalmologic examination is required before starting long-term therapy. This should include visual acuity tests, slit-lamp examination, and funduscopy. These examinations should be repeated periodically, typically every 3 months, for patients on chronic therapy.[10] Patients should be instructed to report any changes in vision immediately.
  • Auditory Monitoring: A baseline audiogram is recommended, with periodic follow-up testing to detect any high-frequency hearing loss, especially in patients on high-dose or prolonged therapy.[10]
  • Renal Function: Serum creatinine and other measures of renal function should be monitored regularly in all patients, as Deferoxamine can cause renal impairment.[18]
  • Pediatric Growth Monitoring: For children receiving Deferoxamine, height and weight must be carefully tracked and plotted on growth charts at least every 3 months to monitor for growth retardation, a known complication of over-chelation or high-dose therapy in this population.[19]

10.0 Special Populations and Overdose Management

The use of Deferoxamine requires special consideration in certain patient populations due to differing risks and benefits. Management of overdose, both of iron (for which Deferoxamine is the treatment) and of Deferoxamine itself, follows specific protocols.

10.1 Pediatric Use

  • Age Limitation: The safety and efficacy of Deferoxamine have not been established in children younger than 3 years of age.[4] Iron mobilization with the drug is relatively poor in this age group unless there is already a significant iron burden.[18] Therefore, it is generally not recommended for these patients unless significant iron excretion (e.g., ≥1 mg/day) can be demonstrated.
  • Growth Retardation: Children are at a high risk for growth retardation and bone changes (metaphyseal dysplasia), particularly if they receive excessive doses (e.g., >40-60 mg/kg/day) or if therapy is initiated before a substantial iron load has accumulated.[1] This underscores the critical importance of careful dosing based on weight and close monitoring of height and weight every 3 months.[27]

10.2 Geriatric Use

  • Dosing and Monitoring: Deferoxamine should be used with caution in elderly patients. Dose selection should generally start at the low end of the dosing range, reflecting the higher frequency of decreased renal, hepatic, or cardiac function in this population.[18] Elderly patients may also be more susceptible to the ocular and auditory toxicities of the drug, requiring vigilant monitoring.[18]

10.3 Pregnancy and Lactation

  • Pregnancy: Deferoxamine is classified as US FDA Pregnancy Category C and Australian TGA Pregnancy Category B3.[47] Animal reproduction studies have shown skeletal anomalies and delayed ossification in the offspring of rabbits and mice at doses exceeding the maximum human dose.[47] There are no adequate and well-controlled studies in pregnant women. Therefore, Deferoxamine should be used during pregnancy only if the potential benefit to the mother clearly justifies the potential risk to the fetus.[18] In cases of severe, life-threatening acute iron intoxication, the risk of untreated poisoning to both the mother and fetus may be greater than the risk posed by the drug itself.[49]
  • Lactation: It is not definitively known whether Deferoxamine is excreted in human milk.[18] However, the drug is known to be very poorly absorbed from the gastrointestinal tract.[21] This pharmacokinetic property suggests that even if small amounts were present in breastmilk, it is unlikely to be absorbed by the infant and reach systemic circulation in significant quantities.[50] Limited case reports of mothers with beta-thalassemia who breastfed while receiving daily subcutaneous Deferoxamine found normal iron levels and no adverse effects in their infants.[50] While some experts advocate for its continued use during breastfeeding, the general recommendation is to exercise caution. A decision should be made to either discontinue breastfeeding or discontinue the drug, taking into account the importance of the drug to the mother. If breastfeeding is continued, monitoring of the infant's serum iron is advised.[19]

10.4 Overdose Management

10.4.1 Overdose of Iron (Acute Iron Poisoning)

Deferoxamine is the specific antidote for severe iron poisoning.

  • Treatment: Management involves supportive care and the administration of Deferoxamine, typically as an intravenous infusion at a rate of 15 mg/kg/hour.[51] The infusion should be continued until the patient is clinically stable, metabolic acidosis has resolved, and serum iron concentrations have fallen to a non-toxic level (e.g., <60 micromol/L or <350 mcg/dL).[51]
  • Role of Hemodialysis: In patients with massive iron overdose who develop renal failure or anuria, the ferrioxamine complex cannot be excreted and will accumulate. In these cases, hemodialysis may be necessary to remove the water-soluble ferrioxamine complex from the blood, which has been shown to be successful in improving clinical status.[51]

10.4.2 Overdose of Deferoxamine

Inadvertent overdose of Deferoxamine can lead to toxicity.

  • Symptoms: The primary manifestations of Deferoxamine overdose are related to its known adverse effects. Rapid infusion can cause severe hypotension and an anaphylactoid reaction.[49] Prolonged high-dose infusion (>24 hours) can lead to ARDS.[27] Acute renal failure and neurological disturbances have also been reported.[42]
  • Management: There is no specific antidote for Deferoxamine overdose. Management is entirely supportive. The infusion should be immediately stopped or the rate significantly reduced. Treatment should focus on managing hypotension with IV fluids and vasopressors if necessary, and providing respiratory support for patients who develop ARDS.

11.0 Future Directions and Research Perspectives

Despite being a mature drug, Deferoxamine remains a subject of active research, with future developments focused on overcoming its limitations and harnessing its newly understood mechanisms for novel therapeutic applications.

11.1 Novel Formulations and Delivery Systems

The most significant drawback of Deferoxamine is its pharmacokinetic profile, which mandates burdensome parenteral infusions. This has been the primary impetus for innovation. The most promising research frontier is the development of novel delivery systems designed to prolong the drug's half-life and improve its bioavailability. A leading example is the creation of deferoxamine-conjugated nanoparticles (DFO-NPs).[23] Preclinical studies in animal models have shown that these nanochelators exhibit a substantially prolonged half-life and favorable bioavailability following subcutaneous administration. This could potentially translate into less frequent dosing regimens (e.g., once weekly instead of daily), which would represent a monumental improvement in convenience and patient compliance, thereby enhancing overall therapeutic efficacy.[26]

11.2 Repurposing for New Indications

The elucidation of Deferoxamine's pleiotropic effects on fundamental cellular pathways has opened up a wealth of possibilities for drug repurposing. Research is actively exploring its potential in several new therapeutic areas:

  • Neuroprotection: Leveraging its ability to inhibit iron-mediated oxidative stress and ferroptosis, Deferoxamine is being investigated as a neuroprotective agent in conditions such as acute spinal cord injury and intracerebral hemorrhage.[1] By chelating the toxic iron released from extravasated blood, it may limit secondary brain and spinal cord injury.
  • Oncology: The anti-proliferative effects of iron deprivation, combined with its ability to induce apoptosis and autophagy in some cancer cells, make Deferoxamine a candidate for repurposing as an anti-cancer agent, either alone or in combination with other chemotherapies.[15]
  • Tissue Regeneration and Wound Healing: The discovery that Deferoxamine induces HIF-1α, a master regulator of angiogenesis, has spurred significant interest in its use for treating chronic, ischemic wounds and mitigating the effects of radiation-induced fibrosis.[8] By promoting the formation of new blood vessels, it could improve oxygen and nutrient delivery to damaged tissues, thereby accelerating healing.

11.3 Optimizing Combination Therapies

As the landscape of iron chelation has evolved to include multiple agents with distinct profiles, a key area of ongoing research is the optimization of combination therapy protocols. Further clinical trials are needed to define the most effective and safest regimens for combining Deferoxamine with oral chelators like Deferiprone and Deferasirox. This includes determining the ideal patient populations, dosing schedules, and monitoring strategies to maximize synergistic effects on organ-specific iron removal while minimizing cumulative toxicity. Studies are even exploring triple-combination therapy (Deferoxamine + Deferiprone + Deferasirox) for the most severely iron-overloaded patients who are refractory to dual-agent treatment.[39]

12.0 Expert Synthesis and Conclusion

Deferoxamine stands as a foundational and life-saving medication in the fields of toxicology and hematology. Its remarkable efficacy is not an accident of synthetic chemistry but a direct inheritance from its natural origin as a high-affinity bacterial siderophore, a molecule perfected by evolution for the singular purpose of sequestering iron. For decades, it was the only available treatment for chronic iron overload, transforming conditions like beta-thalassemia from fatal childhood diseases into manageable chronic illnesses.

While its clinical utility remains undisputed, the primary narrative of Deferoxamine over the past twenty years has been one of challenge and innovation. Its demanding pharmacokinetic profile—characterized by poor oral absorption and a short half-life—necessitates a burdensome daily regimen of prolonged parenteral infusions. This significant treatment burden has been the principal catalyst for innovation in the field, directly driving the research, development, and clinical integration of more convenient oral alternatives, Deferasirox and Deferiprone.

However, Deferoxamine is far from obsolete. Rather, it has found a new, more sophisticated role within the modern therapeutic armamentarium. It remains the undisputed standard of care for acute iron poisoning and a potent option for chronic overload. Its place is no longer as a standalone, one-size-fits-all solution, but as a vital component of personalized, organ-targeted combination therapy. The recognition that different chelators have differential effects on specific organs—with Deferiprone showing superiority for cardiac iron and Deferoxamine retaining robust efficacy for hepatic iron—has ushered in an era of tailored medicine where treatment is matched to the individual patient's pattern of iron deposition.

Looking forward, the story of Deferoxamine is poised for another chapter. The recent elucidation of its pleiotropic mechanisms, including the inhibition of ferroptosis and the induction of HIF-1α, is unlocking its potential for repurposing in entirely new therapeutic areas such as neuroprotection, oncology, and regenerative medicine. Simultaneously, the development of novel delivery systems like nanochelators promises to finally address its long-standing pharmacokinetic limitations. The convergence of these two research paths ensures that this venerable drug, first isolated over sixty years ago, will continue to be a subject of scientific discovery and a source of clinical value for decades to come.

Works cited

  1. Deferoxamine - Wikipedia, accessed August 12, 2025, https://en.wikipedia.org/wiki/Deferoxamine
  2. Deferoxamine | C25H48N6O8 | CID 2973 - PubChem, accessed August 12, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Deferoxamine
  3. HIGHLIGHTS OF PRESCRIBING INFORMATION These highlights do not include all the information needed to use DESFERAL safely and effe - Novartis, accessed August 12, 2025, https://www.novartis.com/us-en/sites/novartis_us/files/desferal.pdf
  4. LA.PHAR.146 - Deferoxamine (Desferal) - Louisiana Healthcare Connections, accessed August 12, 2025, https://www.louisianahealthconnect.com/content/dam/centene/louisiana-health-connect/policies/pharmacy-policies/LA.PHAR.146-Deferoxamine-Desferal.pdf
  5. Deferoxamine - StatPearls - NCBI Bookshelf, accessed August 12, 2025, https://www.ncbi.nlm.nih.gov/books/NBK557654/
  6. Deferoxamine | 70-51-9 - ChemicalBook, accessed August 12, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB8506290.htm
  7. Deferoxamine B: A Natural, Excellent and Versatile Metal Chelator - MDPI, accessed August 12, 2025, https://www.mdpi.com/1420-3049/26/11/3255
  8. Chelating the valley of death: Deferoxamine's path from bench to wound clinic - Frontiers, accessed August 12, 2025, https://www.frontiersin.org/journals/medicine/articles/10.3389/fmed.2023.1015711/full
  9. Chelating the valley of death: Deferoxamine's path from bench to wound clinic - PMC, accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9975168/
  10. Deferoxamine Monograph for Professionals - Drugs.com, accessed August 12, 2025, https://www.drugs.com/monograph/deferoxamine.html
  11. View of Comparison of deferasirox and deferoxamine effects on iron ..., accessed August 12, 2025, https://ijmrr.medresearch.in/index.php/ijmrr/article/view/1499/2788
  12. Compare the Efficacy and Safety of Deferoxamine, Deferasirox, and ..., accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10919752/
  13. Deferoxamine free base| CAS# 70-51-9 | metal binder - MedKoo Biosciences, accessed August 12, 2025, https://www.medkoo.com/products/40753
  14. CAS 70-51-9 - Sigma-Aldrich, accessed August 12, 2025, https://www.sigmaaldrich.com/US/en/search/70-51-9?focus=products&page=1&perpage=30&sort=relevance&term=70-51-9&type=cas_number
  15. Deferoxamine | CAS 70-51-9 | TargetMol | Biomol.com, accessed August 12, 2025, https://www.biomol.com/products/chemicals/biochemicals/deferoxamine-tgm-t124358-1mg
  16. Deferoxamine | 70-51-9 | FD46531 - Biosynth, accessed August 12, 2025, https://www.biosynth.com/p/FD46531/70-51-9-deferoxamine
  17. Deferoxamine for peak identification CRS 1 - Sigma-Aldrich, accessed August 12, 2025, https://www.sigmaaldrich.com/US/en/product/sial/y0001937
  18. Desferal - accessdata.fda.gov, accessed August 12, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/016267s050lbl.pdf
  19. Deferoxamine - Mechanism, Indication, Contraindications, Dosing, Adverse Effect, Interaction, Lactation, Pregnanacy | Drug Index | Pediatric Oncall, accessed August 12, 2025, https://www.pediatriconcall.com/drugs/deferoxamine/1142
  20. Deferoxamine, deferiprone, and deferasirox mechanism of action in the... - ResearchGate, accessed August 12, 2025, https://www.researchgate.net/figure/Deferoxamine-deferiprone-and-deferasirox-mechanism-of-action-in-the-management-of-iron_fig1_360407154
  21. Deferoxamine: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 12, 2025, https://go.drugbank.com/drugs/DB00746
  22. Deferasirox: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 12, 2025, https://go.drugbank.com/drugs/DB01609
  23. Predicted and observed pharmacokinetic profile of deferoxamine. The... - ResearchGate, accessed August 12, 2025, https://www.researchgate.net/figure/Predicted-and-observed-pharmacokinetic-profile-of-deferoxamine-The-circles-represent-the_fig1_283717404
  24. Intravenous infusion pharmacokinetics of desferrioxamine in thalassaemic patients - PubMed, accessed August 12, 2025, https://pubmed.ncbi.nlm.nih.gov/8104123/
  25. Deferoxamine pharmacokinetics - PubMed, accessed August 12, 2025, https://pubmed.ncbi.nlm.nih.gov/11206963/
  26. Pharmacokinetics and Tissue Distribution of Deferoxamine-Based Nanochelator in Rats, accessed August 12, 2025, https://www.researchgate.net/publication/366525001_Pharmacokinetics_and_tissue_distribution_of_deferoxamine-based_nanochelator_in_rats
  27. Deferoxamine (injection route) - Side effects & dosage - Mayo Clinic, accessed August 12, 2025, https://www.mayoclinic.org/drugs-supplements/deferoxamine-injection-route/description/drg-20063362
  28. Clinical Policy: Deferoxamine (Desferal) | Ambetter Health, accessed August 12, 2025, https://www.ambetterhealth.com/content/dam/centene/ar-health-wellness/policies/Ambetter/Ambetter_Pharmacy_Policy/AR.PHAR.146%20deferoxamine%20(Desferal).pdf
  29. Desferal (deferoxamine) dosing, indications, interactions, adverse effects, and more, accessed August 12, 2025, https://reference.medscape.com/drug/desferal-deferoxamine-343722
  30. Clinical Policy: Deferoxamine (Desferal) - PA Health & Wellness, accessed August 12, 2025, https://www.pahealthwellness.com/content/dam/centene/Pennsylvania/policies/pharmacy-policies/Deferoxamine%20_Desferal_%20v1.2024.pdf
  31. Iron Overload Completed Phase 2 Trials for Deferoxamine (DB00746) | DrugBank Online, accessed August 12, 2025, https://go.drugbank.com/indications/DBCOND0027984/clinical_trials/DB00746?phase=2&status=completed
  32. Deferoxamine Completed Phase 3 Trials for Beta-Thalassemia Treatment - DrugBank, accessed August 12, 2025, https://go.drugbank.com/drugs/DB00746/clinical_trials?conditions=DBCOND0027982&phase=3&purpose=treatment&status=completed
  33. Deferoxamine Completed Phase 2 Trials for Sickling Disorder Due to Hemoglobin S Treatment - DrugBank, accessed August 12, 2025, https://go.drugbank.com/drugs/DB00746/clinical_trials?conditions=DBCOND0028117&phase=2&purpose=treatment&status=completed
  34. Deferoxamine - PubMed, accessed August 12, 2025, https://pubmed.ncbi.nlm.nih.gov/32491586/
  35. Optimizing the Use of Deferasirox: Evidence of Synergy When Combined with Deferoxamine | Blood | American Society of Hematology, accessed August 12, 2025, https://ashpublications.org/blood/article/116/21/5163/66757/Optimizing-the-Use-of-Deferasirox-Evidence-of
  36. Deferasirox, deferiprone and desferrioxamine treatment in ..., accessed August 12, 2025, https://haematologica.org/article/view/5849
  37. Deferoxamine - LiverTox - NCBI Bookshelf, accessed August 12, 2025, https://www.ncbi.nlm.nih.gov/books/NBK547998/
  38. Deferiprone vs deferoxamine for transfusional iron overload in SCD ..., accessed August 12, 2025, https://ashpublications.org/bloodadvances/article-abstract/6/4/1243/482836
  39. Efficacy and safety of deferoxamine, deferasirox and deferiprone triple iron chelator combination therapy for transfusion-dependent β-thalassaemia with very high iron overload: a protocol for randomised controlled clinical trial, accessed August 12, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10860110/
  40. Efficacy and safety of iron-chelation therapy with deferoxamine, defer - Dove Medical Press, accessed August 12, 2025, https://www.dovepress.com/efficacy-and-safety-of-iron-chelation-therapy-with-deferoxamine-deferi-peer-reviewed-fulltext-article-DDDT
  41. Deferoxamine Side Effects: Common, Severe, Long Term - Drugs.com, accessed August 12, 2025, https://www.drugs.com/sfx/deferoxamine-side-effects.html
  42. Deferoxamine Uses, Side Effects & Warnings - Drugs.com, accessed August 12, 2025, https://www.drugs.com/mtm/deferoxamine.html
  43. Desferal (Deferoxamine): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed August 12, 2025, https://www.rxlist.com/desferal-drug.htm
  44. Desferal Interactions Checker - Drugs.com, accessed August 12, 2025, https://www.drugs.com/drug-interactions/deferoxamine,desferal.html
  45. Deferoxamine (Desferal): Uses, Side Effects, Dosage & Reviews - GoodRx, accessed August 12, 2025, https://www.goodrx.com/deferoxamine/what-is
  46. reference.medscape.com, accessed August 12, 2025, https://reference.medscape.com/drug/desferal-deferoxamine-343722#:~:text=A%20product%20that%20may%20interact,more%20details%20before%20starting%20deferoxamine.
  47. Deferoxamine (Desferal) Use During Pregnancy - Drugs.com, accessed August 12, 2025, https://www.drugs.com/pregnancy/deferoxamine.html
  48. Desferal® deferoxamine mesylate for injection USP Vials Rx only Prescribing Information - accessdata.fda.gov, accessed August 12, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2007/016267s044lbl.pdf
  49. DEFEROXAMINE | Poisoning & Drug Overdose, 7e | AccessMedicine | McGraw Hill Medical, accessed August 12, 2025, https://accessmedicine.mhmedical.com/content.aspx?bookid=2284§ionid=248385823
  50. Deferoxamine - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed August 12, 2025, https://www.ncbi.nlm.nih.gov/books/NBK500811/
  51. Clinical Practice Guidelines : Iron poisoning - The Royal Children's Hospital, accessed August 12, 2025, https://www.rch.org.au/clinicalguide/guideline_index/iron_poisoning/
  52. Deferoxamine - Ontario Poison Centre, accessed August 12, 2025, https://www.ontariopoisoncentre.ca/siteassets/pdfs/english/faxable_deferoxamine.pdf
  53. Oct 31, 2019: When is Deferoxamine Indicated in Iron Toxicity? | Tennessee Poison Center, accessed August 12, 2025, https://www.vumc.org/poison-control/toxicology-question-week/oct-31-2019-when-deferoxamine-indicated-iron-toxicity
  54. Successful treatment of severe iron intoxication with gastrointestinal decontamination, deferoxamine, and hemodialysis - PubMed, accessed August 12, 2025, https://pubmed.ncbi.nlm.nih.gov/23635030/

Published at: August 12, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.