Next Article in Journal
Quercetin Induces Mitochondrial Apoptosis and Downregulates Ganglioside GD3 Expression in Melanoma Cells
Previous Article in Journal
Assessment of Key Factors Impacting Variability in AAV Vector Genome Titration by Digital PCR
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Puzzle of Aspirin and Iron Deficiency: The Vital Missing Link of the Iron-Chelating Metabolites

by
George J. Kontoghiorghes
Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
Int. J. Mol. Sci. 2024, 25(10), 5150; https://doi.org/10.3390/ijms25105150
Submission received: 2 April 2024 / Revised: 22 April 2024 / Accepted: 6 May 2024 / Published: 9 May 2024
(This article belongs to the Section Molecular Pharmacology)

Abstract

:
Acetylsalicylic acid or aspirin is the most commonly used drug in the world and is taken daily by millions of people. There is increasing evidence that chronic administration of low-dose aspirin of about 75–100 mg/day can cause iron deficiency anaemia (IDA) in the absence of major gastric bleeding; this is found in a large number of about 20% otherwise healthy elderly (>65 years) individuals. The mechanisms of the cause of IDA in this category of individuals are still largely unknown. Evidence is presented suggesting that a likely cause of IDA in this category of aspirin users is the chelation activity and increased excretion of iron caused by aspirin chelating metabolites (ACMs). It is estimated that 90% of oral aspirin is metabolized into about 70% of the ACMs salicyluric acid, salicylic acid, 2,5-dihydroxybenzoic acid, and 2,3-dihydroxybenzoic acid. All ACMs have a high affinity for binding iron and ability to mobilize iron from different iron pools, causing an overall net increase in iron excretion and altering iron balance. Interestingly, 2,3-dihydroxybenzoic acid has been previously tested in iron-loaded thalassaemia patients, leading to substantial increases in iron excretion. The daily administration of low-dose aspirin for long-term periods is likely to enhance the overall iron excretion in small increments each time due to the combined iron mobilization effect of the ACM. In particular, IDA is likely to occur mainly in populations such as elderly vegetarian adults with meals low in iron content. Furthermore, IDA may be exacerbated by the combinations of ACM with other dietary components, which can prevent iron absorption and enhance iron excretion. Overall, aspirin is acting as a chelating pro-drug similar to dexrazoxane, and the ACM as combination chelation therapy. Iron balance, pharmacological, and other studies on the interaction of iron and aspirin, as well as ACM, are likely to shed more light on the mechanism of IDA. Similar mechanisms of iron chelation through ACM may also be implicated in patient improvements observed in cancer, neurodegenerative, and other disease categories when treated long-term with daily aspirin. In particular, the role of aspirin and ACM in iron metabolism and free radical pathology includes ferroptosis, and may identify other missing links in the therapeutic effects of aspirin in many more diseases. It is suggested that aspirin is the first non-chelating drug described to cause IDA through its ACM metabolites. The therapeutic, pharmacological, toxicological and other implications of aspirin are incomplete without taking into consideration the iron binding and other effects of the ACM.

1. Introduction

Iron is one of the essential metal ions and nutrients, and plays an important role for normal physiological functions and maintenance of healthy living in humans. Iron deficiency or overload, as well as other abnormalities of iron metabolism, may cause serious diseases which require treatment for the restoration of iron balance and normal bodily function [1,2,3,4,5,6].
Iron is an integral part of many enzymes and other proteins, as well as a modulator of transcription factors and other co-factors, which ensure the normal function, growth, and development of the human body. Iron also plays a major role in essential physiological processes such as oxygen transport, storage, and utilisation; electron transport; respiration; and other processes involving the metabolism of most natural products such as carbohydrates, lipids, proteins, and nucleic acids, as well as xenobiotic molecules, including drugs [1,2,3,7,8].
The health implications from changes in iron balance and homeostasis are enormous, considering that related diseases such as iron deficiency affect more than a quarter of the world’s population [2,3,7,9,10,11,12,13,14,15] and iron overload in idiopathic haemochromatosis affects one in ten persons of the Caucasian population [2,3,16,17,18,19,20]. Similarly, other iron-related diseases include the haemoglobinopathies, which are the most common group of genetic disorders in humans, with one of them being thalassaemia, identified as the disease with the highest morbidity and mortality rate related to iron or other metal toxicity worldwide [21,22,23,24,25].
Under normal physiological conditions, iron balance can be maintained despite the differences in iron requirements in otherwise healthy individuals. In these cases, the amount of iron absorbed from food is equivalent to the amount of iron excreted or lost from the body [2,7,9,14,15]. However, iron deficiency anaemia (IDA) is a multi-facet disease, which can be caused by many dietary, age, gender, and other factors; in pregnancy and teenagers; and from drugs, traumas, sport, and other activities such as blood donation, vitamin deficiency, malnutrition and different diseases, etc. [9,10,11,12,13,14,15,26,27,28,29,30,31,32,33,34].
The rate of iron absorption in healthy, normal individuals depends mainly in the quantity and quality of iron present in the gastrointestinal tract (GIT). For example, iron deficiency has been widely reported in vegetarian populations, where an insufficient amount of iron is absorbed from vegetarian food containing mostly non-haem iron components, in contrast to meat-containing food, which contains more readily absorbed lipophilic haem iron [7,14,15,28,29,30,31]. Iron deficiency could also be caused through inhibition of iron absorption in the GIT from interactions by many dietary molecules and also orally administered drugs [35,36,37,38,39,40,41,42,43,44]. In particular, the hydrophilic iron-chelating drug deferoxamine is routinely used for the inhibition of iron absorption and in cases of iron poisoning [44,45,46]. In contrast, the lipophilic iron complex of the natural product maltol (ferric iron tri-maltol, or feraccru or accrufer) increases iron absorption and is used for the treatment of IDA [46,47,48].
In contrast to many reports on studies related to the effect of drugs and dietary phytochelators on iron absorption, the effects of drugs on iron excretion have not been fully investigated and are not well documented, except in the case of the iron-chelating drugs deferoxamine, deferiprone, and defarasirox, which are widely used for increasing the excretion of excess iron in iron-loaded patients [7,49,50,51]. The three iron-chelating drugs can also cause iron deficiency in non-iron loaded individuals, as well as zinc deficiency, with other associated side effects following chronic administration [52,53,54,55,56,57,58,59,60]. In almost all the cases of the chelating drugs, natural chelators, and drug metabolites, the available form of iron for chelation and mobilization in humans is protein and non-protein bound and in the ferric state (Fe III) [7,14,43,48,51].
Several clinical investigations have recently reported that the chronic administration of low-dose acetylsalicylic acid or aspirin can cause IDA independent of major bleeding in a large number of elderly, otherwise healthy individuals. The mechanisms and mode of action of aspirin and the cause of IDA in this category of the elderly population have not been identified and are still largely unknown.
Aspirin is one of the most commonly used drugs, which is widely available and inexpensive and can be bought over pharmacy counters or prescribed by physicians. Furthermore, aspirin is one of the oldest drugs and has been in clinical use since 1897, with clinical experience involving different formulations for more than a century [61,62]. It has many uses, including analgesic, anti-inflammatory, antipyretic, and antithrombotic activity [61,62,63]. Different doses are used in general for each condition, including, for example, low doses of about 75–100 mg/day prophylactically for anti-platelet activity and cardiovascular complications, 325 mg or 80 mg twice daily for venous thromboembolism prophylaxis after joint arthroplasty, 300–600 mg every 4–6 h for analgesic function, and 1–2 g every 4–6 h for anti-inflammatory activity with maximum about 2.4–3.6 g/day [64,65,66,67,68]. A low dose of 75–100 mg/day is taken prophylactically by millions of people in the elderly (>60 years) population [64,65,66,67,68]. The main mechanism of action of aspirin is the irreversible inhibition of the iron-containing enzyme cyclo-oxygenase, involved in prostaglandin biosynthesis and platelet aggregation [63,69,70,71].
The pharmacokinetic, metabolic, and therapeutic properties of aspirin can vary between individuals, similarly to other drugs, and depend on absorption, distribution, metabolic, elimination, toxicity (ADMET), genomic, and other characteristics, as well as many factors such as drug dose, drug formulation, GIT dietary content, age and weight of individuals, etc. [72,73,74,75,76,77,78,79,80]. In general, aspirin is rapidly absorbed mainly from the stomach and upper small intestine and mostly hydrolyzed and metabolized to salicylic acid and other metabolites, all of which are excreted in the urine [72,76,81,82].
There is increasing evidence that chronic administration e.g., 3 or more years of low-dose aspirin of about 75–100 mg/day, could cause a 23% higher risk of IDA independent of major bleeding, in a large number of otherwise healthy individuals, usually greater than 65 years of age [83,84,85]. Considering that in addition to millions of normal individuals taking low-dose aspirin for prophylaxis, millions more of patients with cancer, chronic kidney disease, heart failure, diabetes, and many other diseases are also taking low-dose aspirin and are also at risk of IDA. The connection between aspirin use and IDA has not yet been fully characterised. It has recently been suggested that some of the aspirin metabolites have iron-chelating properties (ACM) and may be involved in the cause of IDA [86]. It has also been suggested that the metabolism of aspirin to ACM resembles the cardioprotective chelating pro-drug dexrazoxane, which is metabolized to an EDTA-like chelator [86].
The aim of this review is to examine the iron-binding properties of aspirin, salicylic acid, and other metabolites possessing iron chelating capacity, with emphasis on the prospect of their possible interference regarding iron balance and involvement in the cause of IDA following chronic administration in the aspirin-treated patient populations. Furthermore, it is also intended to examine other factors that may influence iron balance in aspirin-treated individuals, as well as to suggest possible therapeutic and other interventions that may restore iron balance and therapeutic outcomes in these categories of patients, including those with cardiovascular, kidney, cancer, and neurodegenerative diseases.

2. Metabolites of Aspirin with Iron Chelating Properties

There are many variations among individuals regarding the pharmacokinetic and metabolic profile of aspirin, and also other variations as a result of the different doses and formulations used [72,76,77,81,82]. Furthermore, several other factors could influence the pharmacokinetic and metabolic parameters of aspirin users, including age, gender, organ function, and interactions with food substances and other drugs [87,88].
In general, orally administered low-dose aspirin of about 75–100 mg is absorbed within minutes from the stomach and upper intestine with about 60–70% bioavailability. Aspirin is rapidly cleared from the circulation with half-life of about 15–20 min [81,82]. Aspirin is mostly metabolized or de-acetylated through hydrolysis, almost exclusively to salicylic acid, the first major metabolite in plasma, predominantly in the liver but also in the stomach, intestine, and blood [87,88]. Salicylic acid has a longer half-life in comparison to aspirin, of about 2–6 h, and is mostly metabolized in the liver, with only about 10% of unchanged salicylic acid being excreted in the urine [69,81,82,87,88,89,90,91]. The metabolites of salicylic acid are salicyluric acid, two glucuronide conjugates (salicylacyl glucuronide and salicylphenol glucuronide), and two dihydroxybenzoic acids (2,5-dihydroxybenzoic acid and 2,3-dihydroxybenzoic acid or 2,3-DHB) [87,88,89,90,91,92,93,94].
All the salicylate metabolites are cleared through the kidneys and excreted in the urine at different concentrations, with salicyluric acid predominating at about 70%, glucuronide conjugates at about 15%, and much lower quantities of dihydroxybenzoic acids at about 2–3% [81,82,87,88,89,90,91]. Many factors affect the level of aspirin and each of its metabolites in plasma following the metabolic transformation of aspirin [90,95,96,97]. The maximum concentration in plasma of aspirin administered at different doses, as well as its metabolites, depends mainly on the dose. The concentration of each of the aspirin metabolites in plasma also depends on many other factors, including, for example, the enzyme activity involved in each step of the metabolic transformation, such as differences between individuals in glucuronidation and the formation of glucuronide conjugates, as previously observed with other drugs [88,98,99].
The structural chemical characteristics, including potential iron-binding ligands and sites, are important parameters for the determination of the chelation potential of different drugs, metabolites, or other synthetic and natural molecules [7,14,43,48,51]. The chemical structures of aspirin and its metabolites, namely salicylic acid, salicyluric acid, 2,3-DHB, 2,5-dihydroxybenzoic acid (gentisic acid), salicylacyl glucuronide, and salicylphenol glucuronide, are shown in Figure 1a–g.
Chelators are, in general, organic compounds which possess at least two ligands with electron donor atoms such as O and N, which have affinity for binding metal ions and the appropriate proximity between them to form a coordinated covalent bond and, overall, a ring with the metal ion as the closing member. Within this context, there are many organic biological molecules, including some of the aspirin metabolites possessing electron donor atoms on ligands, which can be involved in metal complex formation. The donor atoms can be present in ligands of acidic groups such as -OH, O=C-OH, -NOH, where the proton can be displaced by the metal ion or in Lewis bases such as -C=O and -NH2 [7,14,43,48,51].
In considering the iron-binding capacity of aspirin and its metabolites, it appears from the chemical structures that almost all contain hydroxyl and other ligands with a potential for iron binding. In particular, the principal chelating sites in the aspirin metabolites with the required proximity for chelating iron are oxygen ligands from phenolic (-OH), carboxylate (O=C-OH), and carbonyl (-C=O) groups (Figure 1). Furthermore, an interchange between the iron-binding ligands may also take place in the case of, for example, 2,3-DHB depending on the pH, as previously suggested for similar chelating molecules [99]. In particular, the presence of the 2-hydroxyl (-OH) ligand increases the chelation potential of all metabolites except in the case of salicylphenol glucuronide, where the 2-hydroxyl group is replaced following conjugation with glucuronide, which diminishes the iron-binding potential site of this metabolite (Figure 1g).
In contrast to salicylphenol glucuronide, the metabolite with the highest iron chelating potential is 2,3-DHB, where the two adjacent hydroxyl ligands are similar to the catechol-containing chelators, such as those found in the bacterial siderophore enterobactin, which is a very powerful iron chelator synthesized by bacteria to scavenge iron from the surrounding environment [43,99,100]. Furthermore, aspirin and most of its metabolites contain a carboxylate metal-binding site similar to EDTA and DTPA, two well-known chelating drugs, and also the naturally occurring chelator citric acid, all of which have different iron binding potential [7,43,51,100]. The potential ligands involved in the iron-chelating binding sites in the case of the four most important ACMs are shown in Figure 2.
Following iron binding, a proton is released from the hydroxyl ligand (-OH) of the phenol group of the ACM, allowing the formation of oxygen-iron bond (-O-Fe) (Figure 2). Similarly, on iron binding a proton is also released from the carboxylate group (O=C-OH) of the ACM allowing the formation of oxygen-iron bond (O=C-O-Fe). In the case of salicyluric acid, iron binding is from the adjacent carbonyl (-C=O) group where a pair of electrons from oxygen (-C=O:) group is used for the bond with iron (-C=OFe).
In general, it appears from the chemical structural characteristics that all ACM are likely to be mostly bidentate chelators (Figure 2), which potentially can form a 3 chelator:1 iron molar ratio iron comples at physiological pH, with an octahedral structure similar to other bidentate iron chelating drugs, such as deferiprone [7,14,43,51,100]. However, at lower pH levels and low chelator metabolite concentration, 1:1 and 2:1 chelator: iron molar ratio complexes can also be potentially formed by the ACM [100]. Similarly, the distant phenolic (-OH), carboxylate (O=C-OH), and carbonyl (-C=O) groups present in aspirin and metabolites are also potential ligands for binding iron under certain conditions, including in cases of mixed iron complexes [99,100].
The iron-binding properties and effects of some of the ACMs have been previously reported from in vitro, in vivo, and clinical studies. In particular, at least four of the ACMs (2,3-DHB, 2,5-dihydroxybenzoic acid, salicyluric acid and salicylic acid) are known to be natural plant products involved in many other biological processes (Figure 2) [101,102,103].

3. In Vitro, In Vivo and Clinical Iron Studies of Aspirin and the Aspirin Metabolites

There are many studies reporting different interactions of aspirin and the aspirin metabolites with iron, and also iron metabolism in general. These interactions are similar to other chelating agents or molecules with iron-binding ligands but of different thermodynamic, kinetic, pharmacological, and toxicological parameters in each case [7,14,51,99,100]. In this section, a summary of some of the iron-binding effects of aspirin and the ACM will be introduced, with emphasis on the implications of iron chelation and iron metabolism.

3.1. Interactions of 2,3-Dihydroxybenzoic Acid with Iron

The most widely known and studied ACM for its iron binding properties is the naturally occurring 2,3-DHB [43,99,103]. The iron-binding site of 2,3-DHB, the smallest in production of ACM, are the two adjacent hydroxyl groups, which are involved in the formation of a 3 chelator:1 iron molar ratio iron complex at a physiological pH, similar to other catechol-containing chelators (Figure 2) [99,100]. There are hundreds of investigations, including clinical trials, reported regarding the iron-binding properties and effects of this aspirin metabolite. In particular, 2,3-DHB was selected for clinical use in the treatment of iron overload in thalassaemia patients from a group of 26 benzoic acids following preclinical studies in different animal species [104].
Several clinical investigations have been reported using 2,3-DHB. In one double-blind clinical trial involving 15 regularly red blood cell-transfused thalassaemia patients, the administration of 4 × 25 mg/kg/day of 2,3-DHB for one year resulted in the stabilization of the iron load in the patients [105]. Most importantly, there were no toxic side effects reported during the one-year clinical trial period [105]. In another iron balance clinical study involving 5 iron-loaded thalassaemia patients, 2,3-DHB administered at 25 mg/day for 8 days caused, on average, a 4.5 mg/day increase in iron excretion, while when the dose increased to 4 × 25 mg/kg in 8 patients for 21 days, the iron excretion increased on average to 6.5 mg/day. The drug was well-tolerated, with some mild gastrointestinal complaints [106].
Eventually, further clinical studies on the use of 2,3-DHB in the treatment of transfusional iron overload in thalassaemia were abandoned due to insufficient iron excretion in comparison to the much higher levels of iron intake from regular red blood cell transfusions in most patients [107]. However, although that this natural experimental chelator was not sufficiently effective, its low toxicity may have been useful for its use in combination therapies with other chelating drugs, as previously suggested [108].
More information on the safety and efficacy of 2,3-DHB was obtained from animal studies. In one such study using iron-loaded rats, 2,3-DHB was identified to be effective in mobilizing iron, without causing increases in calcium, magnesium, copper, or zinc excretion [109]. Further toxicity studies have shown that the LD50 for oral 2,3-DHB is more than 3 g/kg in rabbits, while chronic administration of 600 mg/kg in mice and rats has shown no signs of toxicity [109]. Furthermore, its oral administration caused a decrease in radiolabelled (Fe 59) iron absorption in rats, but in another study and under different conditions, a small increase in radiolabelled (Fe 59) absorption and body iron intake in mice was reported [46,109].
There are also many cell, protein, and other in vitro studies involving 2,3-DHB regarding its role as an iron chelator, natural siderophore, and an antioxidant [110,111]. For example, in cell studies, it has been shown to mobilize iron from ferritin, while in transferrin studies, iron could not be mobilized, but instead 2,3-DHB appeared to act as a synergistic anion in the iron binding site of transferrin [112,113]. In different cell studies, no cytotoxic effects on neuroblastoma cells have been shown by 2,3-DHB, while in studies on red blood cells, 2,3-DHB was not effective in transporting iron across the cell membrane, an activity facilitated mostly by other lipophilic chelators such as maltol [100,114,115].
The antioxidant activity of 2,3-DHB has been studied in different models of oxidative stress toxicity, where iron chelation has been considered as the major mechanism of the prevention of oxidative damage [116,117]. For example, the antioxidant effects of 2,3-DHB were examined in free radical toxicity models of the breakdown of deoxyribose, IgG damage due to UV irradiation, and muscle tissue damage due to homogenization. In these studies, the level of antioxidant activity of 2,3-DHB was proportional to its concentration, and usually, levels greater than 0.1 mM appeared to be effective for antioxidant activity in these three different models [118].
In the meantime, many clinical and in vivo studies examining the antioxidant effects of 2,3-DHB in several diseases have been carried out using aspirin as a pro-drug. In particular, increased production of 2,3-DHB and 2,5-dihydroxybenzoic acid was anticipated from aspirin metabolism in conditions with increased production of hydroxyl radical during oxidative stress [119,120,121]. In many cases, the increased production of these two ACMs, and especially 2,3-DHB, was implicated in the increased antioxidant effects observed during the administration of aspirin in inflammatory and other similar conditions [117].
Overall, 2,3-DHB appears to be a generally safe and an effective iron chelator for increasing iron excretion, and to possess high antioxidant activity.

3.2. Interactions of Salicylic Acid with Iron

There is wide interest in the properties and effects of salicylic acid as a natural plant product and dietary component, and as a first major metabolite of aspirin, not only in the context of therapeutic activity but also in different pharmacological, toxicological, ecological, and other aspects related to medicine [101,122,123,124,125,126,127]. In this section, only aspects related to iron will be discussed, despite the fact that the interaction of salicylic acid with iron and the formation of the iron-salicylate complex affects all these and related activities.
Salicylic acid was identified as an iron chelator in about the early 1950s and studied especially for its high iron-binding avidity, including the prospect of its clinical use, as well as its derivatives, in the treatment of transfusional iron overload in thalassaemia [128].
The iron binding site of salicylic acid consists of a carboxylate ligand and an adjacent hydroxyl ligand, as shown in Figure 2. It forms 3 chelator:1 iron molar ratio iron complexes at physiological pH, which are of an octahedral structure, and lower molar ratio chelator:iron complexes at low salicylate concentrations, similar to 2,3-DHB and other bidentate iron-chelating drugs [128,129,130,131].
There are many complexation, physicochemical, photochemical, and other studies regarding the interaction of salicylic acid with iron. In this context, the stability constant of its iron complex was determined (log β3 = 36) and found to be similar to that of transferrin and deferiprone, as well its ability to mobilize iron under physiological conditions (log K eff = 19). However, its ability to solubilize polynuclear iron deposits (log K sol = 1), such as those found in ferritin and haemosiderin has been found to be lower than that of deferiprone (log K sol = 9) [100,128].
The interaction of salicylic acid with iron is also utilized for detecting the use of aspirin under different conditions. In particular, the formation of the coloured ferric salicylate complex has been widely used as a diagnostic tool for the detection of aspirin toxicity or in cases of aspirin poisoning [132,133]. However, it should be noted that this method has many limitations and may not be specific to toxicity associated with aspirin. Furthermore, under experimental conditions using spectrofluorimetry, it has been shown that the formation of the ferric salicylate complex can cause quenching of the fluorescence intensity of salicylic acid [131,134].
Another field of investigation in relation to the interaction of salicylic acid with iron is redox activity, which also involves a diagnostic method for the measurement of increased production of hydroxyl radicals in free radical pathology by determining the increased rate of synthesis of 2,3-DHB from salicylic acid in the presence of iron [119,120]. Furthermore, salicylic acid has been shown to have iron chelating and antioxidant activity for example in attenuating gentamicin ototoxicity [135]. In contrast, under different experimental conditions, the redox activity of the iron–salicylate complex is able to induce lipid peroxidation and has been, for example, implicated in atherosclerosis, with profound effects in intravascular and intrahepatic lipids, and also plasma lipoprotein metabolism [136].
The similarity of the biological and clinical activities of aspirin and its initial main metabolite, salicylic acid, is difficult to separate in most cases, especially considering the rapid metabolism of the former to the latter following oral administration [81,82]. This question has become crucial, especially since many clinical findings have suggested that aspirin administration even at low doses (75 mg/day) can prevent colorectal cancer, other cancers, and metastasis [137]. Similar questions also arise as to whether the iron chelation properties of salicylic acid are implicated in the mode of its anticancer activity in colorectal cancer and also other cancers [95,138,139,140].
It is envisaged that further studies on the iron chelation properties of salicylic acid could provide important information related to the pharmacology of aspirin, its iron metabolic effects, and other similar interactions. Further studies are also needed to investigate the influence of iron and the salicylic acid iron complex on the metabolic transformation of salicylic acid in the rate of production of the other aspirin metabolites. In particular, the formation of iron complexes by salicylic acid may influence the production of glucuronide metabolites, similar to the rate of glucurodination of deferiprone observed in iron-loaded patients [97,98].

3.3. Interactions of 2,5-Dihydroxybenzoic Acid (Gentisic Acid) with Iron

2,5-Dihydroxybenzoic acid or gentisic acid has been reported to be a natural plant, a mammalian product, and also a metabolite of aspirin [81,82,102]. It is found in many fruits and vegetables and has been reported to have anticancer, anti-inflammatory, antioxidant, antimicrobial, hepatoprotective, neuroprotective, and other beneficial health effects [141]. It has iron-binding properties and has been suggested to act as a mammalian siderophore (chelator) in innate immunity against infection by competing with microbial siderophores [142,143,144].
The iron binding site of 2,5-dihydroxybenzoic acid is similar to that of salicylic acid and consists of the carboxylate ligand and its adjacent hydroxyl ligand (Figure 2). The formation of a 3 chelator:1 iron molar ratio iron complex at physiological pH, with an octahedral structure, is anticipated, similar to the other bidentate iron-chelating drugs and lower-molar-ratio chelator:iron complexes (2:1 and 1:1) under different conditions [100,145]. Furthermore, 2,5-dihydroxybenzoic acid is expected to be more hydrophilic and to form more hydrophilic iron complexes than salicylic acid due to the presence of the extra 5-hydroxyl group.
Several studies have been reported related to the interactions of 2,5-dihydroxybenoic acid with other metal ions in addition to iron [146,147]. However, there are not as many reports on the iron-binding and metabolic effects of 2,5-dihydroxybenzoic acid in comparison to those of 2,3-DHB and salicylic acid. In any case, the overall production of 2,5-dihydroxybenzoic acid is higher than 2,3-DHB from the metabolism of aspirin, and in addition to the increased rate of synthesis from salicylic acid during oxidative stress in the presence of iron [148].
In a marine biology study involving, among other topics, the estimation of the stability constant of the iron complex of 2,5-dihydroxybenzoic acid at physiological pH in seawater, it was shown to be slightly lower (log K eff = 18) than salicylic acid (log K eff = 19) [149]. In another study, the estimation of the level of 2,5-dihydroxybenoic acid in blood was developed using its ability to react with iron, forming an iron complex, which could be identified in blood from its specific physicochemical characteristics [150].
There are hundreds of in vitro and in vivo reports regarding the antioxidant properties of 2,5-dihydroxybenzoic acid. In general, the antioxidant effects of 2,5-dihydroxybenzoic acid appear to have more similarities to salicylic acid than 2,3-DHB. In many studies, comparisons of antioxidant effects with other plant phenolic compounds have been carried out [151]. Some of the antioxidant health benefits of 2,5-dihydroxybenzoic acid include mitigation of drug and radiopharmaceutical toxicity, inhibition of glucose autoxidation involved in the modification of low-density lipoproteins (LDLs) implicated in atherosclerosis in diabetes, iron and copper oxidant toxicity, etc. [152,153,154,155,156,157,158].
The interest regarding the anticancer effects of 2,5-dihydroxybenzoic acid, both as an aspirin metabolite and as a mammalian and plant product, has grown especially since epidemiological studies suggested that aspirin protects against colorectal and other cancers, as well as metastasis. In particular, many research investigators have suggested that 2,5-dihydroxybenzoic acid and other aspirin metabolites are responsible for the anticancer effects. In this context, many studies using various models have suggested that 2,5-dihydroxybenzoic acid can exert anticancer effects via different mechanisms [159,160,161].

3.4. Salicyluric Acid as the Major Metabolite of Aspirin in Urine and Interactions with Iron

Salicyluric acid (salicylglycine, or 2-hydroxyhippuric acid) is the major metabolite of aspirin in humans, amounting to about 70% of the metabolites excreted in the urine at doses of 75–100 mg/day [81,82,87,88,89,90,91]. The overall iron-binding capacity of the ACM and related iron metabolic effects are mostly dependent on the concentration of salicyluric acid in biological fluids. In this context, there are many variations in the metabolic transformation of aspirin and the production of salicyluric acid, which depend on many factors such as age, gender, diet, underlying diseases, etc. For example, in one study involving 5 rheumatoid arthritis patients treated chronically with 3.9 g aspirin, which was equivalent to 2.9 g salicylic acid, the mean 24 h urine recovery rates for salicylic acid, 2,5-dihydroxybenzoic acid, and salicyluric acid were 2.9–5.5%, 2.3–2.8%, and 48.9–61.9% respectively [162].
Salicyluric acid is also a natural plant product similar to most of the other ACMs. For example, salicyluric acid has been identified in the urine and serum of normal individuals not taking aspirin. In one study involving 10 individuals not taking aspirin, the levels of salicylic acid and salicyluric acid were estimated in serum using high-performance liquid chromatography (HPLC), with a median concentration of salicylic acid of 0.56 μmol/L and of salicyluric acid of 3.20 μmol/L [163]. Similar results were obtained in another clinical study on non-aspirin users, where the concentration of salicyluric acid in urine was found to be about six times higher than that of salicylic acid [164].
In a comparative clinical study estimating the concentrations of salicylic acid and salicyluric acid in the urine of vegetarians and non-vegetarians, and also patients taking 75 or 150 mg aspirin, it was found that fruit and vegetables are important sources of dietary salicylates. Much higher levels of salicyluric acid (median, 11.01 μmol/24 h) were excreted in the urine of vegetarians than non-vegetarians (median, 3.91 μmol/24), and similarly, overall lower levels for salicylic acid (median, 1.19 μmol/24 h) were found in vegetarians than non-vegetarians (median, 0.31 μmol/24). Interestingly the level of salicylic acid excreted in patients taking 75 or 150 mg aspirin was not different from that found in vegetarians. However, the levels of salicyluric acid excreted in patients taking aspirin was much higher for the 75 mg dose (median, 170.69 μmol/24 h) and the 150 mg dose (median, 165.17 μmol/24 h) than in non-aspirin users [101].
Several studies have been carried out to investigate the affinity of salicyluric acid for iron and other metal ions. The potential ligands involved in salicyluric acid for metal binding are the phenol hydroxyl group and the carbonyl group, as shown in Figure 2. A potential ligand for weaker metal binding is also the distant carboxylate group in salicyluric acid, similar to the carboxylate group of salicylic acid and 2,5-dihydroxybenzoic acid. There have been several studies of iron complex formation with salicyluric acid which suggest that it possesses high affinity for iron [165,166,167]. In particular, in one study, the affinity of salicyluric acid, salicylic acid, and 2,5-dihydroxybenzoic acid for different metal ions was investigated using potentiometric titrations. All three aspirin metabolites were shown to have high affinity for iron (III), aluminium (III), and copper (II) ions, but not for cobalt (II), nickel (II), zinc (II), magnesium (II), or calcium (II) ions. Furthermore, it has been suggested that the iron, aluminium, and copper complexes of these three aspirin metabolites are strong and stable at physiological pH [165].
Several other studies of complex formation with transition metal and other metal ions using salicyluric acid have also been reported [168,169,170,171]. Furthermore, salicyluric acid has also been shown to form mixed complexes with the chelator imidazole and metal ions such as copper and cobalt [168,169]. Similarly, it has been reported to form a terbium binary complex with the macrocycle 1,4,7,10-tetraazacyclododecane-1,7-bisacetate (DO2A), which has been used for the estimation of salicyluric acid in the urine [171].
The iron-binding properties of salicyluric acid and its implication as the major metabolite of aspirin and the ACM with the highest overall iron capacity could increase our understanding of the overall effects on aspirin metabolism and on IDA in chronic aspirin users with different diseases. In this context, further studies are needed, including in vitro, in vivo, and clinical iron mobilization effects, associating the contributory role of salicyluric acid with the overall role of all ACM in IDA.

3.5. Interactions of Aspirin (Acetylsalicylic Acid) and Its Glucuronide Metabolites with Iron

In contrast to the ACM, there have not been many reports regarding the interaction of aspirin or, especially, its glucuronide metabolites with iron. In theory, much weaker iron-binding properties are expected from the structural characteristics of aspirin, and more so of its glucuronide metabolites. In this context, and in comparison, for example, with salicylic acid and 2,3-DHB, the phenolic or the carboxylate ligands needed for chelation are blocked by acetate in the case of aspirin, a glucuronide conjugate in the carboxyate ligand in the case of salicylacyl glucuronide, or a phenolic hydroxyl ligand in the case of salicylphenol glucuronide (Figure 1).
However, despite the apparently weak iron-binding properties of aspirin, several investigators have reported the formation and characterization of the aspirin iron complex, other interactions with iron, and also iron metabolism in general. For example, experimental evidence from UV-Vis spectroscopy and HPLC-MS suggested the formation of a coloured aspirin iron complex following interaction with ferrous gluconate [172]. The formation, isolation, and characterization of an aspirin iron complex have also been reported during studies of oxidative damage to the liver mitochondria by aspirin, where the iron complex of aspirin was implicated in the toxicity [173]. In contrast, a mechanism of antioxidant action through the chelation of intracellular low-molecular-weight iron was proposed in cell studies, where aspirin has been shown to protect endothelial cells from oxidative stress damage caused by hydrogen peroxide [174]. Similarly, chelation of iron by aspirin has been suggested to be related to its antioxidant activity in the improvement of cataracts in diabetic models and other models of oxidative damage [175,176].
The interaction of aspirin with many other iron metabolic pathways, including the effects of aspirin on the expression of iron transport and storage proteins and metabolic pathways such as ferroptosis, which is a newly identified programmed cell death involving iron, free radicals and peroxidation of lipids of the cell membrane are attracting the interest of many investigators in almost all diseases [177,178,179,180,181,182,183]. In particular, several studies have suggested that aspirin is involved in the modulation of ferroptosis in many diseases, including chronic kidney disease and cancer [183,184,185,186]. Most importantly, the modulation of ferroptosis by aspirin has been suggested as a mechanism for the treatment of different untreated cancers, anticancer drug resistance, and metastasis [187,188,189,190,191,192,193,194]. Similarly, iron and other transition metal complexes of aspirin and aspirin-based compounds have been suggested as new anticancer agents [195].
The interaction of aspirin with iron and different pathways of iron metabolism, including both iron absorption and iron excretion, needs further investigation. In particular, the interactions of oral aspirin with many other oral iron-containing drug formulations, in addition to ferrous gluconate, may be related to a general mechanism affecting iron absorption, as has previously been shown with other drugs [39,172,196,197,198,199]. Similar effects on iron metabolism may be envisaged from interactions of aspirin with other oral drugs, and also with ascorbic acid, which is used as a supplement or a dietary constituent [200,201,202,203]. Within this context, increased absorption and body intake of copper has been shown in a rabbit animal model using aspirin copper complexes, suggesting that other metal complexes in addition to iron play a role in the absorption of aspirin and the affected metals [39,204]. Similar effects on aspirin absorption, especially on slow-release preparations of aspirin, are also expected from the state of gut microbiota [205]. In contrast to the effects on iron absorption, the iron excretion-related effects of aspirin are very limited considering its rapid metabolism and clearance from the body following oral administration.
No sufficient information is available on the iron-binding properties or related effects of the glucuronide conjugate metabolites of aspirin. In this context, non-significant effects on iron excretion are expected from the glucuronide conjugate metabolites of aspirin, similar to other glucuronide conjugate metabolites of the chelating drugs deferiprone and deferasirox [51].

4. Factors Causing Iron Deficiency in Otherwise Normal Individuals Treated Long-Term with Aspirin

Millions of elderly people worldwide, including, for example, 50% of older individuals in the USA, are taking low-dose aspirin (75–100 mg/day) for prophylaxis against many diseases, including cardiovascular and cerebrovascular disease and cancer [83,84,85,206,207,208,209]. In several randomized clinical trials involving long-term users of daily aspirin, iron deficiency was observed in a large portion of participants, which was characterized by decreases in haemoglobin and serum ferritin and was unrelated to gastric bleeding [83,84,85,206,208,210,211,212]. Iron deficiency was not observed either in non-aspirin users or users of other anti-inflammatory drugs [83,84,85].
A major development in relation to low-dose daily aspirin users, which has been suggested following several clinical investigations, was the identification of a decrease in cancer risk and increased survival in cancer patients, which was, according to some investigators, linked to iron reduction caused by aspirin [207,209]. Based on this and other clinical evidence, the USA health authorities recommended the use of low-dose aspirin for the chemoprevention of colorectal cancer in patients 50–59 years of age [191]. The exact mechanism of the anticancer activity and iron reduction in low-dose daily aspirin users is still unclear, and further investigations of aspirin users and cancer are in progress [208,209,210,211,212,213].
In an attempt to solve the puzzle of the cause of iron deficiency in long-term, low-dose daily aspirin users, a major role in the iron-chelating activity of the ACM has been preliminarily proposed considering different parameters, including drug biotransformation and posology effects on iron absorption and iron excretion and overall iron chelation activity [86]. The observation that only a portion of the low-dose, long-term aspirin users develops iron deficiency is another parameter of the puzzle which needs further investigation. This parameter could be considered from previous findings regarding the effect of variety of factors influencing iron balance, iron absorption, and iron excretion [14].
The maintenance of iron balance under normal physiological conditions depends primarily on the effective regulatory control of iron, and especially the overall levels between iron absorption and iron excretion. This balance can be affected by dietary, genotypic, age, iatrogenic, lifestyle, and many other factors [1,2,3,4,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,26,27,28,29,30]. Within this context, there is wide variability in the modes of action of drugs and dietary molecules in relation to iron absorption and iron excretion. For example, oral deferoxamine prevents iron absorption, and subcutaneous deferoxamine increases iron excretion mostly in the urine, but also in the faeces. On the other hand, oral deferasirox increases iron absorption and iron excretion in the faeces. Similarly, maltol increases iron absorption, but does not significantly affect iron excretion. The mode of action of oral deferiprone is also different, since it does not significantly affect iron absorption, but increases urinary iron excretion [46,47,50,51]. Another important parameter affecting both iron absorption and iron excretion in relation to drugs with chelating potential, including aspirin and metabolites, are the individual rate of ADMET parameters in each patient, as well as other parameters, such as the iron load, availability, and concentration of iron available for chelation, as well as the length of treatment [53,54,55,56,57,58,59,60,214].
The effects of orally administered aspirin in relation to iron absorption and vice versa have not yet been fully investigated. However, considering the pharmacological and metabolic profile of orally administered aspirin and its partial conversion to salicylic acid in the GIT, it is likely that only a small reduction in dietary iron absorption is caused by both aspirin and salicylic acid binding of dietary iron, which is especially feasible if aspirin is taken during meals or pharmaceutical formulations containing iron are consumed [172].
A different mechanism of iron excretion involving aspirin and ACM is anticipated. In this case, most of the orally administered aspirin seems to be absorbed and metabolically converted to salicylic acid and the other metabolites, with, overall, more than 70% of aspirin associated with the biotransformation to ACM capable of mobilizing iron from different iron pools and causing an increase in iron excretion [7,51,86]. In particular, the mode of chelating activity of aspirin and the ACM resembles the widely used iron chelation combination therapy, where the combination of chelating drugs is more effective in increasing iron excretion than any monotherapy [55,56,57,86,108].
There are several other parameters to be considered regarding the concentration and efficacy of the ACMs. In contrast to chelating drugs such as oral deferiprone and deferasirox, which are rapidly transformed to glucuronide metabolites with no chelating properties, the ACMs are excreted in the urine with no further metabolic transformation (Figure 2) [80,81,82,86]. The drug posology administered, as well as the drug and, in this case, ACM concentration in the blood, are important parameters in determining the level of iron excretion during chelation therapy [86]. For example, several metabolic studies involving iron balance and deferiprone have shown that, in the vast majority of cases of iron-loaded patients, deferiprone is mostly metabolised and excreted as its glucuronide conjugate metabolite, with only 3–20% excreted in the urine as deferiprone iron complex [97,98]. In general, the level of iron excreted by deferiprone and other chelating drugs depend on the drug dose and the iron load of the patient [56,57,58,59,215,216]. Much lower doses of deferiprone and other chelating drugs are used in non-heavily iron-loaded thalassaemia patients, as well as different non-iron-loaded categories of patients with neurodegenerative and other diseases [53,54]. In such cases, regular monitoring of iron parameters is essential to prevent iron deficiency and other toxicities [52,56].
Limited information is available on iron balance studies and the general comparison between iron absorption and iron excretion in otherwise normal elderly individuals taking aspirin and non-aspirin users in the absence of major gastric bleeding. However, in several randomised clinical trials, the use of aspirin has been considered as the major cause or one of the causes of iron deficiency [83,84,85,206,207,208]. A general estimation of the level of changes in iron balance caused by ACM could be considered based on available data from dietary iron absorption levels and iron excretion caused during chelation therapy.
In general, the maintenance of the body’s iron balance is accomplished through the daily absorption of about 2 mg of iron from a typical Western diet and the equivalent daily amount of body iron excretion or loss [1,2,3,13,14,15,16,47]. Despite the fact that iron balance studies have not been carried in individuals using aspirin or ACM, some information is available from related studies on the bidentate chelators deferiprone and 2,3-DHB. In this context, low doses of deferiprone at 0.5 g and 1.0 g of about 7.5 mg/kg and 15 mg/kg caused net iron increases of approximately 0.2 mg and 0.7 mg, respectively, in the urine of a myelodysplasia patient with serum ferritin of 920 μg/L [217]. Low increases in iron excretion (2.7–6.9 mg) were also observed in four myelodysplasia patients (40–75 kg, serum ferritin 750–2040 μg/L) who received 2 g of deferiprone (27–50 mg/kg), and in thalassaemia patients (6.5–22.8 mg iron) who received 1–1.5 g of deferiprone (23–33 mg/kg) [218]. Much lower levels of iron excretion have been reported in normal volunteers and patients with normal iron stores when treated with deferiprone [56,57,58,59,97,98]. For example, administration of deferiprone at 35 mg/kg in a normal volunteer (serum ferritin 210 μg/L) caused a net increase of 0.84 mg of iron [97]. Only 154 mg of a 3.0 g dose of deferiprone was associated with the 0.84 mg increase in urinary iron excretion, while the remaining was excreted as a deferiprone glucuronide [97]. As previously mentioned, the increased iron excretion caused in thalassaemia patients using 2,3-DHB at 25 mg/kg/day was, on average, 4.5 mg iron, and this is expected to be much lower in normal individuals [106]. However, in the case of low-dose aspirin, the overall iron chelating capacity of the ACM, as estimated from the urinary levels, could be about 50 mg, which might be sufficient for a smaller increase in urinary iron excretion [97,106]. It is therefore envisaged that, despite the low level of daily net iron excretion increases by 2,3-DHB and of the other ACM in low-dose aspirin users, the overall incremental iron excretion increases induced over months and years by ACM could be sufficient to cause iron deficiency.
The observation that only a minority (e.g., of about 23% [83]) of the low-dose, long-term aspirin users develop IDA is another parameter of the puzzle of the interaction with iron which needs further investigation. However, considering that the same or similar dilemmas appear in relation to the incidence of IDA in only some sections of the vegetarian population, other parameters should be considered and included in the equation of iron imbalance. In particular, it is suggested that the portion of the low-dose, long-term aspirin users developing IDA may be associated with additional causes or activities, such as those who are also vegetarians or who have dietary habits with vegetarian meals low in iron, or those who are involved in blood donation or intensive sport [26,27,28].
Other factors implicated in iron deficiency affecting aspirin users are dietary habits using food containing phosphates, tannic acid, and other components which inhibit iron absorption [34,35,36,37,38]. In particular, food containing salicylates, including the ACM salicylic acid, 2,5-dihydroxybenzoic acid (b), 2,3-DHB, and salicyluric acid, may inhibit iron absorption and increase iron excretion [163,164]. However, the overall effect of dietary salicylates on iron deficiency is much lower than in aspirin users because of the low content in vegetarian meals in comparison to the ACMs formed from the metabolism of low doses of aspirin [101].
It appears that many factors and mechanisms may participate in the development of iron deficiency in the section of individuals using long-term, low-dose daily aspirin. However, it also appears that, in the majority of cases of aspirin users, any iron losses due to increased iron excretion by ACM and other causes are compensated for by dietary iron sources, thus maintaining iron balance in a mechanism similar to the gradual restoration of iron balance in blood donors (Table 1).
It is therefore suggested by direct and indirect evidence that a primary role in the cause of iron deficiency in aspirin users is played by the ACM. In such cases, the chronic administration of daily doses of aspirin is likely to increase the overall iron excretion in small increments each time, which is caused by the combinatory iron mobilization effect of the ACM formed by aspirin. This effect and the consumption of a vegetarian diet low in iron are likely to be the main contributors to iron deficiency (Table 1). Smaller contributions to iron deficiency may also involve the possible inhibition of iron absorption in GIT by aspirin and salicylic acid, as well as dietary salicylates. Similarly, the dietary salicylates salicylic acid, 2,5-dihydroxybenzoic acid, 2,3-DHB, and salicyluric acid may also cause slight increases in iron excretion and contribute to the overall loss of iron in long-term, low-dose aspirin users.

5. Future Studies and Limitations on the Role of Aspirin in Iron Deficiency and Other Clinical Conditions

There are many different limitations to the mode of action and therapeutic activity of aspirin, including the implications of iron chelation and other effects of the ACMs (Table 1). Some of the limitations and effects are related to differences in the doses and formulations of aspirin use, as well as the underlying conditions, dietary habits, ADMET, and other characteristics of aspirin users. For example, the widely used low doses of 75 mg and 100 mg aspirin are arbitrary in most cases, since they are intended for people of 70 kg, and those of higher weights or obese individuals may not achieve the desired therapeutic aspirin threshold [219,220]. Furthermore, in some cases, the therapeutic benefits of low-dose aspirin for cancer and cardiovascular diseases are usually observed after 5 years or more of daily use [221]. Similar effects and implications are expected from the posology and time period of aspirin use on IDA, since the iron-mobilizing and other effects of ACM are dependent on the concentration and rate of mobilization (Table 1) [219].
Further investigations into the role of aspirin and each of the ACMs are needed in relation to iron deficiency and other diseases of iron metabolism, similar to those used for the development of chelators for the treatment of iron overload and chelator iron complexes for the treatment of iron deficiency [14,51,100,108,222]. The chemical interactions with iron, including the further characterization and stability of the iron complexes, as well as in vitro, in vivo, pharmacological, toxicological, and clinical studies, could provide sufficient information on the overall impact of ACM on iron excretion and the cause of iron deficiency in aspirin users [51,100,108,222]. Many such studies, including clinical trials on iron balance related to increases in iron excretion, which have already been carried out with 2,3-DHB, are also needed for the rest of the ACMs [105,106,107].
The study of the effects of aspirin and ACMs on other essential metal ions in addition to iron, including calcium, zinc, and copper, are also important for possible implications on the general mode of action and toxicity of aspirin [218,223,224]. For example, increases in the excretion of these metal ions or redistribution in the body can result in toxic side effects similar to those observed with DTPA and also other chelating drugs [60,225]. Different metabolic and toxic side effects are also expected from the interactions of aspirin and ACM with proteins and metabolic pathways related to these essential metals.
The relationship of aspirin’s anti-inflammatory activity and its antioxidant potential has not yet been clarified, despite some studies which have previously suggested there may be a link between these two processes [173,174,175,176]. Furthermore, the redox effects of all of the ACMs have not been thoroughly investigated in relation to their iron-binding potential and the Fenton reaction, where iron is the catalytic centre of the formation of very reactive and toxic hydroxyl radicals [116,118,119,148,149,152,154,155,156,157]. In addition, previous studies have shown that deferoxamine, deferiprone, and similar chelators inhibit free radical reactions and the activity of the iron-containing proteins cyclo-oxygenase and lipoxygenase, as well as their metabolic by-products [118,226,227,228]. The mode of action of chelation in this case was suggested to be the reduction in available iron for the turnover of cyclo-oxygenase and lipoxygenase activity [226,227,228]. In this context, further studies are needed to investigate the partial inhibition of cyclo-oxygenase and lipoxygenase through iron chelation in relation to the overall anti-inflammatory activity of aspirin and ACM, as well as the implications of this inhibitory activity in other diseases.
Oxidative stress toxicity damage, which is mostly catalysed by iron catalytic centres, has been identified in almost all diseases in the context of free radical pathology, including those involving organ and tissue damage, cancer, neurodegenerative diseases, and many other conditions [43,118,226]. Chelators, which can have access and bind iron at the iron’s catalytic centre, can effectively inhibit this toxicity [43,226]. In this context, the interactions of the ACMs with the iron catalytic centres involved in this form of toxicity could provide further evidence on the therapeutic activity of aspirin and ACM and other health implications in aspirin users.
A major unexplored area of great importance in pharmacology and medicine is the effects of aspirin and ACM, as well as their iron complexes, on ferroptosis, which is a recently identified type of programmed cell death based on iron-catalysed lipid peroxidation damage of cell membranes [179,180,181,182,183,184]. Ferroptosis has been identified in almost all diseases, including cancer and neurodegeneration [179,180,181,182,183,184,185,186,187,188,189,190]. Several preliminary investigations have already implicated aspirin in the process of ferroptosis via different mechanisms [192,193,194]. In this context, the interaction of aspirin, ACMs, and their iron complexes in ferroptotic cell death may have wider implications in the therapeutic outcome of aspirin in cancer and many other related diseases.
The wide posology selection of aspirin for use in different diseases, which can range from about 50 mg to 3.6 g per day, provides much-needed information regarding the pharmacology, toxicity, and iron chelation efficacy of the ACMs [64,65,66,67,68,229,230,231,232,233]. The prospect of clinical testing of aspirin and ACM at the maximum range of posology for increasing iron excretion in thalassaemia and other iron-loaded diseases, similarly to the clinical studies previously carried out with 2,3-DHB, could further increase the range of clinical applications of aspirin and ACM in different diseases. In particular, such information could increase the prospects of clinical application of aspirin and ACM in colorectal and other cancers, and in the detoxification of some other xenobiotic metals, where higher concentrations of chelating drugs such as deferiprone and EDTA compared to those achieved by low-dose aspirin ACM will be required for effective treatments [223,225,226,234].
Overall, further in vitro, in vivo, and clinical studies are required for selecting aspirin and/or one or more of the ACMs for clinical use in different conditions. Similarly, studies on the selection of the appropriate posology of aspirin and ACM will be necessary for achieving maximum efficacy and low toxicity in each of the associated clinical conditions.

6. Conclusions

Several randomized, double-blind clinical trials of long-term, low-dose aspirin users involving otherwise healthy elderly patients, have shown that a large percentage of such individuals develop IDA in the absence of major gastric bleeding. The cause of iron deficiency has not been identified. In this review, a significant amount of evidence is presented suggesting that most of the metabolites of aspirin have iron-chelating properties, and one of them, 2,3-DHB, has been shown to cause increases in iron excretion in iron-loaded patients. In particular, it has been suggested that the ACMs salicyluric acid, salicylic acid, 2,5-dihydroxybenzoic acid and 2,3-DHB can all act jointly as a form of combination iron chelation therapy and enhance the overall iron excretion in small increments over time, which can result progressively, over many months or years, in iron deficiency in chronic aspirin users. Many other factors may also contribute to iron deficiency in this category of long-term, low-dose elderly aspirin users and especially in cases of individuals who are consuming vegetarian diets, who are low in iron, or who are, for example, involved in intense sport activities. In contrast, it is likely that, in the majority of long-term, low-dose aspirin users who are non-vegetarians, sufficient iron could be absorbed from normal non-vegetarian diets to compensate for the increase in iron excretion caused by the ACM.
Further in vitro, in vivo, and clinical investigations are required for the characterization of the iron-binding effects of the ACM, including iron balance studies and interactions with proteins and metabolic pathways involving iron. Similar investigations are also needed in relation to the therapeutic effects of aspirin and ACM observed in different conditions, including cancer, neurodegeneration, and iron overload, as well as in diseases involving ferroptosis and other diseases related to free radical pathology.

Funding

This research received no external funding.

Acknowledgments

The study was supported from internal funds of the Postgraduate Research Institute of Science, Technology, Environment and Medicine, a non-profit, charitable organization. I thank Christina N. Kontoghiorghe for the comments.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

ACMchelating metabolite
ADMETabsorption, distribution, metabolic, elimination, toxicity
Aspirinacetylsalicylic acid
COXcyclo-oxygenase
2,3-DHB2,3-dihydroxybenzoic acid
DO2A1,4,7,10-tetraazacyclododecane-1,7-bisacetate
DTPAdiethylenetriaminepentaacetic acid
EDTAethylenediaminetriacetic acid
GITgastrointestinal tract
HPLChigh-performance liquid chromatography
IDAiron deficiency anaemia

References

  1. Cairo, G.; Bernuzzi, F.; Recalcati, S.A. A precious metal: Iron, an essential nutrient for all cells. Genes Nutr. 2006, 1, 25–39. [Google Scholar] [CrossRef]
  2. Gozzelino, R.; Arosio, P. Iron Homeostasis in Health and Disease. Int. J. Mol. Sci. 2016, 17, E130. [Google Scholar] [CrossRef] [PubMed]
  3. Anderson, G.J.; Frazer, D.M. Current understanding of iron homeostasis. Am. J. Clin. Nutr. 2017, 106 (Suppl. 6), 1559S–1566S. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, A.; Nemeth, E. New insights into iron regulation and erythropoiesis. Curr. Opin. Hematol. 2015, 22, 199–205. [Google Scholar] [CrossRef]
  5. Galaris, D.; Pantopoulos, K. Oxidative stress and iron homeostasis: Mechanistic and health aspects. Crit. Rev. Clin. Lab. Sci. 2008, 45, 1–23. [Google Scholar] [CrossRef]
  6. Nakamura, T.; Naguro, I.; Ichijo, H. Iron homeostasis and iron-regulated ROS in cell death, senescence and human diseases. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 1398–1409. [Google Scholar] [CrossRef] [PubMed]
  7. Kontoghiorghes, G.J.; Kontoghiorghe, C.N. Iron and Chelation in Biochemistry and Medicine: New Approaches to Controlling Iron Metabolism and Treating Related Diseases. Cells 2020, 9, 1456. [Google Scholar] [CrossRef]
  8. Sargent, P.J.; Farnaud, S.; Evans, R.W. Structure/function overview of proteins involved in iron storage and transport. Curr. Med. Chem. 2005, 12, 2683–2693. [Google Scholar] [CrossRef] [PubMed]
  9. Worwood, M. Inborn errors of metabolism: Iron. Br. Med. Bull. 1999, 55, 556–567. [Google Scholar] [CrossRef]
  10. McLean, E.; Cogswell, M.; Egli, I.; Wojdyla, D.; de Benoist, B. Worldwide prevalence of anaemia, WHO Vitamin and Mineral Nutrition Information System 1993–2005. Public Health Nutr. 2009, 12, 444–454. [Google Scholar] [CrossRef]
  11. Pasricha, S.R.; Tye-Din, J.; Muckenthaler, M.U.; Swinkels, D.W. Iron deficiency. Lancet 2021, 397, 233–248. [Google Scholar] [CrossRef] [PubMed]
  12. Community control of hereditary anaemias: Memorandum from a WHO meeting. Bull. World Health Org. 1983, 61, 63–80.
  13. Tardy, A.L.; Pouteau, E.; Marquez, D.; Yilmaz, C.; Scholey, A. Vitamins and Minerals for Energy, Fatigue and Cognition: A Narrative Review of the Biochemical and Clinical Evidence. Nutrients 2020, 12, 228. [Google Scholar] [CrossRef] [PubMed]
  14. Kontoghiorghes, G.J.; Kolnagou, A. Molecular factors and mechanisms affecting iron and other metal excretion or absorption in health and disease. The role of natural and synthetic chelators. Curr. Med. Chem. 2005, 12, 2695–2709. [Google Scholar] [CrossRef] [PubMed]
  15. Moustarah, F.; Daley, S.F. Dietary Iron. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar] [PubMed]
  16. Barton, J.C.; Edwards, C.Q. (Eds.) Hemochromatosis: Genetics, Pathophysiology, Diagnosis and Treatment; Cambridge University Press: Cambridge, UK, 2000. [Google Scholar]
  17. Adams, P.C. Epidemiology and diagnostic testing for hemochromatosis and iron overload. Int. J. Lab. Hematol. 2015, 37, 25–30. [Google Scholar] [CrossRef] [PubMed]
  18. Pietrangelo, A. Hereditary hemochromatosis—A new look at an old disease. N. Engl. J. Med. 2004, 350, 2383–2397. [Google Scholar] [CrossRef] [PubMed]
  19. Feder, J.N.; Gnirke, A.; Thomas, W.; Tsuchihashi, Z.; Ruddy, D.A.; Basava, A.; Dormishian, F.; Domingo, R., Jr.; Ellis, M.C.; Fullan, A.; et al. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat. Genet. 1996, 13, 399–408. [Google Scholar] [CrossRef]
  20. Dubois, S.; Kowdley, K.V. Targeted screening for hereditary haemochromatosis in high-risk groups. Aliment. Pharmacol. Ther. 2004, 20, 1–14. [Google Scholar] [CrossRef] [PubMed]
  21. Kolnagou, A.; Kontoghiorghe, C.N.; Kontoghiorghes, G.J. Transition of Thalassaemia and Friedreich ataxia from fatal to chronic diseases. World J. Methodol. 2014, 4, 197–218. [Google Scholar] [CrossRef]
  22. Modell, B.; Berdoukas, V. The Clinical Approach to Thalassaemia; Grune and Stratton: New York, NY, USA, 1984; pp. 165–169. [Google Scholar]
  23. Weatherall, D.; Clegg, J.B. Inherited haemoglobin disorders: An increasing global health problem. Bull. World Health Organ. 2001, 79, 704–712. [Google Scholar]
  24. Hokland, P.; Daar, S.; Khair, W.; Sheth, S.; Taher, A.T.; Torti, L.; Hantaweepant, C.; Rund, D. Thalassaemia—A global view. Br. J. Haematol. 2023, 201, 199–214. [Google Scholar] [CrossRef]
  25. Zurlo, M.G.; De Stefano, P.; Borgna-Pignatti, C.; Di Palma, A.; Piga, A.; Melevendi, C.; Di Gregorio, F.; Burattini, M.G.; Terzoli, S. Survival and causes of death in thalassaemia major. Lancet 1989, 2, 27–30. [Google Scholar] [CrossRef] [PubMed]
  26. Sim, M.; Garvican-Lewis, L.A.; Cox, G.R.; Govus, A.; McKay, A.K.A.; Stellingwerff, T.; Peeling, P. Iron considerations for the athlete: A narrative review. Eur. J. Appl. Physiol. 2019, 119, 1463–1478. [Google Scholar] [CrossRef] [PubMed]
  27. Salvin, H.E.; Pasricha, S.R.; Marks, D.C.; Speedy, J. Iron deficiency in blood donors: A national cross-sectional study. Transfusion 2014, 54, 2434–2444. [Google Scholar] [CrossRef] [PubMed]
  28. Savva, S.C.; Kafatos, A. Is red meat required for the prevention of iron deficiency among children and adolescents? Curr. Pediatr. Rev. 2014, 10, 177–183. [Google Scholar] [CrossRef]
  29. Mantadakis, E.; Chatzimichael, E.; Zikidou, P. Iron Deficiency Anemia in Children Residing in High and Low-Income Countries: Risk Factors, Prevention, Diagnosis and Therapy. Mediterr. J. Hematol. Infect. Dis. 2020, 12, e2020041. [Google Scholar] [CrossRef] [PubMed Central]
  30. Wessells, K.R.; Young, R.R.; Ferguson, E.L.; Ouédraogo, C.T.; Faye, M.T.; Hess, S.Y. Assessment of Dietary Intake and Nutrient Gaps, and Development of Food-Based Recommendations, Among Pregnant and Lactating Women in Zinder, Niger: An Optifood Linear Programming Analysis. Nutrients 2019, 11, 72. [Google Scholar] [CrossRef] [PubMed]
  31. Grover, K.; Kumar, T.; Doda, A.; Bhutani, R.; Yadav, S.; Kaushal, P.; Kapoor, R.; Sharma, S. Prevalence of anaemia and its association with dietary habits among pregnant women in the urban area of Haryana. J. Fam. Med. Prim. Care 2020, 9, 783–787. [Google Scholar] [PubMed Central]
  32. Kobayashi, M.; Suhara, T.; Baba, Y.; Kawasaki, N.K.; Higa, J.K.; Matsui, T. Pathological Roles of Iron in Cardiovascular Disease. Curr. Drug Targets 2018, 19, 1068–1076. [Google Scholar] [CrossRef]
  33. Man, C.D.; Maideen, S.F.K.; Rashid, A. Knowledge, Attitude and Practice towards Dietary Iron among Patients with Thalassemia and Their Caregivers in Peninsular Malaysia. Med. J. Malays 2019, 74, 365–371. [Google Scholar]
  34. Zhang, C.; Rawal, S. Dietary Iron Intake, Iron Status, and Gestational Diabetes. Am. J. Clin. Nutr. 2017, 106, 1672–1680. [Google Scholar] [CrossRef] [PubMed]
  35. Milman, N.T. A Review of Nutrients and Compounds, Which Promote or Inhibit Intestinal Iron Absorption: Making a Platform for Dietary Measures That Can Reduce Iron Uptake in Patients with Genetic Haemochromatosis. J. Nutr. Metab. 2020, 2020, 7373498. [Google Scholar] [CrossRef] [PubMed]
  36. Fiesel, A.; Ehrmann, M.; Geßner, D.K.; Most, E.; Eder, K. Effects of polyphenol-rich plant products from grape or hop as feed supplements on iron, zinc and copper status in piglets. Arch. Anim. Nutr. 2015, 69, 276–284. [Google Scholar] [CrossRef] [PubMed]
  37. Jaramillo, Á.; Briones, L.; Andrews, M.; Arredondo, M.; Olivares, M.; Brito, A.; Pizarro, F. Effect of phytic acid, tannic acid and pectin on fasting iron bioavailability both in the presence and absence of calcium. J. Trace Elem. Med. Biol. 2015, 30, 112–117. [Google Scholar] [CrossRef]
  38. Sotelo, A.; González-Osnaya, L.; Sánchez-Chinchillas, A.; Trejo, A. Role of oxate, phytate, tannins and cooking on iron bioavailability from foods commonly consumed in Mexico. Int. J. Food Sci. Nutr. 2010, 61, 29–39. [Google Scholar] [CrossRef] [PubMed]
  39. Djaldetti, M.; Fishman, P.; Notti, I.; Bessler, H. The effect of tetracycline administration on iron absorption in mice. Biomedicine 1981, 35, 150–152. [Google Scholar]
  40. Lu, P.; Sui, M.; Zhang, M.; Wang, M.; Kamiya, T.; Okamoto, K.; Itoh, H.; Okuda, S.; Suzuki, M.; Asakura, T.; et al. Rosmarinic Acid and Sodium Citrate Have a Synergistic Bacteriostatic Effect against Vibrio Species by Inhibiting Iron Uptake. Int. J. Mol. Sci. 2021, 22, 13010. [Google Scholar] [CrossRef]
  41. Duda-Chodak, A.; Tarko, T. Possible Side Effects of Polyphenols and Their Interactions with Medicines. Molecules 2023, 28, 2536. [Google Scholar] [CrossRef]
  42. Paulson, S.K.; Martinez, J.; Sawant, R.; Burke, S.K.; Chavan, A. Effect of Phosphate Binders and a Dietary Iron Supplement on the Pharmacokinetics of a Single Dose of Vadadustat in Healthy Adults. Clin. Pharmacol. Drug Dev. 2022, 11, 475–485. [Google Scholar] [CrossRef]
  43. Kontoghiorghe, C.N.; Kolnagou, A.; Kontoghiorghes, G.J. Phytochelators Intended for Clinical Use in Iron Overload, Other Diseases of Iron Imbalance and Free Radical Pathology. Molecules 2015, 20, 20841–20872. [Google Scholar] [CrossRef]
  44. Cui, S.; Liu, Z.; Nagy, T.; Agboluaje, E.O.; Xiong, M.P. Oral Non-absorbable Polymer-Deferoxamine Conjugates for Reducing Dietary Iron Absorption. Mol. Pharm. 2023, 20, 1285–1295. [Google Scholar] [CrossRef]
  45. Gomez, H.F.; McClafferty, H.H.; Flory, D.; Brent, J.; Dart, R.C. Prevention of gastrointestinal iron absorption by chelation from an orally administered premixed deferoxamine/charcoal slurry. Ann. Emerg. Med. 1997, 30, 587–592. [Google Scholar] [CrossRef]
  46. Kontoghiorghes, G.J. Chelators affecting iron absorption in mice. Arzneimittelforschung 1990, 40, 1332–1335. [Google Scholar] [PubMed]
  47. Kontoghiorghes, G.J.; Kolnagou, A.; Demetriou, T.; Neocleous, M.; Kontoghiorghe, C.N. New Era in the Treatment of Iron Deficiency Anaemia Using Trimaltol Iron and Other Lipophilic Iron Chelator Complexes: Historical Perspectives of Discovery and Future Applications. Int. J. Mol. Sci. 2021, 22, 5546. [Google Scholar] [CrossRef] [PubMed]
  48. Kontoghiorghes, G.J. Advances on Chelation and Chelator Metal Complexes in Medicine. Int. J. Mol. Sci. 2020, 21, 2499. [Google Scholar] [CrossRef] [PubMed]
  49. Konstantinou, E.; Pashalidis, I.; Kolnagou, A.; Kontoghiorghes, G.J. Interactions of hydroxycarbamide (hydroxyurea) with iron and copper: Implications on toxicity and therapeutic strategies. Hemoglobin 2011, 35, 237–246. [Google Scholar] [CrossRef] [PubMed]
  50. Kontoghiorghes, G.J.; Barr, J.; Nortey, P.; Sheppard, L. Selection of a new generation of orally active alpha-ketohydroxypyridine iron chelators intended for use in the treatment of iron overload. Am. J. Hematol. 1993, 42, 340–349. [Google Scholar] [CrossRef] [PubMed]
  51. Kontoghiorghe, C.N.; Kontoghiorghes, G.J. Efficacy and safety of iron-chelation therapy with deferoxamine, deferiprone, and deferasirox for the treatment of iron-loaded patients with non-transfusion-dependent thalassemia syndromes. Drug Des. Devel Ther. 2016, 10, 465–481. [Google Scholar] [CrossRef]
  52. Kontoghiorghes, G.J.; Kolnagou, A.; Peng, C.T.; Shah, S.V.; Aessopos, A. Safety issues of iron chelation therapy in patients with normal range iron stores including thalassaemia, neurodegenerative, renal and infectious diseases. Expert Opin. Drug Saf. 2010, 9, 201–206. [Google Scholar] [CrossRef]
  53. Pandolfo, M.; Hausmann, L. Deferiprone for the treatment of Friedreich’s ataxia. J. Neurochem. 2013, 126 (Suppl. 1), 142–146. [Google Scholar] [CrossRef] [PubMed]
  54. Elincx-Benizri, S.; Glik, A.; Merkel, D.; Arad, M.; Freimark, D.; Kozlova, E.; Cabantchik, I.; Hassin-Baer, S. Clinical Experience With Deferiprone Treatment for Friedreich Ataxia. J. Child Neurol. 2016, 31, 1036–1040. [Google Scholar] [CrossRef]
  55. Kolnagou, A.; Kleanthous, M.; Kontoghiorghes, G.J. Reduction of body iron stores to normal range levels in thalassaemia by using a deferiprone/deferoxamine combination and their maintenance thereafter by deferiprone monotherapy. Eur. J. Haematol. 2010, 85, 430–438. [Google Scholar] [CrossRef] [PubMed]
  56. Aessopos, A.; Kati, M.; Farmakis, D.; Polonifi, E.; Deftereos, S.; Tsironi, M. Intensive chelation therapy in beta-thalassemia and possible adverse cardiac effects of desferrioxamine. Int. J. Hematol. 2007, 86, 212–215. [Google Scholar] [CrossRef] [PubMed]
  57. Kolnagou, A.; Kontoghiorghe, C.N.; Kontoghiorghes, G.J. Prevention of Iron Overload and Long Term Maintenance of Normal Iron Stores in Thalassaemia Major Patients using Deferiprone or Deferiprone Deferoxamine Combination. Drug Res. 2017, 67, 404–411. [Google Scholar] [CrossRef] [PubMed]
  58. Kolnagou, A.; Kontoghiorghes, G.J. Maintenance of normal range body iron store levels for up to 4.5 years in thalassemia major patients using deferiprone monotherapy. Hemoglobin 2010, 34, 204–209. [Google Scholar] [CrossRef] [PubMed]
  59. Kolnagou, A.; Kontoghiorghes, G.J. New golden era of chelation therapy in thalassaemia: The achievement and maintenance of normal range body iron stores. Br. J. Haematol. 2010, 150, 489–490. [Google Scholar] [CrossRef] [PubMed]
  60. Kolnagou, A.; Kleanthous, M.; Kontoghiorghes, G.J. Benefits and Risks in Polypathology and Polypharmacotherapy Challenges in the Era of the Transition of Thalassaemia from a Fatal to a Chronic or Curable Disease. Front. Biosci. (Elite Ed.) 2022, 14, 18. [Google Scholar] [CrossRef] [PubMed]
  61. Montinari, M.R.; Minelli, S.; De Caterina, R. The first 3500 years of aspirin history from its roots—A concise summary. Vascul. Pharmacol. 2019, 113, 1–8. [Google Scholar] [CrossRef] [PubMed]
  62. Desborough, M.J.R.; Keeling, D.M. The aspirin story—From willow to wonder drug. Br. J. Haematol. 2017, 177, 674–683. [Google Scholar] [CrossRef]
  63. Vane, J.R.; Botting, R.M. The mechanism of action of aspirin. Thromb. Res. 2003, 110, 255–258. [Google Scholar] [CrossRef]
  64. Uvodich, M.E.; Siljander, M.P.; Taunton, M.J.; Mabry, T.M.; Perry, K.I.; Abdel, M.P. Low-Dose vs. Regular-Dose Aspirin for Venous Thromboembolism Prophylaxis in Primary Total Joint Arthroplasty. J. Arthroplast. 2021, 36, 2359–2363. [Google Scholar] [CrossRef] [PubMed]
  65. Merkow, D.B.; Tang, A.; Iorio, R.; Slover, J.D.; Bosco, J.A.; Schwarzkopf, R. Low dose aspirin is effective in preventing venous thromboembolism in patients undergoing primary total knee arthroplasty. J. Orthop. 2021, 24, 26–28. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, Y.; Chen, F.; Liao, J.; Han, H.; Li, G.; Zhou, L. Low- or high-dose preventive aspirin use and risk of death from all-cause, cardiovascular disease, and cancer: A nationally representative cohort study. Front. Pharmacol. 2023, 14, 1099810. [Google Scholar] [CrossRef] [PubMed]
  67. Voelker, M.; Schachtel, B.P.; Cooper, S.A.; Gatoulis, S.C. Efficacy of disintegrating aspirin in two different models for acute mild-to-moderate pain: Sore throat pain and dental pain. Inflammopharmacology 2016, 24, 43–51. [Google Scholar] [CrossRef] [PubMed]
  68. Jones, W.S.; Mulder, H.; Wruck, L.M.; Pencina, M.J.; Kripalani, S.; Muñoz, D.; Crenshaw, D.L.; Effron, M.B.; Re, R.N.; Gupta, K.; et al. ADAPTABLE Team. Comparative Effectiveness of Aspirin Dosing in Cardiovascular Disease. N. Engl. J. Med. 2021, 384, 1981–1990. [Google Scholar] [CrossRef] [PubMed]
  69. Dovizio, M.; Bruno, A.; Tacconelli, S.; Patrignani, P. Mode of action of aspirin as a chemopreventive agent. Recent Results Cancer Res. 2013, 191, 39–65. [Google Scholar] [CrossRef]
  70. Patrono, C. Fifty years with aspirin and platelets. Br. J. Pharmacol. 2023, 180, 25–43. [Google Scholar] [CrossRef] [PubMed]
  71. Mekaj, Y.H.; Daci, F.T.; Mekaj, A.Y. New insights into the mechanisms of action of aspirin and its use in the prevention and treatment of arterial and venous thromboembolism. Ther. Clin. Risk Manag. 2015, 11, 1449–1456. [Google Scholar] [CrossRef]
  72. Boelig, R.C.; Kaushal, G.; Rochani, A.; McKenzie, S.E.; Kraft, W.K. Aspirin pharmacokinetics and pharmacodynamics through gestation. Am. J. Obstet. Gynecol. 2023. [Google Scholar] [CrossRef]
  73. Keche, Y.; Gaikwad, N.R.; Wasnik, P.N.; Nagpure, K.; Siddiqui, M.S.; Joshi, A.; Dhaneria, S.; Dewangan, G.; Meher, J.; Das, P. Analysis of Drugs Prescribed to Elderly Patients in a Tertiary Health Care Center in Raipur, Central India: An Observational Study. Cureus 2024, 16, e52770. [Google Scholar] [CrossRef]
  74. Cong, D.; Qi, W.; Liu, X.; Xu, X.; Dong, L.; Xue, W.; Li, K. Pharmacokinetic Study of Enteric-Coated Sustained-Release Aspirin Tablets in Healthy Chinese Participants. Drug Des. Devel Ther. 2023, 17, 2421–2429. [Google Scholar] [CrossRef]
  75. Hofling, U.; Tacconelli, S.; Contursi, A.; Bruno, A.; Mucci, M.; Ballerini, P.; Cohen, S.; Patrignani, P. Characterization of the acetylation of cyclooxygenase-isozymes and targeted lipidomics of eicosanoids in serum and colon cancer cells by the new aspirin formulation IP1867B versus aspirin in vitro. Front. Pharmacol. 2022, 13, 1070277. [Google Scholar] [CrossRef]
  76. Pacławski, A.; Politis, S.; Balafas, E.; Mina, E.; Papakyriakopoulou, P.; Christodoulou, E.; Kostomitsopoulos, N.; Rekkas, D.M.; Valsami, G.; Giovagnoli, S. Development and Pharmacokinetics of a Novel Acetylsalicylic Acid Dry Powder for Pulmonary Administration. Pharmaceutics 2022, 14, 2819. [Google Scholar] [CrossRef]
  77. Atar, D.; Sarkar, S.; Kolev, E.; Mura, C.; Brosstad, F.; Theodorsen, L.; Westen, G.I.; Stribolt-Halvorsen, P.E. Randomized, open-label, crossover trial comparing the pharmacokinetic profile of a novel oral aspirin solution and a chewed aspirin tablet. Int. J. Clin. Pharmacol. Ther. 2022, 60, 430–438. [Google Scholar] [CrossRef] [PubMed]
  78. Del Bianco-Rondeau, M.; Robert-Halabi, M.; Bloom, S.; Rabasa-Lhoret, R.; Tardif, J.C.; Lordkipanidzé, M.; Marquis-Gravel, G. Aspirin for Primary Cardiovascular Prevention in Patients with Diabetes: Uncertainties and Opportunities. Thromb. Haemost. 2022, 122, 1443–1453. [Google Scholar] [CrossRef]
  79. Ray, K. Microbiota modulates aspirin chemoprevention in CRC. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 383. [Google Scholar] [CrossRef]
  80. Rocca, B.; Tosetto, A.; Betti, S.; Soldati, D.; Petrucci, G.; Rossi, E.; Timillero, A.; Cavalca, V.; Porro, B.; Iurlo, A.; et al. A randomized double-blind trial of 3 aspirin regimens to optimize antiplatelet therapy in essential thrombocythemia. Blood 2020, 136, 171–182. [Google Scholar] [CrossRef]
  81. Hutt, A.J.; Caldwell, J.; Smith, R.L. The metabolism of aspirin in man: A population study. Xenobiotica 1986, 16, 239–249. [Google Scholar] [CrossRef]
  82. Patel, D.K.; Notarianni, L.J.; Bennett, P.N. Comparative metabolism of high doses of aspirin in man and rat. Xenobiotica 1990, 20, 847–854. [Google Scholar] [CrossRef]
  83. McQuilten, Z.K.; Thao, L.T.P.; Pasricha, S.R.; Artz, A.S.; Bailey, M.; Chan, A.T.; Cohen, H.J.; Lockery, J.E.; Murray, A.M.; Nelson, M.R.; et al. Effect of Low-Dose Aspirin Versus Placebo on Incidence of Anemia in the Elderly: A Secondary Analysis of the Aspirin in Reducing Events in the Elderly Trial. Ann. Intern. Med. 2023, 176, 913–921. [Google Scholar] [CrossRef]
  84. Vetrano, D.L.; Zucchelli, A.; Marconi, E.; Levi, M.; Pegoraro, V.; Cataldo, N.; Heiman, F.; Cricelli, C.; Lapi, F. Predictors of iron-deficiency anemia in primary care older adults: A real-world European multi-country longitudinal study. Aging Clin. Exp. Res. 2020, 32, 2211–2216. [Google Scholar] [CrossRef]
  85. Milman, N.; Ovesen, L.; Byg, K.; Graudal, N. Iron status in Danes updated 1994. I: Prevalence of iron deficiency and iron overload in 1332 men aged 40-70 years. Influence Of blood donation, alcohol intake, and iron supplementation. Ann. Hematol. 1999, 78, 393–400. [Google Scholar] [CrossRef]
  86. Kontoghiorghes, G.J. Drug Selection and Posology, Optimal Therapies and Risk/Benefit Assessment in Medicine: The Paradigm of Iron-Chelating Drugs. Int. J. Mol. Sci. 2023, 24, 16749. [Google Scholar] [CrossRef]
  87. Levy, G. Clinical pharmacokinetics of salicylates: A re-assessment. Br. J. Clin. Pharmacol. 1980, 10 (Suppl. 2), 285S–290S. [Google Scholar] [CrossRef]
  88. Navarro, S.L.; Saracino, M.R.; Makar, K.W.; Thomas, S.S.; Li, L.; Zheng, Y.; Levy, L.; Schwarz, Y.; Bigler, J.; Potter, J.D.; et al. Determinants of aspirin metabolism in healthy men and women: Effects of dietary inducers of UDP-glucuronosyltransferases. Lifestyle Genom. 2011, 4, 110–118. [Google Scholar] [CrossRef]
  89. Hansen, S.H.; Jensen, M.E.; Bjørnsdottir, I. Assay of acetylsalicylic acid and three of its metabolites in human plasma and urine using non-aqueous capillary electrophoresis with reversed electroosmotic flow. J. Pharm. Biomed. Anal. 1998, 17, 1155–1160. [Google Scholar] [CrossRef]
  90. Lares-Asseff, I.; Flores-Pérez, J.; Juárez-Olguín, H.; Ramírez-Lacayo, M.; Loredo-Abdalá, A.; Carbajal-Rodríguez, L. Influence of nutritional status on the pharmacokinetics of acetylsalicylic acid and its metabolites in children with autoimmune disease. Am. J. Clin. Nutr. 1999, 69, 318–324. [Google Scholar] [CrossRef]
  91. Krivosíková, Z.; Spustová, V.; Dzúrik, R. A highly sensitive HPLC method for the simultaneous determination of acetylsalicylic, salicylic and salicyluric acids in biologic fluids: Pharmacokinetic, metabolic and monitoring implications. Methods Find. Exp. Clin. Pharmacol. 1996, 18, 527–532. [Google Scholar]
  92. Li, J.P.; Guo, J.M.; Shang, E.X.; Zhu, Z.H.; Liu, Y.; Zhao, B.C.; Zhao, J.; Tang, Z.S.; Duan, J.A. Quantitative determination of five metabolites of aspirin by UHPLC-MS/MS coupled with enzymatic reaction and its application to evaluate the effects of aspirin dosage on the metabolic profile. J. Pharm. Biomed. Anal. 2017, 138, 109–117. [Google Scholar] [CrossRef]
  93. Bojić, M.; Sedgeman, C.A.; Nagy, L.D.; Guengerich, F.P. Aromatic hydroxylation of salicylic acid and aspirin by human cytochromes P450. Eur. J. Pharm. Sci. 2015, 73, 49–56. [Google Scholar] [CrossRef]
  94. Nagelschmitz, J.; Blunck, M.; Kraetzschmar, J.; Ludwig, M.; Wensing, G.; Hohlfeld, T. Pharmacokinetics and pharmacodynamics of acetylsalicylic acid after intravenous and oral administration to healthy volunteers. Clin. Pharmacol. 2014, 6, 51–59. [Google Scholar] [CrossRef]
  95. Sankaranarayanan, R.; Kumar, D.R.; Altinoz, M.A.; Bhat, G.J. Mechanisms of Colorectal Cancer Prevention by Aspirin—A Literature Review and Perspective on the Role of COX-Dependent and -Independent Pathways. Int. J. Mol. Sci. 2020, 21, 9018. [Google Scholar] [CrossRef]
  96. Pan, P.; Huang, Y.W.; Oshima, K.; Yearsley, M.; Zhang, J.; Yu, J.; Arnold, M.; Wang, L.S. Could Aspirin and Diets High in Fiber Act Synergistically to Reduce the Risk of Colon Cancer in Humans? Int. J. Mol. Sci. 2018, 19, 166. [Google Scholar] [CrossRef]
  97. Kontoghiorghes, G.J.; Goddard, J.G.; Bartlett, A.N.; Sheppard, L. Pharmacokinetic studies in humans with the oral iron chelator 1,2-dimethyl-3-hydroxypyrid-4-one. Clin. Pharmacol. Ther. 1990, 48, 255–261. [Google Scholar] [CrossRef]
  98. Kontoghiorghes, G.J.; Bartlett, A.N.; Sheppard, L.; Barr, J.; Nortey, P. Oral iron chelation therapy with deferiprone. Monitoring of biochemical, drug and iron excretion changes. Arzneimittelforschung 1995, 45, 65–69. [Google Scholar] [PubMed]
  99. Abergel, R.J.; Warner, J.A.; Shuh, D.K.; Raymond, K.N. Enterobactin protonation and iron release: Structural characterization of the salicylate coordination shift in ferric enterobactin. J. Am. Chem. Soc. 2006, 128, 8920–8931. [Google Scholar] [CrossRef]
  100. Kontoghiorghes, G.J. The Design of Orally Active Iron Chelators for the Treatment of Thalassaemia. Ph.D. Thesis, University of Essex, Colchester, UK, 1982. British Library Microfilm No D66194/86. pp. 1–243. Available online: https://www.pri.ac.cy/files/KGJ_thesis_1982.pdf (accessed on 8 May 2024).
  101. Lawrence, J.R.; Peter, R.; Baxter, G.J.; Robson, J.; Graham, A.B.; Paterson, J.R. Urinary excretion of salicyluric and salicylic acids by non-vegetarians, vegetarians, and patients taking low dose aspirin. J. Clin. Pathol. 2003, 56, 651–653. [Google Scholar] [CrossRef]
  102. Fernández, I.S.; Cuevas, P.; Angulo, J.; López-Navajas, P.; Canales-Mayordomo, A.; González-Corrochano, R.; Lozano, R.M.; Valverde, S.; Jiménez-Barbero, J.; Romero, A.; et al. Gentisic acid, a compound associated with plant defense and a metabolite of aspirin, heads a new class of in vivo fibroblast growth factor inhibitors. J. Biol. Chem. 2010, 285, 11714–11729. [Google Scholar] [CrossRef]
  103. Muljono, R.A.; Darsono, F.L.; Scheffer, J.J.; Verpoorte, R. Assay of 2,3-dihydroxybenzoic acid and related compounds in plant materials by high-performance liquid chromatography. J. Chromatogr. A 2001, 927, 39–45. [Google Scholar] [CrossRef]
  104. Grady, R.W.; Graziano, J.H.; Akers, H.A.; Cerami, A. The development of new iron-chelating drugs. J. Pharmacol. Exp. Ther. 1976, 196, 478–485. [Google Scholar]
  105. Peterson, C.M.; Graziano, J.H.; Grady, R.W.; Jones, R.L.; Vlassara, H.V.; Canale, V.C.; Miller, D.R.; Cerami, A. Chelation studies with 2,3-dihydroxybenzoic acid in patients with beta-thalassaemia major. Br. J. Haematol. 1976, 33, 477–485. [Google Scholar] [CrossRef]
  106. Peterson, C.M.; Graziano, J.H.; Grady, R.W.; Jones, R.L.; Markenson, A.; Lavi, U.; Canale, V.; Gray, G.F.; Cerami, A.; Miller, D.R. Chelation therapy in beta-thalassemia major: A one-year double blind study of 2,3-dihydroxybenzoic acid. Exp. Hematol. 1979, 7, 74–80. [Google Scholar] [PubMed]
  107. Cerami, A.; Grady, R.W.; Peterson, C.M.; Bhargava, K.K. Clinical management of thalassemia. The status of new iron chelators. Ann. N. Y. Acad. Sci. 1980, 344, 425–435. [Google Scholar] [CrossRef] [PubMed]
  108. Kontoghiorghes, G.J. Advances in oral iron chelation in man. Int. J. Hematol. 1992, 55, 27–38. [Google Scholar] [PubMed]
  109. Graziano, J.H.; Grady, R.W.; Cerami, A. The identification of 2, 3-dihydroxybenzoic acid as a potentially useful iron-chelating drug. J. Pharmacol. Exp. Ther. 1974, 190, 570–575. [Google Scholar] [PubMed]
  110. De, M.; Basu, M.; Chakrabartty, P.K. Increased synthesis of dihydroxybenzoic acid in the presence of aluminum by Rhizobium MO1. Acta Microbiol. Pol. 2003, 52, 195–199. [Google Scholar] [PubMed]
  111. Bellaire, B.H.; Elzer, P.H.; Baldwin, C.L. Roop RM 2nd. Production of the siderophore 2,3-dihydroxybenzoic acid is required for wild-type growth of Brucella abortus in the presence of erythritol under low-iron conditions in vitro. Infect. Immun. 2003, 71, 2927–2932. [Google Scholar] [CrossRef] [PubMed]
  112. Crichton, R.R.; Roman, F.; Roland, F. Iron mobilization from ferritin by chelating agents. J. Inorg. Biochem. 1980, 13, 305–316. [Google Scholar] [CrossRef]
  113. Schneider, D.J.; Roe, A.L.; Mayer, R.J.; Que, L., Jr. Evidence for synergistic anion binding to iron in ovotransferrin complexes from resonance Raman and extended X-ray absorption fine structure analysis. J. Biol. Chem. 1984, 259, 9699–9703. [Google Scholar] [CrossRef] [PubMed]
  114. Blatt, J.; Taylor, S.R.; Kontoghiorghes, G.J. Comparison of activity of deferoxamine with that of oral iron chelators against human neuroblastoma cell lines. Cancer Res. 1989, 49, 2925–2927. [Google Scholar] [PubMed]
  115. Kontoghiorghes, G.J. Structure/red blood cell permeability. Activity of iron(III) chelator complexes. Inorg. Chim. Acta 1988, 151, 101–106. [Google Scholar] [CrossRef]
  116. Graziano, J.H.; Miller, D.R.; Grady, R.W.; Cerami, A. Inhibition of membrane peroxidation in thalassaemic erythrocytes by 2,3-dihydroxybenzoic acid. Br. J. Haematol. 1976, 32, 351–356. [Google Scholar] [CrossRef] [PubMed]
  117. Sinswat, P.; Wu, W.J.; Sha, S.H.; Schacht, J. Protection from ototoxicity of intraperitoneal gentamicin in guinea pig. Kidney Int. 2000, 58, 2525–2532. [Google Scholar] [CrossRef]
  118. Kontoghiorghes, G.J.; Jackson, M.J.; Lunec, J. In vitro screening of iron chelators using models of free radical damage. Free Radic. Res. Commun. 1986, 2, 115–124. [Google Scholar] [CrossRef] [PubMed]
  119. Grootveld, M.; Halliwell, B. Aromatic hydroxylation as a potential measure of hydroxyl-radical formation in vivo. Identification of hydroxylated derivatives of salicylate in human body fluids. Biochem. J. 1986, 237, 499–504. [Google Scholar] [CrossRef]
  120. Grinberg, L.N.; Rachmilewitz, E.A.; Kitrossky, N.; Chevion, M. Hydroxyl radical generation in beta-thalassemic red blood cells. Free. Radic. Biol. Med. 1995, 18, 611–615. [Google Scholar] [CrossRef] [PubMed]
  121. Aghabeigi, B.; Haque, M.; Wasil, M.; Hodges, S.J.; Henderson, B.; Harris, M. The role of oxygen free radicals in idiopathic facial pain. Br. J. Oral. Maxillofac. Surg. 1997, 35, 161–165. [Google Scholar] [CrossRef] [PubMed]
  122. Paterson, J.; Baxter, G.; Lawrence, J.; Duthie, G. Is there a role for dietary salicylates in health? Proc. Nutr. Soc. 2006, 65, 93–96. [Google Scholar] [CrossRef]
  123. Blacklock, C.J.; Lawrence, J.R.; Wiles, D.; Malcolm, E.A.; Gibson, I.H.; Kelly, C.J.; Paterson, J.R. Salicylic acid in the serum of subjects not taking aspirin. Comparison of salicylic acid concentrations in the serum of vegetarians, non-vegetarians, and patients taking low dose aspirin. J. Clin. Pathol. 2001, 54, 553–555. [Google Scholar] [CrossRef]
  124. Dotto, C.; Lombarte Serrat, A.; Cattelan, N.; Barbagelata, M.S.; Yantorno, O.M.; Sordelli, D.O.; Ehling-Schulz, M.; Grunert, T.; Buzzola, F.R. The Active Component of Aspirin, Salicylic Acid, Promotes Staphylococcus aureus Biofilm Formation in a PIA-dependent Manner. Front. Microbiol. 2017, 8, 4. [Google Scholar] [CrossRef]
  125. Herrmann, M. Salicylic acid: An old dog, new tricks, and staphylococcal disease. J. Clin. Investig. 2003, 112, 149–151. [Google Scholar] [CrossRef]
  126. Dal Belo, S.E.; Kanoun-Copy, L.; Lambert, C.; Cornillon, C.; Muller, B.; Jouni, H.; Moreau, M.; Palamarchuk, I.; Kerob, D.; Aguilar, L. Efficacy of a multitargeted, salicylic acid-based dermocosmetic cream compared to benzoyl peroxide 5% in Acne vulgaris: Results from a randomized study. J. Cosmet. Dermatol. 2024, 23, 891–897. [Google Scholar] [CrossRef]
  127. Swain, A.R.; Dutton, S.P.; Truswell, A.S. Salicylates in foods. J. Am. Diet Assoc. 1985, 85, 950–960. [Google Scholar] [CrossRef] [PubMed]
  128. Pitt, C.G.; Gupta, G. The design and synthesis of chelating agents for the treatment of iron overload in Cooley’s anemia. In Development of Iron Chelators for Clinical Use; DHEW Publication No. (NIH) 77-994; Anderson, F.W., Hiller, M.C., Eds.; DHEW: Bethesda, MD, USA, 1975; pp. 137–168. [Google Scholar]
  129. McBryde, W.A.E.; Rohr, J.A.; Penciner, J.S.; Page, J.A. Stability constants of three iron(II1) salicylates. Can. J. Chem. 1970, 48, 2574–2586. [Google Scholar] [CrossRef]
  130. Oktavia, B.; Lim, L.W.; Takeuchi, T. Simultaneous determination of Fe(III) and Fe(II) ions via complexation with salicylic acid and 1,10-phenanthroline in microcolumn ion chromatography. Anal. Sci. 2008, 24, 1487–1492. [Google Scholar] [CrossRef]
  131. Pozdnyakov, I.P.; Plyusnin, V.F.; Grivin, V.P.; Oliveros, E. Photochemistry of Fe(III) complexes with salicylic acid derivatives in aqueous solutions. J. Photochem. Photob. A Chem. 2015, 307, 9–15. [Google Scholar] [CrossRef]
  132. Chyka, P.A.; Erdman, A.R.; Christianson, G.; Wax, P.M.; Booze, L.L.; Manoguerra, A.S.; Caravati, E.M.; Nelson, L.S.; Olson, K.R.; Cobaugh, D.J.; et al. Americal Association of Poison Control Centers; Healthcare Systems Bureau, Health Resources and Sevices Administration, Department of Health and Human Services. Salicylate poisoning: An evidence-based consensus guideline for out-of-hospital management. Clin. Toxicol. 2007, 45, 95–131. [Google Scholar] [CrossRef]
  133. Prescott, L.F.; Balali-Mood, M.; Critchley, J.A.; Johnstone, A.F.; Proudfoot, A.T. Diuresis or urinary alkalinisation for salicylate poisoning? Br. Med. J. (Clin. Res. Ed.) 1982, 285, 1383–1386. [Google Scholar] [CrossRef]
  134. Cha, K.W.; Park, K.W. Determination of iron(III) with salicylic acid by the fluorescence quenching method. Talanta 1998, 46, 1567–1571. [Google Scholar] [CrossRef]
  135. Sha, S.H.; Schacht, J. Salicylate attenuates gentamicin-induced ototoxicity. Lab Investig. 1999, 79, 807–813. [Google Scholar] [PubMed]
  136. Brunet, S.; Guertin, F.; Thibault, L.; Gavino, V.; Delvin, E.; Levy, E. Iron-salicylate complex induces peroxidation, alters hepatic lipid profile and affects plasma lipoprotein composition. Atherosclerosis 1997, 129, 159–168. [Google Scholar] [CrossRef]
  137. Yin, M.-J.; Yamamoto, Y.; Gaynor, R.B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of IκB kinase-β. Nature 1998, 396, 77. [Google Scholar] [CrossRef]
  138. Shaukat, A.; Grau, M.V.; Church, T.R.; Baxter, G.; Barry, E.L.; Summers, R.; Sandler, R.S.; Baron, J.A. Serum salicylate levels and risk of recurrent colorectal adenomas. Cancer Epidemiol. Biomark. Prev. 2011, 20, 679–682. [Google Scholar] [CrossRef]
  139. Dachineni, R.; Kumar, D.R.; Callegari, E.; Kesharwani, S.S.; Sankaranarayanan, R.; Seefeldt, T.; Tummala, H.; Bhat, G.J. Salicylic acid metabolites and derivatives inhibit CDK activity: Novel insights into aspirin’s chemopreventive effects against colorectal cancer. Int. J. Oncol. 2017, 51, 1661–1673. [Google Scholar] [CrossRef]
  140. Paterson, J.R.; Lawrence, J.R. Salicylic acid: A link between aspirin, diet and the prevention of colorectal cancer. QJM 2001, 94, 445–448. [Google Scholar] [CrossRef]
  141. Abedi, F.; Razavi, B.M.; Hosseinzadeh, H. A review on gentisic acid as a plant derived phenolic acid and metabolite of aspirin: Comprehensive pharmacology, toxicology, and some pharmaceutical aspects. Phytother. Res. 2020, 34, 729–741. [Google Scholar] [CrossRef]
  142. Devireddy, L.R.; Hart, D.O.; Goetz, D.H.; Green, M.R. A mammalian siderophore synthesized by an enzyme with a bacterial homolog involved in enterobactin production. Cell 2010, 141, 1006–1017. [Google Scholar] [CrossRef]
  143. Liu, Z.; Reba, S.; Chen, W.D.; Porwal, S.K.; Boom, W.H.; Petersen, R.B.; Rojas, R.; Viswanathan, R.; Devireddy, L. Regulation of mammalian siderophore 2,5-DHBA in the innate immune response to infection. J. Exp. Med. 2014, 211, 1197–1213. [Google Scholar] [CrossRef]
  144. Weiss, G. Microbial hijacking of mammalian iron shuttling. J. Exp. Med. 2014, 211, 1009. [Google Scholar] [CrossRef]
  145. Porwal, S.K.; Furia, E.; Harris, M.E.; Viswanathan, R.; Devireddy, L. Synthetic, potentiometric and spectroscopic studies of chelation between Fe(III) and 2,5-DHBA supports salicylate-mode of siderophore binding interactions. J. Inorg. Biochem. 2015, 145, 1–10. [Google Scholar] [CrossRef]
  146. Trindade, V.; Balter, H. Oxidant and Antioxidant Effects of Gentisic Acid in a 177Lu-Labelled Methionine-Containing Minigastrin Analogue. Curr. Radiopharm. 2020, 13, 107–119. [Google Scholar] [CrossRef]
  147. Rubira, L.; Donzé, C.; Fouillet, J.; Algudo, B.; Kotzki, P.O.; Deshayes, E.; Fersing, C. [68Ga]Ga-FAPI-46 synthesis on a GAIA® module system: Thorough study of the automated radiolabeling reaction conditions. Appl. Radiat. Isot. 2024, 206, 111211. [Google Scholar] [CrossRef]
  148. Ingelman-Sundberg, M.; Kaur, H.; Terelius, Y.; Persson, J.O.; Halliwell, B. Hydroxylation of salicylate by microsomal fractions and cytochrome P-450. Lack of production of 2,3-dihydroxybenzoate unless hydroxyl radical formation is permitted. Biochem. J. 1991, 276, 753–757. [Google Scholar] [CrossRef]
  149. Arreguin MLGonzález, A.G.; Pérez-Almeida, N.; Arnone, V.; González-Dávila, M.; Santana-Casiano, J.M. The role of gentisic acid on the Fe(III) redox chemistry in marine environments. Mar. Chem. 2021, 234, 104003. [Google Scholar] [CrossRef]
  150. Cass, M.; Wagner, J.G. A comparison of the determination of gentisic acid in blood by means of the ferric ion-gentisate complex with and without the presence of ferrous ions. J. Am. Pharm. Assoc. 1954, 43, 50–52. [Google Scholar] [CrossRef] [PubMed]
  151. Shingai, Y.; Fujimoto, A.; Nakamura, M.; Masuda, T. Structure and function of the oxidation products of polyphenols and identification of potent lipoxygenase inhibitors from Fe-catalyzed oxidation of resveratrol. J. Agric. Food Chem. 2011, 59, 8180–8186. [Google Scholar] [CrossRef]
  152. Exner, M.; Hermann, M.; Hofbauer, R.; Kapiotis, S.; Speiser, W.; Held, I.; Seelos, C.; Gmeiner, B.M. The salicylate metabolite gentisic acid, but not the parent drug, inhibits glucose autoxidation-mediated atherogenic modification of low density lipoprotein. FEBS Lett. 2000, 470, 47–50. [Google Scholar] [CrossRef]
  153. López, A.; Rico, M.; Santana-Casiano, J.M.; González, A.G.; González-Dávila, M. Phenolic profile of Dunaliella tertiolecta growing under high levels of copper and iron. Environ. Sci. Pollut. Res. Int. 2015, 22, 14820–14828. [Google Scholar] [CrossRef]
  154. Saeedavi, M.; Goudarzi, M.; Fatemi, I.; Basir, Z.; Noori, S.M.A.; Mehrzadi, S. Gentisic acid mitigates gentamicin-induced nephrotoxicity in rats. Tissue Cell 2023, 84, 102191. [Google Scholar] [CrossRef]
  155. Noei Razliqi, R.; Harooni, E.; Neisi, N.; Jafar Sameri, M.; Ahangarpour, A. Hepatoprotective effects of gentisic acid through its anti-oxidant properties in nicotinamide-streptozotocin induced diabetic mice. Iran. J. Basic Med. Sci. 2023, 26, 1409–1415. [Google Scholar] [CrossRef]
  156. Joshi, R.; Gangabhagirathi, R.; Venu, S.; Adhikari, S.; Mukherjee, T. Antioxidant activity and free radical scavenging reactions of gentisic acid: In-vitro and pulse radiolysis studies. Free Radic. Res. 2012, 46, 11–20. [Google Scholar] [CrossRef] [PubMed]
  157. Hang, D.T.N.; Hoa, N.T.; Bich, H.N.; Mechler, A.; Vo, Q.V. The hydroperoxyl radical scavenging activity of natural hydroxybenzoic acids in oil and aqueous environments: Insights into the mechanism and kinetics. Phytochemistry 2022, 201, 113281. [Google Scholar] [CrossRef] [PubMed]
  158. Liu, G.; Zhu, W.; Li, S.; Zhou, W.; Zhang, H.; Wang, J.; Liu, X.; Zhang, J.; Liang, L.; Xu, X. Antioxidant capacity and interaction of endogenous phenolic compounds from tea seed oil. Food Chem. 2021, 376, 131940. [Google Scholar] [CrossRef] [PubMed]
  159. Altinoz, M.A.; Elmaci, I.; Cengiz, S.; Emekli-Alturfan, E.; Ozpinar, A. From epidemiology to treatment: Aspirin’s prevention of brain and breast-cancer and cardioprotection may associate with its metabolite gentisic acid. Chem. Biol. Interact. 2018, 291, 29–39. [Google Scholar] [CrossRef]
  160. Sharma, S.; Khan, N.; Sultana, S. Modulatory effect of gentisic acid on the augmentation of biochemical events of tumor promotion stage by benzoyl peroxide and ultraviolet radiation in Swiss albino mice. Toxicol. Lett. 2004, 153, 293–302. [Google Scholar] [CrossRef]
  161. Sharma, S.; Khan, N.; Sultana, S. Study on prevention of two-stage skin carcinogenesis by Hibiscus rosa sinensis extract and the role of its chemical constituent, gentisic acid, in the inhibition of tumour promotion response and oxidative stress in mice. Eur. J. Cancer Prev. 2004, 13, 53–63. [Google Scholar] [CrossRef] [PubMed]
  162. Owen, S.G.; Roberts, M.S.; Friesen, W.T.; Francis, H.W. Salicylate pharmacokinetics in patients with rheumatoid arthritis. Br. J. Clin. Pharmacol. 1989, 28, 449–461. [Google Scholar] [CrossRef] [PubMed]
  163. Bochner, F.; Graham, G.G.; Polverino, A.; Imhoff, D.M.; Tregenza, R.A.; Rolan, P.E.; Cleland, L.G. Salicyl phenolic glucuronide pharmacokinetics in patients with rheumatoid arthritis. Eur. J. Clin. Pharmacol. 1987, 32, 153–158. [Google Scholar] [CrossRef] [PubMed]
  164. Baxter, G.J.; Lawrence, J.R.; Graham, A.B.; Wiles, D.; Paterson, J.R. Identification and determination of salicylic acid and salicyluric acid in urine of people not taking salicylate drugs. Ann. Clin. Biochem. 2002, 39, 50–55. [Google Scholar] [CrossRef]
  165. Park, M.V. Complex formation between iron (III) and some substituted salicylic acids. Chem. Soc. A 1966, 816–820. [Google Scholar] [CrossRef]
  166. Gonzalez, E.B.; Daeid, N.N.; Nolan, K.B. Complex formation between transition metal ions and salicylglycine, a metabolite of aspirin. Polyhedron 1994, 13, 1495–1499. [Google Scholar] [CrossRef]
  167. Pecci, J.; Foye, W.O. The avidity of salicylic, gentisic, and salicyluric acids for heavy metal cations. J. Am. Pharm. Assoc. Am. Pharm. Assoc. (Sci. Ed.) 1960, 49, 411–414. [Google Scholar] [CrossRef] [PubMed]
  168. Bavoso, A.; Menabue, L.; Saladini, M.; Sola, M. Coordination of 2-hydroxyhippuric acid to the copper(II) ion: A solution and solid state study. Inorg. Chim. Acta 1996, 244, 207–212. [Google Scholar] [CrossRef]
  169. Ferrer, E.G.; González Baró, A.C.; Castellano, E.E.; Piro, O.E.; Williams, P.A. Model complexes with naturally occurring ligands (salicylglycine and imidazol) and the biometals copper and cobalt. J. Inorg. Biochem. 2004, 98, 413–421. [Google Scholar] [CrossRef] [PubMed]
  170. Jancsó, A.; Gajda, T.; Szorcsik, A.; Kiss, T.; Henry, B.; Vankó, G.; Rubini, P. Potentiometric and spectroscopic studies on the dimethyltin(IV) complexes of 2-hydroxyhippuric acid. J. Inorg. Biochem. 2001, 83, 187–192. [Google Scholar] [CrossRef]
  171. Esplin, T.L.; Cable, M.L.; Gray, H.B.; Ponce, A. Terbium-macrocycle complexes as chemical sensors: Detection of an aspirin metabolite in urine using a salicylurate-specific receptor site. Inorg. Chem. 2010, 49, 4643–4647. [Google Scholar] [CrossRef]
  172. Zhang, J. A Reaction of Aspirin with Ferrous Gluconate. AAPS PharmSciTech. 2015, 16, 1495–1499. [Google Scholar] [CrossRef]
  173. Podhaisky, H.P.; Abate, A.; Polte, T.; Oberle, S.; Schröder, H. Aspirin protects endothelial cells from oxidative stress—Possible synergism with vitamin E. FEBS Lett. 1997, 417, 349–351. [Google Scholar] [CrossRef] [PubMed]
  174. Schwarz, K.B.; Arey, B.J.; Tolman, K.; Mahanty, S. Iron chelation as a possible mechanism for aspirin-induced malondialdehyde production by mouse liver microsomes and mitochondria. J. Clin. Investig. 1988, 81, 165–170. [Google Scholar] [CrossRef]
  175. Woollard, A.C.; Wolff, S.P.; Bascal, Z.A. Antioxidant characteristics of some potential anticataract agents. Studies of aspirin, paracetamol, and bendazac provide support for an oxidative component of cataract. Free Radic. Biol. Med. 1990, 9, 299–305. [Google Scholar] [CrossRef]
  176. Aruoma, O.I.; Halliwell, B. The iron-binding and hydroxyl radical scavenging action of anti-inflammatory drugs. Xenobiotica 1988, 18, 459–470. [Google Scholar] [CrossRef] [PubMed]
  177. Xu, Y.X.; Du, F.; Jiang, L.R.; Gong, J.; Zhou, Y.F.; Luo, Q.Q.; Qian, Z.M.; Ke, Y. Effects of aspirin on expression of iron transport and storage proteins in BV-2 microglial cells. Neurochem. Int. 2015, 91, 72–77. [Google Scholar] [CrossRef]
  178. Oberle, S.; Polte, T.; Abate, A.; Podhaisky, H.P.; Schröder, H. Aspirin increases ferritin synthesis in endothelial cells: A novel antioxidant pathway. Circ. Res. 1998, 82, 1016–1020. [Google Scholar] [CrossRef] [PubMed]
  179. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed]
  180. Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tang, D. Ferroptosis: Process and function. Cell Death Differ. 2016, 23, 369–379. [Google Scholar] [CrossRef] [PubMed]
  181. Cao, J.Y.; Dixon, S.J. Mechanisms of ferroptosis. Cell Mol. Life Sci. 2016, 73, 2195–2209. [Google Scholar] [CrossRef] [PubMed]
  182. Liu, L.; Li, L.; Li, M.; Luo, Z. Autophagy-Dependent Ferroptosis as a Therapeutic Target in Cancer. Chemmedchem 2021, 16, 2942–2950. [Google Scholar] [CrossRef] [PubMed]
  183. Mou, Y.; Wang, J.; Wu, J.; He, D.; Zhang, C.; Duan, C.; Li, B. Ferroptosis, a new form of cell death: Opportunities and challenges in cancer. J. Hematol. Oncol. 2019, 12, 34. [Google Scholar] [CrossRef]
  184. Kontoghiorghes, G.J. New Iron Metabolic Pathways and Chelation Targeting Strategies Affecting the Treatment of All Types and Stages of Cancer. Int. J. Mol. Sci. 2022, 23, 13990. [Google Scholar] [CrossRef]
  185. Wu, Z.; Li, D.; Tian, D.; Liu, X.; Wu, Z. Aspirin mediates protection from diabetic kidney disease by inducing ferroptosis inhibition. PLoS ONE 2022, 17, e0279010. [Google Scholar] [CrossRef]
  186. Li, J.; Zheng, S.; Fan, Y.; Tan, K. Emerging significance and therapeutic targets of ferroptosis: A potential avenue for human kidney diseases. Cell Death Dis. 2023, 14, 628. [Google Scholar] [CrossRef]
  187. Jiang, Y.; Yu, Y.; Pan, Z.; Glandorff, C.; Sun, M. Ferroptosis: A new hunter of hepatocellular carcinoma. Cell Death Discov. 2024, 10, 136. [Google Scholar] [CrossRef]
  188. Cheng, X.; Zhao, F.; Ke, B.; Chen, D.; Liu, F. Harnessing Ferroptosis to Overcome Drug Resistance in Colorectal Cancer: Promising Therapeutic Approaches. Cancers 2023, 15, 5209. [Google Scholar] [CrossRef] [PubMed]
  189. Li, B.; Yang, L.; Peng, X.; Fan, Q.; Wei, S.; Yang, S.; Li, X.; Jin, H.; Wu, B.; Huang, M.; et al. Emerging mechanisms and applications of ferroptosis in the treatment of resistant cancers. Biomed. Pharmacother. 2020, 130, 110710. [Google Scholar] [CrossRef] [PubMed]
  190. Zhu, X.; Li, S. Ferroptosis, Necroptosis, and Pyroptosis in Gastrointestinal Cancers: The Chief Culprits of Tumor Progression and Drug Resistance. Adv. Sci. 2023, 10, e2300824. [Google Scholar] [CrossRef]
  191. Grancher, A.; Michel, P.; Di Fiore, F.; Sefrioui, D. Colorectal cancer chemoprevention: Is aspirin still in the game? Cancer Biol. Ther. 2022, 23, 446–461. [Google Scholar] [CrossRef]
  192. Chen, H.; Qi, Q.; Wu, N.; Wang, Y.; Feng, Q.; Jin, R.; Jiang, L. Aspirin promotes RSL3-induced ferroptosis by suppressing mTOR/SREBP-1/SCD1-mediated lipogenesis in PIK3CA-mutant colorectal cancer. Redox Biol. 2022, 55, 102426. [Google Scholar] [CrossRef]
  193. Zhao, L.; Zhou, X.; Xie, F.; Zhang, L.; Yan, H.; Huang, J.; Zhang, C.; Zhou, F.; Chen, J.; Zhang, L. Ferroptosis in cancer and cancer immunotherapy. Cancer Commun. 2022, 42, 88–116. [Google Scholar] [CrossRef]
  194. Wang, Y.F.; Feng, J.Y.; Zhao, L.N.; Zhao, M.; Wei, X.F.; Geng, Y.; Yuan, H.F.; Hou, C.Y.; Zhang, H.H.; Wang, G.W.; et al. Aspirin triggers ferroptosis in hepatocellular carcinoma cells through restricting NF-κB p65-activated SLC7A11 transcription. Acta Pharmacol. Sin. 2023, 44, 1712–1724. [Google Scholar] [CrossRef]
  195. Rubner, G.; Bensdorf, K.; Wellner, A.; Kircher, B.; Bergemann, S.; Ott, I.; Gust, R. Synthesis and biological activities of transition metal complexes based on acetylsalicylic acid as neo-anticancer agents. J. Med. Chem. 2010, 53, 6889–6898. [Google Scholar] [CrossRef]
  196. Campbell, N.R.; Hasinoff, B. Ferrous sulfate reduces levodopa bioavailability: Chelation as a possible mechanism. Clin. Pharmacol. Ther. 1989, 45, 220–225. [Google Scholar] [CrossRef] [PubMed]
  197. Campbell, N.R.; Kara, M.; Hasinoff, B.B.; Haddara, W.M.; McKay, D.W. Norfloxacin interaction with antacids and minerals. Br. J. Clin. Pharmacol. 1992, 33, 115–116. [Google Scholar] [CrossRef] [PubMed]
  198. Schaefer, J.P.; Tam, Y.; Hasinoff, B.B.; Tawfik, S.; Peng, Y.; Reimche, L.; Campbell, N.R. Ferrous sulphate interacts with captopril. Br. J. Clin. Pharmacol. 1998, 46, 377–381. [Google Scholar] [CrossRef] [PubMed]
  199. Geisser, P. In vitro studies on interactions of iron salts and complexes with food-stuffs and medicaments. Arzneimittelforschung 1990, 40, 754–760. [Google Scholar] [PubMed]
  200. Davies, N.M. Clinical pharmacokinetics of oxaprozin. Clin. Pharmacokinet. 1998, 35, 425–436. [Google Scholar] [CrossRef]
  201. Davies, N.M. Clinical pharmacokinetics of ibuprofen. The first 30 years. Clin. Pharmacokinet. 1998, 34, 101–154. [Google Scholar] [CrossRef] [PubMed]
  202. Koomanan, N.; Ko, Y.; Yong, W.P.; Ng, R.; Wong, Y.P.; Lim, S.W.; Salim, A.; Chan, A. Clinical impact of drug-drug interaction between aspirin and prednisolone at a cancer center. Clin. Ther. 2012, 34, 2259–2267. [Google Scholar] [CrossRef]
  203. Johansson, U.; Akesson, B. Interaction between ascorbic acid and acetylsalicylic acid and their effects on nutritional status in man. Int. J. Vitam. Nutr. Res. 1985, 55, 197–204. [Google Scholar] [PubMed]
  204. Sher, M.; Hussain, M.A.; Khan, S.H.; Naeem-Ul-Hassan, M.; Amin, M.; Khan, H. Determination of Copper at Extended Dose Levels of Copper (II)-acetylsalicylate and Pharmacokinetics Applications. Acta Chim. Slov. 2023, 70, 86–90. [Google Scholar] [CrossRef] [PubMed]
  205. Li, T.; Ding, N.; Guo, H.; Hua, R.; Lin, Z.; Tian, H.; Yu, Y.; Fan, D.; Yuan, Z.; Gonzalez, F.J.; et al. A gut microbiota-bile acid axis promotes intestinal homeostasis upon aspirin-mediated damage. Cell Host Microbe 2024, 32, 191–208.e9. [Google Scholar] [CrossRef]
  206. Jobbé-Duval, A.; Bézard, M.; Moutereau, S.; Kharoubi, M.; Oghina, S.; Zaroui, A.; Galat, A.; Chalard, C.; Hugon-Vallet, E.; Lemonnier, F.; et al. Prevalence and determinants of iron deficiency in cardiac amyloidosis. ESC Heart Fail. 2022, 9, 1314–1327. [Google Scholar] [CrossRef]
  207. Fairweather-Tait, S.J.; Wawer, A.A.; Gillings, R.; Jennings, A.; Myint, P.K. Iron status in the elderly. Mech. Ageing Dev. 2014, 136–137, 22–28. [Google Scholar] [CrossRef]
  208. Zacharski, L.R.; Chow, B.K.; Howes, P.S.; Shamayeva, G.; Baron, J.A.; Dalman, R.L.; Malenka, D.J.; Ozaki, C.K.; Lavori, P.W. Decreased cancer risk after iron reduction in patients with peripheral arterial disease: Results from a randomized trial. J. Natl. Cancer Inst. 2008, 100, 996–1002. [Google Scholar] [CrossRef]
  209. Rothwell, P.M.; Fowkes, F.G.; Belch, J.F.; Ogawa, H.; Warlow, C.P.; Meade, T.W. Effect of daily aspirin on long-term risk of death due to cancer:analysis of individual patient data from randomised trials. Lancet 2011, 377, 31–41. [Google Scholar] [CrossRef]
  210. Mascitelli, L.; Goldstein, M.R. Aspirin in the prevention of cancer. Lancet 2011, 377, 1650, author reply 1651–1652. [Google Scholar] [CrossRef] [PubMed]
  211. Michel, P.; Boige, V.; Andre, T.; Aparicio, T.; Bachet, J.B.; Dahan, L.; Guimbaud, R.; Lepage, C.; Manfredi, S.; Tougeron, D.; et al. Aspirin versus placebo in stage III or high-risk stage II colon cancer with PIK3CA mutation: A French randomised double-blind phase III trial (PRODIGE 50-ASPIK). Dig. Liver Dis. 2018, 50, 305–307. [Google Scholar] [CrossRef] [PubMed]
  212. Patrono, C. Cyclooxygenase Inhibitors and Cancer: The Missing Pieces. J. Pharmacol. Exp. Ther. 2023, 386, 181–189. [Google Scholar] [CrossRef] [PubMed]
  213. Tao, D.L.; Tassi Yunga, S.; Williams, C.D.; McCarty, O.J.T. Aspirin and antiplatelet treatments in cancer. Blood 2021, 137, 3201–3211. [Google Scholar] [CrossRef] [PubMed]
  214. Jacobs, A. An intracellular transit iron pool. Ciba Found. Symp. 1976, 51, 91–106. [Google Scholar]
  215. Binding, A.; Ward, R.; Tomlinson, G.; Kuo, K.H.M. Deferiprone exerts a dose-dependent reduction of liver iron in adults with iron overload. Eur. J. Haematol. 2019, 103, 80–87. [Google Scholar] [CrossRef] [PubMed]
  216. Olivieri, N.F.; Koren, G.; Matsui, D.; Liu, P.P.; Blendis, L.; Cameron, R.; McClelland, R.A.; Templeton, D.M. Reduction of tissue iron stores and normalization of serum ferritin during treatment with the oral iron chelator L1 in thalassemia intermedia. Blood 1992, 79, 2741–2748. [Google Scholar] [CrossRef]
  217. Kontoghiorghes, G.J.; Sheppard, L.; Aldouri, M.A.; Hoffbrand, A.V. 1,2-Dimethyl-3-Hydroxypyrid-4-One, an Orally Active Chelator for Treatment of Iron Overload. Lancet 1987, 329, 1294–1295. [Google Scholar] [CrossRef]
  218. Kontoghiorghes, G.J.; Aldouri, M.A.; Hoffbrand, A.V.; Barr, J.; Wonke, B.; Kourouclaris, T.; Sheppard, L. Effective chelation of iron in beta thalassaemia with the oral chelator 1,2-dimethyl-3-hydroxypyrid-4-one. BMJ 1987, 295, 1509–1512. [Google Scholar] [CrossRef] [PubMed]
  219. Rothwell, P.M.; Cook, N.R.; Gaziano, J.M.; Price, J.F.; Belch, J.F.F.; Roncaglioni, M.C.; Morimoto, T.; Mehta, Z. Effects of aspirin on risks of vascular events and cancer according to bodyweight and dose: Analysis of individual patient data from randomised trials. Lancet 2018, 392, 387–399. [Google Scholar] [CrossRef] [PubMed]
  220. Ardeshna, D.; Khare, S.; Jagadish, P.S.; Bhattad, V.; Cave, B.; Khouzam, R.N. The dilemma of aspirin resistance in obese patients. Ann. Transl. Med. 2019, 7, 404. [Google Scholar] [CrossRef] [PubMed]
  221. Cuzick, J.; Thorat, M.A.; Bosetti, C.; Brown, P.H.; Burn, J.; Cook, N.R.; Ford, L.G.; Jacobs, E.J.; Jankowski, J.A.; La Vecchia, C.; et al. Estimates of benefits and harms of prophylactic use of aspirin in the general population. Ann. Oncol. 2015, 26, 47–57. [Google Scholar] [CrossRef]
  222. Kontoghiorghes, G.J. Deferiprone and Iron-Maltol: Forty Years since Their Discovery and Insights into Their Drug Design, Development, Clinical Use and Future Prospects. Int. J. Mol. Sci. 2023, 24, 4970. [Google Scholar] [CrossRef] [PubMed]
  223. Sheppard, L.N.; Kontoghiorghes, G.J. Competition between deferiprone, desferrioxamine and other chelators for iron and the effect of other metals. Arzneimittelforschung 1993, 43, 659–663. [Google Scholar] [PubMed]
  224. Wang, X. A chelate theory for the mechanism of action of aspirin-like drugs. Med. Hypotheses 1998, 50, 239–251. [Google Scholar] [CrossRef] [PubMed]
  225. Pippard, M.J.; Jackson, M.J.; Hoffman, K.; Petrou, M.; Modell, C.B. Iron chelation using subcutaneous infusions of diethylene triamine penta-acetic acid (DTPA). Scand. J. Haematol. 1986, 36, 466–472. [Google Scholar] [CrossRef] [PubMed]
  226. Kontoghiorghes, G.J. Prospects for introducing deferiprone as potent pharmaceutical antioxidant. Front. Biosci. (Elite Ed.) 2009, 1, 161–178. [Google Scholar] [CrossRef] [PubMed]
  227. Barradas, M.A.; Jeremy, J.Y.; Kontoghiorghes, G.J.; Mikhailidis, D.P.; Hoffbrand, A.V.; Dandona, P. Iron chelators inhibit human platelet aggregation, thromboxane A2 synthesis and lipoxygenase activity. FEBS Lett. 1989, 245, 105–109. [Google Scholar] [CrossRef]
  228. Jeremy, J.Y.; Kontoghiorghes, G.J.; Hoffbrand, A.V.; Dandona, P. The iron chelators desferrioxamine and 1-alkyl-2-methyl-3-hydroxypyrid-4-ones inhibit vascular prostacyclin synthesis in vitro. Biochem. J. 1988, 254, 239–244. [Google Scholar] [CrossRef]
  229. Pirmohamed, M.; James, S.; Meakin, S.; Green, C.; Scott, A.K.; Walley, T.J.; Farrar, K.; Park, B.K.; Breckenridge, A.M. Adverse drug reactions as cause of admission to hospital: Prospective analysis of 18 820 patients. BMJ 2004, 329, 15–19. [Google Scholar] [CrossRef] [PubMed]
  230. Derry, S.; Loke, Y.K. Risk of gastrointestinal haemorrhage with long term use of aspirin: Meta-analysis. BMJ 2000, 321, 1183. [Google Scholar] [CrossRef]
  231. Migally, K.; Braunlin, E.A.; Zhang, L.; Binstadt, B.A. Duration of high-dose aspirin therapy does not affect long-term coronary artery outcomes in Kawasaki disease. Pediatr. Res. 2018, 83, 1136–1145. [Google Scholar] [CrossRef] [PubMed]
  232. Gan, R.; Teleg, R.A.; Florento, L.; Bitanga, E.S. Effect of increasing doses of aspirin on platelet aggregation among stroke patients. Cerebrovasc. Dis. 2002, 14, 252–255. [Google Scholar] [CrossRef] [PubMed]
  233. Adams, H.P., Jr.; Bendixen, B.H. Low- versus high-dose aspirin in prevention of ischemic stroke. Clin. Neuropharmacol. 1993, 16, 485–500. [Google Scholar] [CrossRef] [PubMed]
  234. Born, T.; Kontoghiorghe, C.N.; Spyrou, A.; Kolnagou, A.; Kontoghiorghes, G.J. EDTA chelation reappraisal following new clinical trials and regular use in millions of patients: Review of preliminary findings and risk/benefit assessment. Toxicol. Mech. Methods 2013, 23, 11–17. [Google Scholar] [CrossRef]
Figure 1. The metabolic transformation of aspirin. Aspirin or acetyl salicylic acid (a) is metabolized to salicylic acid (b), which is then further metabolized to another five metabolites, which are salicyluric acid, (c) 2,3-dihydroxybenzoic acid, (d) 2,5-dihydroxybenzoic acid (gentisic acid), (e) salicylacyl glucuronide, (f) and salicylphenol glucuronide (g).
Figure 1. The metabolic transformation of aspirin. Aspirin or acetyl salicylic acid (a) is metabolized to salicylic acid (b), which is then further metabolized to another five metabolites, which are salicyluric acid, (c) 2,3-dihydroxybenzoic acid, (d) 2,5-dihydroxybenzoic acid (gentisic acid), (e) salicylacyl glucuronide, (f) and salicylphenol glucuronide (g).
Ijms 25 05150 g001
Figure 2. The potential iron-chelating sites of the aspirin metabolites. All the aspirin-chelating metabolites appear to be potential bidentate chelators. The ligands involved in iron (Fe III) binding and the formation of iron (Fe III) complexes are shown for salicylic acid (a), 2,3-dihydroxybenzoic acid (b), 2,5-dihydroxybenzoic acid (c), and salicyluric acid (d). A similar iron-binding site of (d) is theoretically possible for the salicylacyl glucuronide, both of which are weaker iron chelators in comparison to (a) and (c) and much weaker than (b), which is considered the strongest iron chelator of the aspirin metabolites.
Figure 2. The potential iron-chelating sites of the aspirin metabolites. All the aspirin-chelating metabolites appear to be potential bidentate chelators. The ligands involved in iron (Fe III) binding and the formation of iron (Fe III) complexes are shown for salicylic acid (a), 2,3-dihydroxybenzoic acid (b), 2,5-dihydroxybenzoic acid (c), and salicyluric acid (d). A similar iron-binding site of (d) is theoretically possible for the salicylacyl glucuronide, both of which are weaker iron chelators in comparison to (a) and (c) and much weaker than (b), which is considered the strongest iron chelator of the aspirin metabolites.
Ijms 25 05150 g002
Table 1. The role of aspirin chelating metabolites on iron deficiency and the influence of other factors.
Table 1. The role of aspirin chelating metabolites on iron deficiency and the influence of other factors.
A. The properties and iron binding effects of the aspirin-chelating metabolites
Molecular properties of the aspirin-chelating metabolites
All aspirin metabolites are of low molecular weight, with the smallest being salicylic acid. The ACMs are negatively charged at physiological pH due to the presence of a carboxylate group. All the aspirin metabolites are cleared through the kidneys and eliminated through the urinary route.
Achievement of high concentration of the aspirin-chelating metabolites with high iron capacity
Almost 70% of aspirin is metabolized to ACM, with salicyluric acid predominating (>65%) over the other metabolites. The ACMs have sufficiently long half-lives and can reach optimal concentrations for the mobilization of iron. The combined concentration of the ACM has a high iron binding capacity equivalent to that of iron-chelating drugs.
Evidence of the high affinity for iron by the aspirin-chelating metabolites
The iron complex formation of the ACM, with specific physicochemical characteristics in each case, indicates a high affinity for iron, with high stability constants and the ability to mobilize iron. For example, the iron stability constant of salicylic acid (log β3 = 36) is similar to that of the iron-chelating drug deferiprone.
Iron mobilizing efficacy of the aspirin-chelating metabolites in preclinical studies
The ACMs have been shown to mobilize iron from aqueous solutions, ferritin, cells, and different animal species.
Iron mobilizing efficacy of the aspirin-chelating metabolites in clinical studies
The ACM 2,3-dihydroxybenoic acid has been shown in several clinical trials to be effective in increasing substantially iron excretion in iron-loaded thalassaemia patients. Smaller amounts of iron excretion are expected in aspirin users with normal iron stores.
Cumulative effects on iron mobilization by the aspirin-chelating metabolites
The combinatory effect of the ACM in increasing iron mobilization and iron excretion resembles the combination chelation therapy, which is more effective than monotherapy. Despite the small incremental increases in daily iron excretion caused by ACM, total iron loss over months and years could lead to iron deficiency.
B. Other factors influencing iron deficiency in low-dose daily aspirin users
The influence of dietary iron on iron balance in elderly individuals using low-dose aspirin daily
In the majority of cases of low-dose daily aspirin users, in the absence of major gastric bleeding, iron balance is restored gradually from dietary iron, similarly to blood donors.
Iron deficiency is observed only in a minority of cases of low-dose daily aspirin users in the absence of major gastric bleeding. It is likely that such individuals are vegetarians or receive mostly vegetarian meals with insufficient absorbable content.
Additional factors to dietary iron contributing to iron deficiency in daily low-dose aspirin users
There are additional factors involved in insufficient dietary iron body intake which can contribute to iron deficiency in daily low-dose aspirin users. These include individuals who are blood donors or engaged in heavy sport activities, as well as malabsorption of iron due to complications in the GIT. Minor undetected blood loss may also contribute to iron deficiency.
Cumulative effects contributing to iron deficiency in daily low dose aspirin users
One or more of the aforementioned factors, i.e., low dietary iron intake, blood donation, heavy sports, or minor bleeding, may contribute to the overall increases in body iron loss. The contributory effect of each of these factors is different for each individual.
ACM: aspirin-chelating metabolites. GIT: gastrointestinal tract.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kontoghiorghes, G.J. The Puzzle of Aspirin and Iron Deficiency: The Vital Missing Link of the Iron-Chelating Metabolites. Int. J. Mol. Sci. 2024, 25, 5150. https://doi.org/10.3390/ijms25105150

AMA Style

Kontoghiorghes GJ. The Puzzle of Aspirin and Iron Deficiency: The Vital Missing Link of the Iron-Chelating Metabolites. International Journal of Molecular Sciences. 2024; 25(10):5150. https://doi.org/10.3390/ijms25105150

Chicago/Turabian Style

Kontoghiorghes, George J. 2024. "The Puzzle of Aspirin and Iron Deficiency: The Vital Missing Link of the Iron-Chelating Metabolites" International Journal of Molecular Sciences 25, no. 10: 5150. https://doi.org/10.3390/ijms25105150

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop