throbber
Coordination Chemistry Reviews
`184 (1999) 291–310
`
`Iron chelating agents in clinical practice
`
`Gavino Faa a,*, Guido Crisponi b
`a Dipartimento di Citomorfologia, Sezione di Anatomia Patologica, Via Porcell 2,
`I-09124 Cagliari, Italy
`b Dipartimento di Chimica e Tecnologie Inorganiche e Metallorganiche, Via Ospedale 72,
`I-09124 Cagliari, Italy
`
`Received 24 November 1998; accepted 16 February 1999
`
`Contents
`
`Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`2. Iron overload and toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3. Desferrioxamine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`4. Development of non toxic oral iron chelators . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`4.1 Deferiprone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`5. New indications for iron-chelating therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`5.1 Adult Respiratory Distress Syndrome (ARDS).
`. . . . . . . . . . . . . . . . . . . . . . . .
`5.2
`Iron chelators and myocardial ischemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`5.3
`Iron chelators and cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`5.4 Iron chelators as antimalarials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`6. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`291
`292
`292
`294
`298
`300
`302
`303
`303
`304
`304
`306
`306
`
`Abstract
`
`The relevance of iron chelators in medicine has increased in recent years. Iron is essential
`for life but it is also potentially more toxic than other trace elements. This is due to the lack
`of effective means to protect human cells against iron overload and to the role of iron in the
`generation of free radicals. To protect patients from the consequences of iron toxicity, iron
`chelating agents have been introduced in clinical practice. Unfortunately, the ideal chelator
`for treating iron overload in humans has not been identified yet. The aim of this review is to
`report the experience with desferrioxamine therapy in patients affected by b-thalassemia
`major according to: bioavailability; mechanism of interactions with hepatocellular iron:
`
`* Corresponding author.
`E-mail address: gfaa@vaxca1.unica.it (G. Faa)
`
`0010-8545/99/$ - see front matter © 1999 Elsevier Science S.A. All rights reserved.
`PII: S 0 0 1 0 - 8 5 4 5 ( 9 9 ) 0 0 0 5 6 - 9
`
`
`1 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`292
`
`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`in
`impact of iron chelation on survival
`release of iron chelates and their excretion;
`thalassemia patients and side effects of prolonged therapy. Problems related to the develop-
`ment of non-toxic oral iron chelators are also discussed, with particular emphasis on the
`preliminary data on usefulness and safety of deferiprone (L1), recently evaluated in different
`clinical trials. Iron chelating therapy has been introduced, in recent years, even in the therapy
`of disorders not characterized by iron overload. Here the following new therapeutic
`indications are discussed: adult respiratory distress syndrome, myocardial ischemia, cancer
`and malaria. © 1999 Elsevier Science S.A. All rights reserved.
`
`Keywords: Iron-chelators; Desferrioxamine; Deferiprone; Thalassemia
`
`1. Introduction
`
`Iron chelators are used in medicine to protect patients from the consequences of
`iron overload and iron toxicity in organs and tissues. The ideal chelator for treating
`iron overload in humans should act as a selective depletor of iron, should be
`efficiently absorbed by the gastrointestinal tract, could not cross the blood–brain
`and placental barriers and should lack or have a low toxicity. Such a chelating
`agent has not been identified yet and this goal is at the basis of multiple research
`projects in this field. In this review, the experience with long-term iron chelating
`therapy in patients affected by chronic transfusion-dependent anemias will be
`summarized with particular emphasis on thalassemic patients who are the main
`target of iron-chelating drugs. The experience in thalassemia patients with the well
`established chelator desferrioxamine and with the orally active deferiprone will be
`outlined. This review is also intended to report the most recent development of new
`non-toxic and orally effective iron chelating agents and their possible application in
`clinical use. New indications for iron chelating therapy will also be explored, such
`as the use of iron chelation in oncology to prevent tumor cell growth by the
`inhibition of iron-dependent enzymes, the application of iron-chelating agents in
`the therapy of infectious disease with particular emphasis on their action as
`antimalarials,
`the experience of
`iron-chelating drugs in the therapy of adult
`respiratory distress syndrome and in the therapy of myocardial ischemia. Finally,
`the relevance of the dialogue among clinicians, pathologists, pharmacologists,
`biochemists, chemists, molecular biologists and other experts in metal toxicity in
`order to improve our knowledge on the relationship between the metabolism of
`iron and other trace elements will be discussed. This implies the final goal of
`prolonging survival and improving the quality of life of iron-loaded patients.
`
`2. Iron overload and toxicity
`
`Iron is essential for life: all living cells, whether prokaryotic or eukaryotic, need
`a supply of iron for reduction of oxygen (respiration), reduction of carbon dioxide
`
`
`2 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`293
`
`(photosynthesis), reduction of dinitrogen or other fundamental biological processes
`[1]. Excessive amounts of iron may become very toxic to the human body and,
`eventually, is fatal for vital cell structures [2].
`Iron overload may be defined as an excess in total body iron stores. The normal
`iron concentration in the human body ranges between 40 and 50 mg/kg of body
`weight [3]. Most of this iron is present in hemoglobin and in myoglobin: all the rest
`is stored as ferritin or as its less accessible form, hemosiderin. Only a few hundred
`milligrams of iron are stored in enzymes such as cytochrome c oxidase which,
`however, are essential to human life [1]. Humans have very limited capacity for
`excretion of excess iron: in particular they lack any effective means to protect cells
`and tissues against iron overload. As a consequence, any increase in iron intake
`may cause in a short time an increase in body iron stores [4]. Iron balance is
`normally regulated by controlling iron absorption in the proximal small intestine
`[5]. In women, losing iron in the menstrual cycle could protect against excess iron
`toxicity and it is considered to be, at least in part, responsible for their greater
`longevity than men [1]. The major regulators of mucosal iron absorption are the
`amount of body iron stores and the level of erythropoiesis [6]. Iron overload may
`be caused by two different factors:
`1. parenteral administration of iron, as in chronic transfusion therapy;
`2. increase in iron absorption from the diet, that may be genetically determined
`like in hereditary hemochromatosis [7] or caused by dietary iron overload [8].
`When the accumulation of iron in organs exceeds the body capacity for safe
`storage, potentially lethal tissue damage results [9]. The severity of iron toxicity
`seems to be related to the amount of body iron burden. Recent studies in patients
`with thalassemia major found that the magnitude of the body iron burden was the
`major determinant of the risk of clinical complications and of early death [10,11].
`Target organs for iron-induced injury are the liver, pancreas and heart [12].
`In the category of patients affected by chronic anaemia, who need regular blood
`transfusions in order to sustain their normal growth and development during
`childhood, b-thalassemia major (BTM) constitutes one of the most serious public
`health problems in the Mediterranean area [13], in the Middle East, in the Indian
`subcontinent, in Southeast Asia [14] and, in particular, in the island of Sardinia
`[15]. BTM is an autosomal recessive disease, characterized by absent or decreased
`synthesis of the b globin gene: the number of thalassemic children requiring regular
`blood transfusions and a program of iron chelation has been estimated to be
`100 000 world-wide. In patients with thalassemia major, iron contained in trans-
`fused red cells inexorably accumulates if a concomitant regular chelation therapy is
`not programmed [16]. The major pathological manifestations observed in patients
`with BTM are related to iron overload: chronic liver disease, characterized by
`hepatocytic and Kupffer cell iron storage, fibrosis and, eventually, cirrhosis [17,18];
`dilatative cardiomyopathy with congestive heart failure is nowadays the most
`common cause of death in patients affected by BTM who reach adolescence and
`adulthood [19,20]. Even cardiomyopathy is related to cardiac hemosiderosis: a
`preferential accumulation of iron in the interventricular septum and in the left
`ventricle wall has been observed [21]. The toxic effects of iron overload has been
`
`
`3 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`294
`
`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`demonstrated in cultured heart cells: incubation of rat heart cell cultures with iron
`concentrations from 20 up to 80 g/ml resulted in a marked decrease in amplitude
`and rate of contractions and in gross abnormality in rhythmicity [22].
`
`3. Desferrioxamine
`
`Desferrioxamine is the only chelating agent to have been extensively used in
`clinical practice [23] after its discovery more than 30 years ago. It is a siderophore
`produced by Streptomyces pilosus [24], discovered by the team of Prelog and his
`co-workers Zahner and Keberle.
`
`Desferrioxamine, earlier recognized as an antagonist of the antibiotic ferrimycin,
`was successively identified as an iron chelating agent [25]. It is a trihydroxamic acid
`with three residues of 1-amino-5-N-hydroxy aminopentane, two of succinic acid
`and one of acetic acid organized in a linear array; the free amino group explains its
`very high water solubility. Although it has been synthesized the use of the natural
`product is more economic.
`Initially used in the therapy of acute iron poisoning [26], desferrioxamine was
`later introduced in thalassemia treatment, giving a fresh chance to more than
`100 000 thalassemic patients requiring iron chelating therapy. The widespread use
`of desferrioxamine was firmly established, thanks to British investigators such as
`Berry, Modell, Pippard and Hoffman and to Italian clinicians such as Cao. The
`relevance of desferrioxamine in clinical practice and its role in the progress of the
`therapy of thalassemia patients has been underlined on the occasion of the 1991
`Pharmaceutical of the Year price, awarded to desferrioxamine by the Munchener
`Medizinische Wochenschrift [27]. In order to better understand the value of this
`award, it is useful to observe that it had been previously given to aspirin, cortisone
`and penicillin. Desferrioxamine is the only iron-chelating agent approved for
`clinical use [28]. In the following, we shall focus on
`“ the bioavailability;
`“ the mechanism whereby it interacts with hepatocellular iron, with iron stored in
`macrophages and with iron in transit;
`“ the release of its iron chelates and their subsequent biliary or urinary excretion;
`“ the impact of iron chelation on survival in thalassemia patients;
`“ the side effects of prolonged therapy.
`Bioavailability is defined by Hider [29] as the percentage of absorbed dose of a drug
`which reaches the systemic blood circulation. It depends on the absorption of the
`drug from the gastrointestinal tract and from the extraction by hepatocytes from
`the portal blood supply. Low or high bioavailability may be requested in iron
`chelators, depending on the clinical setting and on the main target of chelating
`
`
`4 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`295
`
`therapy. Thus, when a general systemic action of a chelator is required, a high
`bioavailability is ideal. On the contrary, when liver is the target of iron chelation,
`an efficient absorption of the chelator by hepatocytes and, consequently, a low
`bioavailability is ideal. Two major factors influence the absorption of iron chelators
`from the gastrointestinal tract: the oil/water distribution coefficient [30] and the
`molecular weight. To achieve 70% absorption of an iron chelator, its molecular
`weight [31] needs to be B300. A 70% absorption of the dose is mandatory for iron
`chelators: 50% absorption could leave in the lumen such a level of the chelator that
`might disturb the microbiological flora [29]. Most siderophores, including desfer-
`rioxamine, have a molecular weight from 500 to 900 Da which effectively excludes
`hexadentate ligands, such as desferrioxamine, from consideration as orally active
`chelators. In contrast deferiprone and 1,2-diethyl-3-hydroxy-4-pyridinone, two lig-
`ands of molecular weight 139 and 167 Da, respectively, are both efficiently
`absorbed in man [32]. Since desferrioxamine given orally is poorly absorbed, to be
`effective it must be administered subcutaneously [33], intramuscularly [34] or by
`intravenous infusion with a small portable syringe pump, ideally for 9–12 h each
`day [35]. This difficult regimen of desferrioxamine parenteral treatment easily
`explains why only part of thalassemic patients comply with iron chelating therapy,
`in spite of the knowledge that the advent of treatment with desferrioxamine has
`changed the gloomy prognosis of thalassemia patients [36]. On entering blood by
`intravenous injection, desferrioxamine plasma clearance [37] is generally considered
`rapid, with half life of 5 to 10 min, also if a longer half life (3.0591.30 h) has been
`estimated in a recent study [38]. While only a small part of desferrioxamine is
`inactivated within human plasma, the major part undergoes uptake by hepatocytes.
`The rapid loss of circulating activity of desferrioxamine after intravenous injection
`is the main reason why prolonged infusion results in more efficient iron chelation
`[39]. After injection of desferrioxamine, both fecal and urinary iron excretion are
`observed. Probably the two excretion pathways reflect two different actions of the
`drug. Fecal excretion could only arise through iron chelation within the hepato-
`cytes, followed by excretion in the bile of the iron–desferrioxamine complex [39].
`The source of urinary iron remains more controversial: urinary excretion could
`derive from iron chelation within Kupffer cells and within other monophagocytic
`cells like spleen macrophages [40,41]. A further source to the urine is likely to be
`any nontransferrin bound iron in plasma of patients with a fully saturated plasma
`trasferrin, this kind of iron is reduced by desferrioxamine treatment [42]. The
`picture emerges that the major source of iron excreted through faeces by desferriox-
`amine is iron within hepatocytes, while urinary excretion could reflect an additional
`pathway of chelation, predominantly extracellular, regarding plasma nontransferrin
`bound iron, membrane-related iron of hepatocytes and iron in macrophages [39].
`The mechanism by which desferrioxamine is effective in removing intra and
`extracellular iron is not yet completely understood. To further the understanding of
`how desferrioxamine, as well other chelators, exert their effects on cells, many
`animal [43] and cellular models [44] have been proposed. Experiments on hepato-
`cytes have shown that the hepatocyte plasma membrane possesses a number of
`facilitated transport processes which are particularly efficient for the absorption of
`
`
`5 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`296
`
`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`small peptides as well as of desferrioxamine [29]. Once entered into the hepatocytes,
`the targets of desferrioxamine are the different pools of iron. Ferritin, whose major
`function is the storage of iron, is a protein of molecular weight 450 000 Da which
`can contain ca. 4500 iron atoms [45]. Desferrioxamine has been shown in vitro to
`be able of removing Fe(III) from ferritin: however, iron release requires chelator
`concentrations much higer than expected [45]. Probably the interaction of desfer-
`rioxamine with ferritin-bound iron is indirect. Ferritin is continuosly metabolized
`within lysosomes; the iron released in lysosomes enters a low molecular weight pool
`of iron which is the principal source of iron for desferrioxamine [46], whose main
`target is probably the pool of chelatable free iron in lysosomes. Another possible
`source of iron for desferrioxamine is trasferrin-bound iron. Transferrin is a glyco-
`protein, with a molecular weight of about 80 000 Da, which transports iron from
`the sites of absorption to those of storage and utilization. Desferrioxamine is
`generally unable to remove iron from transferrin; it may chelate transferrin-bound
`iron only in the presence of organic pyrophosphates, whose presence facilitates
`release of iron from transferrin [47]. Once in contact with the cellular membrane of
`hepatocytes, transferrin attaches to specific receptors and it is internalized by
`hepatocytes enclosed within endocytic vesicles in which iron is released. Iron
`released inside endocytic vesicles enters a labile intracellular transit iron pool
`consisting of low molecular iron complexes which may be chelated by desferrioxam-
`ine. A contribution to this intracellular transit iron pool may even derive by
`degradation of iron rich ferritin. Hepatocytes contain, in their cellular membrane,
`receptors for ferritin utilized for the uptake of ferritin released by Kupffer cells,
`following phagocytosis of erythrocytes [48,49]. The ingested ferritin is degraded
`within hepatocytic lysosomes and the released iron enters the chelatable transit iron
`pool, sensitive to the action of desferrioxamine. In patients affected by severe iron
`overload, when complete saturation of transferrin occurs, a low molecular weight
`iron pool, not associated to transferrin, is always present even in plasma. This
`plasmatic fraction of nontransferrin-bound iron may be bound to albumin or
`complexed by amino acids, citrate or sugars. Even this pool is a main target for
`chelation by desferrioxamine. The iron overload observed in hepatocytes from
`patients affected by congenital atransferrinemia [50] has demonstrated the existence
`of systems for the uptake of nontransferrin-bound iron. In vitro studies have shown
`that desferrioxamine may act even at this level, inhibiting the transferrin receptor-
`independent iron uptake systems [51]. Desferrioxamine acts even on macrophages
`and, particularly, on Kupffer cells. An important function of Kupffer cells is
`phagocytosis of senescent erythrocytes. Iron derived from hemoglobin enters a
`labile transit pool which is either returned to plasma transferrin or retained in
`ferritin [45]. Likely desferoxamine competes with apotransferrin for this pool of
`iron, which is on its way out of Kupffer cells and it is considered the major source
`of urinary iron excreted during desferrioxamine therapy [40]; fecal iron excretion on
`the contrary reflects the action of desferrioxamine on the chelatable iron pool
`present in the cytoplasm of the hepatocyte. The intimate mechanism of action of
`desferrioxamine is probably similar to that typical of the majority of iron chelators.
`Desferrioxamine promotes the ferric form of iron and suppresses excellently iron
`
`
`6 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`297
`
`toxicity, inhibiting the formation of hydroxyl radicals and lipid peroxidation [52] by
`its ferroxidase activity [53]. A strong ferroxidase activity is also exerted by transfer-
`rin and by ceruloplasmin, a 132 kDa a-2-glycoprotein containing a trinuclear
`copper cluster responsible for its oxidase activity and radical scavenging [54]. The
`report that patients affected by dietary or genetic deficiency of ceruloplasmin show
`severe accumulation of iron in liver [55] as well in other organs [56] is at the basis
`of recent studies on the relevance of ceruloplasmin in iron metabolism, both in
`physiologic and in iron overloaded patients. One of the multiple functions of
`ceruloplasmin is to aid the release of iron from the hepatocyte to plasma transfer-
`rin. Ceruloplasmin is probably localized inside the endoplasmic reticulum, where it
`acts as a ferroxidase on Fe(II) mobilised from intracellular ferritins which may
`initiate free radical reactions [57]. This is a crucial step in iron release; in fact,
`transferrin may only bind Fe(III) [58]. The relevance of ceruloplasmin in iron
`excretion has been confirmed by the demonstration that infusion of ceruloplasmin
`may rapidly enhance iron release from liver in patients with congenital aceruloplas-
`minemia [55]. In Wilson’s desease, characterized by low plasma levels of ceruloplas-
`min, we have reported high iron levels in the liver in an autoptic case [59] and in
`a percentage of liver biopsies (G.Faa et al., unpublished data). A severe iron
`deposition has been moreover reported in the liver of LEC rats [60], an animal
`model in Wilson’s desease. To our knowledge, the correlation between ceruloplas-
`min status and desferrioxamine has not been extensively studied. On the basis of the
`crucial role of ceruloplasmin in iron release from hepatocytes, a deficiency in
`ceruloplasmin could explain why some thalassemic patients, in spite of their good
`compliance in desferrioxamine therapy, show massive iron storage in the liver and
`in the heart.
`An interesting property of desferrioxamine is that it is not only an excellent iron
`chelator, thereby decreasing the free-radical generating reactions [61], but can also
`directly scavenge some radical species [62].
`Although desferrioxamine has been demonstrated to be a safe drug when
`administered in the presence of an elevated body iron burden [63], serious compli-
`cations may arise as a consequence of long term chelation, mainly in young patients
`with low body iron stores. At the basis of desferrioxamine toxicity is the fact that,
`like other chelators, desferrioxamine is not completely iron-selective. The most
`frequent complications of long-term desferrioxamine treatment are growth retarda-
`tion, described in patients with thalassemia major [64,65]. Other complications
`described include bone changes [66], visual and auditory neurotoxicity [67]. Severe
`limb and metaphyseal deformities have been reported in patients who began
`chelation in the first year of life [64]. Other adverse effects of desferrioxamine, noted
`in patients receiving continuous 24-h infusions, are impairment of vision, with
`defective dark adaption and peripheral field loss [68]. Hearing and vision abnormal-
`ities may occur more readily in patients with low iron stores, but the relationship
`is still not entirely clear and other factors may play a role [69]. Different hypotheses
`have been postulated to explain the adverse effects of desferrioxamine treatment:
`1. depletion of other trace elements, such as zinc, copper, manganese, cobalt;
`2. a direct toxic effect of free desferrioxamine;
`
`
`7 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`298
`
`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`3. depletion of iron from critical iron-dependent enzymes, such as cytochrome c
`oxidase [39].
`A pulmonary syndrome, characterized by diffuse lung infiltration and restrictive
`dysfunction, has been rarely observed in patients with thalassemia major receiving
`intravenous high doses of desferrioxamine [70]. Local skin reaction at the site of
`subcutaneous injection of desferrioxamine is also common, in the form of acute
`inflammation or of subcutaneous nodules persisting for several days [23]. Another
`complication occurring during chronic treatment with desferrioxamine is oppor-
`tunistic Yersinia infection [71]. This complication of desferrioxamine treatment
`probably occurs since the virulence of Yersinia is enhanced by the presence of a
`membrane receptor which binds desferrioxamine [72]. Desferrioxamine toxicity may
`be controlled or prevented: the majority of all the reported adverse effects may be
`reduced or prevented by avoiding high doses and over-early commencement, and by
`long-term clinical monitoring once the overload is significantly reduced [23].
`In conclusion, there is no doubt that desferrioxamine, with all its limitations, has
`changed the quality of life and life expectancy of many patients affected by
`thalassemia [13], by preventing the complications of iron overload [73].
`
`4. Development of non toxic oral iron chelators
`
`Despite the well-acknowledged successes of desferrioxamine in the medical
`treatment of iron overload in thalassemic patients, long-term compliance with a
`regimen of prolonged nightly infusion may be problematic for many patients in the
`developed world [63], while in developing countries such a regimen is impractical
`and unaffordable [28]. On this basis, the search for an orally-active iron chelator as
`an alternative to desferrioxamine in the treatment and prevention of chronic iron
`overload has continued in recent years. To this end, chemists have used their
`knowledge to synthesize a wide variety of chelators often borrowing from examples
`of nature [44]. Desirable properties for an oral iron chelator are:
`1. specificy and affinity for iron;
`2. molecular weight lower than 400 Da, for gastrointestinal absorption;
`3. sufficient lipophilicity for gastrointestinal absorption and intracellular chelation;
`4. sufficient hydrophilicity, to limit liver absorption.
`In the past 20 years a large variety of ligands with oxygen binding groups, above
`all those containing hydroxamates, catechols and hydroxypyridinones has been
`systematically tested as potential orally active drugs. Some of these do not present
`proper pM values for iron chelation (pM was defined by Harris et al. [74] as the
`negative logarithm of free iron(III) concentration in a solution that is 10 mM in
`ligand and 1 mM in metal at pH 7.4. The pM concept is now of common use to
`evaluate the strength of chelates taking into account proton competition). Other
`ligands, while promising from a chemical point of view later proved toxic and could
`not be introduced in clinical practice. Many authors first focused on catecholate
`sequestering agents, which resulted in stronger iron chelators than desferrioxamine
`[75]. Unfortunately, no catechol ligands were found to be orally active and some of
`
`
`8 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`299
`
`them strongly promoted the growth of pathogenic microorganisms, precluding
`designs based on these ligands [76]. Among the molecules showing the more
`interesting properties as oral chelators are HBED [77] ant its derivative dimethyl-
`HBED [78], pyridoxal isonicotinoyl hydrazone [79], desferrithiocin [80] and hydrox-
`ypyridinones [81].
`
`HBED, an analogue of EDTA with two phenolate replacing two carboxylate
`groups shows a log K value much higher than EDTA (36.7 with respect to 25.1);
`nevertheless this value is mitigated by its strong affinity for hydrogen ion resulting
`in a pM 26.74 with respect to 22.3 for EDTA. While it is well absorbed orally and
`it is effective in rodent assays its response in primates and in patient trials was
`remarkably less effective [82]; its dimethyl derivative is now under clinical trials [83].
`
`Due to the high activity of PIH [79] and of its analogues in mobilising iron, their
`chelating properties were extensively studied by Vitolo et al. [84]: the pM values for
`these ligands indicate that they are thermodynamically able to mobilise transferrin-
`bound iron, but a kinetic barrier however inhibits their exchange properties. At any
`rate the response in clinical tests was discouraging under different points of view
`[82].
`
`Desferrithiocin, isolated from S. antibioticus, is one of the few siderophores not
`belonging to hydroxamate or catecholate classes and it forms a stable 2:1 complex
`[85] with iron (K=4×1029 M − 1). This compound proved effective in iron mobili-
`sation both in rat and in primate models but animals exposed to it presented with
`nephrotoxicity [86]. In any case because of the strong chelating properties of
`desferrithiocin investigation is yet performed on its analogues [87].
`Despite their favourable iron binding properties none of the molecules presented
`above has proven completely satisfactory.
`Considering both their binding properties and the results of biological trials
`hydroxypyridinones appear to be the most promising oral chelators; their func-
`tional group was isolated in siderophores from a culture of Pseudomonas alcaligenes
`in 1979 by Barker et al. [88]. Among 1-hydroxy-2-pyridinone, 3-hydroxy-2-pyridi-
`
`
`9 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`300
`
`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`none and 3-hydroxy-4-pyridinone studied by Scarrow [81] with pM values 22.3,
`21.2 and 25.7, respectively,
`
`the third presents the most interesting properties and its 1,2-dimethyl derivative,
`known as deferiprone (L1 or CP20) is at the present the orally active iron chelating
`agent with the broadest clinical experience [63].
`
`4.1. Deferiprone
`
`Deferiprone [29] is a member of the family of hydroxypyridinones, of molecular
`weight 139 Da. By virtue of its low molecular weight, deferiprone possesses high
`absorption efficiency and it is efficiently absorbed from the human intestinal tract
`[32].
`
`Deferiprone was originally synthesized by Robert Hider and his colleagues at Essex
`University, patented in 1982 as an alternative to desferioxamine in the treatment of
`iron overload [89] and the early biological assessments were performed at Univer-
`sity College Hospital in London [36]. Deferiprone, in common with all hydrox-
`ypyridinones, forms 5-membered chelate rings in which iron is bound by two
`oxygen atoms [76]. Little is known about mobilization of iron stored in hepatocytes
`and in Kupffer cells by deferiprone. Deferiprone,
`in common with other low
`molecular weight iron chelators, likely removes Fe(III) from ferritin by penetration
`of the protein shell [90]. In other studies, it has been shown that deferiprone may
`mobilize iron even from hemosiderin, lactoferrin and from transferrin [91]. De-
`feriprone, like other bidentate hydroxypyridones, forms a 3:1 complex with iron; its
`efficiency in chelating iron, determined in iron-loaded monkeys, is significantly less
`than that observed in the same animals with parenteral administration of desfer-
`rioxamine [82]. Deferiprone is metabolized in the liver, where it undergoes rapid
`conversion to nonchelating metabolites. The major metabolite of deferiprone in
`man is the glucuronide which, as a result of conjugation of the 3-hydroxyl function,
`is unable to bind iron [92]. The majority of deferiprone–iron complex is excreted in
`the urine (70%), while little iron is excreted in the feces [93]. Although deferiprone,
`such as other bidentate ligands, has a clear advantage over desferrioxamine with
`respect to oral viability,
`it is potentially more toxic. In fact, by virtue of its
`relatively low molecular weight,
`it may easily penetrate most cell membranes,
`
`
`10 of 20
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1026
`
`

`

`G. Faa, G. Crisponi /Coordination Chemistry Re6iews 184 (1999) 291–310
`
`301
`
`blood–brain barrier and placental barrier. The efficacy of deferiprone in inducing
`and maintaining a negative iron balance in iron loaded transfused patients has been
`demonstrated by the first long-term trials. Iron chelation therapy with deferiprone
`in a large series of subjects affected by thalassemia intermedia showed a reduction
`of liver iron stores and a normalization of serum ferritin levels [94]. This observa-
`tion was followed by the report of a significant decline in liver iron concentration
`in patients affected by thalassemia major, during a 5-year treatment with de-
`feriprone [95]. In b-thalassemic patients who reach adolescence and adulthood, the
`heart has become the core of medical interest, since the main cause of death in adult
`b-thalassemia patients

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket