throbber
revIew
`
`Iron overload thalassemic cardiomyopathy: Iron
`status assessment and mechanisms of mechanical
`and electrical disturbance due to iron toxicity
`Suree Lekawanvijit MD1,3, Nipon Chattipakorn MD PhD2,3
`
`s lekawanvijit, n Chattipakorn. iron overload thalassemic
`cardiomyopathy: iron status assessment and mechanisms of
`mechanical and electrical disturbance due to iron toxicity. Can J
`Cardiol 2009;25(4):213-218.
`
`Patients with thalassemia major have inevitably suffered from complica-
`tions of the disease, due to iron overload. Among such complications,
`cardiomyopathy is the leading cause of morbidity and mortality (63.6% to
`71%). The major causes of death in this group of patients are congestive
`heart failure and fatal cardiac tachyarrhythmias leading to sudden cardiac
`death. The free radical-mediated pathway is the principal mechanism of
`iron toxicity. The consequent series of events caused by iron overload lead
`to catastrophic cardiac effects. The authors review the electrophysiological
`and molecular mechanisms, pathophysiology and correlated clinical
`insight of heart failure and arrhythmias in iron overload thalassemic car-
`diomyopathy.
`
`Key Words: Cardiomyopathy; Cardiovascular death; Iron overload;
`Thalassemia
`
`Thalassemia is the most common monogenic disorder causing
`
`decreased globin, a protein composition of hemoglobin synthesis.
`By clinical manifestation, beta-thalassemia major (TM) is the most
`severe form apart from hemoglobin Bart’s disease, which is always
`fatal. TM patients require intensive blood transfusions due to severe
`anemia from ineffective erythropoiesis. Generally, an increase in body
`iron burden occurs in patients who are not receiving transfusions,
`ranging from 2 g to 5 g/year, compared with 0.0015 g/year in healthy
`individuals (1). Regular transfusions may double this rate of iron accu-
`mulation (2). Hence, the inevitably pursuant complications are from
`iron excess in various organs such as the heart, liver and pancreas.
`Although the heart is not the first target organ, cardiac iron overload
`or iron overload cardiomyopathy is regarded as the most serious condi-
`tion (3,4). In the present review, clinical manifestation of iron over-
`load thalassemic cardiomyopathy and the tools used for its detection
`and monitoring are presented. Mechanisms by which iron toxicity
`causes alterations in cardiomyocytes and cardiac electrophysiology are
`also reviewed and discussed.
`
`CliniCal insight
`The incidence of iron overload cardiomyopathy ranges from 11.4%
`to 15.1% in TM (3,5). Usually it begins at an accumulation of 20 g
`of iron (6). In the early stage, patients are usually asymptomatic.
`Restrictive cardiomyopathy usually occurs before dilated cardiomyo-
`pathy (7), in accordance with diastolic dysfunction, which normally
`happens before systolic dysfunction and overt heart failure (8-12).
`
`la myocardiopathie thalassémique par surcharge
`en fer : l’évaluation de la quantité de fer et des
`mécanismes de perturbation mécanique et
`électrique causés par la toxicité ferreuse
`
`Les patients atteints de thalassémie majeure ont inévitablement souffert de
`complications de la maladie en raison d’une surcharge en fer. Parmi ces
`complications, la myocardiopathie est la principale cause de morbidité et
`de mortalité (63,6 % à 71 %). Les principales causes de décès au sein de ce
`groupe de patients sont l’insuffisance cardiaque congestive et les
`tachyarythmies cardiaques fatales. La voie à médiation par radicaux libres
`est le principal mécanisme de toxicité ferreuse. La série d’événements
`causés par la surcharge en fer a des effets cardiaques catastrophiques. Les
`auteurs ont examiné les mécanismes électrophysiologiques et moléculaires,
`la physiopathologie et les aperçus cliniques connexes d’insuffisance
`cardiaque et d’arythmie en cas de myocardiopathie thalassémique par
`surcharge en fer.
`
`Generally, once the onset of a failing heart occurs, the survival time
`is usually less than three months if left untreated (13). Autopsy
`examinations have found dilated cardiomegaly in patients who died
`from late-stage iron overload cardiomyopathy (7). Although systolic
`dysfunction becomes obvious during the late stage, decreased con-
`tractile function has been demonstrated during the early stage of the
`disease (14-16). Left-sided heart failure is clinically more common
`than right-sided heart failure (12). However, it has been shown that
`right ventricular dysfunction develops earlier in asymptomatic TM
`patients (9,17).
`In addition to congestive heart failure, another major cause of
`death in this group of patients is cardiac tachyarrhythmias, which may
`occur simultaneously with a failing heart, leading to sudden cardiac
`death (12). Kremastinos et al (12) reported that the incidence of sud-
`den death was approximately 11.6% in TM patients with left ven-
`tricular failure, which accounted for approximately 18.5% of total
`cardiac deaths. Similar to mechanical dysfunction, electrophysiologi-
`cal dysfunction varies with the stage of disease. Findings in the early
`stage are usually accidental, including bradycardia, ST-T changes,
`infrequent premature atrial or ventricular contractions, first-degree
`atrioventricular block and evidence of left ventricular hypertrophy
`(18,19). In the late stage, frequent premature atrial or ventricular
`contractions, short runs of supraventricular tachycardia, atrial flutter
`and fibrillation, ventricular tachycardia and second-degree or com-
`plete heart block (including intraventricular block) have been dem-
`onstrated (7,12,19). Among these late electrocardiogram (ECG)
`
`1Department of Pathology; 2Department of Physiology; 3Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai
`University, Chiang Mai, Thailand
`Correspondence: Dr Nipon Chattipakorn, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University,
`Chiang Mai 50200, Thailand. Telephone 66-53-945-329, fax 66-53-945-368, e-mail nchattip@mail.med.med.ac.th
`Received for publication November 1, 2006. Accepted March 4, 2007
`
`Can J Cardiol Vol 25 No 4 April 2009
`
`©2009 Pulsus Group Inc. All rights reserved
`
`Apotex Tech.
`Ex. 2030
`
`213
`
`

`

`Lekawanvijit and Chattipakorn
`
`changes, frequent premature ventricular contraction is most com-
`monly found, while sustained ventricular tachycardia is predominantly
`related to cardiac death (12). Iron levels in the myocardium seem to
`be associated more with arrhythmias and conduction disturbance than
`with the conduction system itself (20,21). It has been shown that
`patients with supraventricular arrhythmias have extensive iron depo-
`sition in the atria, and not in the sinoatrial node (20). In addition,
`patients with atrioventricular block did not have iron deposition in
`the ventricular conduction system (20).
`
`assessment and marKers of
`CardiaC iron status
`Because chelation therapy is an effective way of preventing iron over-
`load cardiomyopathy, the methods and/or markers that can accurately
`assess cardiac iron status and predict the development of cardiac com-
`plications are essential (8,9,11,12,15,16,22-26). Many methods and
`parameters, both direct and indirect, have been investigated. Some
`have been proven to be nonreproducible, some controversial but still
`used, and others are undergoing investigations or being used for
`research work.
`Among serum iron markers, serum ferritin is most commonly used
`as an indirect estimate of body iron store. However, reliance on ferri-
`tin alone can lead to an inaccurate assessment (24). Certain condi-
`tions, such as inflammation, infection and chronic liver disease from
`the hepatitis C virus, which are common complications of thalas-
`semia, may cause an elevation of serum ferritin (27). In view of cardiac
`complications, serum ferritin concentrations have been shown to cor-
`relate poorly with all stages of cardiac dysfunction (diastolic, systolic
`and overt heart failure) (15). Non-transferrin-bound iron (NTBI)
`seems to be the best parameter among serum iron markers. Currently,
`however, it is commonly used only in research studies. Various meth-
`ods and protocols used in the determination of NTBI have been pro-
`posed, but no consensus has been established (28-32).
`Transfusional iron load or the number of blood transfusions is usu-
`ally unreliable in predicting cardiac iron status (25). However, in the
`case of accurate data collection, an increased ratio of the transfusional
`iron load to deferoxamine use (not transfusional iron load alone) has
`been shown to correlate with cardiac dysfunction and death (25).
`Hepatic iron concentration provides the most quantitative
`means of assessing the body iron burden in TM patients (33).
`However, no relationships between hepatic iron concentration and
`cardiac iron status or cardiac dysfunction, have been found
`(16,26,34). Despite being a direct estimation, endomyocardial
`biopsy has not been correlated with cardiac iron status and function,
`possibly due to the pattern of iron deposition and the large sampling
`variation (35). Iron usually deposits primarily in the subepicardial
`layer toward the subendocardial layer and is often patchy (20,36). It
`tends to accumulate in myocardium (especially interventricular sep-
`tum and ventricular free wall) more often than in the conduction
`system. Moreover, clinically, antemortem arrhythmias and conduc-
`tion disturbance have shown no correlation with iron density in the
`conduction system (21). Other assessments, such as antioxidant
`levels – particularly vitamin E, which combats oxidative stress from
`iron toxicity – have been proposed as being indicative of iron over-
`load cardiomyopathy in TM patients (37).
`
`assessment for the development of
`iron overload Cardiomyopathy
`Accurate assessments of cardiac dysfunction and/or cardiac iron sta-
`tus are currently based on imaging techniques. The T2-star magnetic
`resonance (MR-T2*), a novel tool, has been demonstrated to evalu-
`ate cardiac iron status accurately and detect an early global ventricu-
`lar dysfunction (16). In addition, it can be used for monitoring
`myocardial iron levels during iron chelation therapy (22). While
`conventional standard echocardiographic measurements usually
`demonstrate positive findings at the late stage (38), low-dose
`
`214
`
` dobutamine stress echocardiography may be useful for early detection
`of cardiac dysfunction in patients at risk for cardiac hemosiderosis
`(11). Recently, tissue Doppler echocardiography (8) and radionuclide
`angiography (with exercise or low-dose dobutamine stimulation) (9)
`have been shown to detect regional wall motion abnormality, even in
`early-stage thalassemic patients. This finding may also reflect patchy,
`nonhomogeneous deposition of iron in cardiac muscle. However,
`stress echocardiography, tissue Doppler echocardiography and radio-
`nuclide angiography cannot estimate cardiac iron content. MR-T2*
`seems to be superlative (but costly) in terms of noninvasiveness, ear-
`liness (sensitivity) and accuracy (specificity), due to its ability to
`simultaneously assess both cardiac iron and structure, as well as car-
`diac function.
`Although cardiac dysfunction is more clinically obvious than con-
`duction disturbance or arrhythmias in iron-overloaded patients, the
`pathogenesis of arrhythmias may start at the same time, with, or even
`earlier, than contractile dysfunction (39). It has been shown that
`when patients have symptoms of arrhythmias, the mortality rate is
`potentially high (12). Thus, any method or parameter that can deter-
`mine arrhythmogenic risk would be a very useful tool as a guide for
`future therapeutic strategies. Although ECG findings, including 24 h
`Holter monitoring, during the early stage of the disease may trigger
`awareness, they are not specific to iron overload cardiomyopathy.
`Another ECG parameter, QT dispersion, has been proposed to be a
`relevant parameter for the heterogeneity of iron deposition and dura-
`tion of cardiac action potential (23). Kuryshev et al (23) found that
`14 of 24 TM patients with iron overload had increased QT dispersion
`(longer than 60 ms) in their preliminary study.
`Abnormal neurohormonal regulation has been investigated and
`proposed to be a potential contributor to cardiac disease in TM
`patients. Altered sympathovagal balance in TM patients was first
`demonstrated by Veglio et al (18). They investigated heart rate vari-
`ability (HRV) and blood pressure variability in nine asymptomatic
`TM patients and found diminished sympathetic activity. This was
`characterized by the lack of a circadian rhythm of blood pressure and
`heart rate; decreased short-term variability of blood pressure and
`heart rate, particularly at a low frequency range; reduced low to high
`frequency ratio of HRV, suggesting an impaired sympathovagal bal-
`ance; impaired baroreflex gain during a head tilt; and significantly
`decreased plasma norepinephrine. A recent study in iron-overloaded
`rats (40) demonstrated similar findings on baroreflex function.
`Moreover, Franzoni et al (41) demonstrated a significant reduction of
`HRV parameters and increased incidence of ventricular late potential
`(VLP) in asymptomatic TM patients (n=19). In this study, TM
`patients with VLP showed a higher incidence of premature ventricu-
`lar contraction, with episodes of nonsustained ventricular tachycar-
`dia. Recently, De Chiara et al (9) reported that decreased HRV
`parameters can be detected as early as ventricular regional wall
`motion in asymptomatic TM patients (n=20). However, no long-
`term follow-up study has been reported to confirm the validity of
`HRV parameters for predicting cardiac dysfunction and mortality in
`iron overload thalassemic patients.
`In TM patients, renin-angiotensin activity has been shown to
`increase (40). An increased atrial natriuretic peptide level has also been
`proposed as a predictor in asymptomatic TM patients, because it was
`found to be in close correlation with left ventricular diastolic dysfunc-
`tion (42). Taken together, impaired sympathovagal balance and altered
`hormonal regulation may be responsible for the pathophysiology of TM
`patients, who are at risk for iron overload cardiomyopathy.
`
`meChanisms of iron toxiCity
`free radical-mediated iron toxicity
`Tissue toxicity by iron occurs via the free radical-mediated pathway.
`Iron has the capacity to accept and donate electrons, switching
`between Fe2+ and Fe3+, which is known as redox activity. In general, it
`is physiologically essential and present in the plasma in a transferrin-
`bound form. In chronic iron overload, when transferrin is completely
`Apotex Tech.
`Ex. 2030
`
`Can J Cardiol Vol 25 No 4 April 2009
`
`

`

`saturated, NTBI is found in the plasma (43), which is the form of iron
`that causes toxicity. Iron excess in various conditions is harmful by
`engaging in the Fenton-catalyzed Haber-Weiss reaction to yield
`hydroxyl radicals from hydrogen peroxide and superoxide (44). They
`are all called oxygen free radicals. Highly reactive hydroxyl radical
`production, previously demonstrated in an in vivo model of
`iron-overloaded rats (45), is capable of widespread damage to cellular
`lipids, proteins, sugar and DNA (46).
`
`pathways of iron entry into cardiomyocytes
`In in vitro studies using iron-treated, cultured neonatal rat ventricular
`myocytes (NRVMs), NTBI had more than a 300-fold higher rate of
`uptake than transferrin-bound iron (47). The enhancing rate of NTBI
`uptake occurs in a dose- and time-dependent pattern (34), suggesting
`a carrier- or channel-mediated pathway.
`The exact iron uptake pathway is still unclear. The redox form of
`iron uptake is most likely specific to the reduced Fe2+ form, but not
`Fe3+ (48,49). Using the isolated adult rat heart to study iron uptake,
`Tsushima et al (49) demonstrated that iron uptake was diminished
`when cardiac electrical activity was arrested by potassium chloride.
`This finding suggests that iron uptake partly depends on membrane
`potential, indicating that iron enters the cell via voltage-gated chan-
`nels. Many studies have indicated that the sarcolemmal L-type Ca2+
`channel is a major pathway of iron uptake into cardiomyocytes
`(49-51). Using the patch clamp technique, it has been shown that the
`iron current competes with the calcium current, and is inhibited by a
`calcium channel blocker (49). Other in vivo (chronically iron-loaded
`adult mice) studies have demonstrated the inhibitory effect of L-type
`Ca2+ channel blockers on cardiac iron uptake, indicated by decreased
`tissue iron content (50,51), preserved cardiac function and improved
`survival (51). However, using iron-loaded cultured NRVMs, Parkes
`et al (48) demonstrated contradictory results. They reported that iron
`uptake did not occur via the L-type Ca2+ channel, but was preferably
`dependent on the altered redox (Fe2+ to Fe3+) pathway situated on the
`plasma membrane. The discrepancy in these findings may be due to
`differences in the age of animal models. Neonatal myocytes have
`fewer L-type Ca2+ channels with a smaller L-type Ca2+ current than
`adult myocytes (52). It has been proposed that the Ca2+ current inves-
`tigated in cultured neonatal myocytes may be underestimated due to
`less density in beating cells and more myocyte death (52).
`
`evidenCe of iron-mediated
`CardiomyoCyte damage
`In chronic iron overload, iron toxicity is dose-dependent (53). The
`highly toxic hydroxyl radicals, produced via the Fenton-catalyzed
`Haber-Weiss reaction in the setting of iron excess, is well known to
`damage the lipid-rich cell membrane, which is called lipid peroxida-
`tion, or lipoperoxidation. Lipid peroxidation in the presence of ade-
`quate lipids (substrate) and iron (catalyst) seems to be perpetual. This
`reaction is not only restricted to cell membrane phospholipids, but
`also to other membrane-bound cellular organelles. By studying iron
`overload in cultured NRVMs, Link et al (54) demonstrated evidence
`of lipid peroxidation, reduction in polyunsaturated fatty acids (PUFAs)
`and increased products from cellular lipid peroxidation, particularly
`toxic aldehydes (47,53,55,56). The aldehyde products such as malon-
`dialdehyde and 4-hydroxynonenal can form a covalent link to pro-
`teins (aldehyde-protein adducts), rendering the loss of cellular protein
`function (57).
`Loss of cell membrane integrity from lipid peroxidation was
`evidenced by lactate dehydrogenase release (58). As a result, struc-
`tures located on the cell membrane, such as Na+-K+ ATPase and
`5′-nucleotidase, were affected thereafter (59). Oxidative stress-
`mediated iron tocixity also affects other cellular organelles and their
`functions, such as increased lysosomal fragility (60), decreased mito-
`chondrial inner membrane respiratory enzyme activity and ATP (61),
`decreased protective antioxidant enzyme (ie, glutathione peroxidase)
`
`Can J Cardiol Vol 25 No 4 April 2009
`
`Thalassemic cardiomyopathy
`
`activity, decreased myofibrile elements and a decreased number of
`mitochondria (36).
`
`meChanisms of meChaniCal
`disturbanCe in iron overload
`Cardiomyopathy
`Cardiac effects following iron-induced membrane lipid peroxidation
`can be divided into two parts – changes following altered membrane
`lipid components and their metabolites, and changes following altered
`embedding enzymes, ion channels, receptors and other membrane
`proteins. Arachidonic acid (AA) is a 20-carbon PUFA normally
`esterified in membrane phospholipids. It is released through the action
`of cellular phospholipases, which may be activated by chemical stimuli
`or other mediators. AA metabolites, also called eicosanoids, are lipid
`mediators involved in the signal transduction pathway. Both AA and
`eicosanoids have been shown to play a role in iron overload cardio-
`myopathy (62). In cultured NRVMs treated with iron, Mattera et al
`(62) have shown that the rate of AA release is increased, particularly
`after the angiotensin II type I receptor has been activated. AA has
`been shown to activate mitogen-activated protein kinase (63) and
`apoptosis (64). Because the activated angiotensin II type I receptor is
`associated with cardiomyocyte growth and hypertrophy (65,66), the
`outweighing increment of AA release may be a possible mechanism in
`the progression of cardiac hypertrophy to heart failure in iron overload
`cardiomyopathy (62). In addition, cyclooxygenase-2 (COX-2) activ-
`ity also increased and may be responsible for heart failure development
`in iron overload; it is associated with an increased apoptotic rate and
`symptomatic heart failure (67,68).
`Other than the effects of membrane lipid peroxidation causing cel-
`lular environmental changes, the membrane-embedding enzymes, such
`as Na+-K+ ATPase, Ca2+ ATPase and 5′-nucleotidase, may be attacked
`directly by oxygen radicals (59,69,70). It has been shown that modifica-
`tion of the sulfhydryl groups of these enzymes by an oxygen radical
`reduces their activity (70). Alteration of sarcolemmal Na+-K+ ATPase
`activity in an iron-overloaded heart may be one of the important
`mechanisms in the pathophysiology of the disease. A decrease in
`Na+-K+ ATPase activity was shown in both an iron-loading NRVMs
`(59) and a hydroxyl radical-exposed heart (69). Impaired Na+-K+
`ATPase activity renders an increased resting membrane potential
`(membrane depolarization) and changes in ion conductance. Altered
`Na+-Ca2+ exchange can occur following impaired Na+-K+ ATPase
`activity. An increase in intracellular Na+, due to an impaired Na+-K+
`ATPase pump, shifts the Na+ efflux via Na+-Ca2+ exchange, resulting in
`Ca2+ influx. This Ca2+ influx may persist due to impaired Ca2+ extrusion
`caused by iron-inhibited sarcolemmal Ca2+ pump activity and may lead
`to cardiac arrhythmias (70). Furthermore, Ca2+ influx normally causes a
`release of Ca2+ from the sarcoplasmic reticulum (SR), leading to myo-
`cardial contraction. However, iron (Fe2+) itself has a direct inhibitory
`action on ryanodine-sensitive Ca2+ channels on the SR by competing
`with Ca2+, thus contributing to a reduction in Ca2+-induced Ca2+
`release from the SR (71) results in impaired contractility. Oxygen free
`radicals also have a direct effect on decreased cardiac SR Ca2+ ATPase
`activity (72), which may be responsible for diastolic failure. In the
`in vitro study by Zeitz et al (69), hydroxyl radical-exposed rabbit heart
`muscle was demonstrated to have calcium overload (via Na+-Ca2+
`exchange), with an acute diastolic dysfunction. These mechanisms may
`explain the pathophysiology of early cardiac dysfunction in iron over-
`load cardiomyopathy.
`Persistently high iron and free radicals from iron overload also
`affect other cellular constituents, such as altered function (73), struc-
`ture and number of mitochondria (36,74), altered SR function and
`structure, and decreased myofibril elements (36). Mitochondrial dys-
`function results in decreased phospholipid synthesis of all cellular
`membranes, including the mitochondria themselves (75). SR mem-
`brane destruction from lipid peroxidation, together with impaired
`phospholipid synthesis, leads to calcium leakage into the cytoplasm.
`
`Apotex Tech.
`Ex. 2030
`
`215
`
`

`

`Lekawanvijit and Chattipakorn
`
`Further increaseses in intracellular calcium can cause degradation of
`membrane phospholipids by activating endogenous phospholipase and
`the degradation of myofibril elements by activating proteases (76).
`The net effect is toward the end stage of cellular declination, eventu-
`ally leading to systolic dysfunction.
`
`meChanisms of eleCtriCal disturbanCe
`in iron overload Cardiomyopathy
`In ex vivo guinea pig hearts, delayed and blocked electrical conduction
`has been demonstrated to occur earlier in iron-loaded cardiac toxicity
`than impaired contractility (39). An optical mapping study of iron-
`loaded Mongolian gerbils (77) also exhibited pertinent results of slow
`conduction and impulse block, with evidence of re-entry, which may
`lead to cardiac arrhythmias. Iron-overloaded cardiomyocytes have been
`shown to have a smaller overshoot potential and a shorter action poten-
`tial duration than iron-free cardiomyocytes in the same heart (23,58).
`An alteration of ion currents characterized by reduced Na+ currents may
`be an underlying mechanism (23). The severity of Na+ current reduc-
`tion is also associated with higher dose and longer duration of iron
`exposure (23). Reduced overshoot potential ensues as a result of
`decreased rapid phase 0 depolarization (fast sodium current). A reduc-
`tion in the late fast sodium current during the plateau phase may result
`in the rapid shortening of the action potential duration due to the dis-
`turbance of a delicate balance of small currents. Abnormal sodium cur-
`rents may also explain the delayed impulse conduction causing widening
`of the QRS complex, as demonstrated in the intact gerbil heart model
`(77). These electrophysiological heterogeneities, including the patchy
`nature of cardiac iron deposition, may provide substrates for re-entry
`and risk of developing fatal arrhythmias (77). Other arrhythmias
`include PR and QRS interval prolongation (65) and bradycardia in the
`early stage (34,66), and premature ventricular contraction (40), ST-T
`changes (66), QT prolongation (66), second- and third-degree atrio-
`ventricular block (65), and arrhythmias in the late stage (59). These
`changes are similar to those reported in iron overload patients (7,19).
`Further investigations on alterations of ion channels, both quantita-
`tively and structurally, are required to explain the association of molecu-
`lar and electrophysiological changes at the cellular level.
`Changes in calcium homeostasis, as described in the ‘Mechanisms
`of Mechanical Disturbance in Iron Overload Cardiomyopathy’ sec-
`tion, may also lead to cardiac arrhythmias, such as triggered activity
`and sudden cardiac death (78). In addition, increased COX-2 activity,
`due to iron overload, results in enhanced eicosanoid production and
`an altered ratio of eicosanoid products (62). An increased ratio of
`prostaglandin E2 to prostacyclin found in an iron overload condition
`can trigger tachyarrhythmias (79,80). This phenomenon can also
`occur when being stimulated by interleukin-1 alpha, an inflammatory
`cytokine (62). Interleukin-1 alpha-related eicosanoid production
`causing fatal arrhythmias may be a hidden mechanism responsible for
`sudden cardiac death in iron overload thalassemic patients, who die
`suddenly from overwhelming infection (7).
`
`possible future therapeutiC
`modalities
`Iron chelation therapy is accepted worldwide as the most effective
`treatment for iron overload cardiomyopathy. In some cases, iron
`chelation therapy is not suitable due to its high cost, poor compliance
`(because of the route of drug administration) and some adverse effects.
`Calcium channel blockers may be useful in preventing NTBI from
`entering cardiomyocytes via the L-type Ca2+ channel (49-51). Oudit
`et al (51) demonstrated that amlodipine and verapamil reduced intra-
`cellular iron accumulation and oxidative stress without disturbing
`diastolic and systolic function. L-type Ca2+ channel blockers affect
`not only cardiomyocytes, but also pancreatic beta cells and anterior
`pituitary cells, which are important target cells for iron toxicity (52).
`On the other hand, because the pathophysiology of iron overload
` cardiomyopathy depends mainly on oxidative injury, antioxidant
`
`216
`
` therapy is possibly one of the best alternative choices. Vitamin E has
`been demonstrated to be effective for this therapeutic purpose
`(37,54,55). However, vitamin C is not useful because it can promote
`iron toxicity by reducing iron redox status (Fe3+ to Fe2+) and enhanc-
`ing cellular uptake, which may worsen cardiac dysfunction in patients
`at risk for iron overload cardiomyopathy (55).
`Omega-3 PUFAs, abundant in fish oil, have been known for their
`cardioprotective effects (81). Although they are substrates in lipid
`peroxidation, the by-products of lipid peroxidation were not increased
`after omega-3 PUFA supplement (82). However, in intensive oxida-
`tive stress due to iron overload, omega-3 fatty acid dietary supplement
`should be used with caution, unless the antioxidant level is adequate
`(83). Using selective COX-2 inhibitors when an anti-inflammatory
`drug is required in these patients may be beneficial in reducing the
`proarrhythmic effect of the eicosanoid product (prostaglandin E2)
`(62). Further studies of COX-2 inhibitor treatment in iron overload
`cardiomyopathy may be worthwhile.
`Natural products have played a role as alternative therapeutic
`modalities in many diseases, including iron overload thalassemia.
`Curcumin, a constitutent of a type of spice, has been demonstrated to
`have antioxidative (84,85), anti-inflammatory (84) and iron-chelating
`effects (86). These beneficial effects may provide vast benefits to iron
`overload TM patients. However, more interventional studies in ani-
`mal models and humans are needed to confirm their beneficial role in
`iron-overload thalassemic cardiomyopathy.
`
`ConClusion
`The most common cause of morbidity and mortality in TM patients is
`iron overload cardiomyopathy, in which the exact pathophysiology is
`crucial for proper management. NTBI has been accepted as the key
`culprit. Recently, the L-type calcium channel was strongly proposed as
`the pathway of intracellular NTBI uptake in cardiomyocytes.
`Subsequent cellular damage occurs mainly via the free radical-mediated
`mechanism, in which iron acts as a catalytic agent. Lipid peroxidation
`seems to play the most important role for both heart failure and
`arrhythmias, particularly by destroying membrane-embedding enzymes
`(such as ATPase) and being the source of AA metabolites.
`For clinical management, early detection is the goal in asymptom-
`atic patients. Cardiac MR-T2*, tissue Doppler echocardiography,
`radionuclide angiography and stress echocardiography are useful imag-
`ing studies for detecting early cardiac dysfunction. Similarly, many
`electrophysiological parameters, such as decreased HRV, presence of
`VLP and impaired baroreflex function, seem to be excellent predic-
`tors, especially for arrhythmias. In addition, several electrophysiologi-
`cal parameters that have been documented as risk stratifiers of other
`cardiovascular diseases (ie, heart rate turbulence) may be applied to
`iron overload cardiomyopathy. Future investigations of large popula-
`tions with long-term follow-up are needed to warrant the clinical
`benefits in iron overload cardiomyopathy.
`
`funding: This work is supported by the Thailand Research Fund
`grant RMU 4980001 (NC).
`
`referenCes
`1. Pippard MJ, Callender ST, Warner GT, Weatherall DJ. Iron
`absorption and loading in beta-thalassaemia intermedia. Lancet
`1979;2:819-21.
`2. Olivieri NF. The beta-thalassemias. N Engl J Med 1999;341:99-109.
`(Erratum in 1999;341:1407).
`3. Borgna-Pignatti C, Rugolotto S, De Stefano P, et al. Survival and
`disease complications in thalassemia major. Ann NY Acad Sci
`1998;850:227-31.
`4. Zurlo MG, De Stefano P, Borgna-Pignatti C, et al. Survival and
`causes of death in thalassaemia major. Lancet 1989;2:27-30.
`5. Li CK, Luk CW, Ling SC, et al. Morbidity and mortality patterns
`of thalassaemia major patients in Hong Kong: Retrospective study.
`Hong Kong Med J 2002;8:255-60.
`Apotex Tech.
`Ex. 2030
`
`Can J Cardiol Vol 25 No 4 April 2009
`
`

`

`6. Walker JM. The heart in thalassaemia. Eur Heart J 2002;23:102-5.
`7. Engle MA, Erlandson M, Smith CH. Late cardiac complications of
`chronic, severe, refractory anemia with hemochromatosis.
`Circulation 1964;30:698-705.
`8. Vogel M, Anderson LJ, Holden S, Deanfield JE, Pennell DJ,
`Walker JM. Tissue Doppler echocardiography in patients with
`thalassaemia detects early myocardial dysfunction related to
`myocardial iron overload. Eur Heart J 2003;24:113-9.
`9. De Chiara B, Crivellaro W, Sara R, et al. Early detection of cardiac
`dysfunction in thalassemic patients by radionuclide angiography
`and heart rate variability analysis. Eur J Haematol 2005;74:517-22.
`10. Spirito P, Lupi G, Melevendi C, Vecchio C. Restrictive diastolic
`abnormalities identified by Doppler echocardiography in patients
`with thalassemia major. Circulation 1990;82:88-94.
`11. Suarez WA, Snyder SA, Berman BB, Brittenham GM, Patel CR.
`Preclinical cardiac dysfunction in transfusion-dependent children
`and young adults detected with low-dose dobutamine stress
`echocardiography. J Am Soc Echocardiogr 1998;11:948-56.
`12. Kremastinos DT, Tsetsos GA, Tsiapras DP, Karavolias GK, Ladis VA,
`Kattamis CA. Heart failure in beta thalassemia: A 5-year follow-up
`study. Am J Med 2001;111:349-54.
`13. Cohen AR, Galanello R, Pennell DJ, Cunnin

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