throbber
.~
`
`r··
`~ ~\ NIH Public Access
`~
`· EJJ Author Manuscript
`0/:-HEP..~ Ann A 1 .·faad Sci . .\uthor manuscript available in P].·!C 20 lO Juo..:: 28.
`Published in final edited form as.
`Ann NY Acad 9j, 2005 ; 1054: 386-395. doi:lO.l 196/annals.1345.047.
`
`Physiology and Pathophysiology of Iron Cardiomyopathy in
`Thalassemia
`
`JOHN c. woooa,b, CATHLEEN ENRIQUEZa, NILESH GHUGREa,b, MAYA OTTO(cid:173)
`DUESSELb, MICHELLEAGUILARb, MARVIN D. NELSONb, REX MOATsb, and THOMAS D.
`COATESc
`llOivision of Pediatric Cardiology, Childrehs Hospital of Los Angeles, Los Angeles, California 90027,
`USA
`
`bOepartment of Pediatric Radiology! Childrens Hospital of Los Angeles, Los Angeles, California
`90027, USA
`
`coivision of Pediatric Hematology, Childrens Hospital of Los Angeles, Los Angeles, California
`90027, USA
`
`Abstract
`Iron cardiomyopathy remains the leading cause-of death in patients with thalassemia major. Magnetic
`n•sonance imaging (:tvfRI) is ideally suited for monitoring thalassemja patients because it can detect
`cardiac iJ.nd Jiv er iron burdens as well as accurately rn easure left ventricular dimensions and function.
`However, patients with thalassernia have unique physiology that alters their normative data. In this
`article, we review the physiology and pathophysiology of thalassemic heart disease as well as the
`use ofMRI to monitor it. Despite regular transfusions, thalassemia major patients have larger
`ventricular volumes, higher cardiac outputs, and lower total vascular resistances than published data
`for healthy control subjects; these hemodynamic findings are consistent with chronic anemia. Cardiac
`iron overload increases the relative risk of further dilation, arrhythmias, and decreased systolic
`function. However, many patients are asymptomatic despite heavy cardiac burdens. We explore ·
`possible mechanisms behind cardiac iron-function relationships and relate these rn echanism s to
`clinical observations.
`
`Keywords
`iron; heart; 1v!RI; ejection fraction; cardiac function; T2*
`
`INTRODUCTION
`
`Tha!assemia, although relatively uncommon in the United States, is the most common genetic
`disease worldwide.1 With increasing East Asian immigration to the Pacific States in the last
`two decades, thalassemia major is becoming an important domestic as well as international
`health challenge. Regular transfusion therapy, while improving patient qualify of life, creates
`a state of iron overload, a second devastating disease. Once reticuloendothelial stores saturate,
`iron deposition increases in parenchymal tissues such as endocrine glands, hepatocytes, and
`
`© 2005 New York Academy of Sci.ences.
`Address for correspondence: John C. Wood, M.D., Ph.D., Division ofCardiology, Mailstop 34, CbildrensHospital of Los Angeles, 4650
`Sunset Blvd.; Los Angeles, CA 90027. Voice: 323-669-5470; fax: 323-669-731 7.jwood@chla.usc.edu.
`
`Coates
`Thursday, April 26, 2018
`
`Reported by; Elizabeth Borrelli
`CSR 7844, CCRR, CLR
`
`TAR00073173
`
`1 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`WOOD ct al.
`
`Page 2
`
`myocardium. Typically silent for many years, cardiac iron deposition produces arrhythmias,
`systolic and diastolic dysfunction, and congestive heart failure in the s~cond or third life decade.
`
`The introduction of an effective iron chelation agent, deferoxamine, transformed thalassemia
`management in the 1980s. Administered as a continuous subcutaneous infusion, 8-12 hours
`per day, 5-7 days per week, deferoxamine therapy remains an onerous lifeline for thalassemia
`patients . .Although chelation does prolong length and quality of life for thalassemia patients,
`cardiac toxicity remains the leading cause of death, generally striking patients in their third or
`fourth decade. 2 Chelation noncompliance contributes to these deaths; however, some patients
`die despite apparently adequate liver iron chelation. 3 Conventional cardiac surveillance,
`consisting of annual ECG, Holter, and echocardiogram, has proved remarkably ineffective in
`detecting preclinical cardiac iron overload. Electrocardiogram changes reflect primarily left
`ventricular hypertrophy and nonspecific ST-I wave changes from volume overload.
`Conduction abnormalities, consisting primarily of atrioventricular and bundle-branch block,
`typically present after symptomatic disease. Patients may complain of palpitations as the
`earliest clinical symptom; hence Holter monitoring has merit to document atrial and ventricular
`irritability. Iron toxicity arrhythmias are labile and often automatic rather than reentrant in
`nature, typically presenting with polymorphic atrial and ventricular arrhythmias 4 Although
`iron deposition and scarring does occur in the cardiac conduction system, deposits are not
`correlated with clinical presentation.
`
`Abnormalities of ventricular systolic function on echocardiogram are nearly universal but are
`often not detectable until patients are in overt congestive heart failure. Echocardiographic
`assessment of myocardial function may be confounded by segmental wall motion
`abnormalities. As a result, measurements of resting ejection fraction by radionucleide
`angiography and magnetic resonance imaging (:!v1RI) are more robust than echocardiography
`and are better at recognizing preclinical systolic dysfunction. 5 While systolic dysfunction
`carries a grave prognosis, patients can be "rescued" by continuous deferoxamine
`administration, provided they are willing to comply with several years of this therapy. 5' 6
`
`Although many left ventricular filling abnormalities have been described previously in
`thalassemia,7 most reports have failed to acknowledge the critical role .of chronic anemia. 8
`Thalassemia patients have elevated cardiac output and stroke volumes, leading to elevated
`mitral inflow velocities and shorter "deceleration" times, regardless of cardiac iron status. 8
`Restrictive physiology may be observed in advanced disease but is often accompanied by
`systolic dysfunction or severe pulmonary hypertension.8 Impaired myocardial relaxation,
`common in hypertensive and idiopathic hypertrophic cardiomyopathy, has not been well
`documented in thalassemic cardiomyopathy. 7 While diastolic dysfunction measures have the
`potential for earlier diagnosis of cardiac iron toxicity, standard techniques are confounded by
`sensitivity to the volume overload state (discussed in next section).
`
`NORMAL CARDIAC PHYSIOLOGY IN THALASSEMIA MAJOR
`Some of the limitations of conventional cardiac monitoring can be put in perspective by
`considering the normal cardiac physiology of thalassemia patients. Transfusion therapy was
`initiated in thalassemia major to stop the destructive effects of ineffective erythropoiesis and
`marrow expansion. Typically, this can be achieved by keeping pretransfusion hemoglobin
`levels bet\veen 9 and 10 g/dL, leaving patients with a mild chronic anemia.
`
`Because hemoglobin is responsible for oxygen transport, the body compensates for chronic
`anemia in three important ways. 9 Since oxygen delivery represents the product of cardiac
`output, hemoglobin, and hemoglobin saturation (usually >95% regardless of anemia), the body
`can compensate for low hemoglobin levels by increasing cardiac output. This measure can be
`easily documented through :MRI. Table 1 demonstrates cardiac index (cardiac output indexed
`
`Ann NY Acad Sci. Author manuscript; available in PMC 2010 June 28.
`
`TAR00073174
`
`2 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`z
`I
`J
`-0
`)>
`)>
`c:
`.-+
`:::r
`0 ...,
`
`z
`I
`I
`~
`)>
`
`c: -:::r
`
`0 ...,
`
`z -I
`
`I
`
`-0
`)>
`)>
`
`c: -'3"
`0 ...,
`s:
`Dl
`:::J c:
`
`(J)
`()
`::::!.
`"'O
`
`' -·
`
`WOOD et al.
`
`Page 3
`
`to body surface area) in 19 patients with thalassemia major compared with historical control
`subj ects. 1 O,ll Cardiac index was increased nearly 69% in the thalassetn ia patients com pared
`with control subjects, comparable to their degree of anemia, resulting in relatively normal
`oxygen delivery.
`
`Increased cardiac index c·an be achieved either by increased ventricular stroke voh:rrne index
`or by increased heart rate. We compared heart rate, cardiac index, cardiac volumes, and.ejection
`fraction measured by MRI with vital signs and hemoglobin levels measured during routine
`blood transfusion visits (Table 1). Average heart rate was comparable to that of age-matched
`control subjects. Ejection fraction was within reference limits, but end-diastolic, end-systolic,
`and stroke-volume indices were elevated compared with those of controls.
`
`Therefore, thalassem ia major represents a chronic, high-output state, produced by volume(cid:173)
`loaded ventricles rather than increased heart rate. To maintain a norm al mean systemic blood
`pressure in the presence of high cardiac output, the body would have to lower the systemic
`vascular resistance. 9 This response, similar to the physiologic compensation observed during
`exercise, occurs through peripheral arteriolar vasodilation and leads to wide pulse pressures
`and low diastolic pressures. Systolic blood pressures were comparable to those of age-matched
`control subjects; however, diastolic pressures were significantly decreased in our thalassemia
`patients (Tab1e 1).
`
`To summarize, the "normal" heart in thalassemia pumps at larger volumes (pre-load) and
`against lower peripheral resistance (afterload) than a normal heart. As a result, the expected
`cardiac parameters for non~iron-overload thalassemia major patients remains poorly
`characterized. Should the ejection fraction be higher, lower, or the same as that in an age- and
`sex-matched healthy volunteer? For now, the question remains unanswered. Understanding
`the normal physiologic baseline is critical to interpreting cardiac tests in these patients and
`understanding their response to pathologic stimuli .
`
`EARLY DETECTION OF IRON CARDIOMYOPATHY
`
`Since cardiac function remains normal until late in the spectrum of iron cardiomyopathy, other
`tools are necessary to anticipate and prevent iron cardiomyopathy. Liver iron provides a good
`index of total body iron stores, and high levels may convey future cardiac risk. 12 Although
`traditionally estimated by biopsy or SQIBD, liver iron level can now be accurately estimated
`using.MR). Iron shortens the MRI relation parameters T2 and T2* (and lengthens R2 and R2 *)
`in a predictable and reproducible manner. These, MRI techniques can be used to assess iron
`levels in the heart as well. Both myocardial T2 and T2* shorten in thalassemia patients. IO,
`13-15 Patients with a normal T2* have normal function, but the relative prevalence of
`myocardial dysfunction and arrhythmias increases with lower T2* (high iron). 10,14- 16
`Ventricular function is impaired in approximately 10% of patients having a T2* of 10 ms but
`nearly 70% for patients having a T2* of 4 ms.16 Like many other biomaikers, such as serum
`cholesterol level, abnormal T2* conveys only a relative risk; many patients with relatively high
`iron burdens are asymptomatic at the time of study. Thepredictive value of abnormalT2* is
`strongly implied but has. not yet been demonstrated.
`
`Although liver iron level has been used as a surrogate for cardiac iron form any years, 12, 17 the
`link between cardiac iron and liver iron is quite complicated. Some patients develop ventricular
`dysfunction despite low liver iron concentrations. In fact, there is little or no correlation
`between cardiac T2* (or cardiac function) and liver iron level in cross-sectional analyses_ Io,
`14,15 This observation raised concerns that cardiac T2* did not reflect cardiac iron level.
`However, recent work in animals indicates that cardiac T2 and cardiac T2* are determined
`primarily by cardiac iron concentration. 18,19 The apparent paradox between cross-sectional
`
`Ann NY Acad Sci. Author manuscript; available in PMC 2010 June 2.8.
`
`T AR00073175
`
`3 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`z
`I
`I
`'"'D
`)>
`
`l ~ -:::r
`0 ...
`
`WOOD et al.
`
`Page 4
`
`and longitudinal studies of liver iron can be understood in the context of organ-specific iron
`transport and elimination.
`
`PATHOPHYSIOLOGY OF IRON CARDIOMYOPATHY
`
`Figure 1 is a schematic illustration of the pathophysiology of iron cardiomyopathy, dividing
`the disease process into iron uptake, iron storage, and iron interactions. Characterization of
`iron transport mechanisms is impmtant because it may represent an independent therapeutic
`target to complement chelation therapy. Iron uptake occurs primarily through uptake of non,-(cid:173)
`transferrin-bound iron (NTBI). 20-23 Both ferric and ferrous ions can be absorbed in tissue
`culture, and there are membrane-bound enzymes that facilitate conversion from one species to
`the other. 20 Dimethyl transferase 1 (DMTl) levels have been implicated in intestinal iron
`transport but have not been definitively linked to cardiac iron. transport.21 L-Type voltage(cid:173)
`dependent channels (L VDCs) appear to mediate murine cardiac iron transport, accounting for
`at least half of cardiac iron uptake. 23 Interestingly, neonatal rat m yocytes do not use L VDCs,
`but it.is unclear whether this represents species or maturational specificity. 20
`
`In cell culture, NTBI uptake can be quite rapid. 22 Preexposure of m yocytes to iron increases
`their uptake rate dramatically, suggesting a positive-feedback regulatory mechanism 22,24
`Clinically, this finding indicates that relatively brief periods of very poor chelator compliance
`might lead to significant cardiac iron deposition in vulnerable individuals. Abnormal cardiac
`T2* is rarely found before the age of 10 years, even in patients with high liver iron
`concentrations. 10 However, the prevalence of abnormal T2* jumps to more than 50% in late
`adolescence and early adulthood, suggesting relatively "precipitous" iron loading. This
`transition corresponds with the most difficult years for chelation compliance, but one cannot
`exclude contributions from developmental factors such as puberty.
`
`Both the level and duration ofNTBI exposure are probably important components of cardiac
`iron uptake The level of NTBI appears to be a function of trartsferrin saturation and the liver's
`ability to buffer and store iron. Jensen and colleagues demonstrated that liver iron levels in
`excess of 19.5 mg/g led to dramatic increases in NTBI or "chelatable" iron in unchelated
`patients with myelodysplasia, 25 suggesting a "saturation" threshold for the liver. In turn, excess
`chelatable iron.was strongly correlated with cardiac iron loading by 1v1RI (signal intensity ratio
`technique). Similar cardiac-risk "thresholds" for liver iron in this range have been suggested
`by the work ofBrittenham 12 and Mariotti.44 Liver diseases, such as cirrhosis, that modify the
`liver's ability to buffer and store iron could also increase vuln:erability to extrahepatic iron
`deposition. 26
`
`Although high liver iron levels probably increase cardiac risk, low levels do not guarantee
`cardiac safety. Chronic exposure to lower levels ofNTBI may be ·sufficient for cardiac iron
`overload. Labile iron species are suppressed during deferoxamine therapy but rebound within
`a few hours of stopping infusion. 27 As a result, the hours per day of deferoxamine therapy may
`be as important to the heart as the grams per day of the drug.
`
`Once NTBI enters the m yocyte, it is rapidly buffered by ferriti.t)., limiting its potential for redox
`damage or other hannful interactions in the cell. 2s-30 Within hours, the ferritin- iron complexes
`begin to appear in intracellular siderosomes for long-tenn storage. Some studies have suggested
`a "last-in, first-out" pattern of iron accessibility 28 Although this supposition is intuitive, these
`studies are limited to relatively short-term observations in tissue culture.
`
`From a magnetic perspective, the "free-iron" species have little effect on.MRI T2 or T2* values
`at physiologic concentrations. Once bound to ferritin, iron produces greater inhomogeneities
`in the magnetic field, leading to detectable changes in T2 and T2*. However, clusters of ferritin
`molecules or their breakdown products, such as are found in siderosomes, produce much larger
`
`Ann N YAcad Sci. Author manuscript; available in PMC 2010 June 28.
`
`TAR00073176
`
`4 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`z
`::r:
`I
`"'O i
`)>
`)>
`c .....
`:::r
`0 ...,
`
`z
`::r:
`I
`"'O
`)>
`)>
`c ·
`...... .
`:::r .
`co
`...,
`
`z
`::r:
`• "'O
`
`)>
`)>
`c .......
`:::r
`0 ...,
`s: al .
`::l c
`(/)
`0
`::! . .
`"E.
`
`WOOD et al.
`
`Page 5
`
`changes (up to sixfold) in T2 or T2* for the same amount of iron than freely diffusing ferritin
`molecules. 31 Hence, lvfRI is measuring predominantly long-term storage depots of iron rather
`than the functionally active iron. This observation explains why some indiv~duals can have
`massive cardiac iron deposition without cardiac symptoms.
`
`Nonetheless, all buffering systems have limited capacity or can be disrupted by other factors.
`Once this occurs, free iron levels rise within the cell, wreaking havoc through redox reactions,
`gene modulation, and direct interaction with ion channels. 30,32-36 Through the Haber-Weiss
`reaction, iron catalyzes production of free radicals, leading to oxidative membrane damage
`throughout the cell. One membrane target is the siderosomes, increasing their fragility and
`competency. 32,37 This, in turn, could potentially lead to release of additional redox-active iron
`species, setting up the potential for a positive-feedback system. Such a phenomenon may
`explain the -catastrophic hemodynamic collapse seen in some patients.
`
`Other membranes involved include the mitochondrial membranes. Iron is avidly taken up by
`mitochondria_3o, 3s Oxidative phosphorylation is impaired, although the mechanisms of this
`disruption are not completely understood Chr.onic irnpajrrnent of mitochonc:lrial energy
`production causes dilated cardiomyopathy in many diseases and may represent a mechanism
`for the asymptomatic functional abnormalities observed in early iron cardiomyopathy.39
`
`Elevated myocyte iron levels .also lead to alterations in gene expression. 33 V/hether these
`changes represent controlled interactions through iron response elements or nonspecific effects
`from redox damage is unclear. Myocytes appear to tonically suppress fibroblast proliferation,
`but this parac1ine effect is reduced by myocyte iron loading.40 This observation represents one
`potential mechanism for iron-induced cardiac fibrosis in the tha1assemic heart.
`
`Ferrous iron has similar size and charge to that of calcium irons, the major mediator of
`excitation- contraction coupling and a major determinant of the cardiac action potential. Hence
`it is not surprising that iron overload results in arrhythmias and poor cardiac function_ 4,3s,36,
`41 Ferrous iron can directly interact with the ryanodine~sensitive calcium channel in the
`sarcoplasmic reticulum. 34 This channel is responsible for activation of contraction and also
`modulates calcium reuptake in the sarcoplasmic reticulum . Ryanodine channel dysfunction is
`the common denominator for a wide variety of congenital and acquired arrhythmogenic
`cardiomyopathies.42
`
`Intracellular iron also impairs function of membrane-bound fast-sodiwn channels as well as
`delayed-rectifier potassium currents. 36 The form er channels are responsible for the rapid
`upstroke of the cardiac action potentia I. Channel blockage or other interference will slow
`cardiac conduction, broadening the QRS of the EKG36,41 and delayed-action potential spread
`across the myocardium.41 Both calcium and potassium channel modification may be
`responsible forrepolarization abnormalities such as early or delayed afterdepolarizations and
`QTc prolongation. These changes are associated with b.oth triggered ventricular arrhythmias
`and reentrant mechanisms such as t.orsade-de-p.ointes,42
`
`Once arrhythm ias or cardiac dysfunction develops, aggressive chelation must be initiated,
`regardless of the total iron burden. The current standard of care remains continuous
`deferoxamine therapy because it provides a continuous "sink" for free iron species. 5' 6
`Continuous administration also rn ay overcome unfavorable transport kinetics .of deferoxamine
`across the myocyte membrane. Cardiac symptoms typically stabilize in a period of weeks to
`months once the "free" iron levels are consistently suppressed. Sustained recovery, however,
`often necessitates continued therapy for several years, suggesting that cardiac iron stores
`deplete more slowly. 5'6 In fact, it has been demonstrated by MRI that therate of iron elimination
`in the heart is nearly sixfold slower than that in the liver. 15,43 The rate-limiting step in cardiac
`iron excretion is not known but may reflect ferritin turnover.
`
`Ann N YAcadSci. Author manuscript; available in PMC 2010 June 28.
`
`TAR00073177
`
`5 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`WOOD et al.
`
`Page 6
`
`The asymmetry of cardiac iron loading and elimination compared with that in the liver
`effectively weakens or destroys any cross-sectional correJ::ition between liver and cardiac iron
`levels. Thus, an elevated liver iron level has no predictive value for whether the heart is
`currently iron loaded, but it may convey prospective risk for subsequent cardiac iron loading .
`.tv1RI offers a unique tool to prospectively study the interplay between hepatic and extrahepatic
`iron stores.
`
`CONCLUSION
`
`Despite transfusion therapy, thalassemia major represents a chronically anemic condition
`resulting in volume-loaded ventricles and increased peripheral vasodilation. 9 As a result, there
`is a paucity of appropriate normative data for thalassemia patients. The mechanisms and
`kinetics of iron entry and clearance differ markedly in the heart and liver, leading to a
`complicated relationship between the two parameters. 20,22,43 Increased levels of stored iron
`can be detected using MRI and are associated with increased relative risk of poor ventricular
`function. 1o,15
`
`Acknowledgments
`
`This work was rupported by the National Heart, Lung, and Blood Institute of the National Institutes of Health (1 ROI
`HL75592·01Al), the General Clinical Research Center at Childrens Hospital Los Angeks (RR0043-43), Department
`of Pediatrics at diildrens Hospital Los Angeles, and Novartis Pharma AG, Basel, Switzerland.
`
`REFERENCES
`
`L Weatherall DJ. Thalassemia in the next millennium. Keynote address. Ann. N. Y. Acad. Sci
`1998;850:1-9. [PubMed: 9668522]
`2. Borgna- Pignatti C, Rugolotto S, De Stefano P, et al. Survival and complications in patients with
`thalassemia major treated with transfusion and deferoxamine. HaematoJogica 2004;89: 1187-1193.
`[PubMed: 15477202]
`3. Aldouri MA, Wonke B, Hoffbrand AV, et al. High incidence of cardiomyopathy in beta-thalassaemia
`patients receiving regular transfusion and iron chelation: reversal by intensified chelation. Acta
`Haematol 1990;84:113-117. [Pub.Med: 2123060]
`4. Vecchio C, Derchi G. Management of cardiac complications i11 patients with thalassemiamajor. Sernin.
`Hematol 1995;32:288--296. [PubMed: 8560286]
`5. Davis BA, O'SullivanC, Jarritt PH, Porter JB. Value of sequential monitoring ofleftventricular ejection
`fraction in the management ofthalassemia major. Blood 2004;104:263-269. [PubMed: 15001468]
`6. Davis BA, Porter JB. Long-term outcome of continuous 24-hour deferoxan1ine infusion via indwelling
`intravenous catheters in high-risk beta-tha1assemia. Blood 2000;95: 1229-1236. [PubMed: 10666195]
`7. Gharzuddine WS, Kazma HK, Nuwayhid IA, et al. Doppler characterization ofleft ventricular diastolic
`function in beta-thalassaemia major. Evidence for an early stage of impaired relaxation. Eur. J.
`Echocardiogr 2002;3:47-51. [Pub.Med: 12067534]
`8. Krernastinos DT, Tsiapras DP, Tsetsos GA, et al. Left ventricular diastolic Doppler characteristics in
`beta-thalassemia major. Circulation 1993;88: 1127-1135. (Pub.Med: 8353874]
`9. Aessopos A, Farmakis D, Hat.ziliami A, et al . Cardiac status in well-treated patients with thalassemia
`major. Eur. J. Haematol 2004;73:359-366. [PubMed: 15458515]
`l 0. Wood JC, Tyszka TM, Ghugre N, .et al. Myocardial iron loading in transfusion-dependent thalassemia
`and sickle-cell disease. Blood 2004;103: 1934-1936. [Pub.Med: 14630822]
`I.I. Lorenz CH, Walker ES, Morgan VL, et al. Normal human right and left ventricular mass, systolic
`function, and gender differences by cine magnetic resonance imaging. J. Cardiovasc. Magn. Re son
`1999;1 :7-21. [PubMed: 11550343]
`12. Brittenham GM, Griffith PM, Nienhuis AW, et al. Efficacy of deferoxamine in preventing
`complications of iron overload in patients with thalassemia major. N. Engl. J. Med 1994;331:567-
`573. [Pub.Med 8047080]
`
`Ami N YAcadSci . Author manuscript; available in PMC 2010 June 28.
`
`TAR00073178
`
`6 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`WOOD et al.
`
`Page 7
`
`13. Voskaridou E, Douskou M, Terpos E, et al. Magnetic resonance imaging-in the evaluation of iron
`overload in patients with beta thalassaemia and sickle cell disease. Br. J. Haematol 2004) 26:736-
`742. [PubMed: 15327528]
`14. Wood JC, Ghugre N, Carson S, et al. Predictors ofabnorrnal myocardial furiction and T2* in children
`and young adults with thalassemia major. Blood 2003;102:952a.
`15. Anderson LJ, Holden S, Davis B, et al. Cardiovascular T2-star (I'2 *)magnetic resonance for the early
`diagnosis of myocardiaJ iron overload. Eur. HeartJ 2001 )2:2171-2 179. [Pub Med: 11913479]
`16. Westwood MA, Anderson LJ, Tanner MA, Pennell DJ. The relationship between myocardial iron
`deposition and left ventricular dysfunction in tha1assemia using cardiovascular magnetic resonance.
`J. Cardiovasc. Magn. Reson 2005;7:46-47.
`17. Olivieri NF, Brittenham GM. Iron-chelating therapy and the treatment of thalassemia. Blood
`1997;89:739-761. [PubMed: 9028304]
`18. Wood JC, Otto-Duessel M, Aguilar M, eta!. Cardiac MRI.(I'2, T2*) predicts cardiaciranin the gerbil
`model of iron cardiomyopathy. Circulation 2005;112:535-543. [PubMed: 16027257]
`19. Wang ZJ, Lian L, Chen Q, et al. lfT2 and magnetic susceptibility measurements in a gerbil cardiac
`iron overload model. Radiology 2005;234:749- 755. [PubMed: 15734931]
`20. Parkes JG, Olivieri NF, Templeton DM. Characterization of Fe2+ and Fe3+ transport by iron-loaded
`cardiac myocytes. Toxicology 1997;117: 141-151. [PubMed: 905'.7893]
`21. Liu Y, Parkes JG, Templeton DM. Differential accumulation of non-tr(UlSferrin-bound iron by cardiac
`myocytes and fibroblasts. J. Mo!. Cell. Cardiol 2003;35:505-514. [PubMed: 12738232)
`22. Parkes JG, Hussain RA, Olivieri NF,. Templeton DM. Effects ofironJoading on uptake, speciation,
`and chelation of iron in cultured myocardial cells. J. Lab. Clin. Med 1993; 122: 36-47. [Pub Med:
`8320489]
`23. Oudit GY, SlmH, TrivieriMG, et al. L-Type Ca2+ channels provide a major pathway for iron entry
`into cardiomyocytes in iron-overload cardiomyopathy. Nat. Med 2003;9: 1187-1194. [PubMed:

`12937413]
`24. Randell EW, Parkes JG, Olivieri NF, Templeton DM. Uptake ofnon-transferrin-boundiron by both
`reductive and nomeductive processes is modulated by intracellular iron J. Biol. Chem
`1994;269 16046-16053. [PubMed: 8206903]
`25. Jensen PD, Jensen FT, Christensen T, et al. Evaluation of myocardial iron by magnetic resonance
`imaging during iron chelation therapy with deferrioxamine: indication of Close relation between
`myocardial iron content and chelatable ironpool. Blood2003;101 :4632-4639 [PubMed 12576333 ]
`26. BujaLM, Roberts WC. Ironiri the heart. Etiology and cliriical significance. Am. J. Med 1971 ;51:209-
`221. [PubMed: 5095527]
`27. Esposito BP, Breuer W, Sirankapracha P, et al , Labile plasma iron in iron overload: redox activity
`and susceptibility to chelation. Blood 2003;102:2670- 2677. [PubMed: 12805056]
`28. Shiloh H, lancu TC, Bauminger ER, et al. Deferoxarnine-induced iron mobilization and redistribution
`of myocardial iron in cultured rat heart cells: studies oflhe chelatable iron pool by electron
`microscopy and Mossbauer spectroscopy. J. Lab. Clin. Med 1992;11 9: 428-436. [Pub Med: 1583395]
`29. Stuhne-Sekalec L; Xu SX, Parkes JG, et al. Speciationoftissue and cellular iron with on-line detection
`by inductively coupled plasma-mass spectrometry. Anal. Biochem 1992;205:278-284. [PubMed:
`1443573)
`30. Iancu TC, Shiloh H, Link G, et al. Ultrastrnctural patl10logy of iron-loaded rat myocardial cells in
`cil.lture. Br. J. Exp. Pathol 1987;.68:53--65. [PubMed: 3814501]
`31. Wood JC, Fassler J, Meade T. lv1imicking liver iron overload using hposomal ferritin preparations.
`Magn. Reson. Med 2004;51:607--01 l. [PubMed: 15004804]
`32. Link G, Pinson A, Hershko C. Iron loading of cultured cardiac myocytes modifies sarcolernmal
`structure and increases lysosomal fragility. J. Lab. Clin. Med 1993;121:127-134. [PubMed:
`8426074]
`33. Parkes JG, Liu Y, Sima JB, Templeton DM. Changes in gene expression with iron loading and.
`chelation in cardiac myocytes andnon-rnyocyti.c fibro-blasts. J.Mol. Cell. Cardiol 2000;32:233-246.
`[PubMed: 10722800]
`34. Kim E, Giri SN, Pessah IN. lron(II) is a modulator ofryanodine-sensitive calci1lffi channels of cardiac
`muscle sarcoplasrnic reticulum. Toxicol. Appl. Pharmacol l 995;130:57--06. [PubMed: 7530865]
`
`Ann N l' Acad Sci. Author manuscript; available in PMC 2010 June 28.
`
`TAR00073179
`
`7 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`WOOD et al.
`
`Page 8
`
`35. Link G, Athias P, Grynberg A, et al. Effect ofiron loading on transmembrane potential, contraction,
`and automaticity of rat ventricular muscle cells in culture. J. Lab. Clin. Med 1989;113: 103- 111.
`[PubMed: 2909644]
`36. Kuryshev YA, Brittenham GM, Fujioka H, et al. Decreased sodium and increased transient outward
`potassium currents iniron-looded cardiac myocytes. Implications for the arrhythmogenesis ofhinnan
`siderotic heart disease. Circulation 1999;100:675-683. [Pub Med: 10441107]
`37. Selden C, Owen M, Hopkins JM, Peters T J. Studies on the concentration and intracellular localization
`of iron proteins in liver biopsy specimens from patients with iron overload with special reference to
`their role in lysosornal disruption. Br. J. Haematol 1980;44:593-603. [PubMed: 7378318]
`38. Link G, Saada A, Pinson A, et al. Mitochondrial respiratory enzymes are a major target ofiron toxicity
`in rat heart cells. J. Lab. Clin. Med 1998;131:466-474. [PubMed: 9605112]
`39. Russell LK, Finck BN, Kelly DP. Mouse models of mitochondrial dysfunction and heart failure. J.
`Mol. Cell. Cardiol 2005;38:81-91. [PubMed: 15643424]
`40. Liu Y, Templeton DM. The effects of cardiac myocytes on interstitial fibroblasts in toxic iron
`overload. Cardiovasc. Toxicol 2001;1:299-308. [PubMed: 12213968]
`41. Laurita KR, Chuck ET, Yang T, et al. Optical mapping reveals conduction slowing and impulse block
`in iron-overload cardiomyopathy. J. Lab. Cji.n. Med 2003;142:83-89. [PubMed 12960954]
`42. Pogwizd SM, Bers DM. Cellular basis of triggered arrhythmias in heart failure. Trends Cardiovasc.
`Med2004;14:61-66. [PubMed: 15030791]
`43. Anderson LJ, Westwood MA, Holden S, et al. Myocardial iron clearance during reversal of siderotic
`cardiomyopathy with intravenous desferrioxamine: a prospective study using T2 * cardiovascular
`magnetic resonance. Br. J. Haematol 2004;127:348-355. [PubMed: 15491298]
`44. Mariotti. E, /mgelucci E, Agostini A, et al. Evaluation of cardiac status in iron loaded thalassernia
`patients following bone marrow transplantation during reduction in body iron burden. Br. J. Haematol
`1998;103:916-921. [PubMed: 9886301]
`
`z :
`
`Ann NY Acad Sci. Author manuscript; available in PMC 2010 June 28.
`
`TAR00073180
`
`8 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`z
`::c .
`I
`'"U
`)>
`)>
`
`c: -::::r
`
`0 ...,
`
`WOOD et al.
`
`Page 9
`
`Sa; rco· 1'1.!f1nit:
`•:i-1 4xu .. :-~1~11.9 v•-.:
`1'.:q~~
`Exeimt:inn1Cnntrac~1r.-u1 ~
`Co fLin •
`•
`
`Dc.1wlari1.atioo & \
`R po b tizati<Hl
`\
`= :...-..
`l._,~-~~ _::::.- -7 '
`(
`
`lmrneclltdat ion
`t:UflCC rlU'l.'l £ iM t$
`
`Gene Exptess.ion
`lto:n J rncrodil\n\
`
`z
`::c
`I
`'"U
`)>
`)>
`l!:
`......
`::::r
`0 ...,
`
`FIGURE 1.
`Uptake, storage, and toxicity of cardiac iron in a myocyte. Fe2+ andFe3+ enter the cell and are
`rapidly buffered and stored (bold arrows). Only stored iron is :MRI active. Toxicity occurs
`when the iron buffering system is overwhelmed.
`
`z
`::c
`I
`'"U
`)>
`)>
`
`c: -::r
`
`0 ...,
`
`Ann N YAcad Sci. Author manuscript; available in PMC 2010 June 28.
`
`TAR00073181
`
`9 of 10
`
`Taro Pharmaceuticals, Ltd.
`Exhibit 1054
`
`

`

`WOOD et al.
`
`Page 10
`
`Hemodynamic variables in thalassemia major
`
`TABLE 1
`
`Parameter
`
`Cardiac index (L/minlm2)
`
`Pretransfu

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