iron overload and chelation therapy in myelodysplastic syndromes

10
Please cite this article in press as: Temraz S, et al. Iron overload and chelation therapy in myelodysplastic syndromes. Crit Rev Oncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.006 ARTICLE IN PRESS ONCH-1833; No. of Pages 10 Critical Reviews in Oncology/Hematology xxx (2014) xxx–xxx Iron overload and chelation therapy in myelodysplastic syndromes Sally Temraz a , Valeria Santini b , Khaled Musallam a , Ali Taher a,a Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon b Division of Hematology, AOU Careggi, University of Florence, Firenze, Italy Accepted 14 January 2014 Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2. Iron overload. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.1. Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.2. Impact of IO on survival . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.3. Impact of IO on cardiac, hepatic and endocrine function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2.4. Diagnosis and evaluation of IO . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3. Iron chelation therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3.1. Chelation therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3.2. Effect of chelation therapy on survival in MDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3.3. Efficacy of chelation therapy in MDS patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3.4. Hematological improvement with iron chelation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Funding source . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Abstract Iron overload remains a concern in MDS patients especially those requiring recurrent blood transfusions. The consequence of iron overload may be more relevant in patients with low and intermediate-1 risk MDS who may survive long enough to experience such manifestations. It is a matter of debate whether this overload has time to yield organ damage, but it is quite evident that cellular damage and DNA genotoxic effect are induced. Iron overload may play a critical role in exacerbating pre-existing morbidity or even unmask silent ones. Under these circumstances, iron chelation therapy could play an integral role in the management of these patients. This review entails an in depth analysis of iron overload in MDS patients; its pathophysiology, effect on survival, associated risks and diagnostic options. It also discusses management options in relation to chelation therapy used in MDS patients and the impact it has on survival, hematologic response and organ function. © 2014 Elsevier Ireland Ltd. All rights reserved. Keywords: MDS; Myelodysplastic syndrome; Iron chelation; Iron overload; Transfusion-dependent; Deferasirox; Deferoxamine; Deferiprone Corresponding author at: P.O. Box 11 0236, Riad El Solh, 110 72020 Beirut, Lebanon. Tel.: +961 1 350000x5392; fax: +961 350000x7942. E-mail address: [email protected] (A. Taher). 1. Introduction Myelodysplastic syndromes (MDS) represent a group of hematologic disorders characterized by dysplastic and ineffective hematopoiesis. Approximately 80% of patients present with anemia and a substantial percentage of them http://dx.doi.org/10.1016/j.critrevonc.2014.01.006 1040-8428/© 2014 Elsevier Ireland Ltd. All rights reserved.

Upload: ali

Post on 30-Dec-2016

225 views

Category:

Documents


4 download

TRANSCRIPT

Page 1: Iron overload and chelation therapy in myelodysplastic syndromes

O

C

12

3

4

A

miecoo©

K

B

h1

ARTICLE IN PRESSNCH-1833; No. of Pages 10

Critical Reviews in Oncology/Hematology xxx (2014) xxx–xxx

Iron overload and chelation therapy in myelodysplastic syndromes

Sally Temraz a, Valeria Santini b, Khaled Musallam a, Ali Taher a,∗a Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon

b Division of Hematology, AOU Careggi, University of Florence, Firenze, Italy

Accepted 14 January 2014

ontents

. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

. Iron overload. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002.1. Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002.2. Impact of IO on survival . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002.3. Impact of IO on cardiac, hepatic and endocrine function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002.4. Diagnosis and evaluation of IO . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

. Iron chelation therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.1. Chelation therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.2. Effect of chelation therapy on survival in MDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.3. Efficacy of chelation therapy in MDS patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.4. Hematological improvement with iron chelation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00Funding source . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

bstract

Iron overload remains a concern in MDS patients especially those requiring recurrent blood transfusions. The consequence of iron overloaday be more relevant in patients with low and intermediate-1 risk MDS who may survive long enough to experience such manifestations. It

s a matter of debate whether this overload has time to yield organ damage, but it is quite evident that cellular damage and DNA genotoxicffect are induced. Iron overload may play a critical role in exacerbating pre-existing morbidity or even unmask silent ones. Under theseircumstances, iron chelation therapy could play an integral role in the management of these patients. This review entails an in depth analysis

f iron overload in MDS patients; its pathophysiology, effect on survival, associated risks and diagnostic options. It also discusses managementptions in relation to chelation therapy used in MDS patients and the impact it has on survival, hematologic response and organ function.

2014 Elsevier Ireland Ltd. All rights reserved.

; Transf

eywords: MDS; Myelodysplastic syndrome; Iron chelation; Iron overload

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

∗ Corresponding author at: P.O. Box 11 0236, Riad El Solh, 110 72020eirut, Lebanon. Tel.: +961 1 350000x5392; fax: +961 350000x7942.

E-mail address: [email protected] (A. Taher).

1

oip

ttp://dx.doi.org/10.1016/j.critrevonc.2014.01.006040-8428/© 2014 Elsevier Ireland Ltd. All rights reserved.

usion-dependent; Deferasirox; Deferoxamine; Deferiprone

. Introduction

Myelodysplastic syndromes (MDS) represent a group

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

f hematologic disorders characterized by dysplastic andneffective hematopoiesis. Approximately 80% of patientsresent with anemia and a substantial percentage of them

Page 2: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

2 Oncolo

wd

TMropscie

pmty

2

2

tfssHcmcasdwMmiRetpmtoclwiwct

cc

qrfisttptttatt[frufIlteCvlcc(lghaaa

2

tssrtbJplmoIo

S. Temraz et al. / Critical Reviews in

ill become transfusion-dependent during the course of theirisease [1].

Chronic transfusions lead to secondary iron overload (IO).he relatively longer survival of low and intermediate-1 riskDS groups classified by the International Prognostic Sco-

ing System (IPSS) [2,3] places them at an increased riskf damage by IO from prolonged red cell transfusions com-ared to high risk patients who have a markedly reducedurvival [1]. MDS population consists mainly of elderly witho-morbid conditions and a propensity to have cardiac failure,nfection, hemorrhage and hepatic cirrhosis IO may rapidlyxacerbate these pre-existing conditions [4].

In this review, we provide an overview of IO in MDSatients, its pathophysiology, associated risks and manage-ent options as recent and more systematic data on chelation

herapy in MDS patients has become available in the past fewears.

. Iron overload

.1. Pathophysiology

The pathophysiology of IO in MDS patients is related tohat observed in thalassemia syndromes and consists of inef-ective erythropoiesis and hepcidin dysregulation in someubtypes of MDS (primary IO) as well as transfusional sidero-is (secondary IO). There are however some differences.epcidin is a key hormone mediating iron homeostasis. Hep-

idin has a role in the down-regulation of ferroportin, theembrane transporter delivering duodenal iron from entero-

ytes to transferrin, thus resulting in decreased duodenal ironbsorption [5,6]. In disease states of ineffective erythropoei-is, serum hepcidin levels are low that result in unrestraineduodenal iron absorption and subsequent IO [7]. Recently,e measured serum hepcidin by mass-spectrometry in 113DS patients, and found the lowest levels in refractory ane-ia with ring sideroblasts (RARS, 1.43 nM) and the highest

n refractory anemia with excess blasts (RAEB 11.3 nM) [8].ARS patients are particularly refractory to therapy withrythropoietic stimulating factors, and thus are frequentlyransfusion dependent, have a particularly expanded erythro-oiesis, with an extremely frequent presence of acquiredutations in SF3B1, a gene encoding a core component of

he RNA splicing machinery in MDS patients with ring sider-blasts [8,9]. A genetic study has confirmed our findings,orrelating SF3B1 with ring sideroblasts and low hepcidinevels [10]. The growth differentiation factor 15 (GDF15),hich has been shown to suppress the activity of hepcidin

n thalassemia [11], did not seem to regulate hepcidin in ourork and that of our authors. Therefore, low levels of hep-

idin further predispose patients with RARS subtype of MDS

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

o the risk of IO.Anemia affects 80% of MDS patients and in their disease

ourse many of them will receive transfusions. Red bloodell transfusions, while alleviating anemia may improve

brtA

gy/Hematology xxx (2014) xxx–xxx

uality of life and prolong survival, but expose to severalisks [12]. Patients with MDS may have an extremely highrequency of transfusions and may accumulate excess ironn a short time lapse. The rapidity and intensity of transfu-ions accounts for IO more than the total number [13]. Inhis state of iron repletion, ferritin production is increasedo permit adequate storage of iron and transferrin receptorroduction is decreased to prevent further entry of iron intohe cell [14,15]. When the binding capacity of transferrin inhe blood is exceeded, iron is found in the plasma as non-ransferrin bound iron (NTBI) [16]. Since iron cannot bectively excreted out of the body, it initially accumulates inhe reticuloendothelial macrophages and is later deposited inhe parenchymal cells of liver, heart and endocrine organs17]. Intracellular iron can be found in endocytotic vesiclesollowing entry of iron into the cell through the transferrineceptor-1, in ferritin where each ferritin molecule can holdp to 4500 atoms of iron or in association with proteins thatorm prosthetic groups involved in biological reactions [18].n this IO state, NTBI changes to its redox active form termedabile plasma iron (LPI). Patients with low risk MDS havehe higher NTBI levels compared to high risk MDS; high-st levels are seen in RARS followed by 5 q-syndromes andMML, respectively [8]. Entry of LPI into cells is mediatedia several transporters; DMT1 in enterocytes [19], Zrt-Irt-ike protein in hepatocytes [20] and L-type voltage dependentalcium channels in cardiac myocytes [21]. LPI is a toxicompound that enters the cell forming the labile cell ironLCI) [22]. LPI in MDS is high and correlates with ferritinevels quite well. LCI results in the formation of reactive oxy-en species (ROS) that can suppress renewal and number ofematopoietic stem cells [23] and can induce DNA damagend genomic instability [24]. This results in tissue damagend subsequent fibrosis which could result in complicationss cardiomyopathy, cirrhosis and diabetes [25,26].

.2. Impact of IO on survival

Several retrospective studies suggest a negative associa-ion between transfusion dependency in MDS and overallurvival (OS) [27–29]. Whether this is due to the transfu-ions themselves or to the fact that more severe diseasesequire more frequent transfusions is a matter of debate. Fur-hermore, the impact of IO on OS in MDS patients has alsoeen demonstrated. In a retrospective nationwide survey ofapanese patients, Takatoku et al. found that in 37 of 38atients who died of hepatic or cardiac failure had ferritinevels >1000 �g/L suggesting that IO resulted in increased

ortality [30]. On the other hand, in a retrospective analysisf a US database, complications potentially attributable toO in MDS such as cardiac events, diabetes and liver diseaseccurred at a higher frequency in MDS patients receiving

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

lood transfusions [31]. Also, Sanz et al. confirmed theseesults in a large series of 2994 patients where they showedhat IO is an independent prognostic variable of OS and alsoML transformation [32]. However, the results have never

Page 3: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

Oncolo

bcb

2f

pCcptrtwtOcpticom

eprhno[aotrtwtais

2

gt

bwhbte

owiCfba

twvtvwa

cwbpt

oribuMoriMrohiahic

epehahtwtr

S. Temraz et al. / Critical Reviews in

een published in definitive form. Transfusion dependencyoupled by increases in serum ferritin levels further exacer-ates OS [29].

.3. Impact of IO on cardiac, hepatic and endocrineunction

The risk of cardiac events is increased in MDS patients,articularly in those who are transfusion-dependent [33].hronic anemia remains a co-factor in the development ofardiac failure because it necessitates increased cardiac out-ut in order to compensate for impaired tissue oxygenationhereby leading to heart complications. MDS patients areeported to be at higher risk of cardiac events comparedo the general population (73.2% versus 54.5%; P < 0.01)ith transfused MDS patients having a higher risk than non-

ransfused ones (82.4%% versus 67.1%; P < 0.001) [33].nce again, this maybe attributable to cardiac remodeling

orrelated to anemia [34], which is there even in transfusedatients who suffer from fluctuating hemoglobin levels, ando subsequent myocardial hypoxia, as well as to IO. Somenconsistency in fact exists in results from studies employingardiac magnetic resonance imaging T2* (MRI) as measuref IO in MDS. These studies suggest that cardiac iron accu-ulation is not frequent in MDS patients [35–37].Although the majority of MDS patients do not live long

nough to manifest conditions of liver cirrhosis, hepatic com-lication nevertheless have been implicated in IO. In oneetrospective study of 4546 patients with MDS and otherematopoietic disorders, transfusion dependency was sig-ificantly associated with risk of potential complicationsf IO (liver disease P = 0.0008 and diabetes P = 0.0025)38]. Moreover, in another Japanese retrospective study, theuthors report 75 deaths, with cardiac failure noted in 24%f cases and liver failure noted in 6.7% of cases [30]. Ofhese, 97% had ferritin levels >1000 ng/ml. Moreover, a large,etrospective analysis from US Medicare database revealedhat diabetes occurs significantly more frequently in patientsith MDS. When MDS patients were compared based on

ransfusion dependency, those receiving transfusions had andditional increased risk of diabetes than those not receiv-ng transfusions; however, the difference was not statisticallyignificant (44.4% versus 37.1%; P = 0.1) [33].

.4. Diagnosis and evaluation of IO

Most data on modalities for the assessment of IO ineneral come from experience with transfusion-dependenthalassemia patients.

Ferritin: In the general population, IO is generally definedy a serum ferritin level >300 �g/L in men and >200 �g/L inomen. Serum ferritin thresholds of >1000 and >2500 �g/L

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

ave been classically used to flag patients with increased mor-idity risk and to tailor indications for chelation therapy inhe thalassemia population [39]. The aforementioned studiesvaluating the consequences of IO in MDS patients also relied

intf

gy/Hematology xxx (2014) xxx–xxx 3

n a serum ferritin level of >1000 �g/L. Elderly patientsith MDS present frequently with comorbidities, and chronic

nflammation is a common finding. In this group of patients,-reactive protein and other parameters are altered, and so is

erritin. It is thus a parameter which has some disadvantages,ut may help in following a patient, allowing repeated examsnd being rather costless and highly reproducible.

Liver iron: Assessment of liver iron concentration remainshe gold standard for quantification of total body iron [40],ith values up to 1.8 mg Fe/g dry weight considered normal,alues up to 7 mg Fe/g dry weight seen in some non-halassemic populations without apparent adverse effects andalues >15 mg Fe/g dry weight commonly associated withorse prognosis, liver fibrosis progression, or liver function

bnormalities [39].Liver biopsy enables pathologists to determine liver iron

oncentration as well as severity of liver disease in patientsho are at high risk of liver involvement. However, liveriopsy is contraindicated in almost the totality of MDSatients since there is an increased risk of bleeding due tohrombocytopenia and/or platelet dysfunction [7].

MRI: Magnetic resonance imaging using either R2 (1/T2)r R2* (1/T2*) pulse sequences are reliable, internationallyeproducible, and non-invasive methods for assessing liverron concentration, and have been validated against liveriopsy in several diseases complicated by IO [41–43]. Thepper limit to reliably estimate liver iron concentration byRI is approximately 30–40 mg Fe/g dry weight, depending

n the scanner specifications [43]. This method is expensive,equires a skilled radiologist, needs accurate software ands not readily available in all hematologic centers treating

DS patients. Moreover, not all MDS patients on chelationeally require this determination, unless ferritin is increasingr some cardiac failure signs have appeared, or transfusionistory prior to therapy is not clear [8]. Cardiac siderosiss measured using T2* MRI (normal: >20 ms) a functionalnalysis in the end, which correlates with subsequent risk ofeart failure. It is now validated as a true measure of cardiacron, correlating with chemical measurement on post-mortemardiac biopsies [42].

Hepcidin: Serum hepcidin measured by mass spectrom-try or ELISA (EIA-5258) revealed that highly transfusedatients were shown to have significantly higher hepcidin lev-ls while patients with low hepcidin levels had significantlyigher hemoglobin and transferrin, and lower serum ferritinnd transferrin saturation [8,10,44]. A correlation betweenepcidin and OS was seen with a hepcidin level accordingo EIA-5258 at or above the median of 17.5 ng/ml associatedith a significantly worse survival (P = 0.03) [44]. However,

he clinical reliability of hepcidin is still a subject of activeesearch.

Practically, the best approach should be to assess iron

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

ntake by registering all transfusions carefully, which isot frequently done in clinics. For patients with significantransfusion burden, cardiac function (by ultrasound), liverunction, and glucose tolerance test should be performed

Page 4: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

4 S. Temraz et al. / Critical Reviews in Oncology/Hematology xxx (2014) xxx–xxx

Table 1Iron chelators available for the management of IO.

Agent Approval (US, FDA) Administration Schedule and dose Clearance Most common adverse events

Deferoxamine[46]

Chronic IO due totransfusion-dependentanemias

Subcutaneous,intravenous

20–40 mg/kg/d over8–24 h 5–7 d/wk

RenalHepatic

Hypersensitivity reactions,tachycardia, gastrointestinal events,increased transaminases

Deferasirox [50] Chronic IO due toblood transfusions ornon-transfusion-dependentthalassemia

Oral 20–40 mg/kg/d Hepatobiliary Gastrointestinal events involvingdiarrhea, nausea, constipation andabdominal pain; skin rashes andincreased serum creatinine level

Deferiprone [51] Transfusional IO dueto thalassemia

Oral 25–33 mg/kg 3×/d fora total daily dose of

g/kg

Renal Gastrointestinal symptoms,granulocytosis, agranulocytosis and

eocds

3

3

itlleb>t

duAP(DhmFaddadiwoqbT

easslacta

3

cicsh[rt[antpgetcwercr

3

syndromes whenother chelationtherapy is inadequate

75–99 m

very three months. In case of increasing ferritin levels with-ut specific inflammatory or infective causes during ironhelation therapy (ICT), as well as in case of cardiac or liverysfunction, more accurate exams, like cardiac MRI (T2*)hould be performed.

. Iron chelation therapy

.1. Chelation therapy

The rationale behind using ICT is removing the increasedron burden specifically LCI to minimize production of ROShereby decreasing cellular and organ damage. Patients mostikely to benefit from chelation therapy include those withow or intermediate-1 IPSS risk MDS who have a long lifexpectancy and are anticipated to receive more than 20 redlood cell transfusions and/or whose serum ferritin level is1000 �g/L [1,45]. Chelation therapy efficacy can be moni-

ored by monitoring trends and levels of serum ferritin.Multiple drugs have been developed to decrease iron bur-

en from IO states. The three iron chelators approved forse include deferoxamine (DFO, Desferal®, Novartis PharmaG, Basel, Switzerland), deferasirox (Exjade®, Novartisharma AG, Basel, Switzerland) and recently deferiproneFerriprox®, Apotex Inc., Toronto, ON, Canada) (Table 1).FO binds to NTBI or to iron that is found in ferritin andemosiderin but not transferrin forming the ferrioxamineolecule which is later excreted via the kidneys [46] (Fig. 1).errioxamine renders iron unavailable to chemical reactionsnd hence prevents formation of ROS. DFO promotes ferritinegradation in lysosomes by inducing autophagy [47]. Botheferiprone and deferasirox are likely to chelate cytosolic ironnd iron which is extracted from ferritin prior to ferritin degra-ation by proteosomes [47] (Fig. 1). Besides its efficacy as anron chelator, deferasirox administration has been associatedith increased levels of hepcidin that results in the removal

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

f ferroportin from the enterocyte membrane and its subse-uent degradation by lysosomes [48]. Lately, Deferasirox haseen used frequently in the treatment of IO in MDS patients.he oral administration in fact allows higher compliance of

sf

elevation of liver enzymes

lderly patients who do not accept such prolonged infusionsnd may be subject to bleeding at the subcutaneous insertionite. Despite these positive characteristics, in a recent Italiantudy, the percentage of drop out from a chelation study inower risk MDS was pretty high and related to compliancend side effects, even if mild which impaired MDS patientsompliance [49]. Fig. 2 shows one suggested algorithm onhe management of IO in MDS patients with the use of theforementioned iron chelators.

.2. Effect of chelation therapy on survival in MDS

Evidence supporting the fact that IO causes severe compli-ations comes from the demonstration of prolonged survivaln MDS patients undergoing ICT. At present, all the evidenceomes from retrospective studies. These studies demon-trated that low risk MDS patients receiving chelation therapyad a better median OS compared to non-chelated ones52–55]. Recently, significant improvement in survival waseported in low risk MDS patients exhibiting non-RARS fea-ures but the same was not achieved for patients with RARS56]. Median OS in the RARS who received chelation ther-py was 134.4 months versus 99 months; however, it didot reach statistical significance. The retrospective nature ofhese trials does not exclude potential bias that arises from theossibility that patients with a better performance score had areater chance of being started on ICT. This bias can only bexcluded once a prospective randomized placebo controlledrial is undertaken. A large phase 3 trial (TELESTO, Clini-al trials.gov: NCT00940602) is currently recruiting patientsith low and intermediate-1 risk MDS patients to receive

ither deferasirox monotherapy or placebo. Although the ret-ospective studies consistently show a survival advantage ofhelation therapy, these results cannot be confirmed untilesults of this phase 3 trial are available.

.3. Efficacy of chelation therapy in MDS patients

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

Results on the efficacy of chelation therapy in decreasingerum ferritin levels, LPI and liver iron concentrations comerom prospective trials (Table 2). Most available data on the

Page 5: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

S. Temraz et al. / Critical Reviews in Oncology/Hematology xxx (2014) xxx–xxx 5

F n overln a iron;

ubasp

o[

atiie

TE

S

PMG

E

E

U

BNGK

S

ig. 1. Mechanism of action of iron chelators in the management of iroon-transferrin bound iron; TFR-1: transferrin receptor-1; LPI: labile plasm

se of ICT in MDS patients come from deferasirox trials,ecause they are more recent and data were collected in anppropriate and complete manner. Few and relatively smalltudies have been conducted with other iron chelators inatients with MDS.

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

Decreases in liver iron concentration depended on the dosef deferasirox administered and transfusion requirements57,60]. Both Gatterman et al. and Breccia et al. reported

ert

able 2fficacy of the iron chelators deferasirox, deferoxamine and deferiprone in MDS p

tudy Patients Treatment Duration (m

orter et al. [57] 47 Deferasirox 12

etzegeroth et al. [58] 12 Deferasirox 12

reenberg et al. [59] 24 Deferasirox 613

PIC trial, Gattermanet al. [60]

341 Deferasirox 12

Xtend and eXjange,Gatterman et al. [61]

123 Chelationnaïve44 previouslychelated

Deferasirox 12

S03 trial, List et al. [62] 914933

Deferasirox 122436

reccia et al. [63] 40 Deferasirox 12

olte et al. [64] 40 Deferasirox 12

onzalez et al. [65] 28 DFO 12

ersten et al. [66] 18 Deferiprone 12

: significant; NR: not reported; NS: not significant; LPI: labile plasma iron; LIC: l

oad. TBI: transferrin bound iron; ROS: reactive oxygen species; NTBI:DMT1: divalent metal transporter 1.

significant decrease in serum ferritin regardless of chela-ion history [60,63] suggesting that deferasirox is effectiven patients receiving chelation therapy for the first time andn patients switching from other chelation therapies. How-ver, the same result was not achieved in the EXtend and

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

Xjange trials where a significant decrease in serum fer-itin was seen only in patients not receiving prior chelationherapy [61]. Also, the decrease in serum ferritin level was

atients.

onths) Decrease in LIC Decrease inferritin

Decrease in LPI

S S NRS S NRSNS

NSNS

SS

NR S S

NR SNS

NR

NR S S

S NR NRS NR SNS NR NRS NR NR

iver iron concentration; DFO: deferoxamine.

Page 6: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

6 S. Temraz et al. / Critical Reviews in Oncology/Hematology xxx (2014) xxx–xxx

F ontrainc 109/Ld e or anu

aii

3

i

etsni

ig. 2. Suggested algorithm for the use of iron chelators in MDS patients. *Creatinine more than twice the upper limit of normal, platelet counts <50 ×eferasirox. *Contraindications to deferoxamine include: severe renal failur

ssociated with a significant improvement in alanine transam-nase (P < 0.00001), which is an indicator of hepatocellularnjury that can lead to cirrhosis [60].

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

.4. Hematological improvement with iron chelation

Evidence of hematological improvement with the use ofron chelation was reported previously in a study by Jensen

t

r

dications to deferasirox include: creatinine clearance < 40 ml/min or serum, severe hepatic impairment (Child–Pugh class C) and hypersensitivity toria, Clcr < 10 ml/min and hypersensitivity to deferoxamine.

t al. with the use of DFO. DFO was successful in decreasingransfusional requirements and 5/11 patients became transfu-ion independent [67]. Further improvement in platelet andeutrophil counts was also reported. In patients who showedmprovement in erythropoietic output, an increase in serum

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

ransferrin receptor was detected.Similarly, a post hoc analysis evaluated hematologic

esponse to deferasirox in a cohort of iron-overloaded

Page 7: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

Oncolo

pp(aMlsnoata1

tonhahseUtoebsoctopftiaewpelcaHwh

4

ioDi

tb

wetmtcptitshlaeaitt

F

C

TfrPaVC

R

GB

R

S. Temraz et al. / Critical Reviews in

atients with MDS enrolled in the EPIC trial. Erythroid,latelet and neutrophil responses were observed in 21.5%53/247), 13.0% (13/100) and 22.0% (11/50) of the patientsfter a median of 109, 169 and 226 days, respectively.edian serum ferritin reductions were greater in hemato-

ogic responders compared with non-responders at end oftudy, although these differences were not statistically sig-ificant [68]. In another retrospective Italian study, 42.7%f patients receiving chelation with DFO or deferasiroxchieved a hematologic response. Eighteen patients becameransfusion independent; 12 of which received deferasiroxnd the remainder DFO [69]. Median time to response was5 months for DFO and 3 months for deferasirox.

One proposed mechanism is the dynamic, bidirec-ional regulation between erythropoiesis and IO where notnly ineffective erythropoiesis leads to increased intesti-al iron absorption through the down-regulation of theepatic hormone hepcidin [70], but also treatment of IOffects erythropoietic capacity and leads to improvements inemoglobin level and red cell survival [71–73]. In a recenttudy from thalassemia patients, mobilization of hematopoi-tic peripheral progenitors (namely Erythroid Burst-Formingnits) was significantly higher in deferasirox-treated patients

han those receiving other chelators, irrespective or the degreef iron depletion; which may indicate that the observedffects of deferasirox on hematologic outcomes are mediatedy other factors [74]. In fact, it has been recently demon-trated that in MDS patients, IO suppresses the proliferationf erythroid progenitors cells (BFU-E), while the myeloidompartment (CFU-GM) is not affected and it was shownhat ICT may revert such phenomenon [75]. It is a sharedpinion that the ideal MDS patient to receive chelation is aatient with IPSS lower risk score (INT-1 and LOW), trans-usion dependent but with a reasonable expectance of life, sohat ICT may be effective and possibly make some differencen outcome. Nevertheless, it is becoming clearer that there is

subset of IPSS higher risk MDS patients which could ben-fit from ICT [45]. In particular, eligible MDS patients forhom a hematopoietic stem cell transplantation (HSCT) isrogrammed should be chelated because there is growingvidence that high ferritin levels before HSCT are corre-ated with worst outcome in patients receiving myeloablativeonditioning regimen [76,77]. Therefore, it is strongly advis-ble to properly chelate these patients, even shortly beforeSCT [78]. On the other hand, some higher risk patientsho received a therapy modifying their survival and diseaseistory could also take advantage from supportive ICT.

. Conclusion

IO is a reality for transfusion dependent MDS patients. It

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

s a matter of debate whether this overload has time to yieldrgan damage, but it is quite evident that cellular damage andNA genotoxic effect via ROS species are induced. Another

mportant point would be to verify the threshold of RBC

gy/Hematology xxx (2014) xxx–xxx 7

ransfusions leading to IO in MDS, and whether patients maye rescued by appropriate ICT [79].

The consequence of IO may be more relevant in patientsith low and intermediate-1 risk MDS who may survive long

nough. The causes of death in large MDS series may indicatehat IO may play a critical role in exacerbating pre-existing

orbidity or even unmask silent ones, especially because ofhe relative frailty of the large part of MDS patients. Theseonsiderations, together with the concept of “optimal” sup-ortive care, should indicate iron chelation as an integratedool in the management of MDS patients. Nevertheless, theres still ample skepticism in accepting this therapy, maybe dueo low familiarity with the problem of IO and its diverse con-equences. It is to be said that still little is known in ironomeostasis in MDS and that although MDS experts shouldearn the lesson from thalassemia experience, more evidencend data are required to clarify the role of iron in MDS dis-ase manifestation and outcome. A lively debate is going onmong MDS experts, and it seems as if the field was dividedn “a priori” believers in IO and non- believers [80]. Prospec-ive carefully planned randomized studies are the key to solvehis issue.

unding source

None declared.

onflicts of interest

Sally Temraz reported no conflicts of interest. Aliaher reported receiving research funding and honorariarom Novartis Pharmaceuticals; Khaled Musallam reportedeceiving research funding and honoraria from Novartisharmaceuticals and is currently (at the time of submission)n employee of Novartis Pharma AG, Basel, Switzerland;aleria Santini reported receiving honoraria as a speaker fromelegene, Novartis, Janssen and GSK.

eviewers

Photis Beris, Associate Professor, Faculté de Médecine,eneva University, Swiss Internal Medicine, 51 avenuelanc, CH-1202 Genève, Switzerland.

eferences

[1] Steensma DP, Bennett JM. The myelodysplastic syndromes: diagnosisand treatment. Mayo Clin Proc 2006;81(1):104–30.

[2] Greenberg PL. Myelodysplastic syndromes: iron overload conse-

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

quences and current chelating therapies. J Natl Compr Cancer Netw2006;4(1):91–6.

[3] Valent P, Horny HP, Bennett JM, et al. Definitions and standardsin the diagnosis and treatment of the myelodysplastic syndromes:

Page 8: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

8 Oncolo

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

S. Temraz et al. / Critical Reviews in

consensus statements and report from a working conference. Leuk Res2007;31(6):727–36.

[4] Malcovati L, Porta MG, Pascutto C, et al. Prognostic factors andlife expectancy in myelodysplastic syndromes classified accordingto WHO criteria: a basis for clinical decision making. J Clin Oncol2005;23(30):7594–603.

[5] Hentze MW, Muckenthaler MU, Galy B, et al. Two to tango: regulationof Mammalian iron metabolism. Cell 2010;142(1):24–38.

[6] Ganz T, Nemeth E. Iron imports. IV. Hepcidin and regulation ofbody iron metabolism. Am J Physiol Gastrointest Liver Physiol2006;290(2):G199–203.

[7] Gattermann N, Rachmilewitz EA. Iron overload in MDS-pathophysiology, diagnosis, and complications. Ann Hematol2011;90(1):1–10.

[8] Santini V, Girelli D, Sanna A, et al. Hepcidin levels and their deter-minants in different types of myelodysplastic syndromes. PLoS ONE2011;6(8):e23109.

[9] Papaemmanuil E, Cazzola M, Boultwood J, et al. Somatic SF3B1mutation in myelodysplasia with ring sideroblasts. N Engl J Med2011;365(15):1384–95.

10] Ambaglio I, Malcovati L, Papaemmanuil E, et al. Inappropriately lowhepcidin levels in patients with myelodysplastic syndrome carrying asomatic mutation of SF3B1. Haematologica 2013.

11] Tanno T, Bhanu NV, Oneal PA, et al. High levels of GDF15 in tha-lassemia suppress expression of the iron regulatory protein hepcidin.Nat Med 2007;13(9):1096–101.

12] Nilsson-Ehle H, Birgegård G, Samuelsson J, et al. Quality of life, phys-ical function and MRI T2* in elderly low-risk MDS patients treated toa haemoglobin level of ≥120 g/L with darbepoetin alfa ± filgrastim orerythrocyte transfusions. Eur J Haematol 2011;87(3):244–52.

13] Olivieri NF, Brittenham GM. Iron-chelating therapy and the treatmentof thalassemia. Blood 1997;89(3):739–61.

14] Baker E, Baker SM, Morgan EH. Characterisation of non-transferrin-bound iron (ferric citrate) uptake by rat hepatocytes in culture. BiochimBiophys Acta 1998;1380(1):21–30.

15] Wright TL, Brissot P, Ma WL, et al. Characterization of non-transferrin-bound iron clearance by rat liver. J Biol Chem 1986;261(23):10909–14.

16] Hershko C, Link G, Cabantchik I. Pathophysiology of iron overload.Ann N Y Acad Sci 1998;850:191–201.

17] Siah CW, Ombiga J, Adams LA, et al. Normal iron metabolism andthe pathophysiology of iron overload disorders. Clin Biochem Rev2006;27(1):5–16.

18] Brissot P, Ropert M, Le Lan C, et al. Non-transferrin bound iron: akey role in iron overload and iron toxicity. Biochim Biophys Acta2012;1820(3):403–10.

19] Gunshin H, Mackenzie B, Berger UV, et al. Cloning and characteri-zation of a mammalian proton-coupled metal-ion transporter. Nature1997;388(6641):482–8.

20] Liuzzi JP, Aydemir F, Nam H, et al. Zip14 (Slc39a14) mediates non-transferrin-bound iron uptake into cells. Proc Natl Acad Sci U S A2006;103(37):13612–7.

21] Oudit GY, Sun H, Trivieri MG, et al. L-type Ca2+ channels providea major pathway for iron entry into cardiomyocytes in iron-overloadcardiomyopathy. Nat Med 2003;9(9):1187–94.

22] Cabantchik ZI, Breuer W, Zanninelli G, et al. LPI-labile plasma iron iniron overload. Best Pract Res Clin Haematol 2005;18(2):277–87.

23] Ito K, Hirao A, Arai F, et al. Reactive oxygen species act throughp38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med2006;12(4):446–51.

24] Sallmyr A, Fan J, Rassool FV. Genomic instability in myeloid malig-nancies: increased reactive oxygen species (ROS), DNA double strandbreaks (DSBs) and error-prone repair. Cancer Lett 2008;270(1):1–9.

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

25] Hebbel RP. Auto-oxidation and a membrane-associated ‘Fentonreagent’: a possible explanation for development of membrane lesionsin sickle erythrocytes. Clin Haematol 1985;14(1):129–40.

[

gy/Hematology xxx (2014) xxx–xxx

26] Le Lan C, Loréal O, Cohen T, et al. Redox active plasma iron inC282Y/C282Y hemochromatosis. Blood 2005;105(11):4527–31.

27] Malcovati L. Impact of transfusion dependency and secondary ironoverload on the survival of patients with myelodysplastic syndromes.Leuk Res 2007;31(Suppl. 3):S2–6.

28] Cermak J, Kacirkova P, Mikulenkova D, et al. Impact of transfusiondependency on survival in patients with early myelodysplastic syn-drome without excess of blasts. Leuk Res 2009;33(11):1469–74.

29] de Swart L, Smith A, Fenaux P, et al. Transfusion-dependency isthe most important prognostic factor for survival in 1000 newlydiagnosed mds patients with low- and intermediate-1 risk MDS inthe European LeukemiaNet MDS registry. ASH Annu Meet Abstr2011;118(21):2775.

30] Takatoku M, Uchiyama T, Okamoto S, et al. Retrospective nation-wide survey of Japanese patients with transfusion-dependent MDSand aplastic anemia highlights the negative impact of iron overloadon morbidity/mortality. Eur J Haematol 2007;78(6):487–94.

31] Rollison DE, Howlader N, Smith MT, et al. Epidemiology of myelodys-plastic syndromes and chronic myeloproliferative disorders in theUnited States, 2001–2004, using data from the NAACCR and SEERprograms. Blood 2008;112(1):45–52.

32] Sanz G, Nomdedeu B, Such E, et al. Independent impact of iron over-load and transfusion dependency on survival and leukemic evolutionin patients with myelodysplastic syndrome. ASH Annu Meet Abstr2008;112(11):640.

33] Goldberg SL, Chen E, Corral M, et al. Incidence and clinical compli-cations of myelodysplastic syndromes among United States Medicarebeneficiaries. J Clin Oncol 2010;28(17):2847–52.

34] Oliva EN, Dimitrov BD, Benedetto F, et al. Hemoglobin level thresholdfor cardiac remodeling and quality of life in myelodysplastic syndrome.Leuk Res 2005;29(10):1217–9.

35] Konen E, Ghoti H, Goitein O, et al. No evidence for myocardialiron overload in multitransfused patients with myelodysplastic syn-drome using cardiac magnetic resonance T2 technique. Am J Hematol2007;82(11):1013–6.

36] Di Tucci AA, Matta G, Deplano S, et al. Myocardial iron overloadassessment by T2* magnetic resonance imaging in adult trans-fusion dependent patients with acquired anemias. Haematologica2008;93(9):1385–8.

37] Chacko J, Pennell DJ, Tanner MA, et al. Myocardial iron loading bymagnetic resonance imaging T2* in good prognostic myelodysplas-tic syndrome patients on long-term blood transfusions. Br J Haematol2007;138(5):587–93.

38] Delea TE, Hagiwara M, Phatak PD. Retrospective study of the associa-tion between transfusion frequency and potential complications of ironoverload in patients with myelodysplastic syndrome and other acquiredhematopoietic disorders. Curr Med Res Opin 2009;25(1):139–47.

39] Taher AT, Musallam KM, Inati A. Iron overload: consequences, assess-ment, and monitoring. Hemoglobin 2009;33(Suppl. 1):S46–57.

40] Angelucci E, Brittenham GM, McLaren CE, et al. Hepatic iron concen-tration and total body iron stores in thalassemia major. N Engl J Med2000;343(5):327–31.

41] Hankins JS, McCarville MB, Loeffler RB, et al. R2* magnetic res-onance imaging of the liver in patients with iron overload. Blood2009;113(20):4853–5.

42] Carpenter JP, He T, Kirk P, et al. On T2* magnetic resonance and cardiaciron. Circulation 2011;123(14):1519–28.

43] Wood JC. Impact of iron assessment by MRI. Hematol Am Soc HematolEduc Prog 2011;2011:443–50.

44] Zipperer E, Post JG, Herkert M, et al. Serum hepcidin mea-sured with an improved ELISA correlates with parameters of ironmetabolism in patients with myelodysplastic syndrome. Ann Hematol2013;92(12):1617–23.

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

45] Santini V, Alessandrino PE, Angelucci E, et al. Clinical managementof myelodysplastic syndromes: update of SIE, SIES, GITMO practiceguidelines. Leuk Res 2010;34(12):1576–88.

Page 9: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

Oncolo

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

B

sof Beirut Medical Center, Lebanon, where he is also Asso-

S. Temraz et al. / Critical Reviews in

46] Novartis. Desferal® Deferoxamine Mesylate for Injection USP; 2013.http://www.pharma.us.novartis.com/cs/www.pharma.us.novartis.com/product/pi/pdf/desferal.pdf [accessed 15.01.13].

47] De Domenico I, Ward DM, Kaplan J. Specific iron chelators determinethe route of ferritin degradation. Blood 2009;114(20):4546–51.

48] Ghoti H, Fibach E, Westerman M, et al. Increased serum hepcidin lev-els during treatment with deferasirox in iron-overloaded patients withmyelodysplastic syndrome. Br J Haematol 2011;153(1):118–20.

49] Angelucci E, Santini V, Tucci AAD, et al. Deferasirox chelation therapyin transfusion dependent MDS patients. Final report from the GimemaMDS0306 prospective trial. ASH Annu Meet Abstr 2012;120(21):425.

50] Novartis. EXJADE® (deferasirox) Tablets, for Oral Suspension; 2013.http://www.pharma.us.novartis.com/cs/www.pharma.us.novartis.com/product/pi/pdf/exjade.pdf [accessed 15.01.13].

51] Apotex. FERRIPROX® (deferiprone) Tablets, for Oral Use;2013. http://www.accessdata.fda.gov/drugsatfda docs/label/2011/021825lbl.pdf [accessed 15.01.13].

52] Leitch HA, Leger CS, Goodman TA, et al. Improved survival in patientswith myelodysplastic syndrome receiving iron chelation therapy. ClinLeuk 2008;2(3):205–11.

53] Rose C, Brechignac S, Vassilief D, et al. Does iron chelation therapyimprove survival in regularly transfused lower risk MDS patients? Amulticenter study by the GFM (Groupe Francophone des Myélodys-plasies). Leuk Res 2010;34(7):864–70.

54] Neukirchen J, Fox F, Kündgen A, et al. Improved survival in MDSpatients receiving iron chelation therapy – a matched pair analy-sis of 188 patients from the Düsseldorf MDS registry. Leuk Res2012;36(8):1067–70.

55] Lyons RM, Marek BJ, Sharma S, et al. 24-Month analysis of the impactof chelation on clinical outcomes in a 600 patient registry of lower-riskMDS patients. ASH Annu Meet Abstr 2011;118(21):2800.

56] Leitch HA, Chan C, Leger CS, et al. Improved survival with iron chela-tion therapy for red blood cell transfusion dependent lower IPSS riskMDS may be more significant in patients with a non-RARS diagnosis.Leuk Res 2012;36(11):1380–6.

57] Porter J, Galanello R, Saglio G, et al. Relative response of patients withmyelodysplastic syndromes and other transfusion-dependent anaemiasto deferasirox (ICL670): a 1-year prospective study. Eur J Haematol2008;80(2):168–76.

58] Metzgeroth G, Dinter D, Schultheis B, et al. Deferasirox in MDSpatients with transfusion-caused iron overload–a phase-II study. AnnHematol 2009;88(4):301–10.

59] Greenberg PL, Koller CA, Cabantchik ZI, et al. Prospective assessmentof effects on iron-overload parameters of deferasirox therapy in patientswith myelodysplastic syndromes. Leuk Res 2010;34(12):1560–5.

60] Gattermann N, Finelli C, Porta MD, et al. Deferasirox in iron-overloaded patients with transfusion-dependent myelodysplasticsyndromes: results from the large 1-year EPIC study. Leuk Res2010;34(9):1143–50.

61] Gattermann N, Jarisch A, Schlag R, et al. Deferasirox treat-ment of iron-overloaded chelation-naïve and prechelated patientswith myelodysplastic syndromes in medical practice: results fromthe observational studies eXtend and eXjange. Eur J Haematol2012;88(3):260–8.

62] List AF, Baer MR, Steensma DP, et al. Deferasirox reducesserum ferritin and labile plasma iron in RBC transfusion-dependentpatients with myelodysplastic syndrome. J Clin Oncol 2012;30(17):2134–9.

63] Breccia M, Finsinger P, Loglisci G, et al. Deferasirox treatmentfor myelodysplastic syndromes: “real-life” efficacy and safety ina single-institution patient population. Ann Hematol 2012;91(9):1345–9.

64] Nolte F, Höchsmann B, Giagounidis A, et al. Results from a 1-year,open-label, single arm, multi-center trial evaluating the efficacy and

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

safety of oral deferasirox in patients diagnosed with low and int-1risk myelodysplastic syndrome (MDS) and transfusion-dependent ironoverload. Ann Hematol 2013;92(2):191–8.

cRt

gy/Hematology xxx (2014) xxx–xxx 9

65] González FA, Arrizabalaga B, Villegas A, et al. Study of deferoxaminein subcutaneous profusion treatment of iron overload in myelodysplas-tic syndromes. Med Clin (Barc) 2005;124(17):645–7.

66] Kersten MJ, Lange R, Smeets ME, et al. Long-term treatment of trans-fusional iron overload with the oral iron chelator deferiprone (L1): aDutch multicenter trial. Ann Hematol 1996;73(5):247–52.

67] Jensen PD, Heickendorff L, Pedersen B, et al. The effect of iron chela-tion on haemopoiesis in MDS patients with transfusional iron overload.Br J Haematol 1996;94(2):288–99.

68] Gattermann N, Finelli C, Della Porta M, et al. Hematologic responseswith deferasirox therapy in transfusion-dependent myelodysplasticsyndromes patients. Haematologica 2012;97(9):1364–71.

69] Cilloni D, Messa E, Biale L, et al. High rate of erythroid responseduring iron chelation therapy in a cohort of 105 patients affectedby hematologic malignancies with transfusional iron overload: anItalian multicenter retrospective study. ASH Annu Meet Abstr2011;118(21):611.

70] Ginzburg Y, Rivella S. Beta-thalassemia: a model for elucidating thedynamic regulation of ineffective erythropoiesis and iron metabolism.Blood 2011;118(16):4321–30.

71] Nai A, Pagani A, Mandelli G, et al. Deletion of TMPRSS6 atten-uates the phenotype in a mouse model of beta-thalassemia. Blood2012;119(21):5021–9.

72] Li H, Rybicki AC, Suzuka SM, et al. Transferrin therapy amelioratesdisease in beta-thalassemic mice. Nat Med 2010;16(2):177–82.

73] Gardenghi S, Ramos P, Marongiu MF, et al. Hepcidin as a therapeutictool to limit iron overload and improve anemia in beta-thalassemicmice. J Clin Invest 2010;120(12):4466–77.

74] Forni GL, Podesta M, Musso M, et al. Differential effects of the typeof iron chelator on the absolute number of hematopoietic peripheralprogenitors in patients with beta-thalassemia major. Haematologica2012;98(4):555–9.

75] Hartmann J, Braulke F, Sinzig U, et al. Iron overload impairs pro-liferation of erythroid progenitors cells (BFU-E) from patients withmyelodysplastic syndromes. Leuk Res 2013;37(3):327–32.

76] Armand P, Sainvil MM, Kim HT, et al. Pre-transplantation ironchelation in patients with MDS or acute leukemia and iron over-load undergoing myeloablative allo-SCT. Bone Marrow Transplant2013;48(1):146–7.

77] Alessandrino EP, Della Porta MG, Bacigalupo A, et al. Prognosticimpact of pre-transplantation transfusion history and secondary ironoverload in patients with myelodysplastic syndrome undergoing allo-geneic stem cell transplantation: a GITMO study. Haematologica2010;95(3):476–84.

78] Alessandrino EP, Angelucci E, Cazzola M, et al. Iron overload and ironchelation therapy in patients with myelodysplastic syndrome treatedby allogeneic stem-cell transplantation: report from the working con-ference on iron chelation of the Gruppo Italiano Trapianto di MidolloOsseo. Am J Hematol 2011;86(10):897–902.

79] Fenaux P, Adès L. How we treat lower-risk myelodysplastic syndromes.Blood 2013;121(21):4280–6.

80] Mitchell M, Gore SD, Zeidan AM. Iron chelation therapy inmyelodysplastic syndromes: where do we stand? Expert Rev Hematol2013;6(4):397–410.

iography

Dr Ali Taher is a Professor of Medicine at the Divi-ion of Hematology–Oncology of the American University

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

iate Chair for Research and the director of the Residencyesearch Program. In addition, he is a Consultant Hema-

ologist at the Thalassemia Department of the Chronic Care

Page 10: Iron overload and chelation therapy in myelodysplastic syndromes

ARTICLE IN PRESSONCH-1833; No. of Pages 10

1 Oncolo

CloAAHbs�s

lst�itbophilia and bleeding disorders, as well the incidence and

0 S. Temraz et al. / Critical Reviews in

enter in Hazmieh, Lebanon and a fellow of the Royal Col-ege of Physicians and an Adjunct Professor at Emory Schoolf Medicine, Atlanta, USA. Also, he is a member of themerican Society of Hematology, the European Hematologyssociation, the International Society on Thrombosis andemostasis, the Mediterranean League against Thromboem-olic Diseases, and of Alpha Omega Alpha honorary medical

Please cite this article in press as: Temraz S, et al. Iron overload aOncol/Hematol (2014), http://dx.doi.org/10.1016/j.critrevonc.2014.01.0

ociety. His research focuses on hemoglobinopathies, notably-thalassemia and sickle cell disease, as well as thrombo-is and hemostasis. Within thalassemia, his research interest

pi

gy/Hematology xxx (2014) xxx–xxx

ies in the detection of iron overload and the efficacy andafety of novel oral iron chelators. Moreover, he inves-igates the pathophysiology and clinical implications of-thalassemia intermedia and its associated complications

ncluding mostly iron overload and hypercoagulability. Inhrombosis and hemostasis, he investigates inherited throm-

nd chelation therapy in myelodysplastic syndromes. Crit Rev06

rophylaxis of venous thromboembolism across several med-cal and surgical settings.