Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Seminar

Thalassaemia
Ali T Taher, David J Weatherall, Maria Domenica Cappellini

Inherited haemoglobin disorders, including thalassaemia and sickle-cell disease, are the most common monogenic Lancet 2018; 391: 155–67
diseases worldwide. Several clinical forms of α-thalassaemia and β-thalassaemia, including the co-inheritance of Published Online
β-thalassaemia with haemoglobin E resulting in haemoglobin E/β-thalassaemia, have been described. The disease July 31, 2017
http://dx.doi.org/10.1016/
hallmarks include imbalance in the α/β-globin chain ratio, ineffective erythropoiesis, chronic haemolytic anaemia, S0140-6736(17)31822-6
compensatory haemopoietic expansion, hypercoagulability, and increased intestinal iron absorption. The
Department of Internal
complications of iron overload, arising from transfusions that represent the basis of disease management in most Medicine, American University
patients with severe thalassaemia, might further complicate the clinical phenotype. These pathophysiological of Beirut Medical Centre,
mechanisms lead to an array of clinical manifestations involving numerous organ systems. Conventional Beirut, Lebanon
(Prof A T Taher MD); MRC
management primarily relies on transfusion and iron-chelation therapy, as well as splenectomy in specific cases.
Weatherall Institute of
An increased understanding of the molecular and pathogenic factors that govern the disease process have Molecular Medicine, University
suggested routes for the development of new therapeutic approaches that address the underlying chain imbalance, of Oxford, Oxford, UK
ineffective erythropoiesis, and iron dysregulation, with several agents being evaluated in preclinical models and (Sir D J Weatherall MD); and
Department of Clinical Sciences
clinical trials. and Community, University of
Milan, IRCCS Ca’Granda
Introduction sub-Saharan Africa, through the Mediterranean region and Foundation Maggiore
Since the first published description of severe thalass­ Middle East, to the Indian subcontinent and east and Policlinico Hospital, Milan, Italy
(Prof M D Cappellini MD)
aemia over 90 years ago by Cooley and Lee,1 several southeast Asia.8–14 Thus, over 90% of patients with these
Correspondence to:
accounts of the disease have been described and an disorders live in low-income and middle-income countries.
Prof Ali T Taher, Department of
extensive amount has been learnt.2–4 Although the cellular The number of patients with these diseases is expected to Internal Medicine, American
and molecular basis of this group of diseases was initially increase in the coming years as infant mortality from University of Beirut Medical
unknown, in the past 50 years a considerable amount has infectious and nutritional causes declines in many regions Center, PO Box 11–0236,
Beirut 11072020, Lebanon
been discovered to create a substantial body of work.5 of the world. As a result, when discussing management
ataher@aub.edu.lb
Using these resources we now have a refined under­ strategies, we will often focus on the current best practices,
standing of the pathophysiology of the thalassaemia but it is important for clinicians to be aware of the
syndromes. However, despite our understanding of the substantial limitations in implementing even these stan­
pathophysiology, management of these diseases has been dard therapies to many patients around the world. Therefore
complex and is progressing gradually.3,4 Additionally, the we will focus our discussion on the need for improved
available therapeutic routes for thalassaemias and the therapies, as well as on more standardised and easy-to-
complications that result from current treatments are few. implement strategies for use in resource-poor countries. It
Although a great deal of excitement has developed around should also be noted that because of continued migration,
newer therapeutic approaches and potential curative these diseases are now becoming increasingly common in
strategies,6 much remains to be understood about the large multiethnic cities in Europe and North America,
clinical variability of these disorders, the natural history of making it a global health concern.15–18
the thalassaemia syndromes, and the optimal use of the
currently available treatments. In this Seminar, we aim to Molecular and clinical forms
provide an overview of the thalass­aemia syndromes and At the molecular level, haemoglobin synthesis is controlled
comprehensively discuss our current approach to the by two multigene clusters on chromosome 16 (encoding
clinical management of these diseases. As will become
evident, this under­standing is crucial to ensuring that
new therapies can be effectively integrated into the Search strategy and selection criteria
repertoire of existing management strategies. We searched PubMed using the terms “thalassaemia OR
Thalassaemia, sickle-cell disease, and other inherited thalassemia” in combination with “molecular” or
haemoglobin disorders are the most pervasive mono­ ­ “epidemiology” or “diagnosis” or “pathophysiology” or
genic diseases worldwide. The high frequency of in­ “clinical complications” or “treatment OR management”.
herited haemoglobin variants in certain regions reflects We limited our search to publications in English. We mostly
their heterozygote resistance to Plasmodium falciparum selected publications from January, 2006, to May, 2017, but
malaria, and extensive studies7 have shown that this did not exclude frequently referenced and highly regarded
resis­tance is certainly the case for α-thalassaemia, older publications. We also searched the reference lists of
β-­thalassaemia, and haemoglobin E. An estimated 1–5% of articles identified by this search strategy and selected the
the global population are carriers for a genetic thalassaemia most relevant ones. Review articles and book chapters are
mutation.8–14 Although the epidemiology of the various cited to provide readers with more details and more
clinical forms remains poorly recognised, the disease is references than can be addressed in this Seminar.
known to be highly prevalent in the area extending from

www.thelancet.com Vol 391 January 13, 2018 155


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

the α-like globins) and on chromosome 11 (encoding the α-globin to form Hb A (α2β2), the major haemoglobin
β-like globins; figure 1).19,20 The genes in such clusters are component of adult red blood cells. During the first
arranged along the chromosome in the order that they are month of gestation, embryonic haemoglobins β2ε2, α2ε2,
expressed during development to produce different and β2γ2 are formed in erythroid cells located primarily in
haemoglobin tetramers during embryonic, fetal, and adult the yolk sac. During the remainder of fetal life, the sites of
life. The high homology of the α-globin genes can result erythropoiesis gradually shift from the liver and spleen to
in unequal-meiotic cross­over, which is the basis for the the bone marrow, with red blood cells mainly containing
deletions that cause the α-thalassaemias discussed later in Hb F (α2γ2). A switch from γ-globin to β-globin expression
this section. Within the β-globin gene cluster, the ε-gene is begins before birth and completes by the time the baby
expressed only in early embryos, and downstream from reaches 6 months of age. After this time over 95% of the
this gene are two γ-genes, the product of which is found in haemoglobin in normal red blood cells is adult Hb A
fetal haemoglobin (Hb F, α2γ2)—the haemoglobin form (α2β2), with the remaining haemoglobin consisting of two
that predominates throughout most of gestation. The minor components, Hb A2 and Hb F.19,20 Hb F is usually
δ-gene product forms a minor haemoglobin component, confined to a small number of red blood cells known as
Hb A2 (α2δ2), which is useful in the diagnosis of the F cells. Underlying molecular defects in the α-globin or
thalassaemias. The β-gene product combines with β-globin gene clusters form the basis of defective
haemoglobin synthesis and the various inherited forms of
A α-thalassaemias or β-thalassaemias. The type and severity
5’ 3’ of these clinical forms can also rely on additional and
IVS-I IVS-II independent intrinsic and extrinsic factors, which will be
explained in this section. The thalassaemias can also
manifest from co-inheritance of β-thalassaemia and
ξ ψξ ψ α 1 α 2 α 1
5’ 3’ structural haemoglobin variants like haemoglobin S, C,
and E.11,21 Several clinically recognisable forms of
5 kb
16 haemoglobin S/β-thalassaemia and haemoglobin
C/β-thalassaemia exist, each of which have unique
5’ ψβ 2 ε Gγ Αγ ψβ 1 δ β 3’
character­istics and management peculiarities;20 however,
these will not be covered in this seminar.

11 β-Thalassaemia
5’ IVS-I IVS-II 3’
Patients with β-thalassaemia have been typically cate­
gorised as minor, major, or intermedia on the basis of
their α-globin or β-globin chain imbalance, severity of
B
50 anaemia, and clinical picture at presentation. Over
α 200 mutations in the β-globin gene that cause disease
γ β have been recognised, ranging from silent mutations
40
(silent β), to mild mutations that cause a relative reduction
Total globin synthesis (%)

in β-globin chain production (β+), to severe mutations that


30
result in complete absence of β-globin chain synthesis
(β0), with deletions of the gene being uncommon.22
20 ε
β-Thalassaemia minor (trait or carrier) represents the
heterozygous inheritance of a β-thalassaemia mutation,
10 ξ
β with patients often having clinically-asymptomatic-micro­
γ
cytic anaemia, although others can have no identified
6 12 18 24 30 36 1 6 12 18 24 30 36 42 48 haematological abnormalities—so-called silent carriers.
Patients with β-thalassaemia major usually present with
Postconceptual age (weeks) Birth Postnatal age (weeks)
severe anaemia in infancy and become transfusion
Yolk sac Site of erythropoiesis dependent for life, whereas patients with β-thalassaemia
intermedia can present later in life with mild-to-moderate
Liver Bone marrow anaemia and variable transfusion requirements.23–25 Both
β-thalassaemia major and intermedia can result from the
Spleen homozygous or compound-heterozygous inheritance of
mutations in the β-globin gene.
Figure 1: Globin synthesis at the molecular level Several modifications can result in patients having
(A) Organisation of the α-globin family on chromosome 16 and the β-globin family on chromosome 11.
(B) The sites of erythropoiesis and the pattern of globin synthesis during development. ψ designates
β-thalassaemia intermedia rather than major, including
non-expressing pseudogenes. The three exons of the globin genes are shown in pale blue. Reproduced from the extent of the α-globin to β-globin chain imbalance,
Bunn and colleagues19 with permission from McGraw-Hill Education. IVS=intervening segments (or introns). ineffectiveness of erythropoiesis, and the resulting

156 www.thelancet.com Vol 391 January 13, 2018


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

anaemia. The primary modifier is the severity of the Mediterranean region and southeast Asia). The
mutation or mutations in the β-globin gene. Secondary compound heterozygous states for α+-thalassaemia and
modifiers include co-inheritance of α-thalas­saemia, and α⁰-thalassaemia, –α/— or αCSα/—, result in a large excess
increased synthesis of γ-chains and fetal haemoglobin of β-chain production with the formation of β4 tetramers,
production after infancy.22–26 Some rare forms of designated as haemoglobin H. These β4 tetramers are a
β-­thalassaemia intermedia result from deletions removing highly unstable variant of the β-chain and they precipitate
the upstream-regulatory elements, but leaving the globin in red blood cells causing haemoglobin H disease,
genes intact. Other rare mutations linked on the same which is characterised by variably severe haemolysis
chromosome have been important in developing our and consequent anaemia. The homozygous state for
understanding of how the switch from γ-globin to β-globin α⁰-thalassaemia, —/—, results in the production of
gene expression is regulated. All these mutations result in tetramers of γ-chains (γ4) known as haemoglobin Bart’s,
variable concentrations of increased γ-globin expression named after the hospital in London, UK, where it was
and increased concentrations of fetal haemoglobin, leading first discovered. This homozygous state of α⁰-thalassaemia
to what is known as hereditary persistence of fetal is associated with a condition called haemoglobin Bart’s
haemoglobin. Other forms of hereditary persistence of Hydrops fetalis, which is usually characterised by death
fetal haemoglobin result from deletions removing the in utero or just after birth. Rarer causes of α-thalassaemias
adult δ-globin and β-globin genes, but leaving at least include deletions or mutations of regulatory mutations
one γ-globin gene intact (δβ-thalassaemia).19,20 Tertiary involving the α-globin gene cluster.4,17
modifiers include genetic and environmental factors There is another group of α-thalassaemias that, unlike
that alter specific complication rates.23–25,27 β-Thalassaemia those described above, occur in no particular ethnic
intermedia might also result from the increased production groups. They are described under the general title of
of α-globin chains by a triplicated or quadruplicated α-thalassaemia and mental retardation. One thalassaemia
α-genotype associated with β-heterozygosity, or when a in this group is known as ATR-16 syndrome. This
single β-globin locus is affected and the other is completely syndrome involves the whole α-globin gene complex on
normal (dominant inclusion body β-thalassaemia).23–25 chromosome 16 and is associated with mild-to-moderate
When β-thalassaemia is co-inherited with the structural mental retardation. Another member of this group is
variant haemoglobin E (haemoglobin E/β-thalassaemia), known as α-thalassaemia X-linked intellectual disability
which results in β-globin chain synthesis similar to a mild (ATRX) syndrome and results from mutations of the ATRX
β-gene mutation, the resulting clinical forms can also be of gene leading to severe developmental abnormalities, facial
varying severity depending on age and the degree of dysmorphism, and other disorders in addition to its
anaemia at presentation. The clinical forms of haemoglobin α-thalassaemia phenotype. Finally, there is another form of
E/β-thalassaemia are commonly classified as mild, mild hemoglobin H disease that occurs predominantly in
moderate, and severe, with the severe form being similar men and is associated with a form of myelodysplastic
to β-thalassaemia major, and the mild and moderate forms syndrome and pre-leukaemia, which has also been found
being similar to β-thalassaemia intermedia.28,29 The same to be associated with mutations involving ATRX.17
primary, secondary, and tertiary genetic and environmental
modifiers are also relevant in the case of haemoglobin Screening and diagnosis
E/β-thalassaemia, and influence the clinical severity of Screening and prevention programmes for thalassaemia
the disease.30,31 are now widespread, with their adoption being dependent
on regional distribution and cultural factors. Programmes
α-Thalassaemia of nationwide screening, and premarital and neonatal
α-Thalassaemia has two main forms, α+-thalassaemia screenings have been implemented. Confirmation of a
and α⁰-thalassaemia, and their classifications depend thalassaemia diagnosis requires analysis of red blood
on whether one or both of the linked α-globin genes cells combined with haemoglobin electrophoresis, with
are deleted or reduced in activity by mutation. The two DNA analysis required to confirm the diagnosis of
common forms of α+-thalassaemia are designated –α³·⁷ α-thalassaemia and haemoglobin E. Specific guidance on
and –α⁴·² to describe the lengths of the underlying the pathway to establish a thalassaemia diagnosis has
deletions. α+-Thalassaemia has several forms that result been reviewed elsewhere and is available from recent
from point mutations, the most common being caused clinical management guidelines.23,32,33
by the chain-termination mutant haemoglobin Constant
Spring, designated αCSα. In their heterozygous state Clinical classification
these conditions are silent and in their homozygous state Over the past decade, a gradual transition in the labelling
they are characterised by mild-hypochromic anaemia. of the thalassaemias has occurred, moving away from the
α⁰-Thalassaemias are usually caused by the deletion of molecular forms to a more simplified categorisation
both the linked α-globin genes and are designated by the largely based on clinical-management criteria. Transfusion
place of discovery of the first case (eg, –Med and –SEA, therapy remains the basis of management for these
reflecting the first cases to have been found in the disorders, and the frequency and magnitude of transfusion

www.thelancet.com Vol 391 January 13, 2018 157


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

Iron overload Marrow expansion and bone disease

Extramedullary haemopoiesis and organomegaly


Organ damage
α/β-Chain imbalance Ineffective erythropoiesis (heart, liver, endocrine)

Peripheral haemolysis and gall stones

Anaemia Hypercoagulability and vascular disease

Figure 2: Pathophysiology of patients with thalassaemia syndromes

requirements indirectly reflects the underlying severity of having NTDT or TDT, and the role of transfusion therapy
the disease. Moreover, transfusion therapy is not only able in ameliorating much of these pathogenic mechanisms
to control most of the underlying pathophysiological (while introducing other secondary complications) is best
mechanisms but it also contributes a great deal to illustrated in studies showing the different morbidity
secondary morbidity.23,33 Thus, patients today are com­ profile between the two catagories.24,35 The hallmarks of
monly categorised as having transfusion-dependent the disease—with or without treatment—are the α-globin
thalassaemia (TDT)—patients who are not capable of or β-globin chain imbalance leading to ineffective erythro­
producing sufficient haemoglobin to survive without poiesis, an array of subsequent pathophysiological mecha­
blood transfusions—or non-transfusion-dependent nisms, and a multimorbidity profile (figure 2).24,36 In
thalassaemia (NTDT).23,33 However, several points of β-thalassaemia, accumulation of unstable α-globin chain
caution should be highlighted when using such cate­ tetramers in erythroid cells leads to premature cell death
gorisations. For instance, patients with NTDT can still inside (ineffective erythropoiesis) and outside (peripheral
require transfusion therapy sporadically, or even regularly, haemolysis) the bone marrow, facilitated by reactive-
but not for their entire lifetime. This treatment would be oxygen-species formation and structural-membrane de­
for the prevention or management of certain disease formities leading to senescence-antigen exposure.37,38 The
manifestations—and will be illustrated in subsequent immediate resulting manifestation is chronic-haemolytic
sections—whereas patients with TDT require lifelong anaemia that can result in acute compli­cations such as
transfusion treatment for survival. Patients with cholelithiasis and short-term and long-term detrimental
β-thalassaemia intermedia, haemoglobin H disease, and effects on growth, organ, and vascular function.39 In­
mild-to-moderate forms of haemoglobin E/β-thalassaemia creased proliferation of erythroid precursors in the bone
often fall under the classification of NTDT; whereas marrow leads to medullary expansion and subsequent
patients with β-thalassaemia major and severe forms of bone deformities and low bone mass.37 Compensatory
haemoglobin E/β-thalassaemia are classified as having haemo­poietic points out­side the bone marrow are also
TDT.23,24,33 Additionally, it should be noted that some activated, primarily in the spleen (splenomegaly) and liver
patients categorised as having TDT might have been (hepatomegaly), but can also be activated in any body
misclassified. For instance, if transfusions had been tissues with haemopoietic potential, leading to the
started to treat low haemoglobin concentrations as a result formation of extramedullary pseudotumours in high-risk
of an acute complication (eg, an infection) in childhood, anatomic locations such as the spinal canal.40 The ex­
and the patient’s transfusion requirements had not been posure of erythroid cells to senescence antigens, such
revisited and the transfusions thus never halted.34 Also, it as phosphatidylserine, during ineffective erythro­poiesis
should be highlighted that TDT and NTDT are fluid gives them prothrombotic potential. Alongside other
categories based on clinical variables and a patient abnor­ malities in platelets and the coagulation system,
might move from one group to another as a result of hyper­coagulability and associated vascular mani­festations
variations and advances in clinical management, or like venous thrombosis and pulmonary hypertension are
because of changes in other disease modifiers—thus commonly seen in patients with thalassaemia, especially
these designations should primarily represent a patient’s in patients who have been splenectomised and in whom
current clinical status with the understanding that the such markers are further elevated.41 Finally, ineffective
categories are interchangeable.23,24,33,34 erythropoiesis results in increased iron absorption and
primary iron overload mediated by the hepatic hormone
Pathophysiology and associated morbidity hepcidin. An erythroid factor has been postulated as
The underlying disease process in patients with thalass­ communicating to the liver the need of iron for the
aemia remains similar for those patients categorised as incoming red blood cells. This factor would be produced

158 www.thelancet.com Vol 391 January 13, 2018


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

by erythroid cells, especially under the condition of Transfusion


ineffective erythro­poiesis, and acts to suppress hepcidin Transfusion therapy works by supplying normal
synthesis in the liver, leading to increased intestinal iron erythrocytes and suppressing ineffective erythropoiesis,
absorption and increased release of recycled iron from the essentially controlling all downstream pathophysio­
reticulo­endothelial system. Various erythroid factors have logical mechanisms in thalassaemia.49–51 Advances in
been investigated, including growth differentiation factor transfusion and iron-chelation therapy were associated
15 (GDF15), twisted-gastrulation 1 (TWSG1), and erythro­ with improvements in survival in long-term follow-up
ferrone (ERFE); although only ERFE was shown to be studies evaluating different birth cohorts of patients with
increased in animals with β-thalassaemia compared with β-thalassaemia major.52,53 Blood product screening,
those without β-thalassaemia.42,43 Such iron overload preparation, and administration practices have largely
concen­trations can reach clinically significant thresholds improved over the past 3 decades,53,54 and nowadays most
and lead to morbidities in various organ systems, patients with TDT can successfully achieve target
especially the liver.44 Notably, both anaemia and iron haemoglobin concentrations of 90–105 g/L (110–120 g/L
overload can also further worsen ineffective erythropoiesis in those with heart disease); although access to blood for
and complicate patho­ physiology.42,43 In patients with transfusion therapy remains a challenge in resource-
α-thalassaemia, the concepts of α-chain to β-chain poor countries and poses a substantial public health
imbalance and ineffective erythropoiesis is similar to burden.33 In patients with NTDT, however, the indicators
β-thalassaemia. β-Globin chains can form soluble for when blood transfusion is necessary are not as well
tetramers—β4, or haemoglobin H—that precipitate within established, although increased morbidity has been
the cell forming insoluble inclusions (Heinz bodies) that identified in patients with β-thalassaemia intermedia
damage the red blood cell’s membrane. Although who have haemoglobin concentrations of less than
downstream manifestations like primary iron overload 100 g/L.39 Transfusions are sporadically given to patients
and hypercoagulability exist, they tend to be milder in when under acute stress or experiencing a drop in
patients with α-thalassaemia than in patients with haemoglobin concentration, such as during infections,
β-thalassaemia, and the key pathological mechanism is pregnancy, or surgery. Some patients are maintained on
peripheral haemolysis and chronic anaemia, rather than a regular transfusion programme following these
ineffective erythropoiesis, as is seen in patients with incidents—a practice that in many cases might not be
β-thalassaemia.24,36 clinically indicated. Haemoglobin concentrations in
Since adequate and regular transfusion therapy can be isolation should not be an indication for lifelong, regular
considered part of the disease profile of patients with transfusions in patients with NTDT, especially given that
TDT, it is fair to say that the majority of these patho­­ many patients can adapt to their chronic anaemia without
genic mechanisms and associated morbidities are more substantial bone marrow stress.55,56 Nonetheless, a role
commonly encountered in patients with NTDT.24,35 for regular transfusion therapy for a period of months or
Whereas adverse transfusion-related complications, in­ years has been established in patients with NTDT.
cluding secondary-iron overload and subsequent organ Although data have emerged from obser­vational studies
dysfunction, have become hallmarks of clinical mor­ reporting improved growth variables in children, and a
bidity in well transfused patients with TDT.45 Clinical lower proportion of morbidities in regularly transfused
aspects of all such morbidities in patients with thalass­ adults than in those not regularly transfused, especially
aemia are further highlighted in the relevant manage­ in patients with β-thalassaemia intermedia patients,
ment sections below. including leg ulcers, throm­ botic events, pulmonary
hypertension, silent strokes, and extramedullary-­
Conventional management haemopoietic pseudo­tumours.23,34,55–61 Collectively, these
Without intervention, any form of thalassaemia (excluding data might explain the differential clinical picture and
carrier status) is a progressive disease with increased increased proportion of such mor­bidities in patients with
morbidity as the patient advances in age.46 Moreover, the NTDT than in those with TDT.24,25,35 Transfusion therapy,
availability of effective therapeutic options and improved however, does not come without its own side-effects.
patient survival could allow multiple morbidities to Although the risks of alloimmunisation and blood-borne
manifest with age and the quality of life of the patient to infection remain a concern in transfused patients, the
deteriorate.47,48 Adult and elderly patients (older than greatest challenge with regular transfusion therapy is
40 years) with thalassaemia should also be considered at secondary-iron overload.23,33
risk of diseases that are common in non-thalassaemic The human body has no means to excrete iron.
populations, such as cancer and cardiovascular disease. Transfused blood contains around 200–250 mg of iron
Management guidelines for both patients with TDT and, per unit. Patients receiving 2–4 units of blood per month
more recently, NTDT, are available as part of the global will have an annual accumulation of 5000–10 000 mg of
efforts of the Thalassaemia International Federation iron or 0·3–0·6 mg/kg per day.33,62 With continued
to improve the outcomes of patients affected by these transfusions, the reticuloendothelial system—primarily
disorders.23,33 in the liver—can no longer retain all the extra iron.

www.thelancet.com Vol 391 January 13, 2018 159


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

Iron then enters the plasma in amounts that exceed TDT, maintaining concentrations lower than 1000 μg/L of
the transport capacity of circulating transferrin. ferritin is associated with lower morbidity and mortality,
Consequentially, non-transferrin-bound iron exists in and this threshold is most commonly used to indicate the
the plasma as a heterogeneous mixture of iron complexes need for initiation of iron-chelation therapy.33,52,83,84 Serum
that appear to be the major mediators of hepatic and ferritin concentrations consistently higher than 2500 μg/L
extrahepatic tissue damage in transfusional iron are associated with an increased risk of cardiac and
overload.63 Non-transferrin-bound iron enters specific endocrine disease, and thus are commonly used to flag
cells, particularly hepatocytes, cardio­myocytes, anterior the need for dose optimisation or a change in iron-
pituitary cells, and pancreatic β cells. In these cells iron chelation therapy.33,83,84 However, although the association
accumulation leads to the generation of reactive-oxygen of high serum ferritin concentrations with hepatic
species, resulting in damage to lipids, proteins, DNA, siderosis is usually clear, a study85 using imaging tech­
and subcellular organelles, including lysosomes and niques to evaluate organ-specific iron overload failed to
mitochondria. This injury can result in cellular establish an association between serum ferritin concen­
dysfunction, apoptosis, and necrosis that translates into trations and cardiac siderosis. Moreover, single measure­
target organ toxicity and dysfunction.63 The clinical ments of serum ferritin concentration are prone to
burden of iron overload and its management are further mis­interpretation or not being representative of a patient’s
illustrated in the following section. condition, considering their high variability when a patient
is experiencing inflammation, infection, vitamin C
Iron chelation deficiency, hepatic dysfunction, and severe iron overload
Whether iron overload develops from increased intestinal with values higher than 4000 μg/L.33,86 This high variability
absorption—such as in patients with NTDT—or is suggests that, when available and affordable, the use of
secondary to regular transfusions, it can cause substantial imaging to evaluate organ-specific siderosis should be
morbidity and mortality, and so warrants prompt diag­ recommended.33 Worth mentioning is that several case
nosis and effective management. In patients with TDT, reports on patients with NTDT have confirmed that serum
iron accumulation in organ tissues is evident in children ferritin concentrations are commonly lower than in
aged as young as 2–6 years,64–67 with cumulative iron patients with TDT with the same hepatic iron concen­
overload subsequently leading to organ toxic-effects and tration.87 In patients with NTDT (patients with
dysfunction, for instance in the heart, liver, or endocrine β-­thalassaemia intermedia), concentrations of greater
glands.33 Cardiomyopathy has been the leading cause of than 800 μg/L are associated with an increased risk of
death in patients with TDT,33,52 although more recently morbidity, and concentrations of less than 300 μg/L are
large cohort studies68 have also highlighted the high associated with an absence of risk of morbidity. These
incidence of mortality from hepatic causes in patients thresholds are thus used to indicate the need for iron
with TDT. Iron overload in patients with NTDT is a chelation initiation or interruption.23,69
cumulative process with advancing age, and concerns Measurement of liver iron concentration has become
over secondary morbidities start beyond the ages of common practice since the early 2000s, considering the
10–15 years.24,46,69,70 Although cardiac iron overload is strong correlation with total-body iron stores.88 Initial
commonly absent in patients with NTDT,71–74 an association reports relied primarily on the use of liver biopsy, although
between iron overload and hepatic fibrosis has been the technique is now used less frequently and only
observed in patients with β-thalassaemia intermedia,75 reserved for those situations when assessment of hepatic
and the occurrence of hepatocellular carcinoma is histology is needed.89 The use of a Superconducting
increasingly being reported.76 Cumulative iron overload in Quantum Interference Device (SQUID) is also still
patients with NTDT also increases their risk of developing practised, but the technology is only available in few
thrombosis, pulmonary hypertension, silent strokes, centres worldwide.33,90 The introduction of MRI for the
hypo­­thyroidism, hypogonadism, osteo­porosis, and renal non-invasive assessment of liver iron concentration, and
disease, as is evident from studies in patients with later myocardial iron concentration, is considered one of
β-thalassaemia intermedia.61,69,77–81 the most important advances in thalassaemia care over
Several methods are currently available for the diagnosis the past decade. The use of MRI in clinical practice can
and monitoring of iron overload in patients with TDT and allow better tailoring of iron-chelation therapy. For
NTDT. Serum ferritin assessment is widely available and measurement of liver iron concentration, both the R2 and
might be the only assessment that is affordable in T2* techniques are used as they have been validated
resource-poor countries. The assessment of serial serum against liver biopsy measurements of iron.91,92 They are
ferritin concentrations can be a good indicator of iron internationally reproducible and accurately measure liver
chelator effectiveness,33 and available guidelines recom­ iron concentration throughout the clinically relevant
mend measurement of serum ferritin concen­trations range with appropriate calibration and MRI acquisition
every 3 months.23,33,82 Serum ferritin thresholds are techniques, and give a measurement of liver iron
commonly used to indicate the need for initiation or concentration in mg of iron per g of dry liver weight.93,94
modification of iron-chelation therapy. In patients with They are also equally effective in evaluating chronic

160 www.thelancet.com Vol 391 January 13, 2018


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

Deferoxamine Deferiprone Deferasirox


Administration33
Method Subcutaneous or intravenous Oral Oral
Frequency 8–12 h, 5–7 days per week 3 times daily Once daily
Half-life of iron-free drug33 20–30 min 3–4 h 12–16 h
Lipid solubility33 Low Intermediate High
Route of iron excretion33 Urinary and faecal Urinary Faecal
Recommended dose23,33 30–60 mg/kg per day 75–100 mg/kg per day TDT: 20–40 mg/kg per day; NTDT: 5–20 mg/kg per day
TIF guidelines indication
TDT33 >2 years: first-line 2–6 years: no sufficient data; >6 years: second-line† 2–6 years: first-line (USA), second-line (EU); >6 years: first-line
NTDT23 No sufficient data No sufficient data >10 years: first-line
Most relevant clinical data
TDT33 Reduction in serum ferritin and liver Improvement of cardiac T2* in monotherapy or in Reduction in serum ferritin and liver iron concentration after
iron concentration;108 improvement combination with deferoxamine (higher doses than up to 5 years, and cardiac T2* after up to 3 years of therapy,
in cardiac T2*;108 improvement in commonly used in clinical practice)‡;110,111 improvement in even in patients with severe iron overload;115–118 not inferior to
cardiac dysfunction with continuous cardiac dysfunction in combination with deferoxamine†‡;112 deferoxamine for improving cardiac T2*;108 improvements in
infusion109 improvement in endocrine dysfunction in combination hepatic fibrosis and inflammation;119 stabilisation of heart
with deferoxamine or deferasirox113,114 function;108,115 stabilisation of endocrine function120
NTDT23 Data restricted to case series and Data restricted to case series and small studies Significant reduction in serum ferritin and liver iron
small studies concentration after up to 2 years of therapy121
Main adverse events33 Ocular and auditory symptoms, Gastrointestinal symptoms, arthralgia, agranulocytosis or Gastrointestinal symptoms, increased creatinine, increased
bone-growth retardation, local neutropenia hepatic enzymes
reactions, allergy
Pregnancy33 Contraindicated (but has been used Contraindicated Contraindicated
in third trimester)

TDT=transfusion-dependent thalassaemia. NTDT=non-transfusion-dependent thalassaemia. TIF=Thalassaemia International Federation. †In a recent Cochrane review122 it was concluded that in the absence of data
from randomised controlled trials, there is no evidence to suggest the need for a change in the current treatment recommendations. Specifically, deferiprone is indicated for treating iron overload in patients with
thalassaemia major when deferoxamine is contraindicated or inadequate. Intensified deferoxamine treatment, use of other oral iron chelators, or both, remains the established treatment to reverse cardiac
dysfunction due to iron overload. ‡Despite results of some trials110,111 indicating the superiority of deferiprone, or its combination with deferoxamine, compared with deferoxamine monotherapy in improving cardiac
T2*, a meta-analysis123 concluded that deferoxamine and deferiprone monotherapy appeared to reduce myocardial iron by similar amounts. Moreover, the addition of deferiprone to deferoxamine in a recent study124
did not enhance its ability to improve cardiac dysfunction compared to monotherapy. There is an urgent need for high-quality trials to compare the overall clinical efficacy and long-term outcome of treatment with
deferiprone combined with deferoxamine.122

Table 1: Characteristics and evidence on iron chelators for the management of iron overload in thalassaemia

response to iron chelation.95,96 R2 MRI has also been fraction and associated with arrhythmias, whereas values
validated in patients at different stages of liver fibrosis of less than 10 ms are associated with clinical heart failure
and grades of liver inflammation and the technique and mortality.104–106 Thus, these thresholds are commonly
(FerriScan®, Resonance Health, Burswood, WA, used to determine a patient’s iron-overload profile, to
Australia) is approved by the US Food and Drug evaluate their response to iron-chelation therapy in clinical
Administration.94 Cardiac T2* MRI is used for the trials, and to tailor their chelator type and dose in routine
assessment of cardiac iron (values in ms), with recent practice.33,107 In patients with NTDT (β-thalassaemia inter­
validation against myocardial iron concentration in mg of media), liver iron concentration values higher than 5 mg/g
iron per g of dry heart weight from post-mortem cardiac are associated with increased morbidity,78 supporting the
biopsies.97 The use of both techniques to evaluate iron recommendation of iron-chelation initiation in patients
content in other organs—such as the endocrine gland— showing liver iron concentrations greater than 5 mg/g.23 It
has also been investigated, although they are less should be noted that patients with α-thalassaemia do tend
commonly used in routine practice.98–100 Certain liver iron to show slower rates of iron overload than patients with
concentrations and cardiac T2* thresholds have been other NTDT forms, and lower-intensity iron chelation
linked to morbidity in patients with thalassaemia. In standards are usually considered. Most available guide­
patients with TDT, liver iron concentrations greater than lines recommend assessment every 6, 12, or 24 months
7 mg/g are usually used to indicate increased risks of depending on iron-overload severity and iron-chelation
complication, and lower target concentrations are used to adjustment needs.23,33,82
prevent iron-related complications, although this evidence Three iron chelators are currently available for the
has mainly been extrapolated from patients with here­ treatment of iron overload in patients with thalassaemia:
ditary haemochromatosis.101–103 Concentrations greater deferoxamine in subcutaneous or intravenous injection,
than 15 mg/g are associated with progressive liver fibrosis oral deferiprone in tablet or solution form, and oral
and cardiac mortality. Cardiac T2* values of less than deferasirox in dispersible tablet and, more recently, film-
20 ms are indicative of a declining left ventricular ejection coated tablet forms (table 1), and guidelines for their use

www.thelancet.com Vol 391 January 13, 2018 161


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

Splenectomy
Assessment considerations (indication) Management considerations
Splenectomies have traditionally been performed as an
Cardiac dysfunction Echocardiography (routine), As per standard care alternate or adjunct to transfusion therapy. Although
and arrhythmia electrocardiogram (routine)
some studies have found that splenectomies have
Pulmonary Tricuspid regurgitant jet velocity (routine), As per standard care, sildenafil citrate,
hypertension right-heart catheterisation (high risk) bosentan resulted in improvements in growth, quality of life, and
Cerebrovascular MRI and MRA (high risk) As per standard care, antiplatelet
haemoglobin concentration for some patients with
events prophylaxis haemoglobin E/β-thalassaemia.127 The procedure has
Venous thrombosis Standard imaging (suggestive of signs and Anticoagulant therapy, medical and now become almost obsolete in patients with TDT,
symptoms) surgical prophylaxis especially in the context of improved access to and safety
Leg ulcers Physical examination (routine) Topical measures, pentoxifylline, of blood transfusions with regards to blood-borne
hydroxycarbamide, hyperoxygenation infections. The procedure is still more commonly used
Viral hepatitis Viral serology (routine in transfused Hepatitis B vaccination, antiviral in patients with NTDT, because it might be enough to
patients), viral RNA-PCR (if positive serology) therapy
improve mild-to-moderate anaemia and avoid the need
Hepatic fibrosis, Live function tests (routine), ultrasound (high As per standard care
cirrhosis, and risk), α-fetoprotein (high risk), transient for transfusions.25,31,34,57,58 However, data for serious
cancer elastography (investigational) adverse events caused by splenectomies continue to
Endocrine disease Growth retardation (routine), sexual As per standard care accumulate in the thalassaemia patient population,
development (routine), endocrine-function alongside the known risk of infection and sepsis.
tests (routine), bone-mineral density (routine)
Removal of the spleen promotes a hypercoagulable state
Bone disease Bone-mineral density (routine) As per standard care, bisphosphonates
in thalassaemia because of the decreased ability to
Pregnancy As per high-risk pregnancy Revisit iron chelation, anticoagulation scavenge procoagulant red blood cells and activated
prophylaxis, maintenance of
haemoglobin concentration and platelets. Observational studies have further established
heart function that splenectomised NTDT patients are at a 4–5 fold
Extramedullary Physical exam and imaging to rule out Hypertransfusion, radiation, surgery increased risk of overt venous thrombosis and other
haematopoietic compression (suggestive signs and symptoms) vascular manifestations like pulmonary hypertension,
pseudotumours
leg ulcers, and silent strokes than non-splenectomised
Haemolytic crisis Infection screening, electrolytes Adequate hydration, correction of
(haemoglobin H blood electrolytes, control body
patients.60,61 In patients with β-thalassaemia intermedia,
disease) temperature, antibiotics or antivirals the long-term risk of thrombosis is further increased in
those with concomitant high counts of nucleated red
The frequency and age at the start of assessment should rely on the underlying diagnosis and individual patient’s
blood cells (≥300 × 10⁶ cells per L) and platelets (≥500 × 10⁹
needs. Further management details are available in the respective Thalassaemia International Federation guidelines.23,33
MRA=magnetic resonance angiography. platelets per L), and those patients who never received
any transfusions.128 The spleen also usually acts as a
Table 2: Management considerations for specific morbidities in thalassaemia patients beyond
reservoir for scavengers of toxic iron in the body, and so
transfusion and iron chelation
removal of the spleen could also decrease the ability of
these scavengers, thus putting patients at increased risk
are now widely available.23,33,82 Data from several large, of end-organ damage.60 The current recommendations
randomised trials since the development of MRI for splenectomy are restricted to those patients with an
technology showed the efficacy and safety of oral-iron- inability to receive transfusion and iron-chelation therapy,
chelation therapy in removing iron from the liver and as well as those with clinically symptomatic spleno­
heart, which represents a considerable advance in patient megaly or hyper­splenism.23,33 When indicated, appro­
management owing to the greater convenience compared priate treatment with vaccination and antibiotics is
to parenteral deferoxamine. However, parenteral deferox­ recommended, and prophylaxis with aspirin or low-
amine remains the treatment of choice in patients with molecular weight heparins for high-risk patients.23,33
decompensated heart disease, and might be the only
affordable option in resource-poor countries.33 None­ Management of specific morbidities
theless, the search for other novel iron chelators that can Prevention of morbidity in patients with thalassaemia
address the specific efficacy or safety shortcomings of starts with appropriate and adequate application of
these chelators continues.33 Moreover, successful conventional therapies including transfusion or iron
manage­ment of iron overload extends beyond chelator chelation, or both.23,33 Special clinical considerations and
efficacy and safety. Ensuring adherence to treatment is management options beyond conventional therapy are
essential, and applicable to most chelators irrespective of summarised in table 2 for key clinical complications.
their administrative form. Adherence to iron-chelation
therapy correlates with both effective management and Haemopoietic stem-cell transplantation
patient survival, as is evident from several studies.125,126 Replacement of mutant haemopoietic cells using
Moreover, appropriate dose determination and adjust­ haemopoietic stem-cell transplantation is the only
ment of the iron chelator according to baseline and existing curative therapy for thalassaemia and is now an
ongoing iron intake is essential to ensure adequate established approach to correct defective erythropoiesis,
response and avoid overchelation.33 particularly when matched sibling donors are available.

162 www.thelancet.com Vol 391 January 13, 2018


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

Sotatercept TMPRSS inhibitors


Luspatercept
↑ ROS
Iron overload

↑ GDF11
↓ Differentiation Minihepcidins ↓ Hepcidin TMPRSS6

α/β-Chain imbalance Ineffective erythropoiesis Erythroferrone

↑ Proliferation

Anaemia
Gene therapy JAK2 inhibitors ↑ EPO
Gene editing

Figure 3: Novel targets and therapies for patients with β-thalassaemia


EPO=erythropoietin. ROS=reactive-oxygen species.

Haemopoietic stem-cell transplantation is now widely material, such as β-like globin transgenes that can allow
applied with a disease-free survival exceeding 80% for such exogenous gene expression. Following successful
with transplants from HLA-matched-sibling donors.129 application in a few patients with thalassaemia,131 clinical
Moreover, improvements in the management of graft- trials have been initiated.
versus-host disease and inducing graft tolerance have Several approaches are also being developed to address
encouraged the use of unrelated donors and umbilical- the α-globin or β-globin chain imbalance in patients with
cord blood as the haemopoietic stem-cell source for thalassaemia by stimulation of γ-globin production.26
patients who do not have a matched-sibling donor. Although many small trials have examined the use of
Nevertheless, matched-unrelated transplants for high- currently available cytotoxic agents, such as hydroxy­
risk patients with thalassaemia have an overall survival carbamide, there is a scarcity of published results and
of 65%. However, a 5–10% mortality from transplant studies.132 The limitations of the currently available
conditioning, graft-versus-host disease, and graft failure approaches to address the α-globin or β-globin chain
continues to limit the acceptability of this treatment imbalance have motivated a considerable amount of
method. The need for complete myeloablation, which research into better understanding the molecular
can result in infertility and other toxic-effects, is also a regulation of γ-globin. In recent years, human-genetic
concern, although improved approaches with reduced- studies have provided important insights into this area
intensity conditioning are being developed.130 Clinical and have suggested several promising molecular targets,
guidelines specific to patients with thalassaemia are now such as BCL11A, which is a transcription factor capable
available.129 of silencing the γ-globin genes specifically without
affecting production of red blood cells.26 Instead of gene
Future outlook therapy, recent approaches have been developed to
In the past decade several promising targets and directly correct genetic mutations in the endogenous
associated therapeutic options have emerged for patients DNA of the cell or to disrupt specific DNA sequences in
with thalassaemia, though primarily for those with the genome. This approach is known as genome editing
β-thalassaemia, which is reasonable considering the and has been facilitated through the identification of
patient’s more complex and advanced management several enzymes, including CRISPR/Cas9, that can intro­
needs (figure 3).6,42,43 These therapeutic options can be duce DNA breaks in specific regions of the genome.
classified into three major categories on the basis of their Although it is challenging to envision correcting every
attempts to address different aspects of the underlying one of the hundreds–thousands of mutations that cause
pathophysiology of thalassaemia: correction of globin- β-thalassaemia, disruption of factors that silence the
chain imbalance, addressing ineffective erythro­poiesis, γ-globin genes, such as BCL11A, might allow more
and improving how the body handles iron. In this section, immediate treatment of β-thalassaemia with current
we will shortly discuss some of the emerging approaches genome editing approaches.6
in each of these areas. Ineffective erythropoiesis is the major contributor to the
Gene therapy is based on the idea that if there is a defect anaemia observed in patients with β-thalassaemia. In the
in the production of β-globin in β-thalassaemia, exo­ past few years, a group of ligand traps (which include
genous production of β-like globins could correct the sotatercept and luspatercept) have been developed that
disorder. The haemopoietic system is convenient for such have shown potent stimulation of late-stage erythropoiesis
approaches, since haemopoietic stem cells from an and improved ineffective erythropoiesis in patients with
individual with β-thalassaemia can be isolated and trans­ β-thalassaemia.6,133,134 Ongoing clinical studies of these
duced with viruses to introduce exogenous genetic drugs are evaluating their role in reducing transfusion

www.thelancet.com Vol 391 January 13, 2018 163


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

requirements and raising haemoglobin concentrations in Contributors


patients with TDT and NTDT, respectively. JAK2 inhibition, ATT, DJW, and MDC participated in the literature search, manuscript
drafting, and review. All authors approved the final manuscript before
an established therapy in patients with myeloproliferative submission.
disorders, can also ameliorate ineffective erythropoiesis
Declaration of interests
and decrease spleen size in mice with thalassaemia, which ATT and MDC report grants and personal fees from Novartis and Celgene,
has motivated ongoing clinical trials in this area to evaluate outside the submitted work. DJW declares no competing interests.
the role of JAK2 in patients with TDT.43 Acknowledgments
In NTDT mouse models, moderate transgenic hepcidin We thank Khaled Musallam and Vijay Sankaran for their constructive
expression decreased iron loading in the liver, and review of this manuscript. We apologise to those authors whose work
resulted in an extended red blood cell lifespan, increased could not be cited in this review because of space limitations.

haemoglobin concentrations and amelioration of References


1 Cooley TB, Lee P. A series of cases of splenomegaly in children and
splenomegaly.43,135 Recent preclinical studies43,136 have peculiar changes in bones; report of cases. Am J Dis Child 1927;
suggested that long-acting hepcidin analogues (mini­ 34: 347–63
hepcidins) could be beneficial to restrict iron absorption 2 Olivieri NF. The beta-thalassemias. N Engl J Med 1999; 341: 99–109.
and use in the setting of iron overload, with beneficial 3 Rund D, Rachmilewitz E. Beta-thalassemia. N Engl J Med 2005;
353: 1135–46.
effects on ineffective erythropoiesis. If such molecules 4 Piel FB, Weatherall DJ. The α-thalassemias. N Engl J Med 2014;
are developed for clinical use, these could help to further 371: 1908–16.
restrict iron overload either alone or in combination with 5 Sankaran VG, Nathan DG. Thalassemia: an overview of 50 years
of clinical research. Hematol Oncol Clin North Am 2010;
currently available iron chelators. An alternative approach 24: 1005–20.
is to stimulate endogenous hepcidin prod­uction through 6 Sankaran VG, Weiss MJ. Anemia: progress in molecular
the downregulation of a metallo­protease, TMPRSS6, that mechanisms and therapies. Nat Med 2015; 21: 221–30.
plays a key role in hepcidin expression.137 Antisense 7 Weatherall DJ, Williams TN, Allen SJ, O’Donnell A. The population
genetics and dynamics of the thalassemias.
oligonucleotides and siRNAs targeting TMPRSS6 have Hematol Oncol Clin North Am 2010; 24: 1021–31.
been effectively used to stimulate hepcidin, reduce the 8 Weatherall DJ. The inherited diseases of hemoglobin are an
iron burden, and improve in­effective erythropoiesis and emerging global health burden. Blood 2010; 115: 4331–36.
9 Christianson A, Howson CP, Modell B. March of dimes global
red blood cell survival in preclinical iron-overload report on birth defects. The hidden toll of dying and disabled
models138,139—clinical trials are expected soon. children. White Plains, New York: March of Dimes Birth Defects
Foundation, 2006.
Concluding remarks 10 Modell B, Darlison M. Global epidemiology of haemoglobin
disorders and derived service indicators. Bull World Health Organ
Our improved understanding of the pathophysiology 2008; 86: 480–87.
and disease burden in patients with thalassaemia has 11 Weatherall DJ, Clegg JB. The thalassaemia syndromes. 4th edn.
Oxford: Blackwell Science, 2001.
helped optimise disease management and construct a plan
12 Olivieri NF, Pakbaz Z, Vichinsky E. Hb E/beta-thalassaemia:
for the development of novel therapeutics. With several a common & clinically diverse disorder. Indian J Med Res 2011;
treatment options available, the need for comparative and 134: 522–31.
combinative clinical trials is essential to design the best 13 Cohen AR, Galanello R, Pennell DJ, Cunningham MJ, Vichinsky E.
Thalassemia. Hematology Am Soc Hematol Educ Program 2004:
management approach. Nonetheless, the complexity of 14–34.
thalassaemia as a disease might always imply the need for 14 Vichinsky E. Complexity of alpha thalassemia: growing health
individualised therapy, and offering the right treatment problem with new approaches to screening, diagnosis, and therapy.
Ann N Y Acad Sci 2010; 1202: 180–87.
approach to the right patient. The equal prioritisation of 15 Lorey F, Cunningham G, Vichinsky EP, et al. Universal newborn
efforts to promote the prevention of thalassaemia births screening for Hb H disease in California. Genet Test 2001; 5: 93–100.
through nationwide screening programmes and pre­ 16 Lorey F. Asian immigration and public health in California:
thalassemia in newborns in California. J Pediatr Hematol Oncol
marital policy adaptations should also be addressed. 2000; 22: 564–66.
Successful examples of a remarkable decline in disease 17 Harteveld CL, Higgs DR. Alpha-thalassaemia. Orphanet J Rare Dis
incidence from the Mediterranean should aim to be 2010; 5: 13.
reproduced in resource-poor countries where the disease 18 Michlitsch J, Azimi M, Hoppe C, et al. Newborn screening for
hemoglobinopathies in California. Pediatr Blood Cancer 2009;
continues to pose public health concerns. This effort, 52: 486–90.
however, will require challenges attributed to economic, 19 Bunn HF, Nathan DG. Thalassemia. In: Bunn HF, Aster JC, eds.
cultural, and religious differences to be addressed with Pathophysiology of blood disorders. New York: McGraw-Hill, 2011;
93–104.
innovative approaches to premarital and prenatal coun­ 20 Higgs DR, Engel JD, Stamatoyannopoulos G. Thalassaemia.
selling. Expanded-education pro­grammes and health-care- Lancet 2012; 379: 373–83.
system awareness are also needed nowadays in 21 Steinberg MH, Forget BG, Higgs DR, Weatherall DJ. Disorders of
hemoglobin: genetics, pathophysiology, and clinical management.
high-income communities where thalassaemias were, 2nd edn. New York: Cambridge University Press, 2009.
until recently, not commonly present. Lastly, collaborations 22 Danjou F, Anni F, Galanello R. Beta-thalassemia: from genotype to
and partnerships between centres for the exchange of phenotype. Haematologica 2011; 96: 1573–75.
expertise and resources could help improve the diagnosis 23 Taher A, Vichinsky E, Musallam K, Cappellini MD, Viprakasit V.
Guidelines for the management of non transfusion dependent
and management of the thalassaemias, especially in low- thalassaemia (NTDT). Nicosia, Cyprus: Thalassaemia International
income countries where they are most common. Federation, 2013.

164 www.thelancet.com Vol 391 January 13, 2018


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

24 Musallam KM, Rivella S, Vichinsky E, Rachmilewitz EA. 50 Cazzola M, Pootrakul P, Huebers HA, Eng M, Eschbach J, Finch CA.
Non-transfusion-dependent thalassemias. Haematologica 2013; Erythroid marrow function in anemic patients. Blood 1987;
98: 833–44. 69: 296–301.
25 Musallam KM, Taher AT, Rachmilewitz EA. β-thalassemia 51 Cazzola M, De Stefano P, Ponchio L, et al. Relationship between
intermedia: a clinical perspective. Cold Spring Harb Perspect Med transfusion regimen and suppression of erythropoiesis in
2012; 2: a013482. beta-thalassaemia major. Br J Haematol 1995; 89: 473–78.
26 Sankaran VG, Orkin SH. The switch from fetal to adult 52 Borgna-Pignatti C, Rugolotto S, De Stefano P, et al. Survival and
hemoglobin. Cold Spring Harb Perspect Med 2013; 3: a011643. complications in patients with thalassemia major treated with
27 Weatherall D. 2003 William Allan award address. The thalassemias: transfusion and deferoxamine. Haematologica 2004; 89: 1187–93.
the role of molecular genetics in an evolving global health problem. 53 Modell B, Khan M, Darlison M. Survival in β-thalassaemia major in
Am J Hum Genet 2004; 74: 385–92. the UK: data from the UK Thalassaemia Register. Lancet 2000;
28 Galanello R, Origa R. Beta-thalassemia. Orphanet J Rare Dis 2010; 5: 11. 355: 2051–52.
29 Sripichai O, Makarasara W, Munkongdee T, et al. A scoring system 54 Cao A. Quality of life and survival of patients with beta-thalassemia
for the classification of beta-thalassemia/Hb E disease severity. major. Haematologica 2004; 89: 1157–59.
Am J Hematol 2008; 83: 482–84. 55 O’Donnell A, Premawardhena A, Arambepola M, et al. Age-related
30 Fucharoen S, Weatherall DJ. The hemoglobin E thalassemias. changes in adaptation to severe anemia in childhood in developing
Cold Spring Harb Perspect Med 2012; 2: a011734 countries. Proc Natl Acad Sci USA 2007; 104: 9440–44.
31 Olivieri NF. Treatment strategies for hemoglobin E 56 Allen A, Fisher C, Premawardhena A, et al. Adaptation to anemia in
beta-thalassemia. Blood Rev 2012; 26 (suppl 1): S28–30. hemoglobin E-β thalassemia. Blood 2010; 116: 5368–70.
32 Viprakasit V, Tyan P, Rodmai S, Taher AT. Identification and key 57 Olivieri NF, Muraca GM, O’Donnell A, Premawardhena A, Fisher C,
management of non-transfusion-dependent thalassaemia patients: Weatherall DJ. Studies in haemoglobin E beta-thalassaemia.
not a rare but potentially under-recognised condition. Br J Haematol 2008; 141: 388–97.
Orphanet J Rare Dis 2014; 9: 131. 58 Vichinsky E. Advances in the treatment of alpha-thalassemia.
33 Cappellini MD, Cohen A, Porter J, Taher A, Viprakasit V. Blood Rev 2012; 26 (suppl 1): S31–34.
Guidelines for the management of transfusion dependent 59 Taher A, Isma’eel H, Mehio G, et al. Prevalence of thromboembolic
thalassaemia (TDT). 3rd edn. Nicosia, Cyprus: Thalassaemia events among 8,860 patients with thalassaemia major and intermedia
International Federation, 2014. in the Mediterranean area and Iran. Thromb Haemost 2006;
34 Taher AT, Musallam KM, Cappellini MD, Weatherall DJ. 96: 488–91.
Optimal management of β thalassaemia intermedia. Br J Haematol 60 Taher AT, Musallam KM, Karimi M, et al. Overview on practices in
2011; 152: 512–23. thalassemia intermedia management aiming for lowering
35 Taher A, Isma’eel H, Cappellini MD. Thalassemia intermedia: complication rates across a region of endemicity: the OPTIMAL
revisited. Blood Cells Mol Dis 2006; 37: 12–20. CARE study. Blood 2010; 115: 1886–92.
36 Musallam KM, Taher AT, Duca L, Cesaretti C, Halawi R, 61 Musallam KM, Taher AT, Karimi M, Rachmilewitz EA.
Cappellini MD. Levels of growth differentiation factor-15 are high Cerebral infarction in β-thalassemia intermedia: breaking the silence.
and correlate with clinical severity in transfusion-independent Thromb Res 2012; 130: 695–702.
patients with β thalassemia intermedia. Blood Cells Mol Dis 2011; 62 Porter JB. Practical management of iron overload. Br J Haematol 2001;
47: 232–34. 115: 239–52.
37 Rivella S. The role of ineffective erythropoiesis in 63 Brittenham GM. Iron-chelating therapy for transfusional iron
non-transfusion-dependent thalassemia. Blood Rev 2012; overload. N Engl J Med 2011; 364: 146–56.
26 (suppl 1): S12–15. 64 Berdoukas V, Nord A, Carson S, et al. Tissue iron evaluation in
38 Rivella S. Ineffective erythropoiesis and thalassemias. chronically transfused children shows significant levels of iron
Curr Opin Hematol 2009; 16: 187–94. loading at a very young age. Am J Hematol 2013; 88: E283–85.
39 Taher AT, Musallam KM, Saliba AN, Garziadei G, Cappellini MD. 65 Borgna-Pignatti C, Meloni A, Guerrini G, et al. Myocardial iron
Hemoglobin level and morbidity in non-transfusion-dependent overload in thalassaemia major. How early to check? Br J Haematol
thalassemia. Blood Cells Mol Dis 2015; 55: 108–09. 2014; 164: 579–85.
40 Haidar R, Mhaidli H, Taher AT. Paraspinal extramedullary 66 Wood JC, Origa R, Agus A, Matta G, Coates TD, Galanello R. Onset of
hematopoiesis in patients with thalassemia intermedia. Eur Spine J cardiac iron loading in pediatric patients with thalassemia major.
2010; 19: 871–78. Haematologica 2008; 93: 917–20.
41 Cappellini MD, Poggiali E, Taher AT, Musallam KM. 67 Yang G, Liu R, Peng P, et al. How early can myocardial iron overload
Hypercoagulability in β-thalassemia: a status quo. occur in beta thalassemia major? PLoS One 2014; 9: e85379.
Expert Rev Hematol 2012; 5: 505–11; quiz 12.
68 Voskaridou E, Ladis V, Kattamis A, et al. A national registry of
42 Camaschella C, Nai A. Ineffective erythropoiesis and regulation of haemoglobinopathies in Greece: deducted demographics, trends in
iron status in iron loading anaemias. Br J Haematol 2016; 172: 512–23 mortality and affected births. Ann Hematol 2012; 91: 1451–58.
43 Rivella S. β-thalassemias: paradigmatic diseases for scientific 69 Musallam KM, Cappellini MD, Daar S, et al. Serum ferritin level and
discoveries and development of innovative therapies. Haematologica morbidity risk in transfusion-independent patients with
2015; 100: 418–30. β-thalassemia intermedia: the ORIENT study. Haematologica 2014;
44 Musallam KM, Cappellini MD, Wood JC, Taher AT. Iron overload in 99: e218–21
non-transfusion-dependent thalassemia: a clinical perspective. 70 Lal A, Goldrich ML, Haines DA, Azimi M, Singer ST, Vichinsky EP.
Blood Rev 2012; 26 (suppl 1): S16–19. Heterogeneity of hemoglobin H disease in childhood. N Engl J Med
45 Rund D. Thalassemia 2016: modern medicine battles an ancient 2011; 364: 710–18.
disease. Am J Hematol 2016; 91: 15–21. 71 Origa R, Barella S, Argiolas GM, Bina P, Agus A, Galanello R.
46 Taher AT, Musallam KM, El-Beshlawy A, et al. Age-related No evidence of cardiac iron in 20 never- or minimally-transfused
complications in treatment-naïve patients with thalassaemia patients with thalassemia intermedia. Haematologica 2008;
intermedia. Br J Haematol 2010; 150: 486–89. 93: 1095–96.
47 Musallam KM, Khoury B, Abi-Habib R, et al. Health-related quality of 72 Taher AT, Musallam KM, Wood JC, Cappellini MD.
life in adults with transfusion-independent thalassaemia intermedia Magnetic resonance evaluation of hepatic and myocardial iron
compared to regularly transfused thalassaemia major: new insights. deposition in transfusion-independent thalassemia intermedia
Eur J Haematol 2011; 87: 73–79. compared to regularly transfused thalassemia major patients.
48 Haines D, Martin M, Carson S, et al. Pain in thalassaemia: the effects Am J Hematol 2010; 85: 288–90.
of age on pain frequency and severity. Br J Haematol 2013; 73 Roghi A, Cappellini MD, Wood JC, et al. Absence of cardiac
160: 680–87. siderosis despite hepatic iron overload in Italian patients with
49 Cazzola M, Finch CA. Evaluation of erythroid marrow function in thalassemia intermedia: an MRI T2* study. Ann Hematol 2010;
anemic patients. Haematologica 1987; 72: 195–200. 89: 585–89.

www.thelancet.com Vol 391 January 13, 2018 165


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

74 Mavrogeni S, Gotsis E, Ladis V, et al. Magnetic resonance evaluation 95 Wood JC, Enriquez C, Ghugre N, et al. MRI R2 and R2* mapping
of liver and myocardial iron deposition in thalassemia intermedia accurately estimates hepatic iron concentration in
and b-thalassemia major. Int J Cardiovasc Imaging 2008; 24: 849–54. transfusion-dependent thalassemia and sickle cell disease patients.
75 Musallam KM, Motta I, Salvatori M, et al. Longitudinal changes in Blood 2005; 106: 1460–65.
serum ferritin levels correlate with measures of hepatic stiffness in 96 Wood JC, Zhang P, Rienhoff H, Abi-Saab W, Neufeld E. R2 and R2*
transfusion-independent patients with β-thalassemia intermedia. are equally effective in evaluating chronic response to iron
Blood Cells Mol Dis 2012; 49: 136–39. chelation. Am J Hematol 2014; 89: 505–08.
76 Maakaron JE, Cappellini MD, Graziadei G, Ayache JB, Taher AT. 97 Carpenter JP, He T, Kirk P, et al. On T2* magnetic resonance and
Hepatocellular carcinoma in hepatitis-negative patients with cardiac iron. Circulation 2011; 123: 1519–28.
thalassemia intermedia: a closer look at the role of siderosis. 98 Wood JC. Impact of iron assessment by MRI.
Ann Hepatol 2013; 12: 142–46. Hematology Am Soc Hematol Educ Program 2011; 2011: 443–50.
77 Musallam KM, Cappellini MD, Wood JC, et al. Elevated liver iron 99 Noetzli LJ, Panigrahy A, Mittelman SD, et al. Pituitary iron and
concentration is a marker of increased morbidity in patients with volume predict hypogonadism in transfusional iron overload.
β thalassemia intermedia. Haematologica 2011; 96: 1605–12. Am J Hematol 2012; 87: 167–71.
78 Musallam KM, Cappellini MD, Taher AT. Evaluation of the 5mg/g 100 Noetzli LJ, Mittelman SD, Watanabe RM, Coates TD, Wood JC.
liver iron concentration threshold and its association with morbidity Pancreatic iron and glucose dysregulation in thalassemia major.
in patients with β-thalassemia intermedia. Blood Cells Mol Dis 2013; Am J Hematol 2012; 87: 155–60.
51: 35–38. 101 Olivieri NF, Brittenham GM. Iron-chelating therapy and the
79 Ziyadeh FN, Musallam KM, Mallat NS, et al. Glomerular treatment of thalassemia. Blood 1997; 89: 739–61.
hyperfiltration and Pproteinuria in transfusion-independent 102 Angelucci E, Muretto P, Nicolucci A, et al. Effects of iron overload
patients with β-thalassemia intermedia. Nephron Clin Pract 2012; and hepatitis C virus positivity in determining progression of liver
121: c136–43. fibrosis in thalassemia following bone marrow transplantation.
80 Musallam KM, Beydoun A, Hourani R, et al. Brain magnetic Blood 2002; 100: 17–21.
resonance angiography in splenectomized adults with 103 Telfer PT, Prestcott E, Holden S, Walker M, Hoffbrand AV,
β-thalassemia intermedia. Eur J Haematol 2011; 87: 539–46. Wonke B. Hepatic iron concentration combined with long-term
81 Musallam KM, Nasreddine W, Beydoun A, et al. Brain positron monitoring of serum ferritin to predict complications of iron
emission tomography in splenectomized adults with β-thalassemia overload in thalassaemia major. Br J Haematol 2000; 110: 971–77.
intermedia: uncovering yet another covert abnormality. 104 Anderson LJ, Holden S, Davis B, et al. Cardiovascular T2-star (T2*)
Ann Hematol 2012; 91: 235–41. magnetic resonance for the early diagnosis of myocardial iron
82 Musallam KM, Angastiniotis M, Eleftheriou A, Porter JB. Cross-talk overload. Eur Heart J 2001; 22: 2171–79.
between available guidelines for the management of patients with 105 Kirk P, Roughton M, Porter JB, et al. Cardiac T2* magnetic
beta-thalassemia major. Acta Haematol 2013; 130: 64–73. resonance for prediction of cardiac complications in thalassemia
83 Belhoul KM, Bakir ML, Saned MS, Kadhim AM, Musallam KM, major. Circulation 2009; 120: 1961–68.
Taher AT. Serum ferritin levels and endocrinopathy in medically 106 Carpenter JP, Roughton M, Pennell DJ, Myocardial Iron in
treated patients with β thalassemia major. Ann Hematol 2012; Thalassemia Investigators. International survey of T2*
91: 1107–14. cardiovascular magnetic resonance in β-thalassemia major.
84 Olivieri NF, Nathan DG, MacMillan JH, et al. Survival in medically Haematologica 2013; 98: 1368–74.
treated patients with homozygous beta-thalassemia. N Engl J Med 107 Aydinok Y, Porter JB, Piga A, et al. Prevalence and distribution of
1994; 331: 574–78. iron overload in patients with transfusion-dependent anemias
85 Aessopos A, Fragodimitri C, Karabatsos F, et al. Cardiac magnetic differs across geographic regions: results from the CORDELIA
resonance imaging R2* assessments and analysis of historical study. Eur J Haematol 2015; 95: 244–53.
parameters in patients with transfusion-dependent thalassemia. 108 Pennell DJ, Porter JB, Piga A, et al. A 1-year randomized controlled
Haematologica 2007; 92: 131–32. trial of deferasirox vs deferoxamine for myocardial iron removal in
86 Worwood M, Cragg SJ, Jacobs A, McLaren C, Ricketts C, β-thalassemia major (CORDELIA). Blood 2014; 123: 1447–54.
Economidou J. Binding of serum ferritin to concanavalin A: 109 Davis BA, Porter JB. Long-term outcome of continuous 24-hour
patients with homozygous beta thalassaemia and transfusional iron deferoxamine infusion via indwelling intravenous catheters in
overload. Br J Haematol 1980; 46: 409–16. high-risk beta-thalassemia. Blood 2000; 95: 1229–36.
87 Taher A, El Rassi F, Isma’eel H, Koussa S, Inati A, Cappellini MD. 110 Pennell DJ, Berdoukas V, Karagiorga M, et al. Randomized controlled
Correlation of liver iron concentration determined by R2 magnetic trial of deferiprone or deferoxamine in beta-thalassemia major
resonance imaging with serum ferritin in patients with thalassemia patients with asymptomatic myocardial siderosis. Blood 2006;
intermedia. Haematologica 2008; 93: 1584–86. 107: 3738–44.
88 Angelucci E, Brittenham GM, McLaren CE, et al. Hepatic iron 111 Tanner MA, Galanello R, Dessi C, et al. A randomized,
concentration and total body iron stores in thalassemia major. placebo-controlled, double-blind trial of the effect of combined
N Engl J Med 2000; 343: 327–31. therapy with deferoxamine and deferiprone on myocardial iron in
89 Siegel CA, Silas AM, Suriawinata AA, van Leeuwen DJ. Liver biopsy thalassemia major using cardiovascular magnetic resonance.
2005: when and how? Cleve Clin J Med 2005; 72: 199–201, 206, Circulation 2007; 115: 1876–84.
208 passim. 112 Tanner MA, Galanello R, Dessi C, et al. Combined chelation therapy
90 Fischer R, Piga A, Harmatz P, Nielsen P. Monitoring long-term in thalassemia major for the treatment of severe myocardial siderosis
efficacy of iron chelation treatment with biomagnetic liver with left ventricular dysfunction. J Cardiovasc Magn Reson 2008;
susceptometry. Ann N Y Acad Sci 2005; 1054: 350–57. 10: 12.
91 St Pierre TG, Clark PR, Chua-anusorn W, et al. Noninvasive 113 Farmaki K, Tzoumari I, Pappa C, Chouliaras G, Berdoukas V.
measurement and imaging of liver iron concentrations using Normalisation of total body iron load with very intensive combined
proton magnetic resonance. Blood 2005; 105: 855–61. chelation reverses cardiac and endocrine complications of
92 Hankins JS, McCarville MB, Loeffler RB, et al. R2* magnetic thalassaemia major. Br J Haematol 2010; 148: 466–75.
resonance imaging of the liver in patients with iron overload. Blood 114 Farmaki K, Tzoumari I, Pappa C. Oral chelators in
2009; 113: 4853–55. transfusion-dependent thalassemia major patients may prevent or
93 Kirk P, He T, Anderson LJ, et al. International reproducibility of reverse iron overload complications. Blood Cells Mol Dis 2011;
single breathhold T2* MR for cardiac and liver iron assessment 47: 33–40.
among five thalassemia centers. J Magn Reson Imaging 2010; 115 Pennell DJ, Porter JB, Cappellini MD, et al. Deferasirox for up to
32: 315–19. 3 years leads to continued improvement of myocardial T2* in
94 St Pierre TG, El-Beshlawy A, Elalfy M, et al. Multicenter validation patients with β-thalassemia major. Haematologica 2012; 97: 842–48.
of spin-density projection-assisted R2-MRI for the noninvasive 116 Cappellini MD, Bejaoui M, Agaoglu L, et al. Iron chelation with
measurement of liver iron concentration. Magn Reson Med 2014; deferasirox in adult and pediatric patients with thalassemia major:
71: 2215–23. efficacy and safety during 5 years’ follow-up. Blood 2011; 118: 884–93.

166 www.thelancet.com Vol 391 January 13, 2018


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
Seminar

117 Pathare A, Taher A, Daar S. Deferasirox (exjade) significantly 129 Angelucci E, Matthes-Martin S, Baronciani D, et al.
improves cardiac T2* in heavily iron-overloaded patients with Hematopoietic stem cell transplantation in thalassemia major and
beta-thalassemia major. Ann Hematol 2010; 89: 405–09. sickle cell disease: indications and management recommendations
118 Taher A, Cappellini MD, Vichinsky E, et al. Efficacy and safety of from an international expert panel. Haematologica 2014;
deferasirox doses of >30 mg/kg per d in patients with 99: 811–20.
transfusion-dependent anaemia and iron overload. Br J Haematol 130 Angelucci E. Hematopoietic stem cell transplantation in
2009; 147: 752–59. thalassemia. Hematology Am Soc Hematol Educ Program 2010;
119 Deugnier Y, Turlin B, Ropert M, et al. Improvement in liver 2010: 456–62.
pathology of patients with β-thalassemia treated with deferasirox for 131 Cavazzana-Calvo M, Payen E, Negre O, et al. Transfusion independence
at least 3 years. Gastroenterology 2011; 141: 1202–11. and HMGA2 activation after gene therapy of human β-thalassaemia.
120 Casale M, Citarella S, Filosa A, et al. Endocrine function and bone Nature 2010; 467: 318–22.
disease during long-term chelation therapy with deferasirox in 132 Musallam KM, Taher AT, Cappellini MD, Sankaran VG.
patients with β-thalassemia major. Am J Hematol 2014; 89: 1102–06. Clinical experience with fetal hemoglobin induction therapy in
121 Taher AT, Porter JB, Viprakasit V, et al. Deferasirox effectively patients with β-thalassemia. Blood 2013; 121: 2199–212; quiz 2372.
reduces iron overload in non-transfusion-dependent thalassemia 133 Dussiot M, Maciel TT, Fricot A, et al. An activin receptor IIA ligand
(NTDT) patients: 1-year extension results from the THALASSA trap corrects ineffective erythropoiesis in β-thalassemia. Nat Med
study. Ann Hematol 2013; 92: 1485–93. 2014; 20: 398–407.
122 Fisher SA, Brunskill SJ, Doree C, Chowdhury O, Gooding S, 134 Suragani RN, Cadena SM, Cawley SM, et al. Transforming growth
Roberts DJ. Oral deferiprone for iron chelation in people with factor-β superfamily ligand trap ACE-536 corrects anemia by
thalassaemia. Cochrane Database Syst Rev 2013; 8: CD004839. promoting late-stage erythropoiesis. Nat Med 2014; 20: 408–14.
123 Mamtani M, Kulkarni H. Influence of iron chelators on myocardial 135 Gardenghi S, Ramos P, Marongiu MF, et al. Hepcidin as a
iron and cardiac function in transfusion-dependent thalassaemia: therapeutic tool to limit iron overload and improve anemia in
a systematic review and meta-analysis. Br J Haematol 2008; β-thalassemic mice. J Clin Invest 2010; 120: 4466–77.
141: 882–90. 136 Preza GC, Ruchala P, Pinon R, et al. Minihepcidins are rationally
124 Porter JB, Wood J, Olivieri N, et al. Treatment of heart failure in designed small peptides that mimic hepcidin activity in mice and
adults with thalassemia major: response in patients randomised to may be useful for the treatment of iron overload. J Clin Invest 2011;
deferoxamine with or without deferiprone. J Cardiovasc Magn Reson 121: 4880–88.
2013; 15: 38. 137 Nai A, Pagani A, Mandelli G, et al. Deletion of TMPRSS6 attenuates
125 Gabutti V, Piga A. Results of long-term iron-chelating therapy. the phenotype in a mouse model of β-thalassemia. Blood 2012;
Acta Haematol 1996; 95: 26–36. 119: 5021–29.
126 Delea TE, Edelsberg J, Sofrygin O, et al. Consequences and costs of 138 Guo S, Casu C, Gardenghi S, Booten S, Aghajan M, Peralta R, et al.
noncompliance with iron chelation therapy in patients with Reducing TMPRSS6 ameliorates hemochromatosis and
transfusion-dependent thalassemia: a literature review. Transfusion β-thalassemia in mice. J Clin Invest 2013; 123: 1531–41.
2007; 47: 1919–29. 139 Schmidt PJ, Toudjarska I, Sendamarai AK, et al. An RNAi
127 Premawardhena A, Fisher CA, Olivieri NF, et al. Haemoglobin E therapeutic targeting TMPRSS6 decreases iron overload in Hfe(-/-)
beta thalassaemia in Sri Lanka. Lancet 2005; 366: 1467–70. mice and ameliorates anemia and iron overload in murine
128 Taher AT, Musallam KM, Karimi M, et al. Splenectomy and β-thalassemia intermedia. Blood 2013; 121: 1200–08.
thrombosis: the case of thalassemia intermedia. J Thromb Haemost
2010; 8: 2152–58.

www.thelancet.com Vol 391 January 13, 2018 167


Descargado para Anonymous User (n/a) en CES University de ClinicalKey.es por Elsevier en septiembre 17, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.

You might also like