Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

40TH ANNIVERSARY ISSUE

A JH
Thalassemia 2016: Modern medicine battles an ancient
disease
Deborah Rund*

Thalassemia was first clinically described nearly a century ago and treatment of this widespread genetic
disease has greatly advanced during this period. DNA-based diagnosis elucidated the molecular basis of the
disease and clarified the variable clinical picture. It also paved the way for modern methods of carrier
identification and prevention via DNA-based prenatal diagnosis. Every aspect of supportive care, including
safer blood supply, more regular transfusions, specific monitoring of iron overload, parenteral and oral
chelation, and other therapies, has prolonged life and improved the quality of life of these patients.
Significant advances have also been made in allogenic bone marrow transplantation, the only curative
therapy. Recently, there has been a rejuvenated interest in studying thalassemia at the basic science level,
leading to the discovery of previously unknown mechanisms leading to anemia and enabling the
development of novel therapies. These will potentially improve the treatment of, and possibly cure the
disease. Pathways involving activin receptors, heat shock proteins, JAK2 inhibitors and macrophage
targeted therapy, among others, are being studied or are currently in clinical trials for treating thalassemia.
Novel types of genetic therapies are in use or under investigation. In addition to the challenges of treating
each individual patient, the longer survival of thalassemia patients has raised considerations regarding
worldwide control of thalassemia, since prevention is not universally implemented. This review will trace a
number of the original medical milestones of thalassemia diagnosis and treatment, as well as some of the
most recent developments which may lead to innovative therapeutic modalities.
Am. J. Hematol. 91:15–21, 2016. V
C 2015 Wiley Periodicals, Inc.

䊏 Introduction
It is now over 90 years since the first description by Dr. Thomas Cooley of thalassemia in children of Mediterranean origin [1]. Yet, archeologi-
cal evidence [2] and even DNA from remains dating back many generations [3] have suggested that the disease is quite ancient. During the nine
decades following the first clinical description of thalassemia, there have been revolutionary changes in the fields of science and medicine, which
have impacted on the understanding and treatment of the thalassemias. In this review, some of these changes will be discussed, putting into per-
spective the substantial progress which was made both in understanding the pathophysiology of thalassemia and its diagnosis and therapy. This
review will not be all-inclusive, but will touch on the more active and controversial areas, and those with therapeutic implications.

䊏 Diagnosis: From Protein to Genes to Prenatal Diagnosis


Analysis of electrophoresis of hemoglobin provided the first insights into the pathophysiology of the hemoglobinopathies. However, the ability
to analyze genetic information at the level of DNA and RNA led to rapid progress in understanding the disease. By the early 1970s, scientists had
succeeded in isolating mRNA from actual reticulocytes of normal individuals and patients [4], followed by discovery of the quantitative chain
imbalance between the amounts of a- and b-globin RNA [5] in a- and b-thalassemia. Subsequently, restriction enzyme analysis of high molecular
weight DNA with Southern blotting yielded vital information. However, this technique was cumbersome and was unsuitable for analyzing b-
thalassemia, which is generally caused by point mutations. Therefore the development of PCR was truly revolutionary [6]. All of the globin genes
are small, just over 2 kilobases, and as such, are all highly amenable to analysis using PCR-based methodology which has been used to great
advantage for nearly three decades. Various permutations of PCR have revealed hundreds of point mutations causing a- and b-thalassemia, and
gap-PCR can analyze deletions. The large quantity of information amassed on thalassemia mutations has been stored in a freely accessible online
database known as HbVar (http://globin.bx.psu.edu/hbvar). Originally begun in 2001 [7], it was recently updated [8]. This database is one of the
oldest locus-specific databases and is invaluable to clinicians and researchers alike.
The availability of precise genetic diagnostic information enabled progress in diagnosis and furthered understanding the etiology of the varying
severities of thalassemia syndromes. What could crudely be defined as b1- or b0-thalassemia (by the presence of HbA on hemoglobin electropho-
resis) could now be defined by DNA analysis of the specific mutations present in each patient. Subsequently, it became clear that some genotypes
were associated with a variable phenotype, depending on other factors such as fetal hemoglobin levels [9,10]. Furthermore, diseases which are

Hematology Department, Hebrew University-Hadassah Medical Organization, Ein Kerem, Jerusalem, Israel
Conflict of interest: The author has no conflicts of interest to disclose
*Correspondence to: Deborah Rund, Hebrew University-Hadassah Medical Organization, P.O.B. 12000, Ein Kerem, Jerusalem, Israel 91120. E-mail: deborah.
rund@mail.huji.ac.il
Received for publication: 28 September 2015; Revised: 1 November 2015; Accepted: 2 November 2015
Am. J. Hematol. 91:15–21, 2016.
Published online: 4 November 2015 in Wiley Online Library (wileyonlinelibrary.com).
DOI: 10.1002/ajh.24231

C 2015 Wiley Periodicals, Inc.


V

doi:10.1002/ajh.24231 American Journal of Hematology, Vol. 91, No. 1, January 2016 15


Rund 40TH ANNIVERSARY ISSUE

genotypically different could have similar clinical manifestations [11]. thalassemia, but are HLA identical to a previously born affected sib-
This led to reclassification of thalassemia syndromes into transfusion ling [15]. This ensures that the newborn can be a potential transplant
dependent versus nontransfusion dependent thalassemias. The main donor for the sibling patient. Ethical issues have been raised regard-
types of non transfusion dependent thalassemia are b-thalassemia ing the planned selection of such “savior siblings” [19].
intermedia, HbH disease and HbE/ b-thalassemia [11]. While these In summary, advanced sophisticated molecular techniques and the
patients do not require lifelong transfusion, they have significant dis- large database of thalassemia mutations have enabled substantial
ease manifestations requiring therapy. advances in the field of diagnosis and prenatal diagnosis in terms of
DNA-based analysis also facilitated carrier identification, enabling speed, accuracy and noninvasive methodologies. These advances have
determination of couples at risk who could then be referred for early evoked some controversies but have furthered efforts at preventing
and rapid (first trimester) prenatal diagnosis by direct detection of births of affected children.
mutations in fetal DNA. This led to a dramatic fall in birthrates of
patients with severe thalassemia syndromes in many countries where 䊏 Genetic Modifiers
this technology was available [12]. One of the earlier goals of prenatal
diagnosis was to perform it noninvasively using maternal blood sam- Paradoxically the availability of more accurate knowledge at the
ples which contain minute amounts of fetal material. This would DNA level regarding thalassemia mutations led to a clinical conun-
avoid the small but possible risks of fetal injury or even fetal loss dur- drum: patients with the same mutations (sometimes even siblings in
ing invasive methods including amniocentesis and chorionic villus the same family) could have very disparate degrees of clinical severity.
sampling. An early report of the use of maternal peripheral blood This led to the concept of genetic modifiers of thalassemia. Initially,
sampling for prenatal diagnosis of thalassemia was published in 2002 the search for such modifiers was based on linkage analysis of large
[13]. However, this report only screened for one particular mutation family pedigrees [20] or analysis of multiple genes in patients with
which was relatively easy to detect. The methodology was also applied varying thalassemia phenotypes [21]. This subject has been expertly
to deletional a-thalassemia but not sufficiently accurate for most b- reviewed [22]. Besides clarifying and predicting disease severity, the
globin point mutations. Recently, more modern techniques of DNA search for genetic modifiers has another goal: to identify genetic
analysis using next generation sequencing (NGS) were applied toward determinants which can be used for novel therapies, which will be
the goal of noninvasive fetal diagnosis of thalassemia [14]. Using this discussed below.
technique, maternal peripheral blood is drawn at 9 to 10 weeks, Modifiers which directly affect the degree of globin chain imbal-
before chorionic villus sampling can be performed, and analysis is ance are now referred to as “primary modifiers” [22]. These have the
performed for a panel of single nucleotide polymorphisms (SNPs) most notable effects on the level of hemoglobin and on the overall
which are predetermined, using family studies, to be linked to the severity of disease. Ironically, despite advances in genomics, the out-
paternal mutant allele. Using the technique of NGS, the use of four put of the globin genes themselves are still the most potent modifiers.
SNP pairs has been found to be sufficiently accurate to determine if For instance, coinheritance of excess a-globin genes (five or six rather
the fetus is affected. Thus far the technique is only in experimental than four) can worsen the phenotype of individuals who are geneti-
use since it is not diagnostic in all cases and it has required confirma- cally thalassemia trait. These individuals carry only a single b-globin
tion by CVS testing [14]. Yet, it is probable that in the near future, gene mutation but have a phenotype of nontransfusion dependent
this innovative technique will be perfected such that it will be in rou- thalassemia. In contrast, a number of cis or transacting elements
tine use. influence output of the g-globin genes, raising HbF and ameliorating
Accurate DNA diagnosis of fetal DNA has engendered controversy the phenotype of transfusion dependent b-thalassemia [22]. Interest-
regarding the use of preimplantation genetic diagnosis (PGD) of tha- ingly, HbF elevation has been mapped to loci located both inside (the
lassemia [15]. This method involves in vitro fertilization of embryos XmnI polymorphism 5’ to the Gg-globin gene) and outside the b-
and diagnosis using an embryo biopsy or polar body DNA [15]. The globin gene cluster by family and linkage studies [22,23]. More
fetuses identified as healthy are selected for implantation [15]. This recently, the technique of genome wide association studies (GWAS)
method seems ideal for couples with religious or ethical proscriptions has been used to confirm the importance of 2 previously identified
against termination of pregnancy of an affected fetus. However, in loci. One of these is the BCL11A intron 2 polymorphism at chromo-
principle, the same ethical dilemma exists since only a fraction of the some 2p16, and the other is the HBS1L-MYB intergenic polymor-
fertilized embryos are implanted, according to the genetic results. phism at chromosome 6q23 [24]. The advantage of genome wide
Therefore this technique is not permitted in some regions of the association studies is that it enables pinpointing the specific gene
world. Although efforts at perfecting the technique are underway, involved more precisely than linkage studies and furthermore, this
preimplantation genetic diagnosis is not perfect. Using this technique, methodology can potentially reveal associations which were not previ-
the rate of successful pregnancy is at best around 30% [16], which is ously revealed using linkage.
considerably lower than the rate of spontaneous conception in a fer- The subject of genetic modifiers has grown quite complex as vari-
tile couple who would not normally need reproductive intervention
ous disease manifestations have been studied for modifier effects.
other than for embryo selection. Furthermore, it is challenging to
These include the severity of the anemia or of other accompanying
employ this technique for b-thalassemia which is usually due to point
manifestations such as iron overload, the presence of gallstones, and
mutations [17]. Since there are hundreds of such mutations, the
so on. These are referred to as “secondary modifiers” and are related
methodology would need to be perfected for each particular mutation
to any number of genes whose functions relate to various organ sys-
with its different primers and PCR conditions. Since there are only
isolated cells as a template, asymmetric amplification of the different tems [22]. Future studies will determine whether there are treatment
alleles can occur, which can lead to errors in diagnosis. Thus, there implications to these genetic causes for symptom variability.
are intense technical demands on multiple disciplines whose coordi-
nated efforts are required to successfully perform this procedure [18]. 䊏 Conventional Therapy
Furthermore, this technique is considerably less accurate than con-
ventional prenatal diagnosis due to the minute amount of cellular
Prolonging survival: Transfusions and chelation
material available. To add fuel to the controversy, PGD methods have Blood transfusions were given in the 1940s for “Mediterranean
also been used to select fetuses which are not only unaffected by anemia” to prolong the lives of severely anemic children. It was only

16 American Journal of Hematology, Vol. 91, No. 1, January 2016 doi:10.1002/ajh.24231


40TH ANNIVERSARY ISSUE Thalassemia 2016

recognized in 1955 [25] that blood transfusion also suppresses endog- of magnetic resonance imaging (MRI) which can noninvasively mea-
enous hematopoiesis in thalassemia. Since ineffective erythropoiesis is sure liver, cardiac, pancreatic, and even pituitary iron [40]. MRI
one of the major pathogenic mechanisms of disease manifestations measures iron indirectly by the influence of iron on nearby water
(bone deformities, hepatosplenomegaly due to extramedullary hema- molecules. Tissue iron disrupts the magnetic field for a period of time
topoiesis) it became clear about 50 years ago that transfusion was the which can be measured in units of time (designated T2 or T2* by
mainstay of therapy. Since patients are transfusion dependent, they spin echo imaging or gradient echo imaging) or by rate (R2 and R2*
have benefited from advances in blood banking starting with better by spin and gradient echo imaging, respectively) [40]. Quantitation of
crossmatching. Transfusion reactions (some fatal) due to alloantibod- liver iron by R2* has been validated by liver biopsy results [40]. How-
ies were well documented in the older literature. Unfortunately due ever, T2 and T2* have not yet been validated for liver iron concentra-
to differences in ethnicity in donors and recipients of transfusions, tion. A recent meta-analysis [41] found that MRI T2 and T2* are
alloimmunization will undoubtedly continue to persist even in mod- better at excluding liver iron overload than by definitely diagnosing it
ern times [26]. A recent survey found that, even in the industrialized in individual patients. Furthermore, thresholds have not been speci-
world, the practice of transfusion medicine is still variable and could fied and uniformly used, therefore further studies are needed to
be improved and standardized to reduce alloimmunization [27]. standardize T2/T2* protocols for hepatic iron determination [41].
It is clear that thalassemia patients born in the industrialized world Since cardiac mortality is the major cause of mortality in nonchelated
in the last two to three decades have benefited from modern stand- patients, cardiac T2*, which has been validated clinically [40] has
ards of safe blood banking with much reduced pathogen transmis- become the standard of care in industrialized countries. As such, the
sion. Transfusion transmitted diseases have dealt a lifelong blow to use of cardiac MRI has contributed to prolonging life expectancy in
patients who began receiving transfusions prior to testing for hepatitis countries where this tool is available, such as the UK and other parts of
and/or HIV. In the industrialized world, these are either the oldest Europe [42]. An algorithm has been developed for routine use of car-
patient cohort or immigrants who began their transfusion history in diac MRI to guide long term optimum chelation [40]. Ferritin remains
countries where testing for pathogens is not yet standard practice useful in young children, for whom the use of MRI requiring sedation is
[26]. Besides their immediate impact on the patient’s health, transfu- problematic and needs to be weighed individually [33].
sion transmitted infections can lead to late complications such as cir-
rhosis and/or hepatocelluar carcinoma (HCC). Approximately 85% of Hypercoagulability
thalassemia patients with HCC in a recent Italian study showed evi- One of the factors long known to contribute to mortality and mor-
dence of infection with hepatitis B virus or hepatitis C virus [28]. bidity in thalassemia is the chronic hypercoagulability of this disease,
Hemosiderosis was long recognized as a complication of prolonged particularly in splenectomized patients [43]. The contribution of
transfusion therapy and a half a century ago the first reports of the hypercoagulability to the mortality and morbidity of thalassemia
use of desferrioxamine in thalassemia were published [29,30]. Three patients has long been understood [44]. Laboratory studies have eluci-
decades ago, desferrioxamine was shown to prevent many of the dated the many factors contributing to hypercoagulability of this dis-
complications of iron overload [31]. However, desferrioxamine is not
ease [45]. Of concern, hypercoagulability can contribute to cerebral
suitable for all patients and even early on, investigators were working
ischemia which may initially be clinically silent [46]. Nonetheless
on new chelators [32]. At the present time there are three chelators,
despite the importance of these findings, there are still no clearcut
two oral and one parenteral, in routine use, all of which are effective
guidelines on prevention of these complications [45], despite the
to varying degrees and which have differing adverse effects [33,34].
availability of many new antithrombotic agents. Clearly this is an area
The availability of orally active chelators has greatly improved com-
in which further clinical research is urgently needed.
pliance which is still an issue for patients requiring this treatment on
a lifelong basis. New combination therapy approaches and intensifica- Improving quality of life
tion of dosing are promising in terms of increased efficacy [33].
Prolonging life in thalassemia has been accomplished. However, a
While long hypothesized but not absolutely proven [35], recent in
vitro evidence demonstrates synergistic chelation efficacy using a longer life is not necessarily a better life, and unfortunately, the quality
combination of different types of chelators [36]. In summary, half a of life for these patients can be poor. This may be due to discomfort
century of arduous clinical, pharmacological, and laboratory studies from the disease and its therapies, psychological issues, or to low func-
have not yet led to a simple solution to the need for chelation, and tional capacity due to chronic anemia. Recently more attention has
many problems exist in “real life” practice [37]. Even patients with been paid to these types of issues. Among the areas receiving attention
access to all available drugs, chelation is not always optimal [37]. Yet is the chronic bone pain and osteoporosis which can be quite debilitat-
despite these problems, chelation has significantly prolonged the lives ing in adult patients [47]. The pathogenesis is multifactorial, from bone
of thalassemia patients who can obtain this treatment and who com- expansion due to hyperactivity of the bone marrow, to hypogonadism
ply with its use [38], mainly by reducing early death from cardiac [48], and even iron overload is contributory [49]. Recently, an expert
events due to iron overload. panel has published recommendations for surveillance and therapy
Improvements in chelation led to the need for more accurate mon- which are quite welcome, as this area has previously been one of contro-
itoring of the efficacy of iron chelation therapy. Serum parameters versy. In particular, it is encouraging that the use of biophosphonates
(serum iron and transferrin saturation) gave way to the widely avail- shows beneficial results to date [50,51], since in the past, these drugs
able serum ferritin, which is inexpensive and useful for routine clini- have been questioned regarding their safety in thalassemia.
cal assessment. However ferritin is subject to many variations, and Recently, concern has been expressed regarding the presence of
even when measured frequently, may not accurately reflect tissue iron neurological complications of thalassemia. As life expectancy
burden [39]. Recently, scientific studies have led to better understand- increases, chronic hypoxia, thromboembolic phenomena, neurotoxic
ing of iron trafficking, including how iron accumulates in tissues drugs, and other factors, can cause damage which may manifest as
which were not intended to store iron [40]. These studies have eluci- clinically apparent (such as cognitive impairment or peripheral neu-
dated the need to monitor not only serum parameters but also tissue ropathy) or subclinical findings (abnormal evoked potentials) [52].
iron: hepatic iron and even iron in other tissues. Liver biopsy (which This is an important area for future preventative research, in order to
is invasive) was previously considered the “gold standard” but both optimize the level of functioning and quality of life of thalassemia
only measured hepatic iron stores. Currently, there are several types patients.

doi:10.1002/ajh.24231 American Journal of Hematology, Vol. 91, No. 1, January 2016 17


Rund 40TH ANNIVERSARY ISSUE

Bone marrow transplantation to date have generally been disappointing. Recently, greater compre-
hension of the regulatory pathways of HbF expression, including the
Allogeneic bone marrow transplantation (alloBMT) is curative for
crucial influences of BCL11a and MYB, has suggested new therapeutic
thalassemia. Indeed, it has been performed since 1982, with continu-
targets for raising HbF [62]. These include interfering with BCL11a
ous advances in the field in the past decades [53]. There have been
or MYB, which both directly repress g-globin gene expression. These
many issues to resolve in order to achieve successful alloBMT specifi-
potential targets were revealed by studies, mentioned above, of genetic
cally in thalassemia. All patients are heavily transfused leading to
modifier loci on chromosome 6 [66]. However, there are presently no
alloimmunization and a high risk of graft rejection. End organ dam-
specific drugs which can target these pathways. There are several rea-
age from iron overload increases risk of toxicity from conditioning
sons for this, including the broad effects on hematopoiesis exerted by
regimens. The stratification of risk by Lucarelli predicted poor out-
both BCL11a and MYB. Therefore, inhibiting either one could prove
come and enabled selection of those suitable for alloBMT. More
to be problematic. Currently, research in animal models is aimed at
recently, even high risk patients have been successfully transplanted
finding erythroid specific methods of targeting these regulators for
[54]. For patients at high risk, conditioning needs to be titrated to
the treatment of thalassemia [62].
reduce toxicity and at the same time, prevent graft rejection. Condi-
tioning regimens for thalassemia have recently been reviewed [55]. Activin receptor ligand traps
Reduced intensity conditioning had generally had disappointing
Despite all that is known about erythropoiesis, new and important
results due to autologous recovery [56]. However, more favorable
regulatory pathways are still being discovered. Such is the case with
results were recently reported using reduced intensity conditioning
the GDF11 pathway. This gene had been studied predominantly for
[57].
its role in organs such as pancreas, intestine, kidney, skeletal muscle,
In the industrialized world, the limiting factor for alloBMT for tha-
lassemia has been donor availability. Traditionally, fully matched and developing nervous system. Knockout gdf112/2 mice develop
HLA identical sibling donors were used but improved outcome was skeletal and gut abnormalities [67]. A role for this gene in human
reported with matched unrelated [53] and with single locus mismatch erythropoiesis was found indirectly and serendipitously. The first clue
related (not sibling) donors [58]. Recently, success has even been came during a trial of a novel drug designed to treat osteoporosis, in
reported using unrelated cord blood, including with reduced intensity which it was noted that the subjects developed an average of 12% rise
conditioning [59]. The ability to use unrelated cord blood consider- in Hb [68]. This was unexpected and it quickly led to the hypothesis
ably enlarges the donor pool available for each patient. that this drug, sotatercept, could be used to treat anemia. Sotatercept
AlloBMT is costly and has not been available in most developing was developed to treat osteoporosis by targeting activin signaling,
countries for this and other technical reasons. However, recent efforts which inhibits osteoclast dependent bone resorption. Activin belongs
have been made to foster international cooperation between industri- to the TGFb superfamily. It binds to various receptors which initiate
alized and developing countries to increase the utilization of this ther- a cascade culminating in phosphorylation of SMADs which activate
apy in the developing world. It has been suggested that alloBMT is gene expression. Sotatercept acts as an activin IIA receptor ligand
preferable to routine supportive care, since safe blood products and trap. Ligand traps are molecules that inhibit signaling by binding
chelation are not readily available in many parts of the world. In ligands and preventing them from interacting with their receptors. A
addition, it has been suggested that the cost could be reduced to murine version of sotatercept was tested in a mouse model of thalas-
make it economically feasible [60]. Considering the high expenditures semia [69] and the results were remarkable. Not only was there
associated with standard lifelong supportive care, alloBMT could be increased terminal differentiation of erythroblasts, with correction of
financially tenable. International cooperation is required to increase maturation arrest, but RBC morphology improved, with fewer a-
access to alloBMT in areas with the greatest need [61]. globin precipitates on RBC membranes. Furthermore, splenomegaly
decreased. Remarkably, hepcidin levels also increased and reduced
䊏 Novel Therapies Already Here or on iron and transferrin saturation were found, which was independent of
GDF15 expression. This is significant since GDF15 has been sus-
the Horizon pected of being the culprit in downregulating hepcidin in thalassemia,
While thalassemia was once on the forefront of genetic and scien- leading to iron overload [70]. The role of GDF11 in these beneficial
tific research, in the last several decades it fell behind while major effects was elucidated by a series of elegant molecular studies which
strides were made in the comprehension and therapy of malignancies. were initially based on results of gene expression studies in the fetal
In recent years, thalassemia has returned to the forefront of science. livers of mice with thalassemia major. These results, found in an
The study of anemia in general [62] and thalassemia [63] and the b- online database dating 2012 (from the laboratory of Dr. Mitchell
hemoglobinopathies in particular [64], have led to increased under- Weiss), showed that the murine gdf11 gene was the only gene overex-
standing of molecular mechanisms underlying these diseases. Thalas- pressed in the fetal liver of thalassemic mice at embryonic day 14.5.
semia has thus become a paradigm for discoveries regarding the From there, additional experiments and deduction led to the role of
pathophysiology of anemia and potential novel therapies. Three excel- the activin type IIA receptor [69].
lent reviews comprehensively discuss potential (or actual) novel tar- Luspatercept is a compound which is very similar to Sotatercept
gets for therapies for anemia, including thalassemia [62–64]. This and differs in being an activin IIB (not IIA) receptor ligand trap. Lus-
review will only mention seven of these newer therapeutic avenues patercept has been assessed in a Phase I study in normal human vol-
which are aimed at alleviating the anemia and/or iron overload. Some unteers [71] and in a Phase 2 study in thalassemia patients [72]. In
are already in clinical use, others are in clinical trials and some are both normal individuals and patients, hemoglobin rose and side
potential mechanisms which could be targeted for therapeutic pur- effects were tolerable. Further results of ongoing studies of this excit-
poses in the future. ing novel agent are eagerly awaited.
Fetal hemoglobin induction JAK2 inhibitors
HbF induction has been known for nearly four decades. A number Elevation of erythropoietin levels, as seen in thalassemia and other
of agents have been used with varying success to pharmacologically anemias, increase the number of erythroid precursors carrying phos-
raise HbF with tolerable adverse effects (reviewed in Ref. 65). Results phorylated Jak2 [73]. This is seen in murine models and in

18 American Journal of Hematology, Vol. 91, No. 1, January 2016 doi:10.1002/ajh.24231


40TH ANNIVERSARY ISSUE Thalassemia 2016

thalassemia patients, and it is thought to contribute to ineffective (and still open) clinical trials for gene therapy of thalassemia [81].
erythropoiesis, with massive erythroid hyperplasia and extramedullary Unforeseen genetic effects were noted in the first successful patient
hematopoiesis [74]. Therefore it is plausible to administer JAK2 treated: the integrated lentiviral vector caused transcriptional activa-
inhibitors to reduce splenomegaly and ineffective erythropoiesis in tion of HMGA2 in erythroid cells [82], thus this vector had to be
nontransfusion dependent thalassemia [63,73]. While this may seem altered. The modified vector has proven successful in the treatment
counterintuitive, as these drugs can cause anemia and other cytope- of two adolescents with transfusion dependent thalassemia, who have
nias, this approach is currently being tested in clinical trials for been transfusion free for 11 to 14 months following autologous trans-
selected patients. plantation using stem cells corrected with the lentiviral vector [83]
The subject of vectors has been expertly reviewed in great detail [84],
Macrophage targeted therapy including vectors designed for simple gene replacement, newer con-
The concept of the bone marrow microenvironment, known to be structs such as vectors aimed to overcome silencing of the g-globin
important in many hematological diseases, is increasingly understood genes, and vectors for genome editing. This newest genetic modifica-
to be a crucial factor in erythropoiesis [75]. The erythroid island has tion therapy aims to directly repair mutations at the DNA level in
a central macrophage which interacts with surrounding erythroblasts, hematopoietic stem cells. While this would be ideal, this approach
and provides regulatory feedback as erythropoiesis progresses, as well has a low level of efficiency of transfecting hematopoietic stem cells
as supplying nutrients and phagocytosing extruded erythroblast in addition to low efficiency of the homologous recombination neces-
nuclei. The microenvironment has a particularly important role in sary for the correction of the mutation(s). Though gene editing will
the stress erythropoiesis which characterizes thalassemia, with extra- not be clinically applicable in the near future, a recent report suggests
medullary hematopoiesis and expansion of the erythroid progenitor that these shortcomings can be overcome [85].
pool [76]. Unexpectedly, macrophages (previously benevolently
known as “nurse cells”) have been found to exert a negative effect on Therapies to prevent iron overload: Modulation of
hematopoiesis in thalassemia. This has been demonstrated in a recent hepcidin
study in which pharmacological macrophage depletion (by injecting While chelation is an effective and proven therapy, the ideal solu-
clodronate liposomes) resulted in marked improvement of clinical tion for iron overload would be its prevention. In addition to transfu-
features in a murine model of thalassemia intermedia [77]. There was sional iron overload, thalassemia patients have hyperabsorption of
improvement in splenomegaly, reduction in ineffective erythropoiesis, iron due to insufficient production of the peptide hepcidin. Hepcidin
increased Hb with improved RBC morphology (including reduced expression is decreased in response to ineffective erythropoiesis, as is
accumulation of a-globin chains in the RBC membrane), and found in thalassemia. Therefore inappropriately low levels of hepcidin
improved iron kinetics [77]. (Astonishingly, the identical macrophage cause deleterious iron hyperabsorption in thalassemia patients. Several
depletion treatment ameliorated the phenotype of P. vera mice) [77]. tactics have been attempted to compensate for hepcidin insufficiency,
Thus bone marrow macrophages regulate not only normal but patho- including the use of hecpidin mimetic agents (“minhepcidins”), trans-
logical erythropoiesis [78] and could be a target for therapeutic inter- genic overexpression of hepcidin, and repression of TMPRSS6 (phar-
vention in thalassemia. macologically or using genetic methods such as lipid nanoparticle
encapsulated siRNA). TMPRSS6 is one of the key upstream regula-
Heat shock protein 70 (HSP70) tory proteins involved in upregulating hepcidin expression These and
Over 20 years ago, it was noted that thalassemic erythroid precur- other methods have been recently summarized [86]. Interestingly, in
sors underwent accelerated apoptosis while normal precursors did not animal models of thalassemia intermedia, methods leading to
[79]. Studies of normal erythropoiesis have led to increased under- increased hepcidin also had a beneficial effect on ineffective erythro-
standing of these processes. GATA-1, an essential erythroid transcrip- poiesis, with improvement of anemia and reduction of splenomegaly
tion factor, is cleaved by transiently active caspases during late in these animals [86].
erythroid differentiation. In erythroid precursors undergoing normal
terminal differentiation, GATA-1 is protected from premature caspase
action by the chaperon protein, heat shock protein (HSP) 70. This
䊏 The Future of Thalassemia: Will
protein translocates to the nucleus and protects GATA-1 from Modern Medicine Win the Battle?
caspase-mediated proteolysis. However, in b-thalassemia, HSP70 In summary, there are many exciting new developments in the
interacts with free a-globin chains in the cytoplasm, which sequester field of thalassemia at the diagnostic but more importantly, at the
it there and prevent it from translocating to the nucleus to protect therapeutic level. The pace at which new diagnostic and therapeutic
GATA-1. This results in maturation arrest and apoptosis [80]. developments have been developed and clinically applied is rapidly
Genetic manipulations such as transfecting an uncleavable GATA-1 increasing (Table I). Over the last four decades, the life expectancy
or an HSP70 targeted to the nucleus, restored normal maturation of for thalassemia patients in the industrialized world has increased dra-
thalassemia erythroblasts [80]. These findings may be applicable to matically. In the United Kingdom, the average life expectancy in 1970
the development of novel therapies directed at free a-globin chains was 17 years, in 1980 it was 27 years and in 1990, it was 37 years,
or HSP70. more than doubling of the life expectancy in two decades [42]. Since
2000, over 80% of thalassemia patients in the United Kingdom could
Genetic therapies
be expected to live longer than 40 years and the actual upper lifespan
Since the b-globin gene was cloned, scientists have been striving limit cannot be defined due to continued improvements over time
toward the goal of gene therapy to replace nonfunctional b-globin [42]. Lifespan and hopefully, quality of life, should continue to
genes. Though conceptually simple, this has proven to be an onerous improve in the industrialized world.
task due to many initially unforeseen factors. These include factors Unfortunately, most of the techniques for diagnosis, proper treat-
related to the various vectors, to the complex regulation of expression ment, and prevention of thalassemia, utilize sophisticated technology
of globin genes, as well as the very high level expression which was and/or are very costly. It is well known that the world’s distribution
needed to correct the genetic defects. At the present time, a handful of thalassemia patients is predominantly in developing countries
of patients in several different centers have been treated on approved which do not have the resources (financial and technological) to

doi:10.1002/ajh.24231 American Journal of Hematology, Vol. 91, No. 1, January 2016 19


Rund 40TH ANNIVERSARY ISSUE

TABLE I. Thalassemia: From Early Milestones to the Present and Future

Year Milestone Current status

1925 Cooley’s clinical description DNA-based mutational diagnosis


1946 First transfusion clinic for thalassemia Routine transfusions widely available
1973 Parenteral chelation: desferrioxamine Parenteral and oral chelation widely available;
hepcidin modulation for prevention of iron overload
1975 Liver biopsy as indicator of transfusional hemosiderosis No longer needed
1976 Second trimester DNA-based prenatal diagnosis First trimester DNA-based diagnosis widely
for certain genotypes available for virtually all genotypes;
investigational noninvasive diagnosis at
9 to 10 weeks using maternal blood sample
1982 Allogeneic BMT Routinely available for suitable patients
1984 MRI for liver iron overload MRI simultaneous measurement of heart,
liver, and pancreatic iron content
1985 Pharmacologic HbF induction Butyrates in clinical trials, novel agents
under development
1989 Preimplantation genetic diagnosis Available for selected cases
2005 Gene therapy Clinical trials with improved vectors
2014 Novel therapies: activin ligand traps, JAK-2 inhibitors In early clinical trials
2016 Additional novel therapies Under development

Dates are approximate in some cases and refer to the earliest approved human (not animal) use.

adopt the use of most of these modalities. Tragically, there are many with thalassemia syndromes. More patients are surviving into late
countries in which the most basic routine care of transfusion and childhood and adulthood [88], thus increasing the number of thalas-
chelation are either nonexistent or woefully underutilized due to semia patients requiring long term therapy. All of these factors
financial constraints. To those in the industrialized world, it is shock- increase the likelihood that thalassemia will constitute a global health
ing to read that only about 12% of children born with thalassemia problem in the coming decades [88].
worldwide are transfused and of these, only about 40% are adequately Modern medicine can partially alleviate the suffering of thalassemia
chelated [86]. Therefore, the survival of thalassemia patients in low patients and increase the implementation of prevention in regions
income countries at present is similar to that in the Europe and the where the standard of care is already high. However, it cannot van-
United States 50 years ago [87]. Annually about 1.33 million preg- quish thalassemia on a global basis. Thalassemia is, now more than
nancies worldwide are at risk for thalassemia [86] but only a small ever, a disease of imbalance. At the cellular level, there is chain syn-
fraction have access to genetic counseling and prenatal diagnosis. Fur- thesis imbalance, and at the societal level, there is a critical imbalance
thermore, the problem of prevention and treatment of thalassemia is between resource availability and resource need. Ultimately, social
presently a worldwide issue, due to migration of individuals from policy makers and public health practitioners must enact measures to
higher prevalence regions to areas of lower prevalence [88]. Since maximize prevention of the disease, and provide optimal care for tha-
infection has always been a cause of death of thalassemia patients lassemia patients. These are both sorely lacking in much of the world,
[42,88], antibiotic therapy has prevented many deaths of children even in the year 2016.

䊏 References Res 2014;42:D1063–D1069. doi:10.1093/nar/


gkt911
19. Dickens BM. Preimplantation genetic diagnosis
and ’savior siblings.’ Int J Gynaecol Obstet 2005;
1. Cooley T, Lee P. A series of cases of splenomeg- 9. Wainscoat JS, Thein SL, Weatherall DJ. Thalas- 88:91–96.
aly in children with anemia and peculiar bone saemia intermedia. Blood Rev 1987;1:273–279. 20. Thein SL, Sampietro M, Rohde K, et al. Detec-
changes. Trans Am Pediatr 1925;37:29. 10. Rund D, Rachmilewitz EA. Beta thalassemia. N tion of a major gene for heterocellular heredi-
2. Angel JL. Porotic hyperostosis, anemias, Engl J Med 2005;353:1135–1146. tary persistence of fetal hemoglobin after
malarias, and marshes in the prehistoric Eastern 11. Weatherall DJ. The definition and epidemiology accounting for genetic modifiers. Am J Hum
Mediterranean. Science 1966;153:760–763. of non-transfusion-dependent thalassemia. Blood Genet 1994;54:214–228.
3. Filon D, Faerman M, Smith P, Oppenheim A. Rev 2012;26:S3–S6. 21. Rund D, Oron-Karni V, Filon D, et al. Genetic
Sequence analysis reveals a beta-thalassaemia 12. Cao A. Results of programmes for antenatal analysis of beta-thalassemia intermedia in Israel:
mutation in the DNA of skeletal remains from detection of thalassemia in reducing the inci- Diversity of mechanisms and unpredictability of
the archaeological site of Akhziv, Israel. Nat dence of the disorder. Blood Rev 1987;1:169– phenotype. Am J Hematol 1997;54:16–22.
Genet 1995;9:365–368. 176. 22. Thein SL. Genetic association studies in b-
4. Nienhuis AW, Anderson WF. Isolation and 13. Chiu RW, Lau TK, Leung TN, et al. Prenatal hemoglobinopathies. Hematology Am Soc Hem-
translation of hemoglobin messenger RNA from exclusion of beta thalassaemia major by exami- atol Educ Program 2013;2013:354–361.
thalassemia, sickle cell anemia, and normal nation of maternal plasma. Lancet 2002;360: 23. Menzel S, Garner C, Gut I, et al. A QTL influ-
human reticulocytes. J Clin Invest 1971;50: 998–1000. encing F cell production maps to a gene encod-
2458–2460. 14. Papasavva T, van Ijcken WF, Kockx CE, et al. Next ing a zinc-finger protein on chromosome 2p15.
5. Kacian DL, Gambino R, Dow LW, et al. generation sequencing of SNPs for non-invasive Nat Genet 2007;39:1197–1199.
Decreased globin messenger RNA in thalassemia prenatal diagnosis: Challenges and feasibility as 24. Uda M, Galanello R, Sanna S, et al. Genome-
detected by molecular hybridization. Proc Natl illustrated by an application to b-thalassaemia. Eur wide association study shows BCL11A associ-
Acad Sci USA 1973;70:1886–1890. J Hum Genet 2013;21:1403–1410. ated with persistent fetal hemoglobin and ameli-
6. Saiki RK, Bugawan TL, Horn GT, et al. Analysis 15. Berger VK, Baker VL. Preimplantation diagnosis oration of the phenotype of beta-thalassemia.
of enzymatically amplified beta-globin and for single gene disorders. Semin Reprod Med Proc Natl Acad Sci USA 2008;105:1620–1625.
HLA-DQ alpha DNA with allele-specific 2014;32:107–113. 25. Smith CH, Schulman I, Ando RE, Stern G.
oligonucleotide probes. Nature 1986;324:163– 16. Cao A, Kan YW. Prevention of thalassemia. Studies in Mediterranean (Cooley’s) anemia. II.
166. Cold Spring Harb Perspect Med. 2013;3: The suppression of hematopoiesis by transfu-
7. Hardison RC, Chui DH, Giardine B, et al. a011775. sions. Blood 1955;10:707–717.
HbVar: A relational database of human hemo- 17. Ray PF, Kaeda JS, Bingham J, et al. Preimplan- 26. Vichinsky E, Neumayr L, Trimble S, et al.
globin variants and thalassemia mutations at the tation genetic diagnosis of beta-thalassaemia Transfusion complications in thalassemia
globin gene server. Hum Mutat 2002;19:225– major. Lancet 1996;347:1696. patients: A report from the Centers for Disease
233. 18. Traeger-Synodinos J. Preimplantation genetic Control and Prevention (CME). Transfusion
8. Giardine B, Borg J, Viennas E, et al. Updates of diagnosis, an alternative to conventional prena- 2014;54:972–981.
the HbVar database of human hemoglobin var- tal diagnosis of the hemoglobinopathies. Int J 27. Goss C, Giardina P, Degtyaryova D, et al. Red
iants and thalassemia mutations. Nucleic Acids Lab Hematol 2013;35:571–579. blood cell transfusions for thalassemia: Results

20 American Journal of Hematology, Vol. 91, No. 1, January 2016 doi:10.1002/ajh.24231


40TH ANNIVERSARY ISSUE Thalassemia 2016

of a survey assessing current practice and pro- quency and severity. Br J Haematol 2013;160: trolled study of ACE-011 (ActRIIA-IgG1) in
posal of evidence-based guidelines. Transfusion 680–687. postmenopausal women. J Bone Miner Res
2014;54:1773–1781. 48. Wong P, Fuller PJ, Gillespie MT, et al. Thalasse- 2009;24:744–752.
28. Borgna-Pignatti C, Garani MC, Forni GL, et al. mia bone disease: A 19-year longitudinal analy- 69. Dussiot M, Maciel TT, Fricot A, et al. An acti-
Hepatocellular carcinoma in thalassaemia: An sis. J Bone Miner Res 2014;29:2468–2473. vin receptor IIA ligand trap corrects ineffective
update of the Italian Registry. Br J Haematol 49. Tsay J, Yang Z, Ross FP, et al. Bone loss caused erythropoiesis in b-thalassemia. Nat Med 2014;
2014;167:121–126. by iron overload in a murine model: Importance 20:398–407.
29. Romano C, Massimo L, Tortorolo G. Prelimi- of oxidative stress. Blood 2010;116:2582–2589. 70. Nemeth E. Hepcidin and b-thalassemia major.
nary note on the therapeutic use of desferriox- 50. De Sanctis V, Soliman AT, Elsedfy H, et al. Blood 2013;122:3–4.
amine in Cooley’s Disase. Minerva Pediatr 1964; Osteoporosis in thalassemia major: An update 71. Attie KM, Allison MJ, McClure T, et al. A phase
16:711–713. and the I-CET 2013 recommendations for sur- 1 study of ACE-536, a regulator of erythroid
30. Smith RS. Chelating agents in the diagnosis and veillance and treatment. Pediatr Endocrinol Rev differentiation, in healthy volunteers. Am J
treatment of iron overload in thalassemia. Ann 2013;11:167–180. Hematol 2014;89:766–770.
NY Acad Sci 1964;119:776–788. 51. Giusti A. Bisphosphonates in the management 72. Piga AG, Perrotta S, Melpignano A, et al. ACE-
31. Brittenham GM, Griffith PM, Nienhuis AW, of thalassemia-associated osteoporosis: A sys- 536 Increases hemoglobin and decreases transfu-
et al. Efficacy of deferoxamine in preventing tematic review of randomised controlled trials. sion burden and serum ferritin in adults with
complications of iron overload in patients with J Bone Miner Metab 2014;32:606–615. beta-thalassemia: Preliminary results from a
thalassemia major. N Engl J Med 1994;331:567– 52. Nemtsas P, Arnaoutoglou M, Perifanis V, et al. phase 2 study. Blood 2014;124:53a.
573. Neurological complications of beta-thalassemia. 73. Rivella S. Ineffective erythropoiesis and thalasse-
32. Grady RW. The development of new drugs for Ann Hematol 2015;94:1261–1265. mias. Curr Opin Hematol 2009;16:187–94.
use in iron chelation therapy. Birth Defects Orig 53. Gaziev J, Sodani P, Lucarelli G. Hematopoietic 74. Libani IV, Guy EC, Melchiori L, et al. Decreased
Artic Ser 1976;12:161–175. stem cell transplantation in thalassemia. Bone differentiation of erythroid cells exacerbates
33. Aydinok Y, Kattamis A, Viprakasit V. Current Marrow Transplant 2008;42:S41 ineffective erythropoiesis in beta-thalassemia.
approach to iron chelation in children. Br J 54. Gaziev J, Isgro A, Sodani P, et al. Optimal out- Blood 2008;112:875–885.
Haematol 2014;745–755. comes in young class 3 patients with thalasse- 75. Manwani D, Bieker JJ. The erythroblastic island.
34. Sheth S. Iron chelation: An update. Curr Opin mia undergoing HLA-identical sibling bone Curr Top Dev Biol 2008;82:23–53.
Hematol 2014;21:179–185. marrow transplantation. Transplantation, in 76. Ginzburg Y, Rivella S. bthalassemia: A model
35. Link G, Konijn AM, Breuer W, et al. Exploring press. for elucidating the dynamic regulation of inef-
the “iron shuttle” hypothesis in chelation ther- 55. Mathews V, Savani BN. Conditioning regimens fective erythropoiesis and iron metabolism.
apy: Effects of combined deferoxamine and in allo-SCT for thalassemia major. Bone Mar- Blood 2011;118:4321–4330.
deferiprone treatment in hypertransfused rats row Transplant 2014;49:607–610. 77. Ramos P, Casu C, Gardenghi S, et al. Macro-
with labeled iron stores and in iron-loaded rat 56. Kharbanda S, Smith AR, Hutchinson SK, et al. phages support pathological erythropoiesis in
heart cells in culture. J Lab Clin Med 2001;138: Unrelated donor allogeneic hematopoietic stem polycythemia vera and b-thalassemia. Nat Med
130–138. cell transplantation for patients with hemoglobi- 2013;19:437–445.
36. Vlachodimitropoulou Koumoutsea E, Garbowski nopathies using a reduced-intensity conditioning 78. Jacobsen RN, Perkins AC, Levesque JP. Macro-
M, Porter J. Synergistic intracellular iron chela- regimen and third-party mesenchymal stromal phages and regulation of erythropoiesis. Curr
tion combinations: mechanisms and conditions cells. Biol Blood Marrow Transplant 2014;20: Opin Hematol 2015;22:212–219.
for optimizing iron mobilization. Br J Haematol 581–586. 79. Yuan J, Angelucci E, Lucarelli G, et al. Acceler-
2015;170:874–883. 57. King AA, Kamani N, Bunin N, et al. Successful ated programmed cell death (apoptosis) in ery-
37. Piga A, Longo F, Musallam KM, et al. Assess- matched sibling donor marrow transplantation throid precursors of patients with severe beta-
ment and management of iron overload in b- following reduced intensity conditioning in chil- thalassemia (Cooley’s anemia). Blood 1993;82:
thalassaemia major patients during the 21st cen- dren with hemoglobinopathies. Am J Hematol, 374–377.
tury: A real-life experience from the Italian in press. doi: 10.1002/ajh.24183. 80. Arlet JB, Ribeil JA, Guillem F, et al. HSP70
WEBTHAL project. Br J Haematol 2013;161: 58. Gaziev J, Marziali M, Isgr o A, et al. Bone mar- sequestration by free a-globin promotes ineffec-
872–883. row transplantation for thalassemia from alter- tive erythropoiesis in b-thalassaemia. Nature
38. Olivieri NF, Nathan DG, MacMillan JH, et al. native related donors: Improved outcomes with 2014;514:242–246.
Survival in medically treated patients with a new approach. Blood 2013;122:2751–2756. 81. Ghosh S, Thrasher AJ, Gaspar HB. Gene ther-
homozygous beta-thalassemia. N Engl J Med 59. Parikh SH, Mendizabal A, Benjamin CL, et al. A apy for monogenic disorders of the bone mar-
1994;331:574–578. novel reduced-intensity conditioning regimen row. Br J Haematol, in press. doi: 10.1111/
39. Puliyel M, Sposto R, Berdoukas VA, et al. Ferri- for unrelated umbilical cord blood transplanta- bjh.13520.
tin trends do not predict changes in total body tion in children with nonmalignant diseases. 82. Cavazzana-Calvo M, Payen E, Negre O, et al.
iron in patients with transfusional iron overload. Biol Blood Marrow Transplant 2014;20:326–336. Transfusion independence and HMGA2 activa-
Am J Hematol 2014;89:391–394. 60. Faulkner LB, Uderzo C, Masera G. International tion after gene therapy of human b-thalassae-
40. Wood JC. Estimating tissue iron burden: current cooperation for the cure and prevention of mia. Nature 2010;467:318–322.
status and future prospects. Br J Haematol 2015; severe hemoglobinopathies. J Pediatr Hematol 83. Cavazzana M, Ribeil JA, Payen E, et al. Out-
170:15–28. Oncol 2013;35:419–423. comes of gene therapy for b-thalassemia major
41. Sarigianni M, Liakos A, Vlachaki E, et al. Accu- 61. Mehta PA, Faulkner LB. Hematopoietic cell and severe sickle cell disease via transplantation
racy of magnetic resonance imaging in diagnosis transplantation for thalassemia: A global per- of autologous hematopoietic stem cells trans-
of liver iron overload: A systematic review and spective BMT tandem meeting 2013. Biol Blood duced ex vio with a lentiviral beta globin vector.
meta-analysis. Clin Gastroenterol Hepatol 2015; Marrow Transplant 2013;19:S70–S73. Haematologica 2014;99:S466.
13:55–63.e5. 62. Sankaran VG, Weiss MJ. Anemia: Progress in 84. Finotti A, Breda L, Lederer CW, et al. Recent
42. Modell B, Khan M, Darlison M, et al. Improved molecular mechanisms and therapies. Nat Med trends in the gene therapy of b-thalassemia.
survival of thalassaemia major in the UK and 2015;21:221–230. J Blood Med 2015;6:69–85.
relation to T2* cardiovascular magnetic reso- 63. Rivella S. bthalassemias: paradigmatic diseases 85. Genovese P, Schiroli G, Escobar G, et al. Tar-
nance. J Cardiovasc Magn Reson. 2008;10:42– for scientific discoveries and development of geted genome editing in human repopulating
49. innovative therapies. Haematologica 2015;100: haematopoietic stem cells. Nature 2014;510:235–
43. Eldor Lellouche F, Goldfarb A, et al. In vivo 418–430. 240.
platelet activation in beta-thalassemia major 64. Ngo DA, Steinberg MH. Genomic approaches to 86. Modell B, Darlison M. Global epidemiology of
reflected by increased platelet-thromboxane uri- identifying targets for treating b-hemoglobino- haemoglobin disorders and derived service indi-
nary metabolites. Blood 1991;77:1749–1753. pathies. BMC Med Genomics 2015;8:44–57. cators. Bull World Health Organ 2008;86:480–
44. Cappellini MD, Robbiolo L, Bottasso BM, et al. 65. Musallam KM, Taher AT, Cappellini MD, 487.
Venous thromboembolism and hypercoagulabil- Sankaran VG. Clinical experience with fetal 87. Telfer P. Update on survival in thalassemia
ity in splenectomized patients with thalassaemia hemoglobin induction therapy in patients with major. Hemoglobin 2009;33:S76–S80.
intermedia. Br J Haematol 2000;111:467–473. b-thalassemia. Blood 2013;121:2199–2212. 88. Weatherall DJ. The challenge of haemoglobino-
45. Cappellini MD, Poggiali E, Taher AT, Musallam 66. Bauer DE, Kamran SC, Lessard S, et al. An ery- pathies in resource-poor countries. Br J Haema-
KM. Hypercoagulability in b-thalassemia: A sta- throid enhancer of BCL11A subject to genetic tol 2011;154:736–744.
tus quo. Expert Rev Hematol 2012;5:505–511. variation determines fetal hemoglobin level. Sci-
46. Musallam KM, Taher AT, Karimi M, ence 2013;342:253–257.
Rachmilewitz EA. Cerebral infarction in b- 67. McPherron AC. Metabolic functions of myosta-
thalassemia intermedia: Breaking the silence. tin and GDF11. Immunol Endocr Metab Agents
Thromb Res 2012;130:695–702. Med Chem 2010;10:217–231.
47. Haines D, Martin M, Carson S, et al. Pain in 68. Ruckle J, Jacobs M, Kramer W, et al. Single-
thalassaemia: The effects of age on pain fre- dose, randomized, double-blind, placebo-con-

doi:10.1002/ajh.24231 American Journal of Hematology, Vol. 91, No. 1, January 2016 21

You might also like