Sei sulla pagina 1di 11

MED ICA L PR OGR ES S

Review Article

Medical Progress length polymorphisms, or haplotypes, within the


b-globin cluster. A limited number of haplotypes are
found in each population, so that 80 percent of the
mutations are associated with only 20 different hap-
T HE b-T HALASSEMIAS lotypes. This observation has helped demonstrate the
independent origin of b-thalassemia in several pop-
NANCY F. OLIVIERI, M.D. ulations.10 There is evidence that the high frequency
of b-thalassemia throughout the tropics reflects an
advantage of heterozygotes against Plasmodium fal-
ciparum malaria,11 as has already been demonstrated

I
N 1925, Thomas Cooley and Pearl Lee described in a-thalassemia.12
a form of severe anemia, occurring in children
of Italian origin and associated with splenomeg- MOLECULAR PATHOLOGY
aly and characteristic bone changes.1 Over the next Structure and Synthesis of Hemoglobin
decade, a milder form was described independently by The structure and regulation of the human globin
several Italian investigators.2-4 Because all early cases genes have been reviewed elsewhere13; only aspects
were reported in children of Mediterranean origin, with direct relevance to an understanding of the
the disease was later termed thalassemia, from the molecular pathology of the b-thalassemias are out-
Greek word for sea, thalassa.5 Over the next 20 years, lined here.
it became apparent that Cooley and Lee had de- The b-like globin genes, a linked cluster on chro-
scribed the homozygous or compound heterozygous mosome 11, are arranged over approximately 60,000
state for a recessive mendelian disorder not confined nucleotide bases (Fig. 1). Promoter elements up-
to the Mediterranean, but occurring widely through- stream from the initiation codon of each active gene
out tropical countries. In the past 20 years, the two are involved in the initiation of transcription. The
important forms of this disorder, a- and b-thalas- cluster also contains other regulatory elements that in-
semia, resulting from the defective synthesis of the teract to promote erythroid-specific gene expression
a- and b-globin chains of hemoglobin, respectively, and to coordinate the developmental regulation of
have become recognized as the most common mon- each gene.
ogenic diseases in humans.6
This article focuses on the b-thalassemias, the se- Hemoglobin Switching
vere forms of which are by far the most important
As an adaptation to changing oxygen requirements,
of all the thalassemias. The molecular and clinical as-
different hemoglobins, all composed of two differ-
pects of the severe a-thalassemia syndromes have
ent pairs of globin chains each attached to a heme
been reviewed elsewhere.7,8
moiety, are synthesized in the embryo, fetus, and
DISTRIBUTION AND POPULATION adult.14 Severe b-thalassemia usually becomes manifest
AT RISK as a result of the decline in the synthesis of fetal
hemoglobin (a2g 2) during the first year of life (Fig.
The b-thalassemias are widespread throughout the
1). The precise mechanisms that control the switch
Mediterranean region, Africa, the Middle East, the
from the production of fetal hemoglobin to that of
Indian subcontinent and Burma, Southeast Asia in-
adult hemoglobin (a2 b 2) (Fig. 1) are not fully un-
cluding southern China, the Malay Peninsula, and
derstood.13-16
Indonesia. Estimates of gene frequencies range from
3 to 10 percent in some areas.9 Within each popula- Mutations Causing b-Thalassemia
tion at risk for b-thalassemia a small number of
common mutations are found, as well as rarer ones; Nearly 200 different mutations have been described
each mutation is in strong linkage disequilibrium in patients with b-thalassemia and related disorders.
with specific arrangements of restriction-fragment Although most are small nucleotide substitutions
within the cluster, deletions may also cause b-thalas-
semia.9 All the mutations result in either the absence
of the synthesis of b-globin chains (b0-thalassemia)
From the University of Toronto, Toronto. Address reprint requests to or a reduction in synthesis (b+-thalassemia) (Fig. 2).
Dr. Olivieri at the Hospital for Sick Children, 555 University Ave., Toronto,
ON M5G 1X8, Canada, or at noliv@sickkids.on.ca. Mutations in or close to the conserved promoter
1999, Massachusetts Medical Society. sequences and in the 5' untranslated region down-

Volume 341 Numb er 2 99

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
The New Eng land Jour nal of Medicine

Chromosome 11

e G g A g cb d b
Locus-control
region
A

50 a
Globin-Chain Synthesis

b
g
(% of total)

30

e
10 z
d

6 18 30 0 6 18 30 42
Before Birth (wk) Birth After Birth (wk)
B

Figure 1. The b -Globin Gene Cluster on the Short Arm of Chromosome 11.
In Panel A, the b -globinlike genes are arranged in the order in which they are expressed during development. The Gg and Ag genes
are both active genes that produce g -globin chains that differ only at position 136 (glycine is the product of the Gg gene, and alanine
is the product of the Ag gene). The cb gene is a pseudogene, an evolutionary remnant of a previously active b -globinlike gene.
Areas of nucleotide homology upstream from the initiation codon of each active gene, termed promoter elements, are involved in
the initiation of transcription and hence play a vital part in gene regulation. The cluster also contains regulatory elements that in-
teract to promote erythroid-specific gene expression and to coordinate the developmental regulation of each gene, including he-
moglobin switching. These include enhancers, distant regulatory elements that increase gene expression, and a master sequence,
the clusters essential distal regulatory element, the b -globin locus-control region. This is a region that lies 20 kb upstream from
the e-globin gene.13 It encompasses five erythroid-lineagespecific nuclease hypersensitive sites (shown in red) that permit expres-
sion of the downstream genes. In addition to these elements for up-regulation, several suppressor regions, or silencers, have been
defined in the b -globin gene cluster.
Panel B shows the timing of the normal developmental switching of human hemoglobin. Early in fetal life the synthesis of the
embryonic a-globinlike (z) chains switches to that of a-globin, which is produced thereafter. At the same time, the synthesis of
embryonic beta-like (e) chains switches to that of g -globin chains. The a-globin and g -globin chains combine to form fetal hemo-
globin (a2g2), the main b -globinlike globin during the remainder of fetal life and throughout early postnatal life. As a result of the
decline in the synthesis of g -globin chains in patients with b -thalassemia, fetal hemoglobin production becomes insufficient to
compensate for the excess of a-globin chains, the production of which is unaffected in b -thalassemia.

regulate transcription, usually resulting in mild script result in b-thalassemia. In some mutations, no
b+-thalassemia. Transcription is also affected by dele- normal message is produced, whereas other muta-
tions in the 5' region, which completely inactivate tions only slightly reduce the amount of normally
transcription and result in b0-thalassemia. spliced mRNA. Mutations within invariant dinucle-
Both splicing of the messenger RNA (mRNA) pre- otides at intronexon junctions, critical to the remov-
cursor and ineffective cleavage of the mRNA tran- al of intervening sequences and the splicing of exons

100 Jul y 8, 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
MED ICA L PR OGR ES S

Deletions

Intron
Intron
5' 3'
Exon 1 Intron Exon 2 Exon 3 Intron

Other Mutations
Mutation affecting initiation of
transcription
Mutation affecting splicing of
RNA from introns
Polyadenylation-signal mutation
Mutation affecting initiation of
translation
Nonsense mutation
Frame-shift mutation

Figure 2. The Normal Structure of the b-Globin Gene and the Locations and Types of Mutations Resulting in b-Thalassemia.
All b-globinlike genes contain three exons and two introns between codons 30 and 31 and 104 and 105, respectively. The primary
action of all the mutations is to abolish the output of b-globin chains (b0-thalassemia; shown in red) or reduce the output (b+-thal-
assemia; shown in green). The 170 different mutations that act in this way may interfere with the action of the b-globin gene at the
transcriptional level, in the processing of the primary transcript, in the translation of b-globin messenger RNA, or in the post-trans-
lational stability of the b-globin gene product.

to produce functional mRNA, result in b0-thalasse- missense mutations, resulting in the synthesis of un-
mia. Mutations in highly conserved nucleotides flank- stable b-globin chains, cause b-thalassemia.
ing these sequences, or in cryptic splice sites, which
resemble a donor or acceptor splice site, result in PATHOPHYSIOLOGY
severe as well as mild b+-thalassemia. Substitutions
Mechanisms of Anemia
or small deletions affecting the conserved AATAAA
sequence in the 3' untranslated region result in inef- In severe untreated b-thalassemia, erythropoiesis
fective cleavage of the mRNA transcript and cause may be increased by a factor of up to 10, more than
mild b+-thalassemia. 95 percent of which may be ineffective. Ineffective
Mutations that interfere with translation involve the erythropoiesis, the hallmark of b-thalassemia, is a re-
initiation, elongation, or termination of globin-chain sult of the myriad deleterious effects of a relative
production and result in b0-thalassemia. Approxi- excess of a-globin chains.17 This relative excess in-
mately half of all b-thalassemia mutations interfere terferes with most stages of normal erythroid matu-
with translation; these include frame-shift or nonsense ration: both intramedullary death of red-cell precur-
mutations, which introduce premature termination sors through arrest in the G1 phase of the cell cycle18
codons and result in b0-thalassemia. A more recent- and accelerated intramedullary apoptosis of late eryth-
ly identified family of mutations, usually involving roblasts19,20 have been demonstrated. Studies of the
exon 3, results in the production of unstable globin consequences of the accumulation of excess a-globin
chains of varying lengths that, together with a relative chains and their degradation products within the red-
excess of a-globin chains, precipitate in red-cell pre- cell membrane and its skeleton 20-22 have also demon-
cursors and lead to ineffective erythropoiesis, even in strated abnormalities in the ratio of spectrin to band
the heterozygous state. This is the molecular basis for 3 and in the function of band 4.1. This subject has
dominantly inherited (b+) thalassemia. In addition, been thoroughly reviewed recently.20 The observa-

Volume 341 Numb er 2 101

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
The New Eng land Jour nal of Medicine

Excess free Formation of heme


a-globin chains and hemichromes
Denaturation
Degradation

Iron-mediated toxicity

Membrane
Hemolysis Ineffective
binding of
erythropoiesis
IgG and C3 Removal of
damaged red cells

Increased
erythropoietin Reduced tissue Anemia Splenomegaly
synthesis
oxygenation

Skeletal Erythroid
deformities, Increased Iron
marrow overload
osteopenia iron absorption
expansion

Figure 3. Effects of Excess Production of Free a-Globin Chains.


Primary disease processes are indicated in orange, and compensatory mechanisms in yellow. Excess unbound a-globin chains and
their degradation products precipitate in red-cell precursors, causing defective maturation and ineffective erythropoiesis. Hemolysis,
owing to the presence of inclusions in the red cells and damage to their membranes by a-globin chains and their degradation prod-
ucts, also contributes to the anemia. Anemia stimulates the synthesis of erythropoietin, leading to an intense proliferation of the
ineffective marrow, which in turn causes skeletal deformities and a variety of growth and metabolic abnormalities. The anemia is
further exacerbated by hemodilution, caused by the shunting of blood through the expanded marrow, and by splenomegaly result-
ing from entrapment of abnormal red cells in the spleen. Bone marrow expansion also results in characteristic deformities of the
skull and face, severe osteopenia, and increased iron absorption.

tion that the presence of excess membrane iron may progressive splenomegaly exacerbate anemia (Fig. 3).
aggravate membrane changes 22 has led to interest in Increased erythropoietin synthesis may stimulate the
the red-cell membrane as a potential therapeutic tar- formation of extramedullary erythropoietic tissue, pri-
get in b-thalassemia. In a mouse model, increased cel- marily in the thorax and paraspinal region. Marrow
lular rigidity and decreased stability in connection expansion also results in characteristic deformities of
with membrane-associated a-globin chains 23 have re- the skull and face, as well as osteopenia and focal de-
portedly been ameliorated during exposure to agents fects in bone mineralization,25,26 and may aggravate a
that bind membrane iron.24 Further understanding painful periarticular syndrome characterized histo-
of these processes may guide future therapies. logically by microfractures and osteomalacia.27 Marrow
hyperplasia leads ultimately to increased iron absorp-
Clinical Consequences of Anemia tion and progressive deposition of iron in tissues.
The severe ineffective erythropoiesis results in eryth-
roid marrow expansion to as much as 30 times the Cellular Heterogeneity and Fetal Hemoglobin Production
normal level. Both an increase in plasma volume as Although fetal hemoglobin synthesis persists after
a result of shunting through expanded marrow and birth to some degree, its production is insufficient

102 Jul y 8, 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
MED IC A L PR OGR ES S

to compensate for the reduced synthesis of b-globin regularly seen only in countries without resources to
chains and the relative excess of a-globin chains.15 support long-term transfusion programs.
The elevated concentrations of fetal hemoglobin
Relation between Genotype and Phenotype
2 to 4 g per deciliter observed in patients with
b-thalassemia reflect a combination of the selection Several genetic factors may ameliorate the severity
of precursors that produce relatively more fetal he- of b-thalassemia.29 First, the underlying mutations
moglobin and erythroid expansion, which appears to vary widely in their effect on the synthesis of b-globin
favor the production of g-globin chains. Even higher chains.38 Co-inheritance of a-thalassemia may reduce
fetal hemoglobin concentrations are associated with the severity of the globin-chain imbalance. Many dif-
specific b-thalassemia alleles 28,29 or other genetic de- ferent interactions with structural hemoglobin vari-
terminants within or linked to the b-globin com- ants may also result in a complex series of clinical
plex.15 At least two other determinants may affect the phenotypes.29 The interactions of b-thalassemia with
synthesis of fetal hemoglobin, one on chromosome two of these variants, hemoglobin S and hemoglo-
630 and the other on the X chromosome.31,32 bin E, are of global importance.
The clinical consequences of the interaction with
Clinical Forms hemoglobin S depend mainly on the b-thalassemia
The b-thalassemias include four clinical syndromes allele. If inherited with b0-thalassemia or severe
of increasing severity: two conditions are generally b+-thalassemia, the resulting clinical disorder may
asymptomatic, the silent carrier state and b-thalas- be indistinguishable from sickle cell anemia. By con-
semia trait, and usually result from the inheritance of trast, interactions with mild b+-thalassemia alleles
one mutant b-globin gene, and two require medical produce a milder sickling disorder.
management, thalassemia intermedia and thalasse- Although hemoglobin E b-thalassemia is probably
mia major. The more severe forms most often result the most common serious hemoglobinopathy world-
from homozygosity or compound heterozygosity for wide,39 its natural history remains poorly under-
a mutant b-globin allele and, occasionally, from het- stood.29 The mutation that produces hemoglobin E
erozygosity for dominant mutations.33 Homozygous activates a cryptic splice site in exon 1 in the b-globin
or compound heterozygous b-thalassemia usually pre- gene; hence, hemoglobin E is associated with mild
sents no diagnostic problems. The early onset of ane- b-thalassemia. For reasons that are not well under-
mia, characteristic blood changes, and elevated fetal stood,29 the interaction of hemoglobin E and b-thal-
hemoglobin concentrations are found in no other assemia results in a wide spectrum of clinical disorders:
condition. The diagnosis can be confirmed by the some are indistinguishable from thalassemia major,
demonstration of the b-thalassemia trait in both par- and some are much milder and not transfusion-
ents. This condition is characterized by mild anemia, dependent. Finally, a number of acquired and envi-
reduced mean cell volumes and mean cell hemoglobin ronmental factors, including progressive splenomeg-
concentrations,29 and elevated concentrations of the aly, exposure to infections, socioeconomic factors,
normal minor adult component of hemoglobin (usu- and the availability of medical care, may also modify
ally exceeding 3.5 percent), hemoglobin A2 (a2d 2). the severity of the disease.
Thalassemia major and thalassemia intermedia have COMPLICATIONS OF DISEASE
no specific molecular correlate but encompass a wide
spectrum of clinical and laboratory abnormalities.34 Iron Overload
Patients referred to as having thalassemia major are Iron overload of tissue, which is fatal with or with-
usually those who come to medical attention in the out transfusion if not prevented or adequately treated,
first year of life and subsequently require regular is the most important complication of b-thalassemia
transfusions to survive. Those who present later or and is a major focus of management.40 In patients
who seldom need transfusions are said to have thalas- who are not receiving transfusions, abnormally reg-
semia intermedia.35 After thalassemia is diagnosed, ulated iron absorption results in increases in body
patients who appear not to require immediate trans- iron burden ranging from 2 to 5 g per year, depend-
fusion may benefit from a period of observation and ing on the severity of erythroid expansion.41,42 Reg-
folate repletion, particularly if the disease is diagnosed ular transfusions may double this rate of iron accu-
after the age of one year. This approach will allow the mulation. Although most clinical manifestations of
identification of patients in whom early growth and iron loading do not appear until the second decade
development are normal and whose well-compensat- of life in patients with inadequate chelation, evi-
ed anemia may be exacerbated only by infection, folate dence from serial liver biopsies in very young pa-
deficiency, or increasing hypersplenism.34-37 With ad- tients indicates that the deleterious effects of iron are
vancing age, even patients with mild forms may have initiated much earlier than this. After approximately
serious complications, including osteopenia, iron load- one year of transfusions, iron begins to be deposited
ing in tissues, and ectopic marrow expansion. The in parenchymal tissues,43 where it may cause sub-
classic changes of untreated thalassemia major are now stantial toxicity as compared with that within retic-

Volume 341 Numb er 2 103

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
The New Eng land Jour nal of Medicine

uloendothelial cells.44,45 As iron loading progresses, rhea occurs in approximately one quarter of female
the capacity of serum transferrin, the main transport patients over the age of 15 years.63 Even in the mod-
protein of iron, to bind and detoxify iron may be ex- ern era of iron-chelating therapy, diabetes mellitus is
ceeded and a nontransferrin-bound fraction of observed in about 5 percent of adults.63 As the iron
plasma iron may promote the generation of free hy- burden increases and iron-related liver dysfunction
droxyl radicals, propagators of oxygen-related dam- progresses, hyperinsulinemia occurs as a result of re-
age.44,45 The advances in free-radical chemistry that duced extraction of insulin by the liver, leading to ex-
have clarified the toxic properties of these and other haustion of beta cells and reduced circulating insulin
oxygen-derived species generated by iron, which concentrations.64 Studies reporting reduced serum
may cause widespread tissue damage, have recently concentrations of trypsin and lipase65 suggest that the
been summarized.45 Although the body maintains a exocrine pancreas is also damaged by iron loading.
number of antioxidant mechanisms against damage Over the long term, iron deposition also damages
induced by free radicals, including superoxide dis- the thyroid, parathyroid, and adrenal glands66,67 and
mutases, catalase, and glutathione peroxidase, in pa- may provoke pulmonary hypertension, right ventric-
tients with large iron burdens these may not prevent ular dilatation, and restrictive lung disease.51,68-70
oxidative damage.44,45 In most studies, bone density is markedly reduced
In the absence of chelating therapy the accumula- in patients with b-thalassemia, particularly those with
tion of iron results in progressive dysfunction of the hypogonadism. Osteopenia may be related to marrow
heart, liver, and endocrine glands.40 Within the heart, expansion, even in patients who receive transfusions,42
changes associated with chronic anemia are usually or to iron-induced osteoblast dysfunction, diabetes,
present in patients who are not receiving transfusions hypoparathyroidism, or hypogonadism.71-73
and are aggravated by iron deposition. In response
CONTROL AND MANAGEMENT
to iron loading, human myocytes in vitro increase
the transport of nontransferrin-bound iron,46 pos- Prevention Programs and Prenatal Diagnosis
sibly thereby aggravating cardiac iron loading. Exten- Screening programs, aimed at prevention of the
sive iron deposits are associated with cardiac hyper- disease, and prenatal diagnosis have resulted in a
trophy and dilatation, degeneration of myocardial marked reduction in the birth rate of affected chil-
fibers, and in rare cases fibrosis.47 In patients who are dren in Greece, Cyprus, continental Italy, and Sar-
receiving transfusions but not chelating therapy, dinia.74 Widespread use of similar programs in other
symptomatic cardiac disease has been reported with- areas of the world has not yet been possible. Screen-
in 10 years after the start of transfusions48 and may ing for carriers is performed most efficiently by
be aggravated by myocarditis49 and pulmonary hy- measurement of the red-cell indexes and, in samples
pertension.50,51 The survival of patients with b-thal- from persons with reduced mean cell volumes and
assemia is determined by the magnitude of iron mean cell hemoglobin concentrations, estimation of
loading within the heart.52,53 the hemoglobin A2 concentration. The practical
Iron-induced liver disease is a common cause of problems associated with screening for rarer forms
death in older patients54 and is often aggravated by of b-thalassemia and the effect of coexistent a-thal-
infection with hepatitis C virus. Within two years af- assemia on the red-cell indexes have been reviewed
ter the start of transfusions, collagen formation55 recently.29 Prenatal diagnosis, first carried out by fe-
and portal fibrosis56 have been reported; in the ab- tal-blood sampling and assessment of globin-chain
sence of chelating therapy, cirrhosis may develop in synthesis in fetal blood, more recently has involved
the first decade of life.43,57,58 The extent of these direct analysis of fetal DNA obtained by chorionic-
processes may be underestimated if fewer than three villus sampling. This approach is associated with a
cores of liver are sampled at biopsy.59 The risk of he- very slightly increased risk of fetal loss and an error
patic fibrosis is augmented at body iron burdens cor- rate in experienced laboratories of less than 1 per-
responding to hepatic iron concentrations of more cent. The practical aspects of fetal-DNA analysis
than 7 mg per gram of liver, dry weight (Fig. 4).61 have also been recently reviewed.29,74
As in cultured heart cells, in cultured hepatocytes
the transport of nontransferrin-bound iron is in- Medical Therapy
creased,62 possibly aggravating iron loading in vivo. A decision to initiate regular transfusions in pa-
The striking increases in survival in patients with tients with b-thalassemia may be difficult and should
b-thalassemia over the past decade have focused at- be based on the presence and severity of the symp-
tention on abnormal endocrine function, now the toms and signs of anemia, including failure of growth
most prevalent iron-induced complication in older and development. Only rarely is genotyping helpful
patients. Iron loading within the anterior pituitary is in this decision. The goals of transfusion include cor-
the primary cause of disturbed sexual maturation, re- rection of anemia, suppression of erythropoiesis, and
ported in 50 percent of both boys and girls with the inhibition of increased gastrointestinal absorption
condition.63 Furthermore, early secondary amenor- of iron. Hypertransfusion and supertransfusion

104 Jul y 8, 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
MED IC A L PR OGR ES S

15,000 50
Increased risk of complications

Hepatic Iron (mg/g of liver, dry weight)


Hepatic Iron (g/g of liver, wet weight)
Thalassemia major
Adequate iron chelation

Normal hepatic iron 40


concentration

10,000
Homozygous 30
hemochromatosis

20
5,000 Threshold for cardiac disease and early death

Heterozygous
Optimal iron chelation level 10
hemochromatosis

0 0
0 10 20 30 40 50
Age (years)
Figure 4. Hepatic Iron Burden over Time and the Effect of Various Hepatic Iron Concentrations in Patients with Thalas-
semia Major, Homozygous Hemochromatosis, and Heterozygous Hemochromatosis.
Because efforts to maintain normal hepatic iron concentrations in patients with thalassemia major are frequently asso-
ciated with adverse reactions to deferoxamine, an optimal range within which hepatic iron may be safely maintained,
derived in part from clinical experience with the iron-loading disorder hereditary hemochromatosis, has been proposed.
In a proportion of patients who are heterozygous for hemochromatosis, moderate iron loading corresponding to con-
centrations of approximately 3.2 to 7 mg of iron per gram of liver, dry weight (indicated in yellow), is associated with
normal survival without complications of iron overload.60 In patients who are homozygous for this disorder, concentra-
tions exceeding this range, up to approximately 15 mg of iron per gram of liver, dry weight (indicated in blue), are as-
sociated with an increased risk of complications of iron overload,61 whereas maintenance of levels exceeding 15 mg of
iron per gram of liver, dry weight, greatly increases the risk of cardiac disease and early death in patients with thalas-
semia major.52 In patients with thalassemia major who receive regular transfusions, the rate of iron loading is much
more accelerated and death usually occurs before the third decade of life. The goal of treatment is the maintenance of
a body-iron burden corresponding to a hepatic iron concentration of approximately 3.2 to 7 mg of iron per gram of liver,
dry weight, achievable with regular deferoxamine therapy.40 The serum ferritin concentrations corresponding to these
ranges of hepatic iron are not clearly defined.

regimens, which achieve these goals but are associ- crease the risk of clinical disease.52 Nearly normal con-
ated with substantial iron loading,40,75 have been centrations of hepatic iron can be maintained with
supplanted by regimens in which the hemoglobin modern regimens of deferoxamine. Moreover, defer-
concentration before transfusion does not exceed oxamine arrests the progression of hepatic fibrosis to
9.5 g per deciliter.76 These newer regimens are asso- cirrhosis, even when administered in regimens that
ciated with both adequate marrow suppression and stabilize, rather than reduce, the body iron burden.77
relatively lower rates of iron accumulation. The importance of this finding in the seminal study
The beneficial effects of iron-chelating therapy with that ushered in the modern era of deferoxamine ther-
parenteral deferoxamine, the only chelating agent apy is highlighted by evidence that in another form
widely available for clinical use, on the complications of iron overload, hereditary hemochromatosis, pro-
of iron loading have recently been reviewed.40 As a gression of hepatic fibrosis is a critical event associ-
result of programs of deferoxamine therapy, the prog- ated with an increased risk of death.61
nosis for patients in countries able to afford this A favorable effect of a sustained reduction in body
therapy has greatly improved, in contrast to the prog- iron is also suggested by the relatively low prevalence
nosis for patients in developing countries, where wide- of thyroid, parathyroid, and adrenal abnormalities in
spread implementation of this regimen is still awaited. the modern era.78 In parallel, early and intensive de-
Adequate deferoxamine therapy prevents early death feroxamine therapy may increase the incidence of
from cardiac disease: maintenance of body iron bur- normal sexual maturation,78 but it apparently does not
dens corresponding to hepatic iron concentrations of reverse established abnormalities.40 Similarly, although
less than 15 mg per gram, dry weight, greatly de- deferoxamine prevents diabetes mellitus,52 there is

Volume 341 Numb er 2 105

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
The New Eng land Jour nal of Medicine

no evidence that it can reverse this complication. In Experimental Therapies


summary, modern regimens of subcutaneous defer- Chelators Other Than Deferoxamine
oxamine may extend survival free of many complica-
tions of iron overload, if body iron is reduced or Difficulties associated with deferoxamine therapy
maintained below critical concentrations.40,52,53,79 have led to a search for alternatives, including orally
A balance between the effectiveness of deferoxa- active iron chelators. The administration of one
mine and its toxicity the latter observed primarily agent, deferiprone, was reported to have a favorable
in the presence of relatively low body iron burdens80 short-term effect on body iron in the one study in
can be maintained through regular determina- which serial systematic determinations of hepatic iron
tions of body iron burden. In clinical practice, the were obtained.91 Two subsequent long-term studies
serum ferritin concentration is commonly used to have suggested that hepatic iron may stabilize at or
assess the effectiveness of treatment. It is increasing- increase to concentrations associated with an in-
ly recognized that reliance on this test may lead to creased risk of cardiac disease and early death52 in
errors in management; changes in body iron account approximately half of patients.92,93 Previously recog-
for little more than half the variation in serum fer- nized adverse effects of deferiprone included embry-
ritin concentrations.81 By contrast, the measurement otoxicity, teratogenicity, neutropenia, and agranulo-
of hepatic iron stores, whose concentrations are cytosis.94 Long-term treatment has been reported to
highly correlated with total body iron stores,82 pro- be associated with progression of hepatic fibrosis; the
vides the most quantitative, specific, and sensitive odds of progression of fibrosis were estimated to in-
method of evaluating iron burden in patients with crease by a factor of 5.8 with each additional year of
thalassemia. Determination of hepatic iron concen- deferiprone therapy.95 In another study, four deaths
trations in liver-biopsy specimens obtained with ul- due to cardiac failure were reported during long-term
trasonographic guidance is safe and permits rational therapy.92 The results of these clinical trials virtually
adjustments in iron-chelating therapy.40 Magnetic recapitulate those in two animal species, in which
susceptometry provides a direct measure of hepatic deferiprone and a structurally similar compound were
iron stores that is quantitatively equivalent to that shown to increase hepatic and cardiac iron loading,
determined by biopsy of at least 0.6 mg of liver, dry worsen hepatic fibrosis, and induce cardiac and mus-
weight,83 over a range of iron concentrations.84 Mag- culoskeletal fibrosis.95,96 Taken together, these data
netic susceptometry is currently available in only suggest that deferiprone does not adequately control
two centers worldwide. By contrast, the more widely body iron in a substantial proportion of patients and
available technique of magnetic resonance imaging may promote worsening of hepatic fibrosis. These
fails to provide accurate quantitation of hepatic iron studies support cautions previously expressed about
concentrations in patients with severe iron overload, the long-term administration of this agent.97
hepatic fibrosis, or both.85 The results of long-term follow-up of the effec-
tiveness of other modes of administration of defer-
Bone Marrow Transplantation oxamine are awaited. These include deferoxamine
Bone marrow transplantation from HLA-identical attached to high-molecular starch,98 administered in
donors has been successfully performed worldwide in twice-daily subcutaneous boluses,99 and given in a lip-
over 1000 patients with severe b-thalassemia.86 Out- id vehicle, permitting slow release.100
comes after transplantation are greatly influenced by
Augmentation of Fetal-Hemoglobin Synthesis
the presence of hepatomegaly, portal fibrosis, and in-
effective chelating therapy before transplantation.87 Several trials have attempted to augment the syn-
Children without any of these risk factors have rates thesis of fetal hemoglobin in an effort to ameliorate
of survival and disease-free survival exceeding 90 per- the severity of b-thalassemia.101 Administration of in-
cent three years after transplantation. In those with travenous 5-azacytidine was associated with increases
all three risk factors, and in most adults, the rates are in the hemoglobin concentration in a few patients102;
approximately 60 percent. Lower success rates are re- the potential toxicity of the drug later shifted interest
ported at smaller centers.87 Complications include a to less toxic alternatives. Therapy with hydroxyu-
rate of chronic graft-versus-host-disease ranging from rea,103,104 butyric acid compounds,105 and these agents
2 to 8 percent and a variable incidence of mixed chi- in combination106 has reduced or eliminated transfu-
merism.86 Post-transplantation management of preex- sion requirements in some patients. Other studies
isting hepatic iron overload, iron-induced cardiac dys- have reported only small increases in fetal and total
function, and viral hepatitis may prevent progression hemoglobin concentrations during the administra-
of these processes.86 There is interest in experimental tion of hydroxyurea107,108 and both intravenous109,110
approaches to bone marrow replacement in patients and oral111,112 butyrate compounds.
with thalassemia, including cord-blood transplanta- How can the augmentation of fetal hemoglobin
tion,88 the use of unrelated phenotypically matched be optimized? Studies in humans and animal models
donors,89 and in utero transplantation.90 of b-thalassemia, including transgenic mice,113 have

106 Jul y 8, 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
MED IC A L PR OGR ES S

suggested that increases in the production of g-glo- Supported in part by research grants from the Medical Research Council
of Canada, the Ontario Heart and Stroke Foundation, and the Ontario
bin chains may be influenced by the degree of eryth- Thalassemia Foundation. Dr. Olivieri is the recipient of a Scientist Award
roid marrow expansion, sequential administration of from the Medical Research Council of Canada.
specific combinations of agents, the degree to which
the g-globin gene is already partially activated, or all I am indebted to David Nathan, David Weatherall, Gary Brit-
tenham, John Porter, Alan Schechter, Elliott Vichinsky, Brenda Gal-
of these.113-116 Furthermore, the striking clinical re- lie, Helen Chan, Peter Durie, John Dick, Marc Giacomelli, Paul
sponses observed in patients with mutations104-106 that Ranalli, Arthur Schafer, Michele Brill-Edwards, Lori West, Miriam
delete specific sequences within the b-globin gene Kaufman, Michael Langlois, David Kern, Bob Phillips, Maria Mu-
cluster that may have a key role in the silencing of raca, Bill Graham, Rhonda Love, Doug Templeton, John Polanyi,
and Michael Baker, without whose support this review would not have
adjacent genes117 indicate that delineation of some cis been written.
sequences may influence the inducibility of the g-glo-
bin gene. REFERENCES
Although they address a highly desirable, cost-
1. Cooley TB, Lee P. A series of cases of splenomegaly in children with
effective goal in b-thalassemia, therapies to increase anemia and peculiar bone changes. Trans Am Pediatr Soc 1925;37:29-30.
the synthesis of fetal hemoglobin in the disorder have, 2. Rietti F. Ittero emolitico primitivo. Alti Accad Sci Med Nat Ferrara
with few exceptions, proved disappointing to date. 1925;2:14-9.
3. Greppi E. Ittero emolitico familiare con aumento della resistenza dei
Nonetheless, important avenues to be pursued in fur- globuli. Minerva Med 1928;8:1-11.
ther studies include the identification of specific mu- 4. Micheli P, Penati P, Momigliano LG. Ulteriori richereche sulla anemia
tations that may respond to therapy, particularly with ipocromica splenomegalica con poichilocitosi. Atti Soc Ital Ematol Haema-
tol (Pavia) 1935;16:Suppl 1:10-3.
specific combinations of agents. 5. Whipple GH, Bradford WL. Mediterranean disease thalassemia
(erythroblastic anemia of Cooley): associated pigment abnormalities simu-
Gene Therapy lating hemochromatosis. J Pediatr 1936;9:279-311.
6. Weatherall DJ, Clegg JB. Thalassaemia a global public health prob-
Permanent correction of genetic deficit of the lem. Nat Med 1996;2:847-9.
hematopoietic system requires the transfer of genes 7. Higgs DR. a-Thalassaemia. Baillieres Clin Haematol 1993;6:117-50.
8. Chui DH, Waye JS. Hydrops fetalis caused by alpha-thalassemia: an
into stem cells and long-term, high-level, lineage- emerging health care problem. Blood 1998;91:2213-22.
specific expression of these cells after autologous trans- 9. Weatherall DJ. The thalassemias. In: Stamatoyannopoulos G, Nienhuis
plantation; mature cells and committed progenitors AW, Majerus PH, Varmus H, eds. The molecular basis of blood diseases.
2nd ed. Philadelphia: W.B. Saunders, 1994:157-205.
do not have the proliferative capacity to reconstitute 10. Flint J, Harding RM, Boyce AJ, Clegg JB. The population genetics of
the entire hematopoietic system. Over the past dec- the haemoglobinopathies. Baillieres Clin Haematol 1993;6:215-62.
11. Weatherall DJ. Common genetic disorders of the red cell and the ma-
ade, there has been progress in the development of laria hypothesis. Ann Trop Med Parasitol 1987;81:539-48.
transduction methods and vectors.118 Remaining 12. Allen SJ, ODonnell A, Alexander NDE, et al. a+-Thalassaemia pro-
problems include the identification of all sequences tects children against disease caused by other infections as well as malaria.
Proc Natl Acad Sci U S A 1997;94:14736-41.
required for stable, high-level expression of the genes 13. Orkin SH. Regulation of globin gene expression in erythroid cells. Eur
and the development of more effective and safe vec- J Biochem 1995;231:271-81.
tors for the transfer of genes.119 Another approach, 14. Wood WG, Weatherall DJ. Developmental genetics of the human hae-
moglobins. Biochem J 1983;215:1-10.
correction of the defective gene by site-directed re- 15. Wood WG. Increased HbF in adult life. Baillieres Clin Haematol 1993;
combination, is feasible, but current methods lack the 6:177-213.
16. Grosveld F, Dillon N, Higgs D. The regulation of human globin gene
degree of efficiency required.120 expression. Clin Haematol 1993;6:31-55.
17. Nathan DG, Gunn RB. Thalassemia: the consequences of unbalanced
CONCLUSIONS hemoglobin synthesis. Am J Med 1966;41:815-30.
18. Wickramasinghe SN. The morphology and kinetics of erythropoiesis
Among the first diseases to be studied at the mo- in homozygous b-thalassaemia. In: Congenital disorders of erythropoiesis.
lecular level, the b-thalassemias remain a model for Amsterdam: Elsevier, 1976:221-37.
understanding the relation between the molecular 19. Yuan J, Angelucci E, Lucarelli G, et al. Accelerated programmed cell
death (apoptosis) in erythroid precursors of patients with severe beta-thal-
pathology of a disease and its clinical diversity. At assemia. Blood 1993;82:374-7.
the same time, these disorders have become an in- 20. Schrier SL. Pathobiology of thalassemic erythrocytes. Curr Opin He-
matol 1997;4:75-8.
creasingly important part of clinical practice in all 21. Shinar E, Rachmilewitz EA. Haemoglobinopathies and red cell mem-
countries with large populations from the tropics. brane function. Baillieres Clin Haematol 1993;6:357-69.
The marked increase in survival, to the fifth decade 22. Grinberg LN, Rachmilewitz EA. Oxidative stress in b-thalassemic red
blood cells and potential use of antioxidants. In: Beuzard Y, Lubin B, Rosa
of life, of patients with well-managed b-thalassemia J, eds. Sickle cell disease and thalassaemia: new trends in therapy. London:
in developed countries represents one of the most Colloque INSERM/John Libby Eurotext, 1995:519-24.
dramatic alterations in morbidity and mortality asso- 23. Sorensen S, Rubin E, Polster H, Mohandas N, Schrier S. The role of
membrane skeletal-associated alpha-globin in the pathophysiology of beta-
ciated with a genetic disease in this century. Still, thalassemia. Blood 1990;75:1333-6.
nearly 75 years after the fascinating initial descrip- 24. Shalev O, Repka T, Goldfarb A, et al. Deferiprone (L1) chelates patho-
logic iron deposits from membranes of intact thalassemic and sickle RBC
tion of peculiar bone changes and other signs and both in vitro and in vivo. Blood 1995;86:2008-13.
symptoms of the disorder, the b-thalassemias have 25. Rioja L, Girot R , Garabedian M, Cournot-Witmer G. Bone disease
emerged as a huge public health problem worldwide. in children with homozygous beta-thalassemia. Bone Miner 1990;8:69-
86.
They remain a therapeutic challenge for the next 26. Orvieto R, Leichter I, Rachmilewitz EA, Margulies JY. Bone density,
millennium. mineral content, and cortical index in patients with thalassemia major and

Volume 341 Numb er 2 107

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
The New Eng land Jour nal of Medicine

the correlation to their bone fractures, blood transfusions, and treatment 57. Witzleben CL, Wyatt JP. The effect of long survival on the pathology
with desferrioxamine. Calcif Tissue Int 1992;50:397-9. of thalassaemia major. J Pathol Bacteriol 1961;82:1-12.
27. Gratwick GM, Bullough PG, Bohne WHO, Markenson AL, Peterson 58. Jean G, Terzoli S, Mauri R, et al. Cirrhosis associated with multiple
CM. Thalassemia osteoarthropathy. Ann Intern Med 1978;88:494-501. transfusions in thalassaemia. Arch Dis Child 1984;59:67-70.
28. Thein SL. b-Thalassaemia. Baillieres Clin Haematol 1993;6:151-75. 59. Maharaj B, Maharaj RJ, Leary WP, et al. Sampling variability and its
29. Weatherall DJ, Clegg JB. The thalassaemia syndromes. 4th ed. Oxford, influence on the diagnostic yield of the percutaneous needle biopsy of the
England: Blackwell Scientific (in press). liver. Lancet 1986;1:523-5.
30. Thein SL, Sampietro M, Rohde K, et al. Detection of a major gene 60. Cartwright GE, Edwards CQ, Kravitz K, et al. Hereditary hemochro-
for heterocellular hereditary persistence of fetal hemoglobin after account- matosis: phenotypic expression of the disease. N Engl J Med 1979;301:
ing for genetic modifiers. Am J Hum Genet 1994;54:214-28. 175-9.
31. Dover GJ, Smith KD, Chang YC, et al. Fetal hemoglobin levels in sick- 61. Niederau C, Fischer R, Purschel A, Stremmel W, Haussinger D, Stroh-
le cell disease and normal individuals are partially controlled by an X-linked meyer G. Long-term survival in patients with hereditary hemochromatosis.
gene located at Xp22.2. Blood 1992;80:816-24. Gastroenterology 1996;110:1107-79.
32. Chang YC, Smith KD, Moore RD, Serjeant GR, Dover GJ. An analysis 62. Parkes JG, Randell EW, Olivieri NF, Templeton DM. Modulation by
of fetal hemoglobin variation in sickle cell disease: the relative contribu- iron loading and chelation of the uptake of non-transferrin-bound iron by
tions of the X-linked factor, b-globin haplotypes, a-globin gene number, human liver cells. Biochim Biophys Acta 1995;1243:373-80.
gender, and age. Blood 1995;85:1111-7. 63. Italian Working Group on Endocrine Complications in Non-endo-
33. Thein SL, Hesketh C, Taylor P, et al. Molecular basis for dominantly crine Diseases. Multicentre study on prevalence of endocrine complications
inherited inclusion body b thalassemia. Proc Natl Acad Sci U S A 1990; in thalassaemia major. Clin Endocrinol (Oxf ) 1995;42:581-6.
87:3924-8. 64. Cavallo-Perin P, Pacini G, Cerutti F, et al. Insulin resistance and hy-
34. Camaschella C, Cappellini MD. Thalassemia intermedia. Haematolog- perinsulinemia in homozygous b-thalassemia. Metabolism 1995;44:281-6.
ica 1995;80:58-68. 65. Gullo L, Corcioni E, Brancati C, Bria M, Pezzilli R, Sprovieri G. Mor-
35. Cao A. Diagnosis of b-thalassemia intermedia at presentation. In: Fu- phologic and functional evaluation of the exocrine pancreas in b-thalasse-
charoen S, Rowley PT, Paul NW, eds. Thalassemia: pathophysiology and mia major. Pancreas 1993;8:176-80.
management. Part B. Vol. 23 of Birth defects: original article series. New 66. Magro S, Puzzonia P, Consarino C, et al. Hypothyroidism in patients
York: Alan R. Liss, 1988:219-26. with thalassemia syndromes. Acta Haematol 1990;84:72-6.
36. Rund D, Oron-Karni V, Filon D, Goldfarb A, Rachmilewitz E, Op- 67. Sklar CA, Lew LQ, Yoon DJ, David R. Adrenal function in thalasse-
penheim A. Genetic analysis of b-thalassemia intermedia in Israel: diversity mia major following long-term treatment with multiple transfusions and
of mechanisms and unpredictability of phenotype. Am J Hematol 1997;54: chelation therapy: evidence for dissociation of cortisol and adrenal andro-
16-22. gen secretion. Am J Dis Child 1987;141:327-30.
37. Ho PJ, Hall GW, Luo LY, Weatherall DJ, Thein SL. Beta thalassaemia 68. Bacalo A, Kivity S, Heno N, Greif Z, Greif J, Topilsky M. Blood trans-
intermedia: is it possible consistently to predict phenotype from genotype? fusion and lung function in children with thalassemia major. Chest 1992;
Br J Haematol 1998;100:70-8. 101:362-70.
38. Huisman THJ, Carver MFH, Baysal E. A syllabus of thalassemia mu- 69. Factor JM, Pottipati SR, Rappaport I, Rosner IK, Lesser ML, Giardi-
tations. Augusta, Ga.: Sickle Cell Anemia Foundation, 1997:309. na PJ. Pulmonary function abnormalities in thalassemia major and the role
39. Weatherall DJ. Hemoglobin E b-thalassemia: an increasingly common of iron overload. Am J Respir Crit Care Med 1994;149:1570-4.
disease with some diagnostic pitfalls. J Pediatr 1998;132:765-7. 70. Tai DYH, Wang YT, Lou J, Wang WY, Mak KH, Cheng HK. Lungs
40. Olivieri NF, Brittenham GM. Iron-chelating therapy and the treatment in thalassaemia major patients receiving regular transfusion. Eur Respir J
of thalassemia. Blood 1997;89:739-61. [Erratum, Blood 1997;89:2621.] 1996;9:1389-94.
41. Pippard MJ, Callender ST, Warner GT, Weatherall DJ. Iron absorption 71. Anapliotou MLG, Kastanias IT, Psara P, Evangelou EA, Liparaki M,
and loading in beta-thalassaemia intermedia. Lancet 1979;2:819-21. Dimitriou P. The contribution of hypogonadism to the development of os-
42. Pootrakul P, Kitcharoen K, Yansukon P, et al. The effect of erythroid teoporosis in thalassaemia major: new therapeutic approaches. Clin Endo-
hyperplasia on iron balance. Blood 1988;71:1124-9. crinol (Oxf ) 1995;42:279-87.
43. Risdon RA, Flynn DM, Barry M. The relation between liver iron con- 72. Filosa A, Di Maio S, Vocca S, Saviano A, Esposito G, Pagano L. Lon-
centration and liver damage in transfusional iron overload in thalassaemia gitudinal monitoring of bone mineral density in thalassemic patients: ge-
and the effect of chelation therapy. Gut 1973;14:421. netic structure and osteoporosis. Acta Paediatr 1997;86:342-6.
44. Hershko C, Weatherall DJ. Iron-chelating therapy. Crit Rev Clin Lab 73. Olivieri NF. Thalassaemia: clinical management. Baillieres Clin Hae-
Sci 1988;26:303-45. matol 1998;11:147-62.
45. Hershko C, Konijn AM, Link G. Iron chelators for thalassaemia. Br J 74. Cao A, Galanello R, Rosatelli MC. Prenatal diagnosis and screening
Haematol 1998;101:399-406. of the haemoglobinopathies. Baillieres Clin Haematol 1998;11:215-38.
46. Parkes JG, Hussain RA, Olivieri NF, Templeton DM. Effects of iron 75. Fosburg MT, Nathan DG. Treatment of Cooleys anemia. Blood 1990;
loading on uptake, speciation, and chelation of iron in cultured myocardial 76:435-44.
cells. J Lab Clin Med 1993;122:36-47. 76. Cazzola M, Bornga-Pignatti C, Locatelli F, Ponchio L, Beguin Y, De
47. Buja LM, Roberts W. Iron in the heart: etiology and clinical signifi- Stefano P. A moderate transfusion regimen may reduce iron loading in
cance. Am J Med 1971;51:209-21. b-thalassemia major without producing excessive expansion of erythro-
48. Wolfe L, Olivieri N, Sallan D, et al. Prevention of cardiac disease by poiesis. Transfusion 1997;37:135-40.
subcutaneous deferoxamine in patients with thalassemia major. N Engl J 77. Barry M, Flynn DM, Letsky EA, Risdon RA. Long-term chelation
Med 1985;312:1600-3. therapy in thalassaemia major: effect on liver iron concentration, liver his-
49. Kremastinos DT, Tiniakos G, Theodorakis GN, Katritsis DG, Toutou- tology, and clinical progress. BMJ 1974;2:16-20.
zas PK. Myocarditis in b-thalassemia major: a cause of heart failure. Circu- 78. Bronspiegel-Weintrob N, Olivieri NF, Tyler B, Andrews DF, Freed-
lation 1995;91:66-71. man MH, Holland FJ. Effect of age at the start of iron chelation therapy
50. Aessopos A, Stamatelos G, Skoumas V, Vassilopoulos G, Mantzourani on gonadal function in b-thalassemia major. N Engl J Med 1990;323:713-
M, Loukopoulos D. Pulmonary hypertension and right heart failure in pa- 9.
tients with b-thalassemia intermedia. Chest 1995;107:50-3. 79. Gabutti V, Piga A. Results of long-term iron-chelating therapy. Acta
51. Du ZD, Roguin N, Milgram E, Saab K, Koren A. Pulmonary hyper- Haematol 1996;95:26-36.
tension in patients with thalassemia major. Am Heart J 1997;134:532-7. 80. Porter JB. A risk-benefit assessment of iron-chelation therapy. Drug
52. Brittenham GM, Griffith PM, Nienhuis AW, et al. Efficacy of deferox- Saf 1997;17:407-21.
amine in preventing complications of iron overload in patients with thalas- 81. Brittenham GM, Cohen AR, McLaren CE, et al. Hepatic iron stores
semia major. N Engl J Med 1994;331:567-73. and plasma ferritin concentration in patients with sickle cell anemia and
53. Olivieri NF, Nathan DG, MacMillan JH, et al. Survival in medically thalassemia major. Am J Hematol 1993;42:81-5.
treated patients with homozygous b-thalassemia. N Engl J Med 1994;331: 82. Angelucci E, Giardini C, Brittenham GM, Lucarelli G. Hepatic iron
574-8. concentration and body iron stores determined by quantitative phlebotomy
54. Zurlo MG, De Stefano P, Borgna-Pignatti C, et al. Survival and causes in patients cured of thalassemia major by bone marrow transplantation.
of death in thalassaemia major. Lancet 1989;2:27-30. Blood 1997;90:Suppl 1:265a. abstract.
55. Iancu TC, Neustein HB, Landing BH. The liver in thalassaemia ma- 83. Olynyk JK, ONeill R, Britton RS, Bacon BR. Determination of he-
jor: ultrastructural observations. In: Iron metabolism: Ciba Symposium 51. patic iron concentration in fresh and paraffin-embedded tissue: diagnostic
Amsterdam: Elsevier, 1977:293-309. implications. Gastroenterology 1994;106:674-7.
56. Thakerngpol K, Fucharoen S, Boonyaphipat P, et al. Liver injury due 84. Brittenham GM, Farrell DE, Harris JW, et al. Magnetic-susceptibility
to iron overload in thalassemia: histopathologic and ultrastructural studies. measurement of human iron stores. N Engl J Med 1982;307:1671-5.
Biometals 1996;9:177-83. 85. Angelucci E, Giovagnoni A, Valeri G, et al. Limitations of magnetic

108 Jul y 8, 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.
MED IC A L PR OGR ES S

resonance imaging in measurement of hepatic iron. Blood 1997;90:4736- 103. Arruda VR, Lima CSP, Saad STO, Costa FF. Successful use of hy-
42. droxyurea in b-thalassemia major. N Engl J Med 1997;336:964.
86. Giardini C. Treatment of b-thalassemia. Curr Opin Hematol 1997;4: 104. Rigano P, Manfr L, La Galla R, et al. Clinical and hematologic
79-87. response to hydroxyurea in a patient with Hb Lepore/b-thalassemia. He-
87. Lucarelli G, Giardini C, Angelucci E. Bone marrow transplantation in moglobin 1997;21:219-26.
thalassemia. In: Winter JN, ed. Blood stem cell transplantation. Boston: 105. Perrine SP, Ginder CD, Faller DV, et al. A short-term trial of buty-
Kluwer Academic, 1997:305-15. rate to stimulate fetal-globingene expression in the b-globin disorders.
88. Issaragrisil S, Visuthisakchai S, Suvatte V, et al. Transplantation of N Engl J Med 1993;328:81-6.
cord-blood stem cells into a patient with severe thalassemia. N Engl J Med 106. Olivieri NF, Rees DC, Ginder GD, et al. Treatment of thalassemia
1995;332:367-9. major with phenylbutyrate and hydroxyurea. Lancet 1997;350:491-2.
89. Contu L, La Nasa G, Arras M, et al. Successful unrelated bone marrow 107. Rodgers GP, Rachmilewitz EA. Novel treatment options in the severe
transplantation in b-thalassemia. Bone Marrow Transplant 1994;13:329-31. b-globin disorders. Br J Haematol 1995;91:263-8.
90. Westgren M, Ringden O, Eik-Nes S, et al. Lack of evidence of perma- 108. Saxon BR , Rees D, Olivieri NF. Regression of extramedullary hae-
nent engraftment after in utero fetal stem cell transplantation in congenital mopoiesis and augmentation of fetal haemoglobin concentration during
hemoglobinopathies. Transplantation 1996;61:1176-9. hydroxyurea therapy in b thalassaemia. Br J Haematol 1998;101:416-9.
91. Olivieri NF, Brittenham GM, Matsui D, et al. Iron-chelation therapy 109. Sher GD, Ginder GD, Little JA, Wang SY, Dover G, Olivieri NF.
with oral deferiprone in patients with thalassemia major. N Engl J Med Extended therapy with intravenous arginine butyrate in patients with
1995;332:918-22. b-hemoglobinopathies. N Engl J Med 1995;332:1606-10.
92. Hoffbrand AV, al-Refaie F, Davis B, et al. Long-term trial of defer- 110. Atweh GF, Sutton M, Nassif I, et al. Sustained induction of fetal he-
iprone in 51 transfusion-dependent iron overloaded patients. Blood 1998; moglobin by pulse butyrate therapy in sickle cell disease. Blood 1999;93:
91:295-300. 1790-7.
93. Olivieri NF, Brittenham GM, McLaren CE, et al. Long-term safety 111. Collins AF, Pearson HA, Giardina P, McDonagh KT, Brusilow SW,
and effectiveness of iron-chelation therapy with deferiprone in patients Dover GJ. Oral sodium phenylbutyrate therapy in homozygous beta thal-
with thalassemia major. N Engl J Med 1998;339:417-23. assemia: a clinical trial. Blood 1995;85:43-9.
94. Hoffbrand AV. Oral iron chelation. Semin Hematol 1996;33:1-8. 112. Cappellini MD, Graziadei G, Ciceri L, et al. Phase II open study of
95. Carthew P, Smith AG, Hider RC, Dorman B, Edwards RE, Francis oral isobutyramide in patients with thalassemia intermedia. Blood 1996;
JE. Potentiation of iron accumulation in cardiac myocytes during the treat- 88:Suppl 1:311a. abstract.
ment of iron overload in gerbils with the hydroxypyridinone iron chelator 113. Constantoulakis P, Josephson B, Mangahas L, et al. Locus control re-
CP94. Biometals 1994;7:267-71. gion-Ag transgenic mice: a new model for studying the induction of fetal
96. Wong A, Alder V, Robertson D, et al. Liver iron depletion and toxicity hemoglobin in the adult. Blood 1991;77:1326-33.
of the iron chelator deferiprone (L1, CP20) in the guinea pig. Biometals 114. Stamatoyannopoulos JA, Nienhuis AW. Therapeutic approaches to
1997;10:247-56. hemoglobin switching in treatment of hemoglobinopathies. Ann Rev Med
97. Nathan DG. An orally active iron chelator. N Engl J Med 1995;332: 1992;43:497-521.
953-4. [Erratum, N Engl J Med 1995;332:1315.] 115. McDonagh KT, Dover GJ, Donahue RE, et al. Hydroxyurea-induced
98. Olivieri NF, Nisbet-Brown E, Srichairatanakool S, et al. Studies of HbF production in anemic primates: augmentation by erythropoietin,
iron excretion and non-transferrin-bound plasma iron (NTBP1) following hematopoietic growth factors, and sodium butyrate. Exp Hematol 1992;
a single infusion of hydroxyethyl starch-deferoxamine (HES-DFO): a new 20:1156-64.
approach to iron chelation therapy. Blood 1996;88:Suppl 1:310a. ab- 116. Pace B, Li Q, Peterson K, Stamatoyannopoulos G. a-Amino butyric
stract. acid cannot reactivate the silenced gamma gene of the beta locus YAC
99. Borgna-Pignatti C, Cohen AR. Evaluation of a new method of admin- transgenic mouse. Blood 1994;84:4344-53.
istration of the iron chelating agent deferoxamine. J Pediatr 1997;130:86- 117. Kitsberg D, Selig S, Keshet I, Cedar H. Replication structure of the
8. human b-globin gene domain. Nature 1993;366:588-90.
100. Porter JB, Alberti D, Hassan I, et al. Subcutaneous depot desferriox- 118. Verma IM, Somia N. Gene therapy promises, problems and pros-
amine (CGH 749B): relationship of pharmacokinetics to efficacy and drug pects. Nature 1997;389:239-42.
metabolism. Blood 1997;90:Suppl 1:265a. abstract. 119. Higgs DR, Sharpe JA, Wood WG. Understanding a-globin gene ex-
101. Olivieri NF. Reactivation of fetal hemoglobin in patients with b-thal- pression: a step towards effective gene therapy. Semin Hematol 1998;35:
assemia. Semin Hematol 1996;33:24-42. 93-104.
102. Ley TJ, DeSimone J, Anagnou NP, et al. 5-Azacytidine selectively 120. Shesely EG, Kim H-S, Shehee WR, Papayannopoulou T, Smithies O,
increases g-globin synthesis in a patient with b+ thalassemia. N Engl J Med Popovich BW. Correction of a human b S-globin gene by gene targeting.
1982;307:1469-75. Proc Natl Acad Sci U S A 1991;88:4294-8.

Volume 341 Numb er 2 109

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 7, 2014. For personal use only. No other uses without permission.
Copyright 1999 Massachusetts Medical Society. All rights reserved.

Potrebbero piacerti anche