Вы находитесь на странице: 1из 9

Vol 3 | Issue 2 | 2013 | 71-79.

ISSN 2249 - 7641


Print ISSN 2249 - 765X

International Journal of Pharmacology Research


www.ijprjournal.org

-THALASSEMIA-MINI REVIEW
P Sandhya Rani1*, S Vijayakumar1, G Vijay Kumar2, N Chandana1
1
Department of Pharmacy Practice, Vaagdevi College of Pharmacy, MGM Hospital, Warangal, Andhra Pradesh, India.
2
Department of Paediatrics, Kakatiya Medical College/MGM Hospital, Warangal, Andhra Pradesh, India.

ABSTRACT
-thalassemias are a group of hereditary blood disorders characterized by anomalies in the synthesis of the chains
of haemoglobin resulting in variable phenotypes ranging from severe anemia to clinically asymptomatic individuals. The
incidence of symptomatic individual is estimated at 1 in 1,00,000 throughout the world annually. Whereas average incidence
of -thalassemia trait in India is 3.3% with 12 per 1,000 couples being at risk of having an affected offspring each year.
Three main forms have been described: thalassemia major, thalassemia intermedia and thalassemia minor. Individuals with
thalassemia major usually present within the first two years of life with severe anemia, requiring regular red blood cell (RBC)
transfusions. The purpose of this article is to elucidate the diagnosis and management of the -thalassemia patients. The
article briefly describing the epidemiology, types, clinical features, diagnosis, management of the -thalassemia. We describe
use of hematopoietic stem cell transplantation, which has produced cure rates as high as 97%, and the use of cord blood
transplantation. Finally, we touch on prevention and newer therapy that might be effective in the near future.

Keywords: -thalassemias, Anemia, Blood transplantation.

INTRODUCTION individuals. Phenotypically there are two forms of -


The term thalassemia is derived from the Greek thalassemia: 0 - no globin chain synthesis, and + - with
words Thalassa(sea) and Haema (blood) and refers to some globin chain synthesis. Clinically, thalassemia
disorders associated with defective synthesis of or - presents as beta-thalassemia minor (+ or 0), intermedia
globin subunits of haemoglobin (Hb)A(2;2), inherited as (+/+; +/0) or major (0/0). Thalassemia Major, variably
pathologic alleles of one or more of the globin genes referred to as "Cooley's Anemia" and "Mediterranean
located on chromosomes 11 () and 16 ( ). More than 200 Anemia", Thalassemia Intermedia and Thalassemia Minor
deletions or point mutations that impair transcription, also called "beta-thalassemia carrier", "beta-thalassemia
processing or translation of or -globin mRNA have been trait" or "heterozygous beta-thalassemia". Apart from the
identified. The clinical manifestations are diverse, ranging rare dominant forms, subjects with thalassemia major are
from absence of symptoms to profound fatal anemias in homozygotes or compound heterozygotes for beta0 or beta+
utero or if untreated, in early childhood [1]. genes, subjects with thalassemia intermedia are mostly
The thalassemia syndrome is classified according homozygotes or compound heterozygotes and subjects with
to which of the globin chains, or , is affected. These 2 thalassemia minor are mostly heterozygotes [3,4].
major groups, and -thalassemia, are sub classified
according to absent (o and o) or reduced (+ or +) globin Epidemiology
chain synthesis [1,2]. -thalassemia is prevalent in Mediterranean
-thalassemias are a group of hereditary blood countries, the Middle East, Central Asia, India, Southern
disorders characterized by anomalies in the synthesis of the China, and the Far East as well as countries along the north
chains of haemoglobin resulting in variable phenotypes coast of Africa and in South America. The highest carrier
ranging from severe anemia to clinically asymptomatic

Corresponding Author:- P. Sandhya Rani EMail ID: sandhya.pitta16@gmail.com

71 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

frequency is reported in Cyprus (14%), Sardinia (10.3%), The high level of gamma chain production leads to
and Southeast Asia. The incidence of symptomatic a relatively mild degree of globin chain imbalance and
individual is estimated at 1 in 1,00,000 through the world hence these conditions are much milder than the
annually. Whereas average incidence of -thalassemia trait thalassaemia [1]. Homozygous condition is rare, but
in India is 3.3% with 12 per 1,000 couples being at risk of heterozygous form is common and resembles clinically as
having an affected offspring each year. The incidence of - well as haematologically with -thalassemia trait. HbA2
thalassemia in different regions of India varies from 3% to level is normal or reduced and HbF is elevated from 5 to
17%, with a mean prevalence of 4% [5]. It is estimated that 20%. The Hb lepore disorders are forms of delta
there are about 45 million carriers of the -thalassemia gene thalassemia. These conditions result from the production of
in India, while about 15,000 affected infants are born every delta fusion genes which direct the synthesis of delta
year, contributing to about 10% of the total thalassemics fusion chains. They arise by unequal crossing over between
born all over the world [6,12]. the delta and globin genes, depending upon the position
and structure of abnormal crossing over [6,11]. The
TYPES OF BETA THALASSEMIA symptoms of thalassemia intermedia vary from one
a) Heterozygous +thalassemia or thalassemia minor person to another. They are similar to those of
The heterozygous states for thalassemia are thalassemia major but less severe. Some people may have
associated with a mild but significant degree of anaemia. mild anaemia and grow normally while others may need
The haemoglobin values for males ranged from 9 to 15 g/dl regular blood transfusions to help them grow.
with a mean value of 1213 g/dl. In females, the range has
been between 8 and 13 g/dl with mean value of 910 g/dl. Aetiology
The red cell indices are quite characteristic, with reduced Thalassemia is directly linked to genetics and how
MCH values in the 2022 picogram (pg) range and reduced the genes that affect haemoglobin production are inherited.
MCV values in the 60-70 flankolitre (fl) range [1]. It is People with moderate to severe forms received variant
most unusual to find a -thalassemia carrier with an MCH genes from both parents. People who are carriers of the
above 25pg or an MCV of more than 70 fl. The screening disease received variant genes from one parent and normal
for -thalassemia using an electronic cell counter is also genes from the other parent [3].
effective these days in India [2]. The main diagnostic This disorder is caused from a mutation or deletion
feature of this condition is an elevated HbA2 level in the of the globin production genes which leads to decreased
3.57.5% range. The condition is also called -thalassemia production [9]. As a result, the globin protein chains make
trait. Some cases also show a slight elevation of HbF [6]. an abnormal haemoglobin amount which decreases the
synthesis of haemoglobin. The quantity of the disruptions
b) Homozygous 0thalassemia or thalassemia major in the chain synthesis determines the severity of the
This is a serious condition where patients body Thalassemia [1,4].
can't produce enough healthy haemoglobin or red blood
cells. Symptoms start to develop between the age of three Pathophysiology
and six months. These include paleness, shortness of breath The reduced amount (beta+) or absence (beta0) of
and jaundice (where the whites of your eyes and your skin beta globin chains result in a relative excess of unbound
become yellow). This condition usually presents with alpha globin chains that precipitate in erythroid precursors
severe anaemia within the first year of life [3]. Before the in the bone marrow, leading to their premature death and
first transfusion, these patients show a variable degree of hence to ineffective erythropoiesis [9]. The degree of globin
anaemia with marked variation in shape and size of their chain reduction is determined by the nature of the mutation
red cells with hypochromia and a mild reticulocytosis. The at the beta globin gene located on chromosome [11].
bone marrow shows erythroid hyperplasia and many of the Peripheral haemolysis contributing to anemia is
normoblasts contain ragged inclusions after incubation of less prominent in thalassemia major than in thalassemia
the marrow with methyl violet. The haemoglobin pattern in intermedia, and occurs when insoluble alpha globin chains
this condition consists almost entirely of HbF with variable induce membrane damage to the peripheral erythrocytes
HbA2 levels which may be reduced, normal or elevated [8]. [10]. Anemia stimulates the production of erythropoietin
Haemoglobin A is completely absent. Both parents show with consequent intensive but ineffective expansion of the
the features of heterozygous -thalassemia with elevated bone marrow (up 25 to 30 times normal), which in turn
HbA2 levels [2]. causes the typical previously described bone deformities.
Prolonged and severe anemia and increased erythropoietic
c) Thalassaemia (delta) intermedia drive also result in hepatospleenomegaly and
This thalassemia results from complete absence of extramedullary erythropoiesis [3,10][Fig.1].
both - and delta-chain synthesis, in most cases due to
extensive deletion of DNA in the -globin gene complex.

72 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

survival, which leads to so many complications. These


relate to inadequate transfusions, transfusion-related
infections, allow sensitization, iron-overload related
cardiac, endocrine and liver disturbances and toxicities of
iron chelators [22]. Many of these problems are strongly
age dependent. Heart disease is the most important
complication and the main determinant of survival [16]. It
is responsible for more than half of the deaths. It may take
the form of cardiomyopathy, pulmonary hypertension, heart
failure, arrhythmias, pericarditis and myocarditis [21,24].
Although iron over-load is the main cause but other factors;
genetic, immune or infective, may also be important.
Endocrine complications include diabetes mellitus,
hypothyroidism, hypoparathyroidism, hypogonadism and
delayed puberty [14,20]. Endocrine complications along
with osteoporosis, trace elements deficiency and other
metabolic disturbances also lead to growth failure and short
stature [17,15,18]. Less significant complications include
hepatic involvement, neurological complications, and
psychological manifestation [19,7].
Frequent blood transfusion can also lead to
increased chances of transfusion related infections.
Common blood reactions associated with transfusions are
Fig.1. Pathophysiology of -thalassemia non-hemolytic reactions, allergic reactions, acute hemolytic
reactions, autoimmune hemolytic anemia, delayed
transfusion reactions, transfusion related acute lung injury
Hereditary transmission (TRALI) and graft vs. host disease (GVHD), transfusion
The beta-thalassemias are inherited in an transmitted infections are hepatitis B, C, HIV, malaria and
autosomal recessive manner. The parents of an affected syphilis [23][Fig.2].
child are obligate heterozygotes and carry a single copy of a
disease-causing beta globin gene mutation. At conception,
each child of heterozygotes parents has 25% chance of
being affected, 50% chance of being an asymptomatic
carrier, and 25% chance of being unaffected and not carrier.
The parents of the pro-band have a 1 in 4 (25%) risk of
having further affected children in each pregnancy.
Dominant forms of beta-thalassemia, associated
with mutations that result in the production of highly
unstable beta globulin variants and leading to a clinically
manifesting phenotype of beta-thalassemia.

Clinical features of thalassemic syndromes


Fatigue
Weakness
Shortness of breath
Pale appearance
Irritability
Facial bone deformities
Yellow discoloration of the skin (jaundice)
Slow growth
Protruding abdomen
Dark urine

Complications of -thalassemia
-thalassemia major patients may need to have Fig 2. Complication of beta thalassemia
repeated blood transfusions throughout their life for

73 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

DIAGNOSIS MANAGEMENT OF THALASSEMIA


Clinical Diagnosis Transfusion therapy
Thalassemia major is usually suspected in an The goals of transfusion therapy are correction of
infant younger than two years of age with severe microcytic anemia, suppression of erythropoiesis and inhibition of
anemia, mild jaundice and hepatospleenomegaly. gastrointestinal iron absorption, which occurs in non
Thalassemia intermedia presents at a later age with similar transfused patients as a consequence of increased, although
but milder clinical findings [26]. Carriers are usually ineffective, erythropoiesis.
asymptomatic, but sometimes may have mild anemia. The decision to initiate a regular transfusion
program in a child newly diagnosed with thalassemia must
Haematologic Diagnosis take into account both laboratory and clinical findings. An
RBC indices show microcytic anemia. overlap of genotype and phenotype expression make the
Thalassemia major is characterized by reduced Hb level (<7 clinical assessment the most important step in
g/dl), mean corpuscular volume (MCV) > 50 < 70 fl and distinguishing TM from TI. If the child is growing poorly
mean corpuscular Hb (MCH) > 12< 20 pg. Thalassemia and has developed facial or other bone abnormalities,
intermedia is characterized by Hb level between 7 and 10 and/or when Hb levels are 7 g/dL, regular transfusions
g/dl, MCV between 50 and 80 fl and MCH between 16 and will be beneficial.
24pg. Thalassemia minor is characterized by reduced MCV Before the first transfusion, patients RBCs are
and MCH, with increased HbA2 level [3]. typed for Rh and ABO antigens. At the same time,
cytomegalovirus status should be obtained.
Peripheral blood smear Cytomegalovirus-negative blood products are
Affected individuals show RBC morphologic recommended for potential candidates for curative stem cell
changes [microcytosis, hypochromia, anisocytosis, transplantation (SCT). Parents and first-degree relatives
poikilocytosis (spiculated tear-drop and elongatedcells), should not be blood donors for these candidates. Hepatitis
and nucleated RBC (i.e., erythroblasts)]. The number of B vaccination is given before transfusion therapy, as is
erythroblasts is related to the degree of anemia and is hepatitis A vaccine when age appropriate [28].
markedly increased after spleenectomy. Carriers have less Several different transfusional regimens have been
severe RBC morphologic changes than affected individuals. proposed over the years, but the most widely accepted aims
Erythroblasts are normally not seen [25]. at a pretransfusional Hb level of 9 to 10 g/dl and a post-
transfusion level of 13 to 14 g/dl. This prevents growth
Qualitative and quantitative Hb analysis impairment, organ damage and bone deformities, allowing
By cellulose acetate electrophoresis and DE-52 normal activity and quality of life. The frequency of
microchromatography or HPLC identifies the amount and transfusion is usually every two to four weeks. Shorter
type of Hb present. The Hb pattern in beta-thalassemia intervals might further reduce the overall blood
varies according to beta-thalassemia type. In beta0 requirement, but are incompatible with an acceptable
thalassemia, homozygotes HbA is absent and HbF quality of life. The amount of blood to be transfused
constitutes the 92-95% of the total Hb. In beta+ thalassemia depends on several factors including weight of the patient,
homozygotes and beta+/beta0 genetic compounds HbA target increase in Hb level and hematocrit of blood unit. In
levels are between 10 and30% and HbF between 70-90%. general, the amount of transfused RBC should not exceed
HbA2 is variable in beta thalassemia homozygotes and it is 15 to 20ml/kg/day, infused at a maximum rate of 5
enhanced in beta thalassemia minor. ml/kg/hour, to avoid a fast increase in blood volume. To
Hb electrophoresis and HPLC also detect other monitor the effectiveness of transfusion therapy, some
haemoglobinopathies (S, C, E, Lepore) that may interact indices should be recorded at each transfusion, such as pre-
with beta-thalassemia. and post-transfusion Hb, amount and hematocrit of the
blood unit, daily Hb fall and transfusional interval.
Molecular Genetic Analysis A clinical record of all transfusion events should
The prevalence of a limited number of mutations be monitored annually to identify hyperspleenism. A record
in each population has greatly facilitated molecular genetic of weight, the amount of blood transfused at each visit, and
testing. Commonly occurring mutations of the beta globin the pretransfusion Hb level is needed to calculate the annual
gene are detected by PCR-based procedures. The most transfusion requirement. Although red cell transfusions are
commonly used methods are reverse dot blot analysis or life savers for patients with thalassemia, they are
primer-specific amplification, with a set of probes or responsible for a series of complications and expose the
primers complementary to the most common mutations in patients to a variety of risks. Iron overload is the most
the population from which the affected individual relevant complication associated with transfusion therapy
originated. If targeted mutation analysis fails to detect the [29].
mutation, beta globin gene sequence analysis can be used to
detect mutations in the beta globin gene [27].

74 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

Iron overload assessment and treatment but it should be pointed out that many patients with
As a consequence of repeated long-life red blood moderate or even severe cardiac iron may have normal
cell transfusions, essential for survival, patients with cardiac function, suggesting that T2* is able to identify
thalassemia major accumulate over time iron in the body. preclinical cardiac dysfunction [33,30].
Excess iron is toxic to many tissues, including the liver,
endocrine organs and heart, leading to a series of Iron Chelators
complications which cause morbidity and mortality in these As the body has no effective means for removing
patients. Hypogonadism (35-55% of the patients), iron, the only way to remove excess iron is to use iron
hypothyroidism (9-11%), hypoparathyroidism (4%), binders (chelators), which allow iron excretion through the
diabetes (6-10%), liver fibrosis, and heart dysfunction urine and/or stool. As a general rule, patients should start
(33%) [14, 20]. Iron status should be accurately assessed in iron chelation treatment once they have had 10-20
order to evaluate its clinical relevance, the need for transfusions or when ferritin levels rise above 1000 ng/ml.
treatment, and the timing and monitoring of chelation
therapy [30]. Important properties for an ideal iron chelator
There is variability in iron deposition between and High and specific affinity for Fe3+
within different organs. Serum ferritin is the most High chelating efficiency
commonly used indirect estimate of body iron stores, but Slow rate of metabolism
this reflects only 1% of the total iron storage pool. In Tissue and cell penetration
addition, interpretation of ferritin values may be No iron redistribution
complicated by a variety of conditions. Reliance on serum Relatively non-toxic
ferritin alone can lead to an inaccurate assessment of body Achievement of negative iron balance
iron stores in individual patients. The hepatic iron Low cost
concentration by liver biopsy is considered the most Oral availability [9]
accurate and sensitive method for determining the body
iron burden. However, this is an invasive technique with a Deferoxamine
low but recognized complication rate; the result is affected The first drug available for treatment of iron
by hepatic fibrosis (especially in small biopsies),which is overload was deferoxamine (DFO). Deferoxamineis not
common in thalassemia as a result of increased liver iron orally absorbed and thus needs parenteral administration,
and HCV infection, and iron has been shown to be usually as a subcutaneous 8 to12 hour nightly infusion, 5-7
unevenly distributed in the thalassemic liver even in the nights a week. Average dosage is 20-40 mg/kg body weight
non-cirrhotic stages. Endomyocardial biopsy can evaluate for children and 30-50 mg/kg body weight for adults. In
iron deposition in the heart in thalassemia, but is not a high risk cases, continuous administration of DFO via an
routine part of patient management due to the invasiveness implanted delivery system or subcutaneously, at doses
of the technique and the heterogeneous deposition of iron in between 50 and 60 mg/kg per day, were the only options to
the heart [31, 32]. In addition, iron has been reported to be intensify the chelation treatment before the advent of the
absent from the right ventricular sub endocardium in some combined therapy with DFO and deferiprone [34].
patients with cardiac iron overload. Implanted delivery systems are associated with risk of
There are non-invasive techniques that can assess thrombosis and infection. With DFO, iron is excreted both
tissue iron. Both magnetic resonance (MR) and magnetic in faeces (about 40%) and in urine. The most frequent
susceptometry are significantly affected by iron, and both adverse effects of DFO are local reactions at the site of
show changes in iron overload. However, to date only MR infusion, such as pain, swelling, in duration, erythema,
can be applied to a moving organ such as the heart. burning, pruritus, wheals and rash, occasionally
Fortunately there has been substantial progressing the speed accompanied by fever, chills and malaise. Other
and image quality of cardiovascular MR in recent years, complications, mainly associated with high doses of DFO
and therefore we investigated its use in assessing in young patients and low ferritin values are:
myocardial iron in thalassemia. Iron shortens T2*
relaxation time, which has become the most sensitive, Sensorineural hypoacusia, particularly at high
robust and therefore most used measure of cardiac iron. In frequencies
2001 Anderson et al.61 published a fundamental paper Ocular toxicity (night-blindness, blurred vision,
describing the relationship between cardiac T2* and left decreased visual acuity, impairment of colour vision,
ventricular ejection fraction in patients with thalassemia cataract and other disturbances of the eye)
major. Cardiac T2* greater than 20 milliseconds (ms) Retarded growth and skeletal changes with a
indicates normal iron content, while values between 10 and disproportionately short trunk and dysplasia of the long
20 ms or lower than 10 ms indicate moderate or severe iron bones
overload respectively. As T2* decreases below 20 ms, Infections by Yersinia Enterocolitica, and other
increases the risk of having a decline in ejection fraction, pathogens (Klebsiella Pneumoniae).

75 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

The use of DFO decreases morbidity and mortality among reported following the post marketing use of DFX [30].
those who are able to comply with regular prolonged [Table 1]
infusions. However, because of the side effects and the
inconvenient parenteral administration, a consistent Table 1. Comparison of the 3 leading iron-chelating
proportion of patients is non-compliant, limiting the drugs in the management of thalassemia
Compound
usefulness of this chelator [36]. Deferoxamine Deferiprone Deferasirox

Molecular weight, Da 657 139 373


Deferiprone Recommended dose 30-60 mg/kg 75-100 mg/kg
20-40 mg/kg per
day
Deferiprone or L1 (DFP) is currently the only Subcutaneous or
Oral 3 times
orally active iron chelator available for clinical use. It was Delivery intravenous 8-12 h, 5-7
d/wk
daily
Oral once daily

licensed first in India in 1995 and subsequently in Europein Half-life 8-10 min 1.5-4 h 12-18 h
1999. At present, DFP is licensed in Europe for patients for Excretion 40%-60% faecal 90% urinary 90% faecal
whom treatment with DFO is inadequate. Gastrointestinal
Overall DFP is currently available in Gastrointestinal
upset, rash,
Ocular, auditory ocular, auditory
approximately 50 countries. Studies have demonstrated a Adverse effects
toxicity, growth
upset,
arthralgia,
toxicity,
retardation, local reversible
stable or declining mean serum ferritin and liver iron reactions, allergy
neutropenia,
increases in
Agranulocytosis
concentration during long-term therapy in most transfusion creatinine,
hepatitis
dependent patients, although iron accumulation continues
in others. Agranulocytosis is the most serious side effect, Combined chelation therapy
with a reported incidence of 0.6 per 100 patient years. More DFP and DFO can be given to the same patient
common but less serious side effects are gastrointestinal with different regimes: in combination on the same day,
symptoms (e.g., nausea, vomiting, gastric discomfort), either simultaneously (i.e. DFP given before breakfast,
arthralgia, zinc deficiency, and fluctuating ALT levels, lunch and dinner and DFO infused during the day) or
particularly in anti-hepatitis C virus (HCV)-positive sequentially (i.e. DFP as above and DFO infused
patients. Retrospective studies in patients treated with DFP overnight), or as alternate treatment (i.e. one or the other
have shown significant improvement in cardiac magnetic chelator is given on different days). Combination therapy is
resonance imaging (MRI), consistent with a reduction in considered an intensive chelation regimen and usually DFP
cardiac iron overload and improved cardiac function, in is administered every day, while subcutaneous DFO is
comparison with patients treated with DFO [8,9]. These given 2 to 7 days/week, according to the severity of iron
observations support a potential cardioprotective role of overload [34,35]. In patients with heart failure, to reinforce
DFP that needs to be confirmed in prospective randomized chelation, DFO can be given intravenously 24 h/day.
trials [30,3].
The potential advantages of the combined chelation are
Deferasirox Access to different iron pools
Deferasirox (DFX) is a once-daily, orally Prevention of non transferrin bound iron (NTBI)
administered iron chelator that a large program of clinical accumulation
trials has shown to be effective in adults and children. It Increased efficacy
received European Union marketing authorization as an
Decreased toxicity
orphan drug from the EMEA in 2002 and was authorized
Better compliance
for marketing in most countries in 2006. The recommended
starting dose of DFX for most patients is 20 mg/kg/day, Improved quality of life
although this can be modified to 10 or 30 mg/kg/day
depending on the number of transfusions a patient is Bone marrow transplantation (BMT)
receiving and whether the therapeutic goal is to decrease or Bone marrow transplantation (BMT) remains the
maintain body iron levels. The most frequent adverse only definitive cure currently available for patients with
events reported during treatment with DFX include thalassemia. The outcome of BMT is related to the
transient, mild-to-moderate gastrointestinal disturbances pretransplantation clinical conditions, specifically the
and skin rash. These events rarely require drug presence of hepatomegaly, extent of liver fibrosis, history
discontinuation and most resolve spontaneously. Mild, of regular chelation and hence severity of iron
usually nonprogressive increases in serum creatinine accumulation [37]. In patients without the above risk
(generally within the upper limit of normal) has been factors, stem cell transplantation from an human leukocyte
observed in approximately a third of patients. Creatinine antigen (HLA) identical sibling has a disease free survival
levels returned spontaneously to baseline in most of rate over 90%. The major limitation of allogenic BMT is
patients and data from up to 3.5 years of treatment in more the lack of an HLA-identical sibling donor for the majority
than1000 patients have confirmed that creatinine increase is of affected patients. In fact, approximately 25-30% of
non progressive. However, cases of renal failure have been thalassemic patients could have a matched sibling donor.

76 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

BMT from unrelated donors has been carried out proved unsuccessful due to the limitation of available gene
on a limited number of individuals with beta-thalassemia. transfer vectors. The newer approaches to overcome these
Provided that selection of the donor is based on stringent limitations, includes the introduction of lentiviral vectors.
criteria of HLA compatibility and that individuals have New approaches have also focused on targeting the specific
limited iron overload, results are comparable to those mutation in the globin genes, correcting the DNA sequence
obtained when the donor is a compatible sib. However, or manipulating the development in DNA translocation and
because of the limited number of individuals enrolled, splicing to restore globin chain synthesis [41,40].
further studies are needed to confirm these preliminary
findings. If BMT is successful, iron overload may be PREVENTION
reduced by repeated phlebotomy, thus eliminating the need Creating awareness
for iron chelation. Chronic graft versus-host disease Creating awareness about thalassemia to the
(GVHD) of variable severity may occur in 5-8% of general population, government and medical communities
individuals [41, 38]. by holding seminars, workshops and writing articles in the
daily newspapers, broadcasting in television and radio is of
Cord blood transplantation prime importance.
Cord blood transplantation from a related donor
offers a good probability of a successful cure and is Genetic counselling
associated with a low risk of graft versus- host disease Provides information for individuals and at risk
(GVHD) [3]. For couples who have already had a child couples (i.e. both carriers) regarding the mode of
with thalassemia and who under take prenatal diagnosis in a inheritance, the genetic risk of having affected children and
subsequent pregnancy, prenatal identification of HLA the natural history of the disease including the available
compatibility between the affected child and an unaffected treatment and therapies under investigation [42].
foetus allows collection of placental blood at delivery and
the option of cord blood transplantation to cure the affected Prenatal diagnosis
child. On the other hand, in cases with an affected fetus and Acceptance of prenatal diagnosis and termination
a previous normal child, the couple may decide to continue of affected foetuses are dependent on the early
the pregnancy and pursue BMT later, using the normal identification of couples at risk. Prenatal diagnosis for
child as the donor. pregnancies at increased risk is possible by analysis of
DNA extracted from foetal cells obtained by amniocentesis,
FUTURE THERAPIES usually performed at approximately 15-18weeks' gestation
Stimulating HbF production or chorionic villi sampling at 11 weeks' gestation. Both
Individuals with Hereditary persistence foetal disease-causing alleles must be identified before prenatal
haemoglobin (HPFH) demonstrate that preventing or testing can be performed. New technology using foetal
reversing the switch from foetal to adult haemoglobin DNA obtained from maternal plasma or maternal peripheral
would provide efficacious therapy for thalassemia and blood has also been developed but is not routinely available
various other haemoglobinopathies. It has been observed [39].
that some patients recovering from cytotoxic therapy have
reactivated HbF synthesis. Several therapeutic agents such CONCLUSION
as erythropoietin, hydroxyurea, cytarabine and butyrate Thalassaemia major imposes highly clinical,
analogs have produced an increase in HbF synthesis in the psychological burden on the patients and economical
thalassemic patients by stimulating the HbF-producing burden their family and hence our article briefly describing
progenitor cell population [1,4]. the epidemiology, types, clinical features, diagnosis,
management of the -thalassemia. We describe use of
Gene therapy hematopoietic stem cell transplantation, which has
Thalassemia can be cured by introducing or produced cure rates as high as 97%, and the use of cord
correcting a gene into the hematopoietic compartment or a blood transplantation. Finally, we touch on prevention and
single stem cell. Initial attempts at gene transfer have newer therapy that might be effective in the near future.

REFERENCES
1. Eliezer AR and Patricia JG. How I treat thalassemia. Blood Journal Hematology library Org, 118, 2011, 3479-3488.
2. Amit KM, Archana Tiwari. - Thalassaemia - A Fatal Blood Disorder. Int. J. Rev. Life. Sci, 1(2), 2011, 83-87.
3. Renzo Galanello and Raffaella Origa. Beta-thalassemia. Galanello and Origa Orphanet Journal of Rare Diseases, 5, 2010,
11.
4. Alain J, Marengo-Rowe MD. The thalassemias and related disorders. Proc (Bayl Univ Med Cent), 20, 2007, 2731.
5. Sood SK, Madan N, Colah R, Sharma S, Apte SV. Collaborative Study on thalassaemia. Report of ICMR Task Force Study.
Indian Council of Medical Research, 1993, 20-27.

77 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

6. Balgir RS. The burden of haemoglobin opathies in India and the challenges ahead. Current Science, 79, 2000, 1536-1547.
7. Deborah Rund MD and Eliezer Rachmilewitz MD. Medical progress beta-thalassemia. N Engl J Med, 353, 2005, 1135-46.
8. Hunt T. Control of globin synthesis in haemoglobin: structure, function and synthesis. Br Med Bull, 1976, 32(3), 257261.
9. Alan RC, Renzo G, Dudley JP, Melody JC, et al. Thalassemia. American Society of Hematology, 2004, 14-34.
10. Stanley L, Schrier MD. Pathophysiology of thalassemia. Current Opinion in Hematology, 2002, 9, 123126.
11. Weatherall DG, Clegg JB. The thalassemia syndromes, 4thed, Oxford, UK, Blackwell, 2000, 1-10.
12. Ishwar CV, Renu S & Sudha K. Past, present & future scenario of thalassemic care &control in india. Indian J Med Res,
134, 2011, 507-521.
13. Emanuele Angelucci MD, Gary M.Brittenham MD, Christine E. Mclaren. Hepatic iron concentration and total body iron
stores in thalassemia major. Nejm.org, 343, 2000, 327-331.
14. Leila JN, Steven DM, Richard MW. Pancreatic iron and glucose dysregulation in thalassemia major. Am.J.Hematil, 87,
2012, 155-160.
15. Anita S. Growth Retardation in Thalassemia Major Patients. Int J Hum Genet, 3(4), 2003, 237-246.
16. Dimitrios T, Kremastinos. Heart failure in -thalassemia.CHF, 7, 2001, 312-314.
17. Meropi Toumba and Nicosskordis. Osteoporosis syndrome in thalassemia major: an overview. Journal of osteoporosis,
2010, 1-7.
18. Akm Amirul M, Shahnoor I, Afiqul I. Growth status and serum zinc level in patients with haemoglobin-E--thalassemia.
Bangladesh J Child Health, 2012, 3(2), 76-81.
19. Manoj J, Abhay SB, Ashok P. Psychosocial problems in thalassemic adolescents and young adults. Chronicles of young
scientists, 2013, 21-23.
20. Ayfer GP, Turgay DG, et al. Assessment of thyroid function in children aged 1-13 years with beta-thalassemia major. Iran J
Pediatr, 21(1), 2001, 77-82.
21. Eldor A, Rachmilewitz EA. The hypercoagulable state in thalassemia. Blood, 99(1), 2009, 3643.
22. Agarwal MB. Advances in management of thalassaemia (Editorial). Indian Pediatrics, 41, 2004, 989-92.
23. Cunningham MJ, Macklin EA, Neufeld EJ, Cohen AR. Thalassemia Clinical Research Network. Complications of beta-
thalassemia major in North America. Blood, 2004, 104(1), 3439.
24. Samira ZS, Basma AA, Amr SA. The early cardiac involvement in patients with -thalassemia major. The Egyptian Heart
Journal, 2013, 1-7.
25. Ram Mohan J, Ashina S, Shashikant A, Tandra C. Screening of -thalassemia trait by means of red cell indices and derived
formulae. Medical Journal of Dr. D. Y. Patil University, 6(1), 2013, 71-74.
26. Weatherall DJ. The diagnostic features of the different forms of thalassaemia: In the Thalassaemias. Chur-chill Livingstone,
Edinburgh, 1983, 1-9.
27. Vrettou C, Traeger-Synodinos J, Tzetis M, Malamis G, Kanavakis E. Rapid screening of multiple betaglobin gene mutations
by real-time PCR on the Light Cycler: application to carrier screening and prenatal diagnosis of thalassemia syndromes.
Clin Chem, 49, 2003, 769-776.
28. Di Marco V, Capra M, Angelucci E et al., Management of chronic viral hepatitis in patients with thalassemia:
recommendations from an international panel. Blood, 116(16), 2010, 2875-2883.
29. Cohen AR, Glimm E, Porter JB. Effect of transfusional iron intake on response to chelation therapy in beta-thalassemia
major. Blood, 2008, 111(2), 583-587.
30. Antonio C, Paolo M, Renzo G. Recent advances in -thalassemias. Pediatric Reports, 3, 2011, e17.
31. Leilla J, Susan MC, Anne SN, Thomas DC and John CW. Longitudinal analysis of heart and liver iron in thalassemia major.
Blood, 112, 2008, 2973-2978.
32. Leilla JN, Thomas DC and John CW. Pancreatic iron loading predicts cardiac iron loading in thalassemia major. Blood, 114,
2009, 4021-4026.
33. Anderson LJ, Holden S, Davis B et al., Cardiovascular T2-star (T2*) magnetic resonance for the early diagnosis of
myocardial iron overload. Eur Heart J, 22, 2001, 2171-9.
34. MS Ramezani, A Tamaddoni. Comparison between Deferoxamine and combined therapy with Deferoxamine and
Deferiprone in iron overloaded thalassemia patients. Iranian Red Crescent Medical Journal, 12(6), 2010, 655-659.
35. Nishi M, Sunil G, Rrohit S. Comparative efficacy of desferrioxamine, Deferiprone and in combination on iron chalation in
thalassemia children. Indian paediatrics, 41, 2004, 21-27.
36. Gary M, Brittenham MD, Patricia M, Griffith RN, Arthur W, Nienhuis MD et al., Efficacy of Deferoxamine in preventing
complications of iron overload in patients with thalassemia major. Nejm.org, 331, 2013, 567-573.
37. Emanuele A. Hematopoietic stem cell transplantation in thalassemia. American society of hematology, 2010, 456-462.
38. Khaled M, Maria DC and Ali Taher. Challenges associated with prolonged survival of patients with thalassemia: Transition
from children to adulthood. Pediatrics, 121, 2008, 1426-1429.

78 | P a g e
Vol 3 | Issue 2 | 2013 | 71-79.

39. Colah RB, Gorakshakar RC & Nadkarni AH. Invasive & non-invasive approaches for prenatal diagnosis of
haemoglobinopathies: Experiences from india. Indian J Med Res, 134, 2011, 552-560.
40. Susan PP. Fetal globin induction can it cure -thalassemia?. American society of hematology 2005, 38-44.
41. Raja JV, Rachchh MA, Gokani RH. Recent advances in gene therapy for thalassemia. J Pharm Bioall Sci, 4, 2012, 194-201.
42. Rahman MJ, Rahman MH. Prevention & control strategy of thalassemia in Bangladesh. The ORION Medical Journal, 16,
2003, 121-122.
43. Thalassaemia major imposes highly clinical, psychological burden on the patients and economical burden.

79 | P a g e

Вам также может понравиться