Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

A Review of Iron Overload in Beta-Thalassemia Plasmatology

Volume 16: 1–9


Major, and a Discussion on Alternative Potent © The Author(s) 2022
Article reuse guidelines:
Iron Chelation Targets sagepub.com/journals-permissions
DOI: 10.1177/26348535221103560

Piyush Kumar Yadav and Ajay Kumar Singh


Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, India.

ABSTRACT: For years, arrhythmias have been well documented in the medical arena as a cardiovascular consequence of iron overload (IO).
They are thought to be linked to the accumulation of iron in the myocardium. Iron is the earth’s fourth most abundant element and the second
most plentiful metal (after aluminium). When it comes to biology, iron fills two roles: it’s necessary and it’s poisonous. It is necessary as a
trace iron element since it is found in hemoproteins such as haemo-globin, but it is poisonous in excess amounts of the ability to produce
free radicals, which can harm the biological system. The high prevalence of cardiomyopathy in patients with hemosiderosis, particularly in
cases of transfusional iron overload, strongly suggests that iron deposition in the heart plays a key role in the development of heart failure.
Thalassemia major, which necessitates blood transfusion as a treatment, absorbs a large amount of iron in the patient’s duodenum. Moreover,
Iron Overload causes a threat to vital organs such as the liver and, initiates events of the pathologic progression involving apoptosis, fibrosis,
and ultimately cardiac dysfunction. Furthermore, we discuss the iron overload issue as it relates to beta-thalassemia major patient blood trans-
fusion treatment, as well as key individuals accountable for iron excess that ultimately leads to cardiomyopathy.

KEYWORDS: Thalassemia major, Iron overload, Chelation Methods

RECEIVED: November 18, 2022. ACCEPTED: May 11, 2022. DECLARATION OF CONFLICTING INTERESTS: The author(s) declared no potential conflicts
of interest with respect to the research, authorship, and/or publication of this article.
TYPE: Review
CORRESPONDING AUTHOR: Ajay Kumar Singh, Department of Bioinformatics, Central
FUNDING: This work is supported by the Indian Council of Medical Research, New Delhi India University of South Bihar, Gaya, Bihar, India.
under the ICMR Grant (ISRM/12(22)/2019, ID No. 2019-0370) for this manuscript. Email: ajaysingh@cusb.ac.in

Introduction by total body iron stores. Standard concentrations of ferritin


Thalassemia is a multi-genetic hereditary condition, namely alpha differ by sex and age.9 Ferritin concentration tends to increase
thalassemia, beta-thalassemia, delta beta-thalassemia and some at the age of around one year and even grow in adulthood.
others.1 Thalassemia is a hereditary disease, which means that at Males, however, have a higher level of concentration values
least one parent must be a carrier of the disorder. To be affected than females.10
by the disorder, a child must receive one abnormal gene from As NTDT has indeed been significantly investigated and
each parent.2 It is caused either by a genetic disorder or by the researched over the past few years, it has been found in patients
deletion of certain main segments of the gene. In a cluster of that do not rely on regular blood transfusion, iron overload dif-
the beta-globin gene on chromosome 11,3 and the 16 chromo- ferentially affects the liver rather than the myocardium. This
somes on alpha-globin gene cluster,4 molecular defects result in was apparent from retrospective studies that demonstrated a
inaccurate production of hemoglobin.5 Thalassemia conditions lack of cardiovascular siderosis in patients with extreme
with multiple clinical symptoms, phenotypes and therapeutic hepatic iron excess.11 Physiologically there is a lack of a mech-
options are focused on a continuum of severity. TDT (transfusion- anism to remove excess iron load caused by blood transfusion
dependent thalassemia) and NTDT (non-transfusion dependent from the human body. 200-250 mg is elemental iron present
thalassemia) are two types of thalassemia.6 In both transfusion- in each transfused unit of packed red blood cells. Transfusion
dependent thalassemia and non-transfusion dependent thalassemia, iron normally amounts to 03 to 0.6 mg/kg per day for TDT
iron overload is associated with high morbidity. Iron overload is patients, with an estimated monthly average transfusion
caused by an excess accumulation of intestinal iron confirmed volume of red blood cells filled with 2 to 4 units. This excess
by inadequate erythropoiesis.7 Excess iron deposition, which iron accounts for the disruption of major vital organs’ function-
begins in the first year of routine blood transfusion, causes ing and causes irreversible damage.
harm to many vital organs. Cardiomyopathy, often caused by iron overload, is a frequent
Hemochromatosis, also known as iron overload, is charac- and preventable form of heart failure.8 With atrial and ventricular
terized by improper iron accumulation in the functional parts tachyarrhythmias, iron accumulation in the heart tissue promotes
of an organ, which results in organ damage and failure.8 non-homogeneous electrical conduction and repolarization.12
Human bodies store iron, mainly in the form of ferritin. The entire cardiac conduction system, particularly the atrioven-
Limited content of ferritin is secreted into bloodstreams. The tricular node, may be affected by iron deposition. A permanent
blood ferritin concentration is checked, in the absence of pacemaker may be required if a complete atrioventricular block
inflammation, this blood ferritin is linked positively corrected, is caused by iron deposition.13

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0
License (https://creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without
further permission provided the original work is attributed as specified on the SAGE and Open Access page (https://us.sagepub.com/en-us/nam/open-access-at-sage).
2 Plasmatology

In almost every nation on the planet, including Northern in iron overload, especially in individuals with NTDT
Europe, where thalassemia was historically absent, migration (Non-transfusion-dependent Thalassemia) and contributes to
and marriages between different ethnicities induced thalassemia the transfusion overload of Fe in individuals with TDT
in humans. Beta-thalassemia has been reported to be around (Transfusion-dependent Thalassemia).
1.5 per cent of the world’s population (80-90 million people)
carriers, with about 60,000 serving as symptoms born globally, Therapeutic drugs used for the chelation of iron
the overwhelming majority in the developing countries in par-
Iron chelation therapy is used to minimize the accumulation of
ticular. The internationally estimated average annual frequency
iron deposition in the patient’s vital organs such as the liver and
of symptomatic people is 1 in 100,000.14 Beta-thalassemia with
heart. The most common FDA-approved iron chelators are
irregular Hb or structural Hb form with thalassemia character-
Deferoxamine (DFO), intravenous administration for the treat-
istics has been the most common combination within the
ment of transfusion-induced acute iron overdose and chronic
region of South East Asia with a carrier level of about 50%.2
iron excess overload. Deferiprone (DFP), an oral iron chelator
A keyword search of Cardiac arrhythmia was undertaken in
with a 3:1 molar ratio of iron-binding, prevent iron buildup but
the literature databases Pubmed, Gene Reviews, and Scientific
does not adequately prevent iron-induced organ dysfunction,
Research Journals from 1980 to 2021 for this review paper.
and Deferasirox (DFX/DFRA) is a tridentate oral iron chelator
With indications of a relationship between arrhythmia and
that binds specifically to ferric iron to mobilise stored iron.19
iron overload, primarily in beta-thalassemia major and other
A severe complication of iron overload in beta-thalassemia is
blood diseases requiring transfusions, such as hemochromatosis
cardiac dysfunction, which results in a 71 per cent mortality rate
and severe anaemia.
due to iron accumulation in the myocardium.20 It is critical to
reduce LPI (Labile Plasma Iron) and eliminate excess iron to
Etiology
avoid major consequences from iron overload.21 Phlebotomy
Thalassemia is autosomal recessive, which simply means both
is impossible in individuals with severe beta-thalassemia and
parents need to be affected with or carrier for the disease to
hereditary hemochromatosis because they are anaemic. As a
pass it on to the next generation. It is caused by mutations of
result, iron chelation therapy is indeed the best option for treat-
the Hb genes, resulting in alpha or beta chains being under-
ing iron overload.
produced or not present. More than 200 mutations are
known as the causes of thalassemia. Alpha thalassemia and
Efficacy of iron-chelating drugs
beta-thalassemia are caused by the deletion of alpha-globin
and beta-globin genes respectively caused by a point mutation Chelating medicines in general have a variety of qualities that
on chromosome 11 in the splice site and promoter regions of can impact iron elimination along with intake, and they can
the beta-globin gene.15 be useful in the treatment of a variety of iron-metabolism dis-
eases, including Beta-Thalassemia, and other Iron-Loaded
Diagnosis Non-Transfusion Dependent Thalassemia. In each situation,
a risk-benefit evaluation for the chosen therapy is indicated,
Patients with thalassemia disorder are found to have an inci-
based on the individual differences as well as the chelating med-
dence count when mild microcytic anemia is observed in their
ication’s long-term effectiveness, safety, and expense.
standard blood samples. Thalassemia, iron deficiency, chronic
Deferasirox (DFX/DFRA) has been reported in iron meta-
sideroblastic anemia and lead poisoning (also known as plum-
bolic balance tests to be effective for increasing faecal, but not
bism) are responsible for microcytic anemia.16
urinary, iron excretion, as well as lowering liver iron and
serum ferritin values in some patients using the recommended
Metabolism of thalassemia and iron
doses of 10–30 mg/kg/day. Medications of 10–40 mg/kg have
Erythropoiesis (from Greek “erythro” meaning “red” and been observed in iron-loaded thalassemia patients to generate a
“poiesis’ meaning “to make”) and iron (Fe) metabolism are steady rise in iron excretion, which would have been dose-
closely related. The hormone that regulates the absorption of dependent but not adequate to create a negative iron balance
iron is hepcidin, synthesized in the liver.17 Hepcidin synthesis (>15-20 mg iron excretion) in the number of patients at 20
is regulated by transferrin saturation, the concentration of mg/kg or even in most cases at 30 mg/kg/day. In most patients,
iron, inflammation and erythropoiesis demand. Several eryth- medication of 40 mg/kg of deferasirox is more effective, with a
roid factors affect hepcidin development, eg growth differenti- mean iron excretion rate of 28 mg/day per 50 kg man body
ation factor (GDF-15) and erythroferrone (Erfe). Increased IE weight.22
(interleukin), anemia, and inflammation lead to a rise in DFX/DFRA is now used by millions of transfusional iron-
GDF-15 development leading to suppression of hepcidin.18 loaded patients. Many current types of research and assess-
This, in turn, results in increased absorption of Fe from the ments have been published indicating that DFRA is a relatively
intestine. High intestine absorption of Fe leads to an increase effective, well-tolerated, and safe medicine, raising the hopes of
Yadav and Singh 3

many patients for a more effective treatment.23 Acute renal Iron Overload in beta-Thalassemia
failure caused mortality, as well as additional major toxic side Standard blood transfusions are required in the most severe form
effects such as Diarrhea, nausea, constipation, and abdominal of beta-thalassemia to reduce the risk of anaemia. Iron overload is
pain; skin rashes; and a rise in serum creatinine level24 caused caused by RBC (red blood cell) count being boosted by monthly
by the prolonged use of DFX/DFRA. DFX/DFRA’s effective- blood transfusions, hemolysis, and increased absorption of iron
ness is also challenged, as there is no indication that it can achieve from the duodenum and proximal jejunum.33 Cardiac stiffness is
negative iron balance or eliminate excess cardiovascular iron. the major cause of death in thalassemia patients who have received
The elimination rate of DFO from plasma is faster and is blood transfusions.34 1-2 mg of iron is excreted from the human
dependent on the route of drug administration. Intravenous body in a day, whereas a transfused red blood cell unit contains
DFO has a half-life of 5-10 min while intramuscular DFO about 200 mg of iron.35 In absence of any iron chelation
has a half-life of roughly 60 min. The elimination of DFO therapy, an individual getting 25 blood units a year requires 5 g
from blood is faster (5-10 min) than that of its iron complex of iron each year. Iron toxicity is highly harmful to all cells in
(90 min). It is expected that during the 8-12 h long subcutaneous the body and can cause severe and permanent organ failure,
DFO dose, DFO is eliminated relatively quickly and a threshold such as liver disease, diabetes, cardiac failure and testosterone defi-
level in plasma is reached after about 4 h.25 The most frequent ciency.34 The iron surcharge in the bloodstream can be measured
major toxic adverse effects of DFO include ocular, auditory, using, urinary iron excretion, hepatic iron content, serum ferritin
and bone problems. The side of the DFO injection causes hard- and TIBC (Total Iron Binding Capacity) levels.7 The iron toxicity
ness, edoema (excess fluid trapped in the body’s tissues), and dis- threshold values are concentration of, serum ferritin > 2500 ng/mL,
comfort in over 80% of thalassaemia patients. To increase DFO urinary iron discharge> 20 mg/day, concentration of liver iron
compliance and efficacy, different formulations and administra- more than 440 mmol/g and transferrin congestion> 75%.36
tion strategies have been used in the past. The oral formulation Estimating the concentration of hepatic iron by MRI (magnetic
of DFO did not affect iron excretion.26 resonance imaging) is a widely used method in beta-thalassemia
In iron-loaded patients, dosages of 75-120 mg/kg/day of major to assess the overburden of iron37 T2* Magnetic
Deferiprone (DFO/L1) are usually enough to produce a nega- Resonance Imaging (T2* MRI) is a non-invasive approach for
tive iron balance.27 Gastrointestinal symptoms, granulocytosis, determining heart and liver iron overload.38 T2* MRI is a
agranulocytosis, and hepatic enzyme increase.28 The combin- highly reproducible and sensitive approach for detecting cardio-
ation of DFO and L1 is especially beneficial for patients who vascular iron overload. Serum ferritin did not affect the T2* MRI
are experiencing toxicity from either medicine or who are not reading (p-value: 0.464).39 Iron over-burden has additionally
fully cooperating with DFO therapy, because compliance been seen in patients with NTDT (non-transfusion-subordinate
with L1 is substantially higher than compliance with DF. In thalassemia).40 Beta-thalassemia with histidine to aspartic acid
terms of toxicities, the use of combination therapy is especially (H63D) experiences mutation in carriers patients, and iron over-
beneficial for decreasing high doses of subcutaneous or intra- load at codon 63 of the HFE gene which indicates that the
venous DFO, which are known to produce auditory and H63D mutation can influence the absorption of iron.41 Iron
ocular toxicity in a large percentage of patients, as well as L1 overabundance increases the risk of hepatitis, (swollen liver),
dosages that may cause arthropathy.29 fibrosis, and or irreversible damage to the liver due to scarring
iron excess also leads the chance of abnormal heart rhythms/
Role of Hepcidin in Iron Metabolism arrhythmias, or, and cardiac obstruction.24
Iron metabolism is a closely controlled biological process with little
redundancies, and its disruption frequently results in iron deficiency
Signaling pathways that regulate Hamp/Hepcidin expression
or iron overload. Anemia, caused by an iron deficiency, is a major
public health hazard that affects up to a billion people globally. Stat3 signaling pathways- Stat3 signaling pathway is a regulator
Iron, on the other hand, has the potential to be toxic.30 It produces for hepcidin, mostly through inflammation.42 This contributes to
reactive oxygen species (ROS) when it combines with oxygen, the development of IL-6 and IL-22 but not to IL-1β and
causing cell damage. Three regulatory mechanisms manage iron factor-α tumour necrosis (TNF-α).43 The Stat3-binding site
metabolism in mammals: one controls cellular iron metabolism and the SiRNA- mediated Stat3 knockdown contain a promoter
through iron regulatory proteins (IRPs), which bind iron- from Hamp which significantly lower the transcription of hepci-
responsive elements (IREs) in regulated mRNAs.31 In humans, din. Stat3 signaling pathway is important not only under condi-
low hepcidin levels cause iron overload (IO). As a result of low hep- tions of inflammation but also under expression of baseline
cidin levels and ferroportin overexpression, iron overload is caused hepcidin.43 The activation of IL-6-mediated hepcidin is based
by increased iron export from enterocytes and macrophages. Iron on the signaling pathway IL-6-Stat3.IL-6 binds to the IL-6
builds up in vital organs including the heart, liver, and pancreas, receptor (IL-6R), then to glycoprotein 130 (gp130), causing
causing oxidative stress and conditions like cirrhosis, cancer, dia- Stat3 phosphorylation, which facilitates Stat3 translocation to
betes, and cardiomyopathy.32 As shown in “[ Figure 1]”. the nucleus and triggers Hamp transcriptions.44
4 Plasmatology

Figure 1. (A) Elevated iron levels beta thalassemia major, a result of continuous blood transfusions received from packed red blood cell (PRCB) which around
200-250 mg of iron. A small amount of iron is secreted from the excretion route and through menstrual bleeding in adolescent girls and adult females. (B) Hepcidin, a
major iron regulator which is severely monitored, IL-6 activates hepcidin via the IL-6 receptor (IL-6R) and JAK2-STAT3 signalling pathways. In active BMP-SMAD
pathway enables a normal hepcidin expression which inhibits the expression of iron stores and manages the iron metabolism. Transferrin binds iron and transports it
through the bloodstream. The majority of iron is needed for erythropoiesis. (C) An active BMP-SMAD is required for full hepcidin activation. In beta thalassemia
major, an inactive BMP-SMAD pathway causes decreased production of hepcidin, which is unable to block iron reserves transferred from FPN in macrophages and
Entrocytes, resulting in iron overload which leads oxidative stress and cellular damages to major vital organs such as heart, liver and intestine.

Other signaling pathways that regulate Hamp/Hepcidin signaling of SFKs responsible for the regulation of Hamp
expression expression is still unclear,46 and needs further study.
In Hamp promoter, estrogen was found as a sex hormone to
Hepcidin is regulated by BMP/SMAD protein pathway
down-regulate Hamp expression via an estrogen receptor
element (ERE).45 A report indicated that the membrane com- There are approximately 20 mammalian bone morphogenetic
ponent, PGRMC1 (membrane-bound progesterone proteins (BMPs), among which BMP2, BMP4, BMP5,
receptor-1), also contributes to the modulation of hepcidin by BMP6, BMP7 and BMP9 can cause Hamp expression.47
Src-family tyrosine kinases (SFKs).46 The downstream Studies have shown that the most important among all of the
Yadav and Singh 5

bone morphogenetic proteins (BMPs) is BMP6 which is an resulting in cell death and fibrosis, as well as reduced systolic
important factor in the regulation of hepcidin.47 To activate and diastolic function. Cell damages caused by iron are not
Smad1/5/8 phosphorylation, it attaches to the BMP receptor limited to cases of systemic iron overload. Unfair iron distribu-
(BMPR), and then it translocates to the nucleus together tion within organs or tissues, as well as among cellular compart-
with Smad4 and binds to the Hamp promoter to promote ments, has been shown to impact cell integrity and longevity.
Hamp transcription.48 BMPR type I expresses only Ischemic heart disease or myocardial infarction is not connected
Activin-Like Kinase 2/3mammals.49 Unlike many other with cardiac hemochromatosis,55 thus there is a lack of knowl-
members of the BMP protein family, the BMP signaling edge regarding the basic mechanism of iron overload-induced
pathway is the essential iron-mediated regulator of hepcidin arrhythmia.56
as BMP6 is sensitive to iron concentrations. BMP6 is devel-
oped in non-parenchymal liver cells and stimulated by iron; Iron Excretion
its expression is relative to the amount of hepatic iron. The amount of iron in the human body is calculated through
Furthermore, neutralizing the BMP6 antibody may reduce dietary consumption of iron, not through iron excretion. The
hepcidin expression and cause serum iron concentrations to removal of iron from the human body is handled at a very
increase in mice. small rate. The faecal secretion of iron is equivalent to the con-
sumption of iron by diet, and zero in the case of iron by trans-
Cardiac Dysfunction and its Relation with Iron fusion.57 However, several cases such as blood transfusion in
Toxicity beta-thalassemia which suppress in production of hepcidin
Iron is a two-pronged element, its role as an oxygen carrier plays may lead to an overload of iron. Unintentional exposures can
a critical role in catalyzing enzyme reactions, and disruption in cause the majority of serious medical problems, such as
iron homeostasis results in excessive iron intake and storage, cardiac arrhythmia and liver failure. As a result, iron absorption
which is harmful to various tissues and organs. Toxic levels of is continuously monitored to prevent cell damage. The toxifica-
ferrous iron are primarily exhibited as cardiac dysfunction and tion of iron mainly occurs where it is stored i.e, in the liver.
failure, as well as liver dysfunction and cirrhosis and diabetes Biliary iron excretion estimated a significant amount of excess
mellitus in the endocrine system. These conditions are generally iron extraction through the bile and remaining to be processed
detected in the late stages of their recurrence when conse- for the reuse of hemoglobin synthesis.58 Urinary iron excretion
quences have already been developed. Arrhythmias and increas- a usual routine excretion of iron in the urine is a large propor-
ing systolic dysfunction are commonly caused by iron tion of the overall daily iron depletion.59
accumulation in the heart muscle tissue. A dilated cardiomyop-
athy with poor left ventricular ejection fraction (LVEF) is the Therapeutic Targets in Beta-Thalassemia
most common manifestation, which can be reversed only if Mini-hepcidin
recognized and treated in the early stages of diagnosis.50 This
condition is often asymptomatic. Iron deposition can occur Since low hepcidin levels are associated with greater Fe absorp-
throughout the cardiac conduction system, particularly in the tion in the intestine; inhibitors of hepcidin in thalassemia
atrioventricular node. Severe atrioventricular block due to iron patients may boost the iron load. Small peptides such as
accumulation.13 Non-homogeneous electrical conduction and Mini-hepcidin mimetics are necessary to induce hepcidin
repolarization induce cardiomyopathy with atrial and ventricu- actions; hence, serum iron levels decrease and iron overload
lar tachyarrhythmia and iron buildup in heart muscle.12 improves. By using this mini-hepcidin, iron overload and
Tf-bound iron is not a limiting factor under normal settings, damage to the erythroid cells are significantly reduced.60
and the absorption of iron into cells is regulated by TfR1
expression.51 As a result, iron overload occurs only when Tf’s Apo-transferrin administration
binding ability is exhausted and non-transferrin-bound iron Apo-transferrin administration can decrease labile plasma iron
(NTBI) or labile plasma iron (LPI), the portion of NTBI concentrations, leading to a normalization of RBC survival and
that may infiltrate cells and is chelatable, is present.52 increased production of Hb. The protein also exists during the
Although NTBI is linked to tissue siderosis, the exact mechan- early phases of a clinical study.61 As the Hamp negative regula-
ism by which it enters the cell is unknown, owing to a lack of tor, 17b estradiol (E2) therapy reduced Hamp expression in cell
complete characterization of the iron species involved, which lines HuH7 and Hep G2, which could be blocked by ICI
have a variable composition under different pathological condi- 182780, an estrogen receptor antagonist.45
tions and change their association with macromolecules. Several
plasma components, including citrate, phosphates and proteins,
JAK2 inhibitors
are said to be linked to NTBI.53 Iron toxicity in cells is caused
by its ability to catalyze the reactive oxygen species (ROS),54 These agents have substantial antiproliferative and anti-
which induce lipid peroxidation and organelle damage, inflammatory activity in patients with b-thalassemia, to regulate
6 Plasmatology

splenomegaly and extra-modular hematopoiesis.62 Through regions could mediate β-globin gene silence. Whether this is
suppressing the EPO’s key signaling pathway on progenitor a direct result of the suppression of BCL11A or an indirect
erythrocytes, similar agents can have therapeutic effects on effect due to the activation of γ-globin by another process is
splenomegaly and secreted blood from the spleen, resulting in uncertain.
fewer transfusion needs. These agents can be used in place of
splenectomy, once proven to be safe and to decrease additional Therapies Under Investigation for Iron Overload
medullary hematopoiesis. Hepcidin deficiency treatment
Activin receptor Ii ligand traps Hepcidin, the hormone which regulates the accumulation of
absorption of iron and its abnormal transportation are a cause
These model ligands (Luspatercept/Sotatercept) suppress over of iron overload in nearly every form of hereditary hemo-
activated SMAD signals in erythroid precursors which inhibit chromatosis and non-transfused iron overloading anemia.69
the effect of GDF-11 cytokine on differentiation and matur- Analogues of hepcidin have also been seen reducing the toxicity
ation of the advanced stage erythrocytes. These ligands have of the iron-mediated tissue in mouse models.69 Agonists of
been shown to mitigate complications from IE (ineffective hepcidin known as Mini-hepcidin are based on peptides that
erythropoiesis) diseases, minimizing the excess iron and redu- are rationally planned, based on the hepcidin field engaging
cing bone disease deformities in animal models.63 Ongoing with the ferroportin.60 Mini-hepcidin may be helpful in iron
clinical trials are being carried out which show improvements excessive conditions used for treatment or chelation treat-
in 12 weeks of studies on the impact of these drugs on IE ment.70 Analogues of normal hepcidin and mini-hepcidin are
and transfusion-dependent beta-thalassemia patients.64 studied for preventing the overloading of iron in hemochroma-
tosis and beta-thalassemia. As deficiency in hepcidin causes an
Transferrin overload of iron It would be expected that agents capable of
The main protein in the transportation of iron is transferrin, mimicking hepcidin action or potentiating its endogenous pro-
which binds to Fe3 + molecules, transported iron is bound to duction would inhibit iron.71
(TfR1) receptor 1 and (TfR2) receptor 2. Transferrin is endo-
cytosed, the lysosomal, acid framework Fe3 + is produced from Gene therapy management
transferrin and is reduced to Fe2 + and is then entered into the
cytosol through divalent metallic transporter 1. Whereas TfR1 The treatment by gene therapy of genetic conditions such as
with high transferrin saturation is down-regulated.TfR2 is spe- sickle cell anemia and beta-thalassemia will prevent blood
cific in the liver and intestine, because of high liver iron concen- transfusions and reduce iron overload in the tissues.72 In indi-
tration (LIC), TfR2 lacks an iron-responsive feature and the viduals with hereditary hemochromatosis and beta-thalassemia
iron charge in the liver continues. In most tissues, including including the DMT-1 activation and gene expression of ferro-
the erythroid, a liver and myocardial precursor, TfR1 is portin in enterocytes Over-expression of the wild-type HFE
expressed.65 gene in enterocytes and overexpression of the iron regulatory
peptide hepcidin in the liver are other therapeutic strategies
that could be studied.73 The HFE genotype may influence
TMPRSS6
the survival of myelodysplastic syndrome patients and tests
TMPRSS6 is a membrane binding serine protease in hepato- need to be carried out if these patients are to be treated with
cytes, it is also known as matriptase-2,66 which modulates the effective iron chelation therapy.41
production of hepcidin negatively.67 TMPRSS6 cleaves HJV
(hemojuvelin) from plasma membrane favourable in vitro con-
Discussion
ditions.67 This means that TMPPRSS6 down-regulates the
In the Indian subcontinent, the common genetic disorder is
critical BMP/SMAD signalling route important for iron-
Thalassemia. Hyper-transfusion has improved the expected
dependent hepcidin transcription regulations.
lifespan of thalassemic patients over the decades, but the exces-
sive number of blood transfusions ensures iron overload is an
BCL11A (B cell lymphoma-leukaemia 11a)
inevitable complication major in thalassemia patients.74
A decent approach for gene editing strategies is the BCL11a Several studies have concluded that cirrhosis of the liver is asso-
(TF controlling the transition from HbF to HbA). It is postu- ciated with increased levels of serum ferritin.75 Iron is an essen-
lated that the production of HbF in thalassemia patients can be tial component in biochemical and biological processes,
triggered by suppressing the BCL11a. Deletions in the however, when excess, oxidative stress can lead to tissue
BCL11a erythroid enhancer are a promising approach, which damage. Excess iron in the body can cause damage to organs
is being investigated.68 The Long-dimensional interactions such as the liver, spleen, liver, bone marrow, pancreas, pituitary
across the β-globin gene locus between BCL11A and several gland and nervous system.
Yadav and Singh 7

In the last 20 years, thalassemia major management has devel- found in intestine duodenal cells, macrophages, and placental
oped to the point where the life expectancy of patients is as high as cells. The absence of ferroportin on the cell membrane prevents
normal inhabitants. Therapies that reduce transfusion demands in iron from entering the bloodstream. Low transferrin saturation
TDT and remove insufficient erythropoiesis in NTDT may in results from less iron entrance into plasma and less iron is supplied
this way be promising. Specific disruptions in factors like to the erythroblast. Higher cell surface ferroportin and increased
BCL11A that may potentially lead to γ-globin genes being kept iron absorption resulted from decreased hepcidin expression. By
suppressed may result in simpler and safer treatment of this hepcidin and its interacting constituents (ferroportin, transfer-
β-thalassemia, depending on transfusions or non-transfusions.7 ring) became a potent target for tapping the iron overload
The developmental stage-specific BCL11A repressor regulates problem. These are discussed briefly in the review article.
the expression of hemoglobin. The use of strengthened activin Clinicians utilize the classic iron chelation approach (use of
receptors to enhance delay erythropoiesis by functioning as drugs ie, Deferoxamine, Deferiprone and Deferasirox) to treat
ligand traps for the participants in the transformation of superfam- iron overload in patients who get regular blood transfusions, even
ily growth factors is also a promising method, currently being though this method has a significant risk of side effects in later
tested in clinical studies.65 Therapy with these agents aims to life of patients. There is no insightful topic in agreement on the
increase hemoglobin levels and reduce the need for transfusion excretion of excess iron; nevertheless, if the study focuses on
in both NTDT and TDT patients. excess iron excretion from the body system, individuals with thal-
assemia who require regular blood transfusions may be cured and
Conclusion lead a longer and healthier life. The use of computational biology
In thalassemia major, cardiac illness is still the leading cause of and drug design methods is important in the quest for a powerful
death. Transfusion and iron chelation treatments have greatly inhibitor of iron overload, and the results can be confirmed in vivo
improved survival and reduced morbidity in thalassemic facility. An alternative to iron chelation that has fewer adverse
patients. In the past patients died by the age of 16, while now effects on the human body can be designed. New emerging tech-
80 per cent live to be at least 40 years of age. This is of a nologies (Drug Designing and Lead Identification) could help find
kind improvement, as no other previously fatal genetic flaw a better-designed inhibitor in iron overload.
has shown such a benefit. Heart complications, on the other
hand, continue to be a substantial cause of morbidity and mor- Acknowledgements
tality in transfusion-dependent thalassemia (TM) patients. Author(s) would like to acknowledge the Department of
Some tests can detect the possibility of thalassemia in neonates; Bioinformatics, Central University of South Bihar, Gaya
however, depending on the severity of the thalassemia, blood Bihar and the Indian Council of Medical Research, New
transfusions may be necessary at a very early stage of life. In Delhi India for all technical and financial support.
the human body, iron is essential for the creation of heme
enzymes and other iron-containing enzymes involved in elec- Author Contributions
tron transfer and oxidation reductions, as well as the synthesis The authors have contributed equally to the manuscript.
of oxygen transport proteins such as haemoglobin and myoglo-
bin. The majority of iron is delivered via haemoglobin, which ORCID iD
circulates in erythrocytes. However, 15% of iron is found in Ajay Kumar Singh https://orcid.org/0000-0003-4544-3596
myoglobin, which is found in muscle tissue and a variety of
other tissues. Transferrin binds to iron and transports it REFERENCES
throughout the body, where it is stored in ferritin molecules. 1. Marengo-Rowe AJ. The thalassemias and related disorders. Baylor Univ Med Cent
There is no physiologic mechanism for excreting excess iron Proc. 2007;20(1)(Jan):27-31.
2. Galanello R, Origa R. Beta-thalassemia. Orphanet J Rare Dis. 2010;5(Aug):1-15.
from the body once it has been absorbed, other than blood 3. Thein SL. Molecular basis of β thalassemia and potential therapeutic targets. Blood
loss, such as during pregnancy, menstruation, or other periods Cells, Mol Dis. 2018;70(May):54-65.
4. Farashi S, Harteveld CL. Molecular basis of α-thalassemia. Blood Cells, Molecules,
of bleeding. As a result, unabsorbed iron can lead to liver and Diseases. 2018;70(May):43-53.
disease, cardiac troubles, diabetes, and problems with human 5. Muncie HL, Campbell JS. Alpha and beta thalassemia. Am Fam Physician.
2009(Aug):339–344.
growth. The percentage of iron absorbed from the total 6. Musallam KM, Rivella S, Vichinsky E, Rachmilewitz EA. Non-transfusion-dependent
amount consumed is normally low, ranging from 5 to 35 per thalassemias. Haematologica. 2013;98(6)(Jun):833-844. doi: 10.3324/haematol.2012.
cent based on the circumstances and type of iron consumed. 066845
7. Mishra AK, Tiwari A. Iron overload in Beta thalassaemia major and intermedia
Hepcidin is a peptide hormone released by the liver that plays patients. Maedica (Buchar). 2013;8(4)(Sep):328-332.
an important function in iron homeostasis management. It is a 8. Aronow WS. Management of cardiac hemochromatosis. Arch Med Sci.
2018;14(Apr):560-568. doi: 10.5114/aoms.2017.68729.
potent inhibitor of systemic iron homeostasis, coordinating iron 9. Domellöf M, Dewey KG, Lönnerdal B, Cohen RJ, Hernell O. The diagnostic criteria
uptake, usage, and storage. Hepatocytes produce it, and it’s a for iron deficiency in infants should be reevaluated. J Nutr. 2002;132(Dec):3680-3686.
doi: 10.1093/jn/132.12.3680.
negative regulator of iron absorption into the bloodstream. 10. Gibson RS. Principles of Nutritional Assessment. 2nd. Editor Gibson RS ed. Oct.
Hepcidin specifically binds to ferroportin, an iron transporter Oxford University Press Inc.; 2005.
8 Plasmatology

11. Taher AT, Musallam KM, Wood JC, Cappellini MD. Magnetic resonance evalu- 39. Alvi N, et al. Burden of cardiac siderosis in a thalassemia-Major endemic population:
ation of hepatic and myocardial iron deposition in transfusion-independent thalas- a preliminary report from Pakistan. J Pediatr Hematol Oncol. 2016;38(Jul):378-383.
semia intermedia compared to regularly transfused thalassemia major patients. Am doi: 10.1097/MPH.0000000000000574
J Hematol. 2010;85(Apr):288-290. doi: 10.1002/ajh.21626. 40. Musallam KM, Cappellini MD, Taher AT. Iron overload in beta-thalassemia inter-
12. Wu V-C, Huang J W, Wu M-S, et al. The effect of iron stores on corrected QT media: an emerging concern. Curr Opin Hematol. 2013;20(May):187-192. doi: 10.
dispersion in patients undergoing peritoneal dialysis. Am J Kidney Dis Off J Natl 1097/MOH.0b013e32835f5a5c.
Kidney Found. 2004;44(Oct):720-728. 41. Lucijanic M, et al. Hemochromatosis gene mutations may affect the survival of
13. Aronow WS, Meister L, Kent JR. Atrioventricular block in familial hemochromatosis patients with myelodysplastic syndrome. Hematology. 2016;21(Apr):170-174. doi:
treated by permanent synchronous pacemaker. Arch Intern Med. 1969;123(Apr):433- 10.1080/10245332.2015.1101964
435. 42. Pigeon C, et al. A new mouse liver-specific gene, encoding a protein homologous to
14. Vichinsky EP. Changing patterns of thalassemia worldwide. Ann N Y Acad Sci. human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol
2005;1054:18-24. doi: 10.1196/annals.1345.003. Chem. 2001;276(Mar):7811-7819. doi: 10.1074/jbc.M008923200
15. Jalil T, Yousafzai Y M, Rashid I, et al. Mutational analysis of Beta thalassaemia by 43. Verga Falzacappa MV, et al. STAT3 Mediates hepatic hepcidin expression and its
Multiplex Arms-Pcr in Khyber Pakhtunkhwa, Pakistan. J Ayub Med Coll Abbottabad. inflammatory stimulation. Blood. 2007;109(Jan):353-358. doi: 10.1182/blood-2006-
2019;31(Jan-Ma):98-103. 07-033969
16. Brancaleoni V, Di Pierro E, Motta I, Cappellini MD. Laboratory diagnosis of thal- 44. Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 signalling in
assemia. Int J Lab Hematol. 2016;38(May):32-40. doi: 10.1111/ijlh.12527. cancer: new and unexpected biological functions. Nat Rev Cancer.
17. Sangkhae V, Nemeth E. Regulation of the iron homeostatic hormone Hepcidin. Adv 2014;14(Nov):736-746. doi: 10.1038/nrc3818.
Nutr. 2017;8(Jan):126-136. doi: 10.3945/an.116.013961 45. Yang Q, Jian J, Katz S, Abramson SB, Huang X. 17beta-Estradiol Inhibits iron
18. Pagani A, Nai A, Silvestri L, Camaschella C. Hepcidin and Anemia: a tight relation- hormone hepcidin through an estrogen responsive element half-site. Endocrinology.
ship. Front Physiol. 2019;10(Oct):1294. doi.org/10.3389/fphys.2019.01294 2012;153(Jul):3170-3178. doi: 10.1210/en.2011-2045
19. Mobarra N, Shanaki M, Ehteram H, et al. A review on iron chelators in treatment of 46. Li X, et al. Progesterone receptor membrane component-1 regulates hepcidin bio-
iron overload syndromes. Int J Hematol Stem Cell Res. 2016;10(Oct):239-247. synthesis. J Clin Invest. 2016;126(Jan):389-401. doi: 10.1172/JCI83831
20. Brittenham GM, Griffith P M, Nienhuis A W, et al. Efficacy of deferoxamine in 47. Andriopoulos BJ, et al. BMP6 Is a key endogenous regulator of hepcidin expression
preventing complications of iron overload in patients with thalassemia major. and iron metabolism. Nat Genet. 2009;41(Apr):482-487. doi: 10.1038/ng.335
N Engl J Med. 1994;331(Sep):567-573. doi: 10.1056/NEJM199409013310902. 48. Wang R-H, et al. A role of SMAD4 in iron metabolism through the positive regu-
21. Xiao W, Beibei F, Guangsi S, et al. Iron overload increases osteoclastogenesis and aggra- lation of hepcidin expression. Cell Metab. 2005;2(Dec):399-409. doi: 10.1016/j.
vates the effects of ovariectomy on bone mass. J Endocrinol. 2015;226(Sep):121-134. doi: cmet.2005.10.010
10.1530/JOE-14-0657 49. Core AB, Canali S, Babitt JL. Hemojuvelin and bone morphogenetic protein
22. Nisbet-Brown E, Olivieri N F, Giardina P J, et al. Effectiveness and safety of (BMP) signaling in iron homeostasis. Front Pharmacol. 2014;5(MAY):1-9.
ICL670 in iron-loaded patients with thalassaemia: a randomised, double-blind, 50. Sudmantait V, Celutkien J, Glaveckaite S, Katkus R. Difficult diagnosis of cardiac
placebo-controlled, dose-escalation trial. Lancet (London, England). haemochromatosis: a case report.. Eur Heart J Case Rep. 2020;4(Jan):1–6. doi: 10.
2003;361(May):1597-1602. doi: 10.1016/S0140-6736(03)13309-0 1093/ehjcr/ytaa012
23. Nick H, Acklin P, Lattmann R, et al. Development of tridentate iron chelators: from 51. Frazer DM, Anderson GJ. The regulation of iron transport. Biofactors. 2014;40(Mar-
desferrithiocin to ICL670. Curr Med Chem. 2003;10(May):1065-1076. doi: 10. Apr):206-214. doi: 10.1002/biof.1148.
1016/S0140-6736(03)13309-0. 52. Cabantchik ZI. Labile iron in cells and body fluids: physiology, pathology, and pharma-
24. Taher AT, Porter J, Viprakasit V, et al. Deferasirox reduces iron overload signifi- cology. Front Pharmacol. 2014;5(MAR):1-11. doi: 10.3389/fphar.2014.00045
cantly in nontransfusion-dependent thalassemia: 1-year results from a prospective, 53. Patel M, Ramavataram DVSS. Non transferrin bound iron: nature, manifestations and
randomized, double-blind, placebo-controlled study. Blood. 2012;120(Aug):970- analytical approaches for estimation. Indian J Clin Biochem. 2012;27(Oct):322-332. doi:
977. doi: 10.1182/blood-2012-02-412692. 10.1007/s12291-012-0250-7
25. Keberle H. The biochemistry of desferrioxamine and its relation to iron metabolism. 54. Brissot P, Ropert M, Le Lan C, Loréal O. Non-transferrin bound iron: a key role in
Ann N Y Acad Sci. 1964;119(Oct):758-768. doi: 10.1111/j.1749-6632.1965.tb54077.x. iron overload and iron toxicity. Biochim Biophys Acta. 2012;1820(Mar):403-410.
26. Pippard MJ, Callender ST, Weatherall DJ. Intensive iron chelation therapy with 55. Candore G, et al. Association between HFE mutations and acute myocardial infarc-
desferrioxamine in iron loading anaemias. Clin Sci Mol Med. 1978;54(Jan):99-106. tion: a study in patients from northern and southern Italy. Blood Cells Mol Dis.
doi: 10.1042/cs0540099. 2003;31(Jul-Aug):57-62. doi: 10.1016/s1079-9796(03)00065-2.
27. Kontoghiorghes GJ, Aldouri M A, Hoffbrand A V, et al. Effective chelation of iron 56. Shizukuda Y, Rosing DR. Iron overload and arrhythmias: influence of confounding
in beta thalassaemia with the oral chelator 1,2-dimethyl-3-hydroxypyrid-4-one. Br factors. J Arrhythmia. 2019;35(Aug):575-583. doi: 10.1002/joa3.12208
Med J (Clin Res Ed). 1987;295(Dec):1509-1512. doi: 10.1136/bmj.295.6612.1509 57. Mercadante CJ, et al. Gastrointestinal iron excretion and reversal of iron excess in a
28. Taher AT, Saliba AN. Iron overload in thalassemia: different organs at different rates. mouse model of inherited iron excess. Haematologica. 2019;104(Apr):678-689. doi:
Hematology. 2017;2017(Dec):265-271. doi: 10.1182/asheducation-2017.1.265 10.3324/haematol.2018.198382
29. Kellenberger CJ, et al. Radiographic and MRI features of deferiprone-related arthropathy 58. Hultcrantz R, Angelin B, Björn-Rasmussen E, Ewerth S, Einarsson K. Biliary excretion
of the knees in patients with β-thalassemia. Am J Roentgenol. 2004;183(OCT):989-994. of iron and ferritin in idiopathic hemochromatosis. Gastroenterology. 1989;96(Jun):1539-
30. Liberal Â, Pinela J, Vívar-Quintana AM, Ferreira ICFR, Barros L. Fighting iron- 1545. doi: 10.1016/0016-5085(89)90524-6.
deficiency anemia: innovations in food fortificants and biofortification strategies. 59. Wahidiyat PA, et al. Urinary iron excretion for evaluating iron chelation efficacy in
Foods. 2020;9(Dec):1-19. doi: 10.3390/foods9121871 children with thalassemia major. Blood Cells Mol Dis. 2019;77(Jul):67-71. doi: 10.
31. Muckenthaler MU, Galy B, Hentze MW. Systemic iron homeostasis and the iron- 1016/j.bcmd.2019.03.007.
responsive element/iron-regulatory protein (IRE/IRP) regulatory network. Annu 60. Preza GC, et al. Minihepcidins are rationally designed small peptides that mimic
Rev Nutr. 2008;28(Aug):197-213. doi: 10.1146/annurev.nutr.28.061807.155521 hepcidin activity in mice and may be useful for the treatment of iron overload.
32. Chifman J, Laubenbacher R, Torti SV. A systems biology approach to iron metab- J Clin Invest. 2011;121(Dec):4880-4888. doi: 10.1172/JCI57693.
olism. Adv Exp Med Biol. 2014;844:201-225. 61. Calzolari A, et al. Transferrin receptor 2 is frequently and highly expressed in glio-
33. Gulec S, Anderson GJ, Collins JF. Mechanistic and regulatory aspects of intestinal blastomas. Transl Oncol. 2010;3(Apr):123-134. doi: 10.1593/tlo.09274
iron absorption. Am J Physiol. Gastrointest. Liver Physiol. 2014;307(Aug):G397- 62. Libani IV, et al. Decreased differentiation of erythroid cells exacerbates ineffective erythropoi-
G409. doi: 10.1152/ajpgi.00348.2013. esis in β-thalassemia. Blood. 2008;112(Aug):875-885. doi: 10.1182/blood-2007-12-126938
34. Ginzburg Y, Rivella S. β-thalassemia: a model for elucidating the dynamic regulation 63. Dussiot M, et al. An activin receptor IIA ligand trap corrects ineffective erythropoi-
of ineffective erythropoiesis and iron metabolism. Blood. 2011;118(Oct):4321-4330. esis in β-thalassemia. Nat Med. 2014;20(Apr):398-407. doi: 10.1038/nm.3468
doi: 10.1182/blood-2011-03-283614 64. Piga A, et al. ACE-536 Increases hemoglobin and decreases transfusion burden and
35. Lymboussaki A, et al. The role of the iron responsive element in the control of fer- Serum ferritin in adults with Beta-thalassemia: preliminary results from a phase 2
roportin1/IREG1/MTP1 gene expression. J Hepatol. 2003;39(Nov):710-715. doi: study. Blood. 2014;124(Dec):53. https://doi.org/10.1182/blood.V124.21.53.53
10.1016/s0168-8278(03)00408-2. 65. Motta I, Scaramellini N, Cappellini MD. Expert opinion on investigational drugs
36. Hershko C. Pathogenesis and management of iron toxicity in thalassemia. Ann N Y investigational drugs in phase I and phase II clinical trials for thalassemia. Expert
Acad Sci. 2010;1202(Aug):1-9. doi: 10.1111/j.1749-6632.2010.05544.x. Opin Investig Drugs. 2017(26;7). DOI: 10.1080/13543784.2017.1335709
37. Henninger B, et al. Evaluation of MR imaging with T1 and T2* mapping for the 66. Wang C-Y, Meynard D, Lin HY. The role of TMPRSS6/matriptase-2 in iron regu-
determination of hepatic iron overload. Eur Radiol. 2012;22(Nov):2478-2486. doi: lation and anemia. Front Pharmacol. 2014;5:114.
10.1007/s00330-012-2506-2 67. Silvestri L, et al. The serine protease matriptase-2 (TMPRSS6) inhibits hepcidin
38. Wood JC. Magnetic resonance imaging measurement of iron overload. Curr Opin activation by cleaving membrane hemojuvelin. Cell Metab. 2008;8(Dec):502-511.
Hematol. 2007;14(May):183-190. doi: 10.1097/MOH.0b013e3280d2b76b doi: 10.1016/j.cmet.2008.09.012.
Yadav and Singh 9

68. Bauer DE, et al. A erythroid enhancer of BCL11A subject t genetic variation. 72. Payen E, Leboulch P. Advances in stem cell transplantation and gene therapy in the beta-
Science. 2014;342(Oct 11):253-257. doi: 10.1126/science.1242088 hemoglobinopathies. Hematol Am Soc Hematol Educ Progr. 2012;2012(Dec):276-283. doi:
69. Viatte L, et al. Chronic hepcidin induction causes hyposideremia and alters the pattern 10.1182/asheducation-2012.1.276.
of cellular iron accumulation in hemochromatotic mice. Blood. 2006;107(Apr):2952- 73. Ezquer F, Nunez MT, Rojas A, Asenjo J, Israel Y. Hereditary hemochromatosis: an
2958. doi: 10.1182/blood-2005-10-4071 opportunity for gene therapy. Biol Res. 2006;39(Jun):113-124. doi: 10.4067/s0716-
70. Ramos E, et al. Minihepcidins prevent iron overload in a hepcidin-deficient mouse 97602006000100014.
model of severe hemochromatosis. Blood. 2012;120(Nov):3829-3836. doi: 10.1182/ 74. Riaz H, et al. Serum ferritin levels, socio-demographic factors and desferrioxamine therapy
blood-2012-07-440743. in multi-transfused thalassemia major patients at a government tertiary care hospital of
71. Xiao JJ, et al. Pharmacokinetics of anti-hepcidin monoclonal antibody Ab 12B9 m Karachi, Pakistan. BMC Res Notes. 2011;4(Aug):287. doi: 10.1186/1756-0500-4-287
and hepcidin in cynomolgus monkeys. AAPS J. 2010;12(Dec):646-657. doi: 10. 75. Knovich MA, Storey JA, Coffman LG, Torti SV, Torti FM. Ferritin for the clin-
1208/s12248-010-9222-0 ician. Blood Rev. 2009;23(May):95-104. doi: 10.1016/j.blre.2008.08.001

You might also like