Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Review Article

Hepatocellular Carcinoma as an Emerging Morbidity in the


Thalassemia Syndromes: A Comprehensive Review
Hassan M. Moukhadder, MD1; Racha Halawi, MD, MS2; Maria Domenica Cappellini, MD3,4; and
Ali T. Taher, MD, PhD, FRCP1

The incidence of hepatocellular carcinoma (HCC) in patients with thalassemia is on the rise. The 2 well recognized HCC risk factors in
thalassemia are iron overload and chronic viral infection with hepatitis C. The carcinogenicity of iron is related to its induction of oxi-
dative damage, which results in genotoxicity, and to immunologic dysregulation, which attenuates cancer immune surveillance. Chron-
ic hepatitis B and C infections lead to necroinflammation, which can prompt progression to HCC, but an independent role of hepatitis
B virus in hepatic carcinogenesis among patients with thalassemia has not been demonstrated. Screening patients who have thalasse-
mia using magnetic resonance imaging-based liver iron concentration measurement and liver ultrasound is recommended for early
detection of iron overload and HCC, respectively. Prevention primarily resides in hepatitis B vaccination, donor blood screening, hepa-
titis treatment, and iron chelation. Although solid data is lacking on the outcomes of HCC treatment in patients with thalassemia, a
personalized approach tailored to the individual patient’s comorbidities remains necessary for treatment success. Treatment modali-
ties for HCC include surgical resection, chemoembolization, and liver transplantation, among others. Multicenter studies are needed
to better explore therapeutic targets that can improve the prognosis of these patients. Cancer 2017;123:751-8. V C 2016 American

Cancer Society.

KEYWORDS: hepatitis B virus, hepatitis C virus, hepatocellular carcinoma, iron overload, thalassemia.

INTRODUCTION
The Thalassemia Syndromes
Thalassemia is a genetically transmitted, quantitative disorder of the hemoglobin.1 It is divided into 2 main categories,
a-thalassemia and b-thalassemia, in which the defect resides in a-chain and b-chain synthesis, respectively. Thalassemia is
a spectrum of syndromes classified according to clinical severity into asymptomatic carriers (a-thalassemia or
b-thalassemia minor), nontransfusion-dependent thalassemias (NTDTs) (which include b-thalassemia intermedia [TI]),
and transfusion-dependent thalassemias (TDTs) (which include b-thalassemia major [TM]).2

Hepatocellular Carcinoma in Thalassemia


Hepatocellular carcinoma (HCC) was not a common complication of thalassemia in the past, likely because patients with
thalassemia used to die at a young age secondary to anemia and heart failure.3 Better outcomes, particularly prevention of
heart disease after the advent of highly effective iron-chelating medicines, have greatly prolonged the survival of patients
with thalassemia, and patients currently live long enough to develop adverse complications like HCC.3,4 Although both
hepatitis B virus (HBV) and hepatitis C virus (HCV) infections are established risk factors for the development of HCC
in the general population,5 the direct role of HBV in hepatic carcinogenesis in thalassemia is not well defined. In addition
to chronic hepatitis C, the other well recognized HCC risk factor in thalassemia is iron overload,6,7 which is the result of
chronic transfusional iron accumulation in patients with TDT as opposed to increased absorption of iron from the intes-
tine and release of recycled iron from the reticuloendothelial system in patients with NTDT.8 In this review, we describe

Corresponding author: Ali T. Taher, MD, PhD, FRCP, Department of Internal Medicine, American University of Beirut Medical Center, Cairo Street, PO Box 11-
0236, Riad El Solh 1107 2020, Beirut, Lebanon; Fax: (011) 961-1-370814; ataher@aub.edu.lb
1
Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon; 2Department of Internal
Medicine, Emory University School of Medicine, Atlanta, Georgia; 3Department of Internal Medicine, Foundation of the Scientific Institute for Research, Hospitali-
zation, and Health Care (IRCCS), Ca’ Granda Ospedale Maggiore Polyclinic, Milan, Italy; 4Department of Clinical Sciences and Community Health, University of Mi-
lan, Milan, Italy

The first 2 authors contributed equally to this work.

DOI: 10.1002/cncr.30462, Received: September 16, 2016; Revised: October 14, 2016; Accepted: October 17, 2016, Published online December 1, 2016 in Wiley
Online Library (wileyonlinelibrary.com)

Cancer March 1, 2017 751


Review Article

existing knowledge on the epidemiology and pathophysi- also promote malignant transformation in the liver
ology of HCC in thalassemia, and we outline the most re- through the acceleration of fibrosis to cirrhosis by activa-
cent recommended approaches to screening, prevention, tion of stellate cells and through the profibrogenic effects
and management of this emerging morbidity. of lipid peroxidation.7 Conversely, recent data supports
the finding that free iron is independently hepatocarcino-
INCIDENCE genic, ie, in the absence of cirrhosis. For example, in a re-
The incidence of HCC in patients with thalassemia has cently described animal model, dietary iron overload was
been increasing with time. For example, the number of associated with iron-free preneoplastic nodules and HCC
cases diagnosed in Italy steadily increased from 8 to 31 be- in the absence of cirrhosis, which may be caused by iron-
tween 1993 to 1997 and 2008 to 2012.9 A prospective mediated oxidative stress, as detailed above.19 A recent
study of 105 adults with TM identified a 2% incidence of randomized trial demonstrated a decrease in the risk of
HCC during a 1-year observation period, which is almost new visceral malignancy in a group of individuals who did
the same as the risk of HCC in the general population.10 not have thalassemia or any other iron-loading disorder
It is noteworthy that the younger age at HCC diagnosis in but who underwent iron reduction compared to a control
patients with thalassemia suggests the presence of risk fac- group (hazard ratio 5 0.65).20
tors unique to this population.11 In the general popula- It also has been suggested that iron overload induces
tion, the median age at HCC onset in developed countries immunologic aberrancies, which may contribute to cancer
is approximately 70 years, with a 2.4:1 male-to-female formation. Growing evidence highlights the negative ef-
ratio12,13; on the other hand, most patients with thalasse- fect of iron and ferritin on the tumoricidal function of
mia develop HCC at age <50 years, and there is no macrophages in mice,21 in which it was specifically dem-
discernible difference in incidence between men and onstrated that iron overload decreased antibody-mediated
women in this group of patients.9,10,14,15 It is worth men- and mitogen-stimulated phagocytosis by monocytes and
tioning that HCC seems to be more common in patients macrophages.22 Moreover, lymphocyte proliferation is
with TI than TM, with a male-to-female ratio ranging inhibited independently by both nontransferrin-bound
from 2:1 to 9.5:1.14-16 One possible explanation is that iron and ferritin.19,23 Figure 1 summarizes the mecha-
patients who have TI usually have improved survival com- nisms by which iron overload is believed to contribute to
pared with those who have TM, which enables them to the pathogenesis of HCC in patients with thalassemia.
live long enough to develop HCC.14 Several reports in the literature have suggested the
hepatocarcinogenic role of iron in patients with thalasse-
RISK FACTORS mia. In fact, a number of case reports and case series have
Iron Overload described the development of HCC in patients with thal-
Iron balance depends on intake and losses, and iron over- assemia who were negative for hepatitis B and C testing
load is generally the result of an overly positive iron bal- but had significant hepatic iron overload.15,24,25 An
ance. Hepcidin, a key regulator of iron balance, functions updated survey on the prevalence of HCC in Italian
by decreasing iron absorption from the gut and release patients with thalassemia highlighted the observation that
from the reticuloendothelial system.17 In states of abnor- 4 of the 62 reported patients had no evidence of exposure
mally low hepcidin levels, such as NTDT, excess iron is to either HCV or HBV.9 All 4 patients had TI, and, in 3
absorbed into the system and released into the circulation, of them, severe siderosis was reported in the biopsied liver
causing depletion of macrophage iron and preferential tissue.
portal and hepatocyte iron loading.18 The ultimate result
is an increase in free or nontransferrin-bound iron in the HBV and HCV
blood, leading to end-organ damage, such as cardiomyop- Another risk factor for HCC in patients with thalassemia
athy, endocrinopathies, cirrhosis, and even cancer.9 The is chronic viral infection, namely, with HCV or HBV, re-
first mechanism by which free iron is believed to trigger lated to blood transfusion exposure. This applies mostly
malignant transformation is the generation of reactive ox- to patients in their 30s or older because the risk of viral
ygen species (ROS), which causes peroxidation of mem- transmission through blood transfusion was greatly re-
brane fatty acids and subsequent formation of toxic duced after the 1980s through blood donor screening.
byproducts that impair protein synthesis and disrupt The prevalence of anti-HCV antibodies in patients with
DNA, leading to mutations in tumor suppressor genes thalassemia ranges from 4.4% to 87%, with the highest
(such as p53) and DNA repair genes.19 Iron overload may rates being reported from Italy.9,26-28 In an Italian study,9

752 Cancer March 1, 2017


Hepatocellular Carcinoma in Thalassemia/Moukhadder et al

Figure 1. The pathophysiology of iron-mediated toxicity in hepatocellular carcinoma in thalassemia is illustrated. NTBI indicates
nontransferrin-bound iron; ROS, reactive oxygen species.

87% and 69% of patients were positive for HCV anti- several reports in the literature. In a recent report on
bodies and HCV-RNA, respectively. Data from the previ- HCC incidence in a Greek thalassemia unit, HCC devel-
ous decade about the prevalence of HCC among multi- oped in 2 patients with TM who were not iron overloaded
transfused thalassemia patients in Italy and the United but had HCV-related cirrhosis that failed to respond to
Kingdom showed that 70–80% of individuals infected antiviral treatment.16 The results from this study suggest
with HCV will develop chronic hepatitis, approximately that HCV infection is 1 of the main culprits of HCC de-
20% of whom will develop cirrhosis with a risk that is di- velopment in patients with TM. Indeed, transfusion-
rectly related to the age at infection.14,26,29,30 Another related hepatitis C is a key factor in liver damage among
transfusion-transmitted infection is hepatitis B. The on- patients with TM through the increased risk of HCV-
cogenicity of HBV was first noted in the 1970s, when associated cirrhosis.32 However, it was observed that
chronic HBV infection was associated with an increased HCC also developed in a cirrhosis-free liver in an HCV-
incidence of HCC in the general population.31 Infection infected female with TI in the presence of iron overload10;
with HBV is also encountered in patients with thalasse- and, in another account, HCC was witnessed in a noncir-
mia, in whom, for example, 5% of patients reported in rhotic patient with thalassemia in the absence of iron over-
the updated Italian registry were positive for hepatitis B load.33 These observations suggest that HCV infection
surface antigen (HBsAg) and 58% had evidence of past might be an independent HCC risk factor in patients
HBV infection.9 with thalassemia. It is important to mention that chronic
The direct role of HCV infection in progression to hepatitis C and liver iron overload have been proposed to
HCC among patients with thalassemia is suggested by work in synergy to increase the HCC risk, which is

Cancer March 1, 2017 753


Review Article

reflected by the fact that hepatic iron is often increased in the procedure’s inconvenience and potential side effects,
patients who have chronic HCV infection secondary to an in favor of noninvasive LIC quantification with R2 or R2*
HCV-induced decrease in serum hepcidin levels.34,35 Al- magnetic resonance imaging (MRI).9 However, the serum
though chronic HBV infection accounts for approximate- ferritin assay, which is an easy and inexpensive method
ly one-half of all HCC cases worldwide,36 the direct role compared with LIC measurement, remains to be heavily
of chronic hepatitis B in hepatic carcinogenesis is not well relied upon in resource-poor areas where MRI technology
established for patients with thalassemia. To the best of is not available.45 Although this marker is reflective of
our knowledge, there are no reported cases in the litera- iron stores in patients with TDT,46 serum ferritin levels
ture of HCC in HBV-infected patients with thalassemia underestimate the true iron burden in NTDT patients,47
in the absence of HCV serologic markers or significant which can be explained by the finding that hyperabsorbed
iron overload. iron in the latter group is accumulated in hepatocytes,
usually leading to lower serum ferritin levels.47,48 There-
Transfusion-Associated Immunomodulation fore, because LIC and total body iron are linearly related,
Increasing evidence suggests that blood transfusions have yearly LIC assessment using noninvasive MRI methodol-
negative effects on the recipient’s immune system, most ogies is the mainstay for accurately estimating total body
likely as a result of chronic antigenic stimulation by the iron levels in all patients with thalassemia, but it may be
transfused blood.37,38 Some of the reported immune ab- done on a more or less frequent basis, depending on the
normalities related to blood transfusions are impaired extent of iron overload and its control.49
B-lymphocyte differentiation and defective chemotaxis and In addition to screening for iron overload, HCC can
phagocytosis, which are important mechanisms in antiviral be secondarily prevented in thalassemia if high-risk lesions
immunity and anticancer immune surveillance.39-41 Several are detected early by timely radiographic testing. The
studies have demonstrated the contributory effect of trans- American Association for the Study of Liver Diseases rec-
fusions to cancer progression or recurrence, particularly in ommends surveillance of cirrhotic patients in the general
colorectal and lung cancers.42-44 However, to date, the pre- population using biannual abdominal ultrasound, and it
cise role of transfusion-related immunomodulation in the recognizes that a-fetoprotein (AFP) measurement lacks
development of HCC in thalassemia has not been elucidat- the sensitivity and specificity required for optimal HCC
ed, and further studies are required for a better understand- detection.50-52 The inadequate sensitivity of AFP for
ing of this association. HCC screening is also apparent in patients with thalasse-
mia, in whom AFP levels were normal in 44% of patients
MANAGEMENT in an Italian study9 and in 100% of those in another study
The management of HCC in patients with thalassemia by Greek investigators.16
includes screening and prevention plans in addition to The lack of clear-cut HCC screening guidelines for
treatment strategies that target the HCC risk factors. thalassemia might be problematic, especially because the
HCC risk factors in these patients are different from those
Screening in the general population. Various professional organiza-
The emergence of HCC as a pressing morbidity in thalas- tions have identified target populations that might benefit
semia suggests the need for structured HCC screening from HCC surveillance plans, but the risk factors in the
programs. Borgna-Pignatti et al reported nonspecific or thalassemia population are not clearly outlined in those
even completely absent symptoms in 82% of patients who recommendations. On the basis of current evidence, it is
had thalassemia and were diagnosed with HCC, which suggested that high-risk patients with thalassemia should
supports the usefulness of effective screening regimes tar- undergo biannual liver ultrasound for HCC screening,
geting early HCC detection.9 Iron overload, as discussed with high-risk patients identified as patients with HCV
above, is associated with a host of complications in and/or HBV infection, NTDT with LIC 5 mg Fe/g dry
patients with thalassemia, including carcinogenesis, sug- weight (dw), TDT with LIC 7 mg Fe/g dw or serum fer-
gesting that HCC surveillance strategies for patients with ritin 1000 ng/mL, or advanced cirrhosis.3,9,11,53-55
thalassemia should include close monitoring of increased
iron burden which might put them at risk for developing Prevention
HCC. Although liver biopsy was previously used in The development of HCC can be avoided by preventing
patients with thalassemia to estimate liver iron concentra- or treating the HCC risk factors, namely, chronic viral
tion (LIC), this practice has been abandoned, because of hepatitis and iron overload.3 In the general population,

754 Cancer March 1, 2017


Hepatocellular Carcinoma in Thalassemia/Moukhadder et al

TABLE 1. Iron-Chelating Drugs in Transfusion-Dependent and Nontransfusion-Dependent Thalassemia

Iron-Chelating Drug

Variable Deferoxamine Deferiprone Deferasirox

Route of administration Parenteral Oral Oral


Indication
TDT  Ferritin level  Ferritin level  Ferritin level
consistently  1000 ng/mL consistently  1000 ng/mL consistently  1000 ng/mL
 10-15 transfusions  10-15 transfusions  10-15 transfusions
 LIC  7 mg Fe/g dw  LIC  7 mg Fe/g dw  LIC  7 mg Fe/g dw
 Children aged > 6 y and adults
only if other chelators are not
tolerated or ineffective
NTDT Not approved Not approved  LIC  5 mg Fe/g dw
 Ferritin level  800 ng/mL in
patients aged >10 y (or > 15 y in
those with hemoglobin H disease)

Abbreviations: dw, dry weight; LIC, liver iron concentration; NTDT, nontransfusion-dependent thalassemia; TDT, transfusion-dependent thalassemia.

HCC caused by HBV-related cirrhosis can be prevented prospective study demonstrated that LIC did not correlate
with vaccination.9 In addition, HCC caused by with virologic response to antiviral treatment in chronic
transfusion-transmitted hepatitis B or C can be prevented HCV-infected patients who had TM,66 which suggests
by donor blood screening with nucleic acid testing (NAT) against delaying antiviral treatment in patients with thal-
technology, the utility of which in developed countries is assemia. Regarding the treatment of HBV in patients with
minimal because of the low prevalence of HBV and HCV thalassemia, this has not been reported in the literature.
infections in these nations.9 In the United States, HBsAg Currently approved drugs for chronic hepatitis B treat-
and antibody against hepatitis B core antigen (anti- ment in the general population include interferons and
HBcAg) screening are mandatory for whole blood dona- oral nucleoside/nucleotide analogs,67,68 but the safety and
tions based on current US Food and Drug Administration efficacy of these agents in thalassemia remain to be
recommendations, which also state that the use of NAT is investigated.
optional and cannot replace HBV serologic testing.56 Patients with thalassemia who are iron overloaded
Efforts to eradicate HCV infection should be imple- should receive chelation therapy.3 Table 1 outlines the
mented once hepatitis C has developed.53 The recent in- different iron-chelation strategies and their indications in
troduction of direct-acting antivirals (DAAs) for HCV the TDT and NTDT syndromes.2,54,55,69-71 It is also im-
treatment has represented an enormous leap in the ap- portant to mention that blood transfusions, in light of
proach to HCV infection, with high sustained viral re- their putative immunomodulatory effects, should be re-
sponse rates of approximately 90% to 95%.57 However, stricted to guidelines in TDT and only to instances when
all HCV-infected patients with thalassemia reported in they are indicated in NTDT. In addition, donor blood
the literature had received pegylated interferon with or should be leukoreduced to decrease its antigenic poten-
without ribavirin, which used to be the standard treat- tial,71 but this is only a hypothesis given the lack of studies
ment for chronic HCV infection before the advent of establishing a correlation between blood transfusions and
DAAs.58-60 Although some degree of efficacy has been HCC.
demonstrated in most of the trials investigating the use of
interferon alone or with ribavirin for the treatment of Treatment
chronic hepatitis C in thalassemia,3 the use of ribavirin in Table 2 outlines the HCC cases in the literature among
thalassemia patients remains controversial because of patients with thalassemia reported to date along with the
ribavirin-associated increased transfusion require- therapies that were tried in them.9,14,16,53,72-74 It is im-
ment.61,62 Another concern in HCV treatment is the need portant to note that, although little data has been pub-
to delay antiviral therapy pending appropriate iron chela- lished on HCC treatment in thalassemia, the following
tion, as suggested by some studies indicating that iron modalities have been proven both safe and effective in se-
overload reduces the response to interferon-a or com- lected patients with thalassemia: 1) surgical resection, 2)
bined interferon plus ribavirin.63-65 Conversely, a recent chemoembolization, and 3) simultaneous percutaneous

Cancer March 1, 2017 755


Review Article

TABLE 2. Summary of Hepatocellular Carcinoma in Patients With Thalassemia Reported in the Literature
and the Therapeutic Modalities Received

No. of
Reference Patients Type of Thalassemia Therapy

Zurlo 198972 1 TM Not mentioned


Borgna-Pignatti 200414 22 TM (n 5 8), TI (n 5 11), ST (n 5 3) Surgery (n 5 2); palliative therapy (n 5 20)
Modell 200873 2 Unspecified thalassemia Not mentioned
Fragatou 201016 5 TM (n 5 2) and TI (n 5 3) Combined chemotherapy (n 5 3); chemoembolization (n 5 2)
Maakaron 201353 2 TI Palliative therapy (n 5 1); percutaneous radioablation, sur-
gery, and palliative therapy (n 5 1)
Ansari 201374 1 Unspecified thalassemia Chemotherapy
Borgna-Pignatti 20149a 62 TM (n 5 32), TI (n 5 28), ST (n 5 2) Chemoembolization alone; thermoablation; surgery preced-
ed or followed by chemoembolization or thermoablation;
liver transplantation; palliative therapy; sorafenib

Abbreviations: TI, thalassemia intermedia; TM, thalassemia major; ST, sickle thalassemia.
a
In the patients described by Borgna-Pignatti et al,9 all attempted treatments are outlined; some patients received more than 1 treatment strategy, and the
authors did not mention which patient received which treatment(s).

radiofrequency thermoablation and ethanol injec- cating the course of the disease. It is noteworthy that the
tion.3,10,33 Although it has been demonstrated that the ki- significantly younger age at the diagnosis of HCC among
nase inhibitor sorafenib significantly improves prognosis patients who have thalassemia compared with the general
in early stage HCC,75,76 it is not known whether this drug population makes this a unique entity in which the con-
is efficacious in treating HCC in patients with thalasse- stellation of iron overload, chronic viral hepatitis, and
mia, because it was tried in 3 Italian thalassemia patients possibly transfusion-related immunomodulation are the
but produced unclear outcomes.9,15 main risk factors for hepatic carcinogenesis.
Thalassemia was previously considered a contraindi- Current approaches to screening for HCC in both
cation to liver transplantation, which has led to only a few TDT and NTDT patients include liver ultrasonography
patients undergoing this procedure.11 In fact, liver trans- every 6 months in individuals with at least 1 HCC risk
plantation has long been considered an HCC treatment factor (ie, HCV and/or HBV infection, iron overload, or
option that improves survival in the general population.5 advanced cirrhosis) in addition to annual MRI-based LIC
In an Italian case series of patients with TDT and NTDT, measurement in all patients.3,9,11,53-55 Biannual liver ul-
the survival of the only patient who underwent transplan- trasonography is a shared recommendation with the gen-
tation was 69 months, whereas the survival of the eral population, but yearly LIC assessment is particular to
8 untransplanted patients was 25.25 6 23.65 months patients who have thalassemia because of their high risk of
(range, 3-64 months).15 Two other patients with TDT iron overload, which is an established HCC risk factor as
from a different Italian study underwent successful liver described above. HCC prevention in patients with thalas-
transplantation with satisfactory post-transplantation out- semia might differ slightly from that in patients without
comes.3 Borgna-Pignatti et al reported 2 deaths among 3 thalassemia, and extra care must be taken to ameliorate
patients who underwent transplantation but for reasons the cancer risk factors unique to the former subgroup,
not related to thalassemia, including 1 death from cirrhot- namely, chelation therapy for increased iron burden and
ic liver failure possibly related to HCV recurrence and 1 hepatitis treatment only with drugs that have been proven
from meningococcal sepsis.9 Given the promising results both safe and efficacious in this patient population. To
highlighted above, thalassemia should no longer be con- date, there is no data supporting a different approach for
sidered a contraindication to liver transplantation in the HCC treatment, including liver transplantation, in
absence of significant comorbidities like heart disease and patients who have thalassemia compared with those who
pulmonary hypertension.3 do not, provided that the treatment options are tailored to
the individual patient’s clinical status and coexisting mor-
CONCLUSION bidities.3 It is paramount to stress the importance of
HCC is an emerging complication in patients with thalas- adopting a multidisciplinary approach that values the
semia, but data is lacking with regard to its precise burden. patient’s quality of life, in which the hematologist, gastro-
The incidence of HCC in the thalassemia syndromes enterologist, medical oncologist, surgeon, and mental
reflects an evolving morbidity that is increasingly compli- health provider all work together to optimize treatment

756 Cancer March 1, 2017


Hepatocellular Carcinoma in Thalassemia/Moukhadder et al

outcomes. On the other hand, ongoing research on HCC 9. Borgna-Pignatti C, Garani MC, Forni GL, et al. Hepatocellular car-
cinoma in thalassaemia: an update of the Italian Registry. Br J Hae-
in the thalassemia realm is lacking, although multiple matol. 2014;167:121-126.
aspects about HCC in thalassemia are a nidus for future 10. Mancuso A, Sciarrino E, Concetta Renda M, Maggio A. A prospec-
tive study of hepatocellular carcinoma incidence in thalassemia. He-
investigation, such as the exact role of transfusional im- moglobin. 2006;30:119-124.
mune dysregulation in hepatic carcinogenesis, the use of 11. Mancuso A, Perricone G. Time to define a new strategy for manage-
DAAs in treating HCV-infected thalassemia patients, and ment of hepatocellular carcinoma in thalassaemia? Br J Haematol.
2015;168:304-305.
the utility of anti-HBV treatments in patients with thalas- 12. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics,
semia who concurrently have hepatitis B. Multicenter in- 2002. CA Cancer J Clin. 2005;55:74-108.
13. El-Serag HB, Mason AC. Rising incidence of hepatocellular carcino-
ternational studies will be valuable to strengthen available ma in the United States. N Engl J Med. 1999;340:745-750.
data on the aforementioned aspects and pave the way for 14. Borgna-Pignatti C, Vergine G, Lombardo T, et al. Hepatocellular
carcinoma in the thalassaemia syndromes. Br J Haematol. 2004;124:
novel therapeutic targets in this special population, with 114-117.
particular emphasis on the long-term outcomes of such 15. Restivo Pantalone G, Renda D, Valenza F, et al. Hepatocellular car-
treatment modalities as tumor resection, percutaneous lo- cinoma in patients with thalassaemia syndromes: clinical characteris-
tics and outcome in a long term single centre experience. Br J
cal ablation, transarterial chemoembolization, liver trans- Haematol. 2010;150:245-247.
plantation, and palliative therapy in the subgroup of 16. Fragatou S, Tsourveloudis I, Manesis G. Incidence of hepatocellular
carcinoma in a thalassemia unit. Hemoglobin. 2010;34:221-226.
patients with HCC who also have thalassemia. An inter- 17. Rivella S. The role of ineffective erythropoiesis in non-transfusion-
national registry tracking all HCC cases in patients with dependent thalassemia. Blood Rev. 2012;26(suppl 1):S12-S5.
thalassemia reported in the literature to date and incorpo- 18. Nemeth E. Hepcidin in b-thalassemia. Ann N Y Acad Sci. 2010;
1202:31-35.
rating additional accounts in the next 5 to 10 years may be 19. Kew MC. Hepatic iron overload and hepatocellular carcinoma. Can-
especially useful in outlining pathophysiologic and thera- cer Lett. 2009;286:38-43.
20. Zacharski LR, Chow BK, Howes PS, et al. Decreased cancer risk af-
peutic trends unique to this patient population. ter iron reduction in patients with peripheral arterial disease: results
from a randomized trial. J Natl Cancer Inst. 2008;100:996-1002.
21. Matzner Y, Hershko C, Polliack A, Konijn AM, Izak G. Suppressive
FUNDING SUPPORT effect of ferritin on in vitro lymphocyte function. Br J Haematol.
No specific funding was disclosed. 1979;42:345-353.
22. Walker EM, Walker SM. Effects of iron overload on the immune
system. Ann Clin Lab Sci. 2000;30:354-365.
CONFLICT OF INTEREST DISCLOSURES 23. Green R, Esparza I, Schreiber R. Iron inhibits the nonspecific
Maria Domenica Cappellini and Ali T. Taher report grants from tumoricidal activity of macrophages. Ann N Y Acad Sci. 1988;526:
Novartis Pharmaceuticals and Celgene Corporation outside the 301-309.
24. Borgna-Pignatti C, De Stefano P, Sessa F, Avato F. Hepatocellular
submitted work. carcinoma in thalassemia major. Med Pediatr Oncol. 1986;14:327-
328.
AUTHOR CONTRIBUTIONS 25. Maakaron JE, Musallam KM, Ayache JB, Jabbour M, Tawil AN,
Taher AT. A liver mass in an iron-overloaded thalassaemia interme-
Hassan M. Moukhadder and Racha Halawi wrote the article. dia patient [serial online]. Br J Haematol. 2013;161:1.
Maria Domenica Cappellini critically reviewed the article. Ali T. 26. Wonke B, Hoffbrand A, Brown D, Dusheiko G. Antibody to hepa-
Taher edited and critically reviewed the article. All authors read titis C virus in multiply transfused patients with thalassaemia major.
and approved the final draft. J Clin Pathol. 1990;43:638-640.
27. Mirmomen S, Alavian SM, Hajarizadeh B, et al. Epidemiology of
hepatitis B, hepatitis C, and human immunodeficiency virus infec-
REFERENCES tions in patients with beta-thalassemia in Iran: a multicenter study.
1. Weatherall DJ, Clegg JB. The Thalassaemia Syndromes. 4th ed. Ox- Arch Iran Med. 2006;9:319-323.
ford, UK: Blackwell Scientific Publications; 2001. 28. Prati D, Zanella A, Farma E, et al. A multicenter prospective study
2. Musallam KM, Rivella S, Vichinsky E, Rachmilewitz EA. Non-trans- on the risk of acquiring liver disease in anti-hepatitis C virus nega-
fusion-dependent thalassemias. Haematologica. 2013;98:833-844. tive patients affected from homozygous b-thalassemia. Blood. 1998;
3. Mancuso A. Hepatocellular carcinoma in thalassemia: a critical re- 92:3460-3464.
view. World J Hepatol. 2010;2:171-174. 29. Prati D. Benefits and complications of regular blood transfusion in
4. Borgna-Pignatti C, Rugolotto S, De Stefano P, et al. Survival and patients with beta-thalassaemia major. Vox Sang. 2000;79:129-137.
complications in patients with thalassemia major treated with trans- 30. Minola E, Prati D, Suter F, et al. Age at infection affects the long-
fusion and deferoxamine. Haematologica. 2004;89:1187-1193. term outcome of transfusion-associated chronic hepatitis C. Blood.
5. Mancuso A. Management of hepatocellular carcinoma: enlightening 2002;99:4588-4591.
the gray zones. World J Hepatol. 2013;5:302-310. 31. Szmuness W. Hepatocellular carcinoma and the hepatitis B virus: ev-
6. Bruix J, Sherman M, Llovet JM, et al. EASL Panel of Experts on idence for a causal association. Prog Med Virol. 1978;24:40-69.
HCC. Clinical management of hepatocellular carcinoma. Conclu- 32. Chakravarti A, Verma V, Kumaria R, Dubey A. Anti-HCV seroposi-
sions of the Barcelona-2000 EASL conference. European Association tivity among multiple transfused patients with beta thalassaemia.
for the Study of the Liver. J Hepatol. 2001;35:421-430. J Indian Med Assoc. 2005;103:64-66.
7. Kowdley KV. Iron, hemochromatosis, and hepatocellular carcinoma. 33. Mancuso A, Rigano P, Renda D, et al. Hepatocellular carcinoma on
Gastroenterology. 2004;127(5 suppl 1):S79-S86. cirrhosis-free liver in a HCV-infected thalassemic. Am J Hematol.
8. Ginzburg Y, Rivella S. b-Thalassemia: a model for elucidating the 2005;78:158-159.
dynamic regulation of ineffective erythropoiesis and iron metabolism. 34. Girelli D, Pasino M, Goodnough JB, et al. Reduced serum hepcidin
Blood. 2011;118:4321-4330. levels in patients with chronic hepatitis C. J Hepatol. 2009;51:845-852.

Cancer March 1, 2017 757


Review Article

35. Angelucci E, Muretto P, Nicolucci A, et al. Effects of iron overload 57. Jimenez-Perez M, Gonzalez-Grande R, Espanaa Contreras P, Pinazo
and hepatitis C virus positivity in determining progression of liver fi- Martinez I, de la Cruz Lombardo J, Olmedo Martin R. Treatment
brosis in thalassemia following bone marrow transplantation. Blood. of chronic hepatitis C with direct-acting antivirals: the role of resis-
2002;100:17-21. tance. World J Gastroenterol. 2016;22:6573-6781.
36. Harnois DM. Hepatitis C virus infection and the rising incidence of 58. Hernandez N, Mescia G, Pollio C, Robaina G, Quintana L, Bianchi
hepatocellular carcinoma. Mayo Clin Proc. 2012;87:7-8. C. Effectiveness of pegylated interferon alfa 2a and ribavirin for
37. Vento S, Cainelli F, Cesario F. Infections and thalassaemia. Lancet chronic hepatitis C in clinical practice [article in Spanish]. Acta Gas-
Infect Dis. 2006;6:226-233. troenterol Latinoam. 2010;40:117-121.
38. Refaai MA, Blumberg N. Transfusion immunomodulation from a 59. NIH Consensus Statement on management of hepatitis C: 2002.
clinical perspective: an update. Expert Rev Hematol. 2013;6:653-663. NIH Consens State Sci Statements. 2002;19:1-46.
39. Dua D, Choudhury M, Prakash K. Altered T and B lymphocytes in 60. Yu ML, Chuang WL. Treatment of chronic hepatitis C in Asia:
multitransfused patients of thalassemia major. Indian Pediatr. 1993; when East meets West. J Gastroenterol Hepatol. 2009;24:336-345.
30:893-896. 61. Saffar MJ, Saffar H, Khalilian AR, Naqshvar F. Combination thera-
40. Skoutelis AT, Lianou E, Papavassiliou T, Karamerou A, Politi K, py with interferon and ribavirin for chronic hepatitis C infection in
Bassaris H. Defective phagocytic and bactericidal function of poly- beta-thalassaemia major. East Mediterr Health J. 2009;15:785-791.
morphonuclear leucocytes in patients with b-thalassaemia major. 62. Inati A, Taher A, Ghorra S, et al. Efficacy and tolerability of pegin-
J Infect. 1984;8:118-122. terferon alpha-2a with or without ribavirin in thalassaemia major
41. Matzner Y, Goldfarb A, Abrahamov A, Drexler R, Friedberg A, patients with chronic hepatitis C virus infection. Br J Haematol.
Rachmilewitz EA. Impaired neutrophil chemotaxis in patients with 2005;130:644-646.
thalassaemia major. Br J Haematol. 1993;85:153-158. 63. Burt MJ, Cooksley WG. The influence of iron on chronic hepatitis
42. Burrows L, Tartter P. Effect of blood transfusions on colonic malig- C. J Gastroenterol Hepatol. 1998;13:330-333.
nancy recurrence rate [letter]. Lancet. 1982;320:662. 64. Distante S, Bjoro K, Hellum K, et al. Raised serum ferritin predicts
43. Blumberg N. Deleterious clinical effects of transfusion immunomo- non-response to interferon and ribavirin treatment in patients with
dulation: proven beyond a reasonable doubt. Transfusion. 2005;45(2 chronic hepatitis C infection. Liver. 2002;22:269-275.
suppl):33S-39S; discussion 39S-40S. 65. Lebray P, Zylberberg H, Hue S, et al. Influence of HFE gene poly-
44. Landers DF, Hill GE, Wong K, Fox IJ. Blood transfusion-induced morphism on the progression and treatment of chronic hepatitis C.
immunomodulation. Anesth Analg. 1996;82:187-204. J Viral Hepat. 2004;11:175-282.
45. Puliyel M, Sposto R, Berdoukas VA, et al. Ferritin trends do not 66. Jafroodi M, Asadi R, Heydarzadeh A, Besharati S. Effect of hepatic
predict changes in total body iron in patients with transfusional iron
iron concentration and viral factors in chronic hepatitis C-infected
overload. Am J Hematol. 2014;89:391-394.
patients with thalassemia major, treated with interferon and ribavi-
46. Fischer R, Harmatz PR. Non-invasive assessment of tissue iron over-
rin. Int J Gen Med. 2011;4:529-533.
load. Hematology Am Soc Hematol Educ Program. 2009;2009:215-
67. Lok AS, McMahon BJ. Chronic hepatitis B: update 2009. Hepatol-
221.
ogy. 2009;50:661-662.
47. Taher A, El Rassi F, Isma’eel H, Koussa S, Inati A, Cappellini MD.
68. Gish RG. Hepatitis B treatment: current best practices, avoiding re-
Correlation of liver iron concentration determined by R2 magnetic
resonance imaging with serum ferritin in patients with thalassemia sistance. Cleve Clin J Med. 2009;76(suppl 3):S14-S19.
intermedia. Haematologica. 2008;93:1584-1586. 69. Vichinsky E. Non-transfusion-dependent thalassemia and thalassemia
48. Pakbaz Z, Fischer R, Fung E, Nielsen P, Harmatz P, Vichinsky E. intermedia: epidemiology, complications, and management. Curr
Serum ferritin underestimates liver iron concentration in transfusion Med Res Opin. 2016;32:191-204.
independent thalassemia patients as compared to regularly transfused 70. Cappellini MD, Cohen A, Piga A, et al. A phase 3 study of defera-
thalassemia and sickle cell patients. Pediatr Blood Cancer. 2007;49: sirox (ICL670), a once-daily oral iron chelator, in patients with
329-332. b-thalassemia. Blood. 2006;107:3455-3462.
49. Angelucci E, Brittenham GM, Mclaren CE, et al. Hepatic iron con- 71. Cappellini MD, Cohen A, Porter J, Taher A, Viprakasit V, eds.
centration and total body iron stores in thalassemia major. N Engl J Guidelines for the Management of Transfusion Dependent Thalas-
Med. 2000;343:327-331. saemia (TDT) [internet]. Nicosia, Cyprus: Thalassaemia Internation-
50. Bruix J, Sherman M. Management of hepatocellular carcinoma: an al Federation; 2014.
update. Hepatology. 2011;53:1020-1022. 72. Zurlo M, De Stefano P, Borgna-Pignatti C, et al. Survival and
51. Bruix J, Sherman M. Management of hepatocellular carcinoma. Hep- causes of death in thalassaemia major. Lancet. 1989;334:27-30.
atology. 2005;42:1208-1236. 73. Modell B, Khan M, Darlison M, Westwood MA, Ingram D,
52. Fitzmorris P, Singal AK. Surveillance and diagnosis of hepatocellular Pennell DJ. Improved survival of thalassaemia major in the UK and
carcinoma. Gastroenterol Hepatol (N Y). 2015;11:38-46. relation to T2* cardiovascular magnetic resonance [serial online].
53. Maakaron JE, Cappellini MD, Graziadei G, Ayache JB, Taher AT. J Cardiovasc Magn Reson. 2008;10:42.
Hepatocellular carcinoma in hepatitis-negative patients with thalasse- 74. Ansari S, Azarkivan A, Halagi F. Incidence of hepatocellular carcino-
mia intermedia: a closer look at the role of siderosis. Ann Hepatol. ma in patients with thalassemia who had hepatitis C. Acta Med Iran.
2013;12:142-146. 2013;51:404-407.
54. Borgna-Pignatti C, Marsella M. Iron chelation in thalassemia major. 75. Rampone B, Schiavone B, Martino A, Viviano C, Confuorto G.
Clin Ther. 2015;37:2866-2877. Current management strategy of hepatocellular carcinoma. World J
55. Saliba AN, Harb AR, Taher AT. Iron chelation therapy in Gastroenterol. 2009;15:3210-3216.
transfusion-dependent thalassemia patients: current strategies and fu- 76. Poon D, Anderson BO, Chen LT, et al. Asian Oncology Summit.
ture directions. J Blood Med. 2015;6:197-209. Management of hepatocellular carcinoma in Asia: consensus state-
56. Kuhns MC, Busch MP. New strategies for blood donor screening ment from the Asian Oncology Summit 2009. Lancet Oncol. 2009;
for hepatitis B virus. Mol Diagn Ther. 2006;10:77-91. 10:1111-1118.

758 Cancer March 1, 2017

You might also like