Download PDF
Review  |  Open Access  |  4 Dec 2021

Hepatocellular carcinoma in transfusion dependent thalassemia patients: a review from a clinical perspective

Views: 1556 |  Downloads: 627 |  Cited:   0
Hepatoma Res 2021;7:77.
10.20517/2394-5079.2021.118 |  © The Author(s) 2021.
Author Information
Article Notes
Cite This Article

Abstract

Survival in patients with transfusion-dependent thalassemias (TDT) has increased, and complications such as hepatocellular carcinoma (HCC) are emerging. Risk factors include viral infection, mainly hepatitis C virus (HCV), iron overload, the presence of cirrhosis, and immune dysregulation. Median survival after HCC occurrence has been estimated at 12 months, while data regarding the incidence of HCC in this population are minimal. Implementing effective hepatitis B virus (HBV)/HCV antiviral treatment and universal HBV vaccination programs is expected to decrease the risk for hepatocarcinogenesis substantially. Significant hemosiderosis and hepatic fibrosis are common in patients with TDT despite chelation therapy and have been correlated with HCC development. Thus, iron overload should be monitored with liver iron concentration and ferritin levels, and effective chelation therapy should be applied. In addition, all TDT patients, particularly those with cirrhosis, should be under surveillance every six months with abdominal ultrasound ± alpha-fetoprotein levels, as this combination seems to provide better sensitivity for early HCC detection.

Keywords

Transfusion-dependent thalassemias, hepatocellular carcinoma, hepatitis B virus, hepatitis C virus, iron overload, liver iron concentration, liver stiffness measurement

INTRODUCTION

Beta thalassemia is caused by mutations in the hemoglobin beta-globin gene that leads to impaired production of beta-globin chains. Beta thalassemia major (TM) is the most severe form of beta-thalassemia, characterized by minimal to no beta-globin chain production. Individuals with beta-thalassemia major have profound and lifelong transfusion-dependent anemia (TDT), which leads to several organ pathological conditions, mainly due to iron overload.

However, as survival in patients with TDT has increased over time, mainly due to efficient iron chelation therapy, it seems that the complications from the heart that were common and fatal are reduced, and other previously rarer complications such as hepatocellular carcinoma (HCC) are emerging[1].

Traditionally, the risk of HCC development in these patients has been linked to hepatitis B virus (HBV) and hepatitis C virus (HCV) infections mainly transmitted by blood transfusions[2]. However, recent data highlight the risk of HCC development in HCV- and HBV-negative patients, indicating the crucial role of iron overload[3-5].

This review aims to investigate the epidemiological data and the possible mechanisms involved in developing HCC in patients with TDT, including viral hepatitis, the role of iron overload, and the immunological disturbances that characterize these patients.

HCC IN TDT EPIDEMIOLOGY

HCC is the seventh most frequently occurring cancer globally and the second most common cause of cancer mortality[6]. Its incidence rises progressively with advancing age, showing a higher prevalence among males[7]. Significant risk factors for HCC include HBV, HCV, chronic alcohol consumption, obesity, type II diabetes, and non-alcoholic steatohepatitis (NASH). In general, the prevalence and etiology of HCC present a heterogeneous geographic distribution according to the predominant risk factor of each region. Thus, most cases are associated with HBV in Western Africa, Latin America, and East Asia, while most cases are associated with HCV in North America and Western Europe[8]. However, non-alcoholic fatty liver disease (NAFLD)/NASH is rapidly increasing and is expected to become the predominant risk factor for HCC in high-income regions[9]. In the United States, the incidence rates per 100,000 persons are 13.8 in men and 4.9 in women[10].

Data concerning etiological factors and treatment outcomes of HCC appear to be lacking in patients with TDT. Moreover, data regarding the incidence of HCC in this population are minimal. The majority of HCC cases are from population-based studies from Italy and Greece[11-15]. The evaluation of the total number of patients included in the updated Italian registry revealed a cumulative incidence of HCC in patients with TDT of 1.02%[12]. Overall, the study showed 60 new cases of HCC between 2002 and December 2012, among 5855 thalassemia patients who have been followed. A Greek study evaluated 57 patients with TM and thalassemia intermedia[14]. The incidence of HCC in patients with TM has been calculated as 3.5%. Data from a prospective study with 105 adults with TDT reveal a 2% incidence of HCC during a one-year observation period[16]. Furthermore, preliminary data regarding HCC survey from 1327 thalassemic patients, aged > 30 years from 13 centers, enrolled in the International Network of Clinicians for Endocrinopathies in Thalassemia and Adolescent Medicine (ICET-A), reveal a prevalence of 1.66% in TDT patients and 1.96% in non-TDT patients[17] [Table 1].

Table 1

Data regarding clinical characteristics, incidence, prevalence, and survival in TDT patients with HCC

StudyBorgna-Pignatti et al.[11], 2004Borgna-Pignatti et al.[12], 2014Fragatou et al.[14], 2010Papadopoulos, et al.[15], 2020Mancuso et al.[16], 2006De Sanctis et al.[17], 2020
N236057 (2 patients with HCC)42108 (2 patients with HCC)1022 (17 patients with HCC)
Mean/median age at diagnosis, years45 ± 11NA49 (48-50)45.5 ± 5.851 (39-63)41 (36-59)
Males, n (%)15 (65)37 (60)1 (50)27 (64.5)1 (50)11 (65)
HBsAg (+), n/N (%)
2/22 (9)3/60 (5)0/2 (0)0/48 (0)0/2 (0)0/17 (0)
HCV-RNA (+), n/N (%)17/22 (77.3)43/60 (71.5)2/2 (100)25/42 (59.5)2/2 (100)10/17 (59)
Mean/median level of serum ferritin (μg/l)1926.4 (369-6300)1041 (124-6200)1100 (310-1890)NANANA
Mean LIC values-9 mgFe/g-48.6 ± 29.5 μmolFe/g NA7.9 mgFe/g
Cirrhosis, n/N (%)NANA2/2 (100)33/42 (78.5)1/2 (50)6/17 (35.3)
Incidence (%)-1.023.5-2-
Prevalence (%)-----1.66
Median survival, (months)3.5 (1-26)11.5 (1.4-107.2)5.5 (3-8)12 (2-96)--

Based on current reports, it is clear that, compared to the general population, HCC appears at a younger age in TDT patients, suggesting the presence of multiple risk factors operating early in life for this population[3]. More precisely, the median age at diagnosis is 45 years among patients with TDT compared to 64 years among the general population[7,10,12]. Moreover, as discussed above, until 2000, HCC was relatively uncommon in TDT patients, as they died younger from cardiac problems related to iron overload[18,19]. In recent years, effective iron chelation treatment has led to a significant prolongation of survival thanks to the prevention of cardiac complications. However, many TDT patients still have liver iron overload with concomitant liver fibrosis[20]. Median survival after HCC occurrence has been estimated at 12 months (range 2-96 months) in a Greek study and was mainly dependent on the Barcelona Clinic Liver Cancer grading system[15]. In addition, data from Italy indicate a similar median survival of 11.5 months (range 1.4-107.2 months)[12].

In most countries, incidence rates of HCC among men in the general population are two- to four-fold higher than rates among women, possibly due to the higher incidence of cirrhosis, higher levels of smoking, and greater alcohol intake[7]. Interestingly, this difference seems to be less profound between men and women in TDT patients. It is well known that the effects of testosterone may increase signaling androgen receptors in men, promoting the proliferation of hepatocytes[21]. As hypogonadism has been revealed in a significant proportion of TDT patients, we may assume that lower testosterone levels could explain the almost equal occurrence of HCC between men and women[12].

HCC IN TDT - HBV INFECTION

Chronic HBV infection is the leading cause of global liver cancer incidences and deaths[22]. The incidence rate of HCC in patients with chronic HBV infection has been estimated to be 0.6% for those without cirrhosis and 3.7% for those with compensated cirrhosis[23]. Furthermore, the overall mortality rate among all cases of chronic HBV infection has been estimated as 30%-50%[24]. Besides the increased risk of HBV-related HCC in patients with cirrhosis, it is well known that HBV initiates the process of hepatic carcinogenesis by integrating into the host genome, thus leading to chromosomal instability and cell proliferation[25] [Figure 1]. The oncogenic nature of HBV per se is further demonstrated by the fact that the risk of developing HCC is not eliminated even in non-cirrhotic patients receiving high-barrier to resistance antiviral treatment[26].

Hepatocellular carcinoma in transfusion dependent thalassemia patients: a review from a clinical perspective

Figure 1. Hepatocarcinogenesis in patients with transfusion-dependent thalassemias (TDT). HCC: Hepatocellular carcinoma; HBV: hepatitis B virus; HCV: hepatitis C virus; ROS: reactive oxygen species; DNA: deoxyribonucleic acid.

Worldwide, 0.3%-5.7% of thalassemia patients are hepatitis B surface antigen (HBsAg) positive[19]. In the Italian updated registry data, the HBsAg positivity reached 5% among 60 TDT patients with HCC. In contrast, no HBsAg positivity was revealed in a Greek study with 42 TDT patients with HCC[12,15]. Thus, HBV infection does not play a significant role in HCC development in these patients, at least in Western countries. However, since the prevalence of HBV infection presents a large heterogeneous geographical distribution, its role is likely to remain important in high endemicity areas such as Asia and Africa. The most effective method to control HBV transmission is universal vaccination. Successful outcomes of nationwide vaccination programs implemented by Taiwan in 1984 proved that HBV infection and HCC incidence rates could be substantially reduced by a rigorous vaccination program[27]. Thus, HBV-related HCC, including in high-endemic areas, is expected to decrease in the coming years, even in TDT patients[28].

HBV treatment improves survival and quality of life by preventing liver disease progression and HCC. The indications for treatment in TDT patients do not differ from those recommended in the general population and are primarily based on the combination of three criteria; HBV-DNA levels, serum alanine aminotransferase levels, and severity of liver disease[29].

HCC IN TDT - HCV INFECTION

Based on previously published data, HCV infection has been proposed as the leading risk factor for liver fibrosis in TDT patients[30]. TDT patients transfused before the universal blood donors screening, which was introduced in 1992 after the discovery of HCV, were at higher risk of blood-borne HCV infection. Although the prevalence of HCV infection among these patients has dramatically decreased during the last twenty years, it remains higher than the general population. It is still a significant problem in underdeveloped countries. Anti-HCV positive was present in 87% of thalassemic patients based on an Italian registry, 39.6%-74.4% in two Greek studies, and 18%-70% in studies from the Middle East[12,15,31,32]. However, whether HCV plays a direct role or merely an indirect one through cirrhosis in the pathogenesis of HCC remains uncertain[33]. It seems that oxidative stress production enhanced by the HCV core protein would partly contribute to the development of HCC, even rarely in the absence of liver cirrhosis[34] [Figure 1]. In a recent study from Greece, which reported all cases of malignant neoplastic disorders occurring in 3652 thalassemic patients diagnosed between 1985 and 2018, a strong positive association between HCV and HCC was found[13]. Moreover, it has been identified that the most frequent HCV genotype in TDT patients is 1b, which seems to play an essential role in HCC development according to a recent meta-analysis[35,36].

The new interferon-free regimens with direct-acting antiviral (DAA) agents have changed the treatment landscape of HCV infection. The indication for HCV treatment in this population is similar to that of the general population[37]. In a real-life study, DAAs achieved a sustained virological response rate of > 90% without any additional adverse events or drug-drug interactions with iron-chelating drugs[38]. However, it is well established that, in patients treated with DAAs, the absolute risk of HCC remains in patients with cirrhosis or other concomitant etiological factors[39].

HCC IN TDT - CIRRHOSIS

It is well known that cirrhosis is the dominant risk factor for the development of HCC. In the Western world, up to 90% of HCC arises in the presence of cirrhosis[40]. It was estimated that up to one third of patients with cirrhosis would develop HCC during their lifetime[41]. The annual incidence of HCC depends on the etiology of cirrhosis. Thus, it has been estimated as 3.23% for HBV, 4.81% for HCV, and 1.2% for genetic hemochromatosis[42].

Significant hemosiderosis and hepatic fibrosis were common in patients with TM despite chelation therapy[43]. Worldwide available data indicate that the prevalence of cirrhosis among patients with thalassemia ranges 11.1%-78.5%[14-17].

The co-existence of specific risk factors such as iron overload and HCV or HBV infection are additive factors that increase the risk of cirrhosis in this group of patients. However, the HCC development in patients with TDT is not inextricably linked to the presence of cirrhosis[16].

Since HCC incidence is higher in patients with more advanced cirrhosis, surveillance for early HCC detection with abdominal ultrasound (US) every six months is a reasonable and cost-effective route to reduce mortality in these patients[44]. Moreover, recently published data indicate that alpha-fetoprotein levels (with 20 ng/mL as a cut-off) in combination with US is better in detecting patients with HCC, providing a sensitivity of more than 95%[45].

HCC IN TDT - IRON OVERLOAD

It is well known that hereditary hemochromatosis (HH) is associated with a 20-200-fold increased risk for HCC[46]. The overall standardized incidence ratio of HCC in patients with HH was estimated as 1.7 (95% confidence interval: 1.5-2.0) in a Swedish study[47].

While iron is essential for normal human physiology, its excess is toxic as there is no physiological pathway for removal from the body. In addition, free iron accelerates the Fenton reaction that generates noxious reactive oxygen species (ROS), which severely damage cells and tissues and promote fibrogenesis[48].

Iron overload is implicated in the development of HCC in other hereditary disorders such as TDT. Kountouras et al.[49] investigated the role of iron overload and HCV in the severity of liver disease in a cohort of 211 adult Greek patients with beta-thalassemia major. Based on the findings from 109 patients with liver biopsy, they demonstrated that advanced fibrosis was present with even minimal hemosiderosis. Moreover, the presence of fibrosis was independent of ferritin values or HCV history. Consistent with other reports, iron seems to have a leading role in liver disease initiation and progression[50]. Liver stiffness measurement by transient elastography, as hepatic fibrosis assessment, is closely related to the degree of hepatic siderosis in TDT patients, indicating that chelation therapy is mandatory to prevent liver disease progression[51,52].

As discussed above, iron overload in TDT patients leads to denaturation of ferritin, producing an excess of ROS into the cytoplasm of the hepatocytes, thus provoking DNA damage and genomic instability[53] [Figure 1]. Free iron overproduction in hepatic tissue could be responsible for carcinogenesis, overcoming the protective effect of activation of tumor suppressor genes such as p53, which regulates cell cycle arrest, apoptosis, and senescence in response to cellular stress[54,55]. In addition, free intracellular ferrous ions react with hydrogen peroxide and activate the lipoxygenase that induces the peroxidation of polyunsaturated fatty acids in cell membranes[56,57]. This mechanism, called ferroptosis, provides evidence about the role of iron in HCC even in the absence of liver cirrhosis[57,58].

There is sufficient evidence that iron chelation therapy improves or stabilizes liver fibrosis independently of liver iron concentration or HCV prevalence[59]. However, it is uncertain whether iron chelation treatment may reduce the risk of HCC, although there is evidence that it may reduce the risk of developing HCC or even suppresses HCC growth in experimental studies[60,61]. Thus, monitoring of iron overload in patients with TDT is crucial.

Nevertheless, iron plays a significant role in the pathogenesis of liver disease and HCC in TDT patients, and effective chelation therapy is mandatory.

Iron overload measurements include liver iron concentration (LIC) using magnetic resonance imaging, which accurately reflects the total body iron stores and ferritin levels that may predict changes in the total body iron levels[62]. Annual targets of LIC 2-5 mg/g dry weight and ferritin levels < 1000 ng/mL appear to be a reasonable strategy in TDT patients[63].

HCC IN TDT - OTHER OR COMBINED RISK FACTORS

Immune deficiency

Transfusion-related immunomodulation was reported several years ago and has been linked with several adverse effects in oncology[64]. Some of the reported immune abnormalities include quantitative and functional defects involving impaired B-lymphocyte function, defective chemotaxis and phagocytosis, and impaired immunoglobulin production, all of which are essential mechanisms in anticancer immune surveillance[65,66] [Figure 1]. It also has been suggested that iron homeostasis is an important determinant of valid T cell-mediated immune response, as either iron overload or iron deficiency induces immunologic aberrancies[67].

“Second-hit” mechanisms

The iron-related oxidative damage can be further enhanced by several factors such as HCV infection and NAFLD, promoting liver injury and fibrosis synergistically as a “second hit” phenomenon[53,68,69].

In a landmark study, Angelucci et al.[53] described that the combination of iron overload and HCV substantially increases the progression of liver fibrosis.

Dysmetabolic iron overload syndrome is now a frequent finding in the general population, as it is detected in about one-third of patients with NAFLD[70,71]. Moreover, hepatocellular iron accumulation was associated with a higher risk of fibrosis than the absence of siderosis in patients with NAFLD, and, once again, the mechanism involves increased oxidative stress[71].

CONCLUSIONS

HCC development in patients with TDT is a complex phenomenon. Several factors such as HBV and HCV infections, iron overload, and immune dysfunction are of paramount importance. Moreover, NAFLD represents a newly emerging risk factor. The efficient antiviral treatment for HCV and HBV infections, the preventive anti-HBV immunization, and, most importantly, the monitoring and treatment of iron overload are expected to reduce HCC in these patients in the future. In all patients, particularly those with cirrhosis, surveillance programs for HCC are mandatory for early detection and application of more effective and curative treatment.

DECLARATIONS

Authors’ contributions

Analyzed the data: Papadopoulos N

Wrote the paper: Papadopoulos N, Koskinas J

Designed and directed the project: Koskinas J

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

Both authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2021.

REFERENCES

1. Camaschella C, Nai A. Ineffective erythropoiesis and regulation of iron status in iron loading anaemias. Br J Haematol 2016;172:512-23.

2. Vichinsky E, Neumayr L, Trimble S, et al. CDC Thalassemia Investigators. Transfusion complications in thalassemia patients: a report from the Centers for Disease Control and Prevention (CME). Transfusion 2014;54:972-81; quiz 971.

3. Marsella M, Ricchi P. Thalassemia and hepatocellular carcinoma: links and risks. J Blood Med 2019;10:323-34.

4. Mancuso A. Evidence-based medicine and management of hepatocellular carcinoma in Thalassemia. BMC Gastroenterol 2020;20:409.

5. Mangia A, Bellini D, Cillo U, et al. Hepatocellular carcinoma in adult thalassemia patients: an expert opinion based on current evidence. BMC Gastroenterol 2020;20:251.

6. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424.

7. McGlynn KA, Petrick JL, El-Serag HB. Epidemiology of hepatocellular carcinoma. Hepatology 2021;73 Suppl 1:4-13.

8. Singal AG, Lampertico P, Nahon P. Epidemiology and surveillance for hepatocellular carcinoma: new trends. J Hepatol 2020;72:250-61.

9. Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:6.

10. Cancer stat facts: liver and intrahepatic bile duct cancer. Available from: https://seer.cancer.gov/statfacts/html/livibd.html [Last accessed on 25 Nov 2021].

11. Borgna-Pignatti C, Vergine G, Lombardo T, et al. Hepatocellular carcinoma in the thalassaemia syndromes. Br J Haematol 2004;124:114-7.

12. Borgna-Pignatti C, Garani MC, Forni GL, et al. Hepatocellular carcinoma in thalassaemia: an update of the Italian Registry. Br J Haematol 2014;167:121-6.

13. Kourakli A, Diamantidis MD, Skafidas M, et al. Hepatitis C virus infection, but not hepatic iron overload is the dominant risk factor for the manifestation of hepatocellular carcinoma among Greek thalassemic patients. Blood 2018;132:2347.

14. Fragatou S, Tsourveloudis I, Manesis G. Incidence of hepatocellular carcinoma in a thalassemia unit. Hemoglobin 2010;34:221-6.

15. Papadopoulos N, Kountouras D, Malagari K, Tampaki M, Theochari M, Koskinas J. Characteristics and prognosis of hepatocellular carcinoma in multi-transfused patients with β-thalassemia. Experience of a single tertiary center. Mediterr J Hematol Infect Dis 2020;12:e2020013.

16. Mancuso A, Sciarrino E, Renda MC, Maggio A. A prospective study of hepatocellular carcinoma incidence in thalassemia. Hemoglobin 2006;30:119-24.

17. De Sanctis V, Soliman AT, Daar S, et al. A concise review on the frequency, major risk factors and surveillance of hepatocellular carcinoma (HCC) in β-thalassemias: past, present and future perspectives and the ICET-A experience. Mediterr J Hematol Infect Dis 2020;12:e2020006.

18. Mancuso A. Hepatocellular carcinoma in thalassemia: a critical review. World J Hepatol 2010;2:171-4.

19. Finianos A, Matar CF, Taher A. Hepatocellular carcinoma in β-thalassemia patients: review of the literature with molecular insight into liver carcinogenesis. Int J Mol Sci 2018;19:4070.

20. Aydinok Y, Porter JB, Piga A, et al. Prevalence and distribution of iron overload in patients with transfusion-dependent anemias differs across geographic regions: results from the CORDELIA study. Eur J Haematol 2015;95:244-53.

21. Pok S, Barn VA, Wong HJ, et al. Testosterone regulation of cyclin E kinase: a key factor in determining gender differences in hepatocarcinogenesis. J Gastroenterol Hepatol 2016;31:1210-9.

22. Akinyemiju T, Abera S, Ahmed M, et al. Global Burden of Disease Liver Cancer Collaboration. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the global burden of disease study 2015. JAMA Oncol 2017;3:1683-91.

23. Fattovich G, Bortolotti F, Donato F. Natural history of chronic hepatitis B: special emphasis on disease progression and prognostic factors. J Hepatol 2008;48:335-52.

24. Huang YT, Jen CL, Yang HI, et al. Lifetime risk and sex difference of hepatocellular carcinoma among patients with chronic hepatitis B and C. J Clin Oncol 2011;29:3643-50.

25. Ghouri YA, Mian I, Rowe JH. Review of hepatocellular carcinoma: epidemiology, etiology, and carcinogenesis. J Carcinog 2017;16:1.

26. Papatheodoridis G, Dalekos G, Sypsa V, et al. PAGE-B predicts the risk of developing hepatocellular carcinoma in Caucasians with chronic hepatitis B on 5-year antiviral therapy. J Hepatol 2016;64:800-6.

27. Chien YC, Jan CF, Kuo HS, Chen CJ. Nationwide hepatitis B vaccination program in Taiwan: effectiveness in the 20 years after it was launched. Epidemiol Rev 2006;28:126-35.

28. Franco E, Bagnato B, Marino MG, Meleleo C, Serino L, Zaratti L. Hepatitis B: epidemiology and prevention in developing countries. World J Hepatol 2012;4:74-80.

29. European Association for the Study of the Liver, Electronic address: easloffice@easloffice.eu, European Association for the Study of the Liver. EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J Hepatol 2017;67:370-98.

30. Di Marco V, Capra M, Gagliardotto F, et al. Liver disease in chelated transfusion-dependent thalassemics: the role of iron overload and chronic hepatitis C. Haematologica 2008;93:1243-6.

31. Triantos C, Kourakli A, Kalafateli M, et al. Hepatitis C in patients with β-thalassemia major. A single-centre experience. Ann Hematol 2013;92:739-46.

32. Alavian SM, Tabatabaei SV, Kamran BL. Epidemiology of HCV infection among thalassemia patients in eastern Mediterranean countries: a quantitative review of literature. Iran Red Crescent Med J 2010;12:365-76.

33. Koike K. Pathogenesis of HCV-associated HCC: dual-pass carcinogenesis through activation of oxidative stress and intracellular signaling. Hepatol Res 2007;37 Suppl 2:S115-20.

34. Koike K. Hepatitis C virus contributes to hepatocarcinogenesis by modulating metabolic and intracellular signaling pathways. J Gastroenterol Hepatol 2007;22 Suppl 1:S108-11.

35. Origa R, Ponti ML, Filosa A, et al. Italy for THAlassemia and hepatitis C Advance - Società Italiana Talassemie ed Emoglobinopatie (ITHACA-SITE). Treatment of hepatitis C virus infection with direct-acting antiviral drugs is safe and effective in patients with hemoglobinopathies. Am J Hematol 2017;92:1349-55.

36. Raimondi S, Bruno S, Mondelli MU, Maisonneuve P. Hepatitis C virus genotype 1b as a risk factor for hepatocellular carcinoma development: a meta-analysis. J Hepatol 2009;50:1142-54.

37. Association for the Study of the Liver, Electronic address: easloffice@easloffice.eu, Clinical Practice Guidelines Panel: Chair:, EASL Governing Board representative, Panel members. EASL recommendations on treatment of hepatitis C: Final update of the series. J Hepatol 2020;73:1170-218.

38. Sinakos E, Kountouras D, Koskinas J, et al. Treatment of chronic hepatitis C with direct-acting antivirals in patients with β-thalassaemia major and advanced liver disease. Br J Haematol 2017;178:130-6.

39. Kanwal F, Kramer J, Asch SM, Chayanupatkul M, Cao Y, El-Serag HB. Risk of hepatocellular cancer in HCV patients treated with direct-acting antiviral agents. Gastroenterology 2017;153:996-1005.e1.

40. Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet 2018;391:1301-14.

41. Ioannou GN, Splan MF, Weiss NS, McDonald GB, Beretta L, Lee SP. Incidence and predictors of hepatocellular carcinoma in patients with cirrhosis. Clin Gastroenterol Hepatol 2007;5:938-45, 945.e1.

42. Tarao K, Nozaki A, Ikeda T, et al. Real impact of liver cirrhosis on the development of hepatocellular carcinoma in various liver diseases-meta-analytic assessment. Cancer Med 2019;8:1054-65.

43. Li CK, Chik KW, Lam CW, et al. Liver disease in transfusion dependent thalassaemia major. Arch Dis Child 2002;86:344-7.

44. European Association for the Study of the Liver, Electronic address: easloffice@easloffice.eu., European Association for the Study of the Liver. EASL Clinical Practice Guidelines: management of hepatocellular carcinoma. J Hepatol 2018;69:182-236.

45. Colli A, Nadarevic T, Miletic D, et al. Abdominal ultrasound and alpha-foetoprotein for the diagnosis of hepatocellular carcinoma in adults with chronic liver disease. Cochrane Database Syst Rev 2021;4:CD013346.

46. Bradbear RA, Bain C, Siskind V, et al. Cohort study of internal malignancy in genetic hemochromatosis and other chronic nonalcoholic liver diseases. J Natl Cancer Inst 1985;75:81-4.

47. Elmberg M, Hultcrantz R, Ekbom A, et al. Cancer risk in patients with hereditary hemochromatosis and in their first-degree relatives. Gastroenterology 2003;125:1733-41.

48. Sharp P, Srai SK. Molecular mechanisms involved in intestinal iron absorption. World J Gastroenterol 2007;13:4716-24.

49. Kountouras D, Tsagarakis NJ, Fatourou E, et al. Liver disease in adult transfusion-dependent beta-thalassaemic patients: investigating the role of iron overload and chronic HCV infection. Liver Int 2013;33:420-7.

50. Siagris D, Giannakoulas N, Christofidou M, et al. Virological, immunological and histological aspects in adult beta-thalassemic patients with chronic hepatitis C virus infection. Liver Int 2004;24:204-9.

51. Maira D, Cassinerio E, Marcon A, et al. Progression of liver fibrosis can be controlled by adequate chelation in transfusion-dependent thalassemia (TDT). Ann Hematol 2017;96:1931-6.

52. Delicou S, Maragkos K, Tambaki M, Kountouras D, Koskinas J. Transient elastography (TE) is a useful tool for assessing the response of liver iron chelation in sickle cell disease patients. Mediterr J Hematol Infect Dis 2018;10:e2018049.

53. Angelucci E, Muretto P, Nicolucci A, et al. Effects of iron overload and hepatitis C virus positivity in determining progression of liver fibrosis in thalassemia following bone marrow transplantation. Blood 2002;100:17-21.

54. Kim E, Viatour P. Hepatocellular carcinoma: old friends and new tricks. Exp Mol Med 2020;52:1898-907.

55. Fargion S, Valenti L, Fracanzani AL. Role of iron in hepatocellular carcinoma. Clin Liver Dis (Hoboken) 2014;3:108-10.

56. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012;149:1060-72.

57. Li J, Cao F, Yin HL, et al. Ferroptosis: past, present and future. Cell Death Dis 2020;11:88.

58. Kew MC. Hepatic iron overload and hepatocellular carcinoma. Cancer Lett 2009;286:38-43.

59. Deugnier Y, Turlin B, Ropert M, et al. Improvement in liver pathology of patients with β-thalassemia treated with deferasirox for at least 3 years. Gastroenterology 2011;141:1202-11, 1211.e1-3.

60. Kato J, Miyanishi K, Kobune M, et al. Long-term phlebotomy with low-iron diet therapy lowers risk of development of hepatocellular carcinoma from chronic hepatitis C. J Gastroenterol 2007;42:830-6.

61. Ba Q, Hao M, Huang H, et al. Iron deprivation suppresses hepatocellular carcinoma growth in experimental studies. Clin Cancer Res 2011;17:7625-33.

62. St Pierre TG, Clark PR, Chua-anusorn W, et al. Noninvasive measurement and imaging of liver iron concentrations using proton magnetic resonance. Blood 2005;105:855-61.

63. Porter J, Viprakasit V, Kattamis A. . Chapter 3: Iron overload and chelation. Guidelines for the management of transfusion dependent thalassaemia (TDT). Nicosia (CY): Thalassaemia International Federation; 2014.

64. Cata JP, Wang H, Gottumukkala V, Reuben J, Sessler DI. Inflammatory response, immunosuppression, and cancer recurrence after perioperative blood transfusions. Br J Anaesth 2013;110:690-701.

65. Moukhadder HM, Halawi R, Cappellini MD, Taher AT. Hepatocellular carcinoma as an emerging morbidity in the thalassemia syndromes: a comprehensive review. Cancer 2017;123:751-8.

66. Farmakis D, Giakoumis A, Polymeropoulos E, Aessopos A. Pathogenetic aspects of immune deficiency associated with beta-thalassemia. Med Sci Monit 2003;9:RA19-22.

67. Shen L, Zhou Y, He H, et al. Crosstalk between macrophages, T Cells, and iron metabolism in tumor microenvironment. Oxid Med Cell Longev 2021;2021:8865791.

68. Asare GA, Bronz M, Naidoo V, Kew MC. Synergistic interaction between excess hepatic iron and alcohol ingestion in hepatic mutagenesis. Toxicology 2008;254:11-8.

69. Sekine S, Ito K, Watanabe H, et al. Mitochondrial iron accumulation exacerbates hepatic toxicity caused by hepatitis C virus core protein. Toxicol Appl Pharmacol 2015;282:237-43.

70. Dongiovanni P, Fracanzani AL, Fargion S, Valenti L. Iron in fatty liver and in the metabolic syndrome: a promising therapeutic target. J Hepatol 2011;55:920-32.

71. Valenti L, Fracanzani AL, Bugianesi E, et al. HFE genotype, parenchymal iron accumulation, and liver fibrosis in patients with nonalcoholic fatty liver disease. Gastroenterology 2010;138:905-12.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Papadopoulos N, Koskinas J. Hepatocellular carcinoma in transfusion dependent thalassemia patients: a review from a clinical perspective. Hepatoma Res 2021;7:77. http://dx.doi.org/10.20517/2394-5079.2021.118

AMA Style

Papadopoulos N, Koskinas J. Hepatocellular carcinoma in transfusion dependent thalassemia patients: a review from a clinical perspective. Hepatoma Research. 2021; 7: 77. http://dx.doi.org/10.20517/2394-5079.2021.118

Chicago/Turabian Style

Papadopoulos, Nikolaos, John Koskinas. 2021. "Hepatocellular carcinoma in transfusion dependent thalassemia patients: a review from a clinical perspective" Hepatoma Research. 7: 77. http://dx.doi.org/10.20517/2394-5079.2021.118

ACS Style

Papadopoulos, N.; Koskinas J. Hepatocellular carcinoma in transfusion dependent thalassemia patients: a review from a clinical perspective. Hepatoma. Res. 2021, 7, 77. http://dx.doi.org/10.20517/2394-5079.2021.118

About This Article

© The Author(s) 2021. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
1556
Downloads
627
Citations
0
Comments
0
6

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 7 clicks
Like This Article 6 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Hepatoma Research
ISSN 2454-2520 (Online) 2394-5079 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/