169
Views
9
CrossRef citations to date
0
Altmetric
Review

Thalassemia and hepatocellular carcinoma: links and risks

& ORCID Icon
Pages 323-334 | Published online: 17 Sep 2019

References

  • Vitrano A, Calvaruso G, Lai E, et al. The era of comparable life expectancy between thalassaemia major and intermedia: is it time to revisit the major-intermedia dichotomy? Br J Haematol. 2017;176(1):124–130. doi:10.1111/bjh.1438127748513
  • Taher AT, Mussalam K, Cappellini MD. Guidelines for the Management of Non Transfusion Dependent Editors of the 2nd Edition. TIF; 2018.
  • Ricchi P, Ammirabile M, Costantini S, et al. Splenectomy is a risk factor for developing hyperuricemia and nephrolithiasis in patients with thalassemia intermedia: a retrospective study. Blood Cells Mol Dis. 2012;49(3–4):133–135. doi:10.1016/j.bcmd.2012.05.01222705193
  • Ricchi P, Ammirabile M, Costantini S, et al. A useful relationship between the presence of extramedullary erythropoeisis and the level of the soluble form of the transferrin receptor in a large cohort of adult patients with thalassemia intermedia: a prospective study. Ann Hematol. 2012;91(6):905–909. doi:10.1007/s00277-011-1385-y22167341
  • Modell B, Khan M, Darlison M, Westwood MA, Ingram D, Pennell DJ. Improved survival of thalassaemia major in the UK and relation to T2* cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2008;10(1):1–8. doi:10.1186/1532-429X-10-42
  • Ricchi P, Ammirabile M, Spasiano A, et al. Combined chelation therapy in thalassemia major with deferiprone and desferrioxamine: a retrospective study. Eur J Haematol. 2010;85(1):36–42.20331740
  • Pepe A, Meloni A, Rossi G, et al. Cardiac and hepatic iron and ejection fraction in thalassemia major: multicentre prospective comparison of combined deferiprone and deferoxamine therapy against deferiprone or deferoxamine monotherapy. J Cardiovasc Magn Reson. 2013;15(1):1–11. doi:10.1186/1532-429X-15-123324167
  • Dessi C, Leoni G, Moi P, et al. Thalassemia major between liver and heart: where we are now. Blood Cells Mol Dis. 2015;55(1):82–88.25976473
  • 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(3):244–253.25418187
  • Petrick JL, Kelly SP, Altekruse SF, McGlynn KA, Rosenberg PS. Future of hepatocellular carcinoma incidence in the United States forecast through 2030. J Clin Oncol. 2016;34(15):1787–1794. doi:10.1200/JCO.2015.64.741227044939
  • Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–E386. doi:10.1002/ijc.2921025220842
  • Globocan. Liver fact sheet. Available from: http://gco.iarc.fr/today/data/factsheets/cancers/11-Liver-fact-sheet.pdf. Accessed 8, 22 2019.
  • Borgna-Pignatti C, De Stefano P, Sessa F, Avato F. Hepatocellular carcinoma in thalassemia major. Med Pediatr Oncol. 1986;14(6):327–328.2431257
  • Moukhadder HM, Halawi R, Cappellini MD, Taher AT. Hepatocellular carcinoma as an emerging morbidity in the thalassemia syndromes: a comprehensive review. Cancer. 2017;123(5):751–758. doi:10.1002/cncr.3046227911488
  • 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(12):4070. doi:10.3390/ijms19124070.
  • Zurlo MG, De Stefano P, Borgna-Pignatti C, et al. Survival and causes of death in thalassaemia major. Lancet (London, England). 1989;2(8653):27–30. doi:10.1016/s0140-6736(89)90264-x
  • Borgna-Pignatti C, Garani MC, Forni GL, et al. Hepatocellular carcinoma in thalassaemia: an update of the Italian registry. Br J Haematol. 2014;167(1):121–126. doi:10.1111/bjh.1300924992281
  • Ansari S, Azarkivan A, Halagi F. Incidence of hepatocellular carcinoma in patients with thalassemia who had hepatitis C. Acta Med Iran. 2013;51(6):404–407.23852846
  • Borgna-Pignatti C, Vergine G, Lombardo T, et al. Hepatocellular carcinoma in the thalassaemia syndromes. Br J Haematol. 2004;124(1):114–117. doi:10.1046/j.1365-2141.2003.04732.x14675416
  • Fragatou S, Tsourveloudis I, Manesis G. Incidence of hepatocellular carcinoma in a thalassemia unit. Hemoglobin. 2010;34(3):221–226. doi:10.3109/03630269.2010.48507120524812
  • Maakaron JE, Cappellini MD, Graziadei G, Ayache JB, Taher AT. Hepatocellular Carcinoma in Hepatitis-Negative Patients with Thalassemia Intermedia: A Closer Look at the Ann Hepatol. 2013;12(1):142–146.
  • Maakaron JE, Musallam KM, Ayache JB, Jabbour M, Tawil AN, Taher AT. A liver mass in an iron-overloaded thalassaemia intermedia patient. Br J Haematol. 2013;161(1):1. doi:10.1111/bjh.12195
  • Mancuso A, Rigano P, Renda D, et al. Hepatocellular carcinoma on cirrhosis-free liver in a HCV-infected thalassemic. Am J Hematol. 2005;78(2):158–159. doi:10.1002/ajh.20289
  • Mancuso A, Sciarrino E, Renda MC, Maggio A. A prospective study of hepatocellular carcinoma incidence in thalassemia. Hemoglobin. 2006;30(1):119–124. doi:10.1080/0363026050045556516540424
  • Moukhadder HM, Roumi JE, Bou-Fakhredin R, Taher AT. Hepatocellular carcinoma in a beta-thalassemia intermedia patient: yet another case in the expanding epidemic. Hemoglobin. 2018;42(1):58–60. doi:10.1080/03630269.2018.143419729493312
  • Restivo PG, Renda D, Valenza F, et al. Hepatocellular carcinoma in patients with thalassaemia syndromes: clinical characteristics and outcome in a long term single centre experience. Br J Haematol. 2010;150:226–248. doi:10.1111/j.1365-2141.2010.08179.x20433679
  • Musallam KM, Cappellini MD, Wood JC, Taher AT. Iron overload in non-transfusion-dependent thalassemia: a clinical perspective. Blood Rev. 2012;26(Suppl 1):S16–S19. doi:10.1016/S0268-960X(12)70006-122631036
  • Globocan. Estimated age-standardized incidence and mortality rates for liver cancer (Southern Europe) in 2018, all ages. Available from: http://gco.iarc.fr/today/online-analysis-dual-bars-2. Published 2018 Accessed 8, 22 2019.
  • Leong TY-M, Leong AS-Y. Epidemiology and carcinogenesis of hepatocellular carcinoma. HPB (Oxford). 2005;7(1):5–15. doi:10.1080/1365182041002402118333156
  • Mahmoudvand S, Shokri S, Taherkhani R, Farshadpour F. Hepatitis c virus core protein modulates several signaling pathways involved in hepatocellular carcinoma. World J Gastroenterol. 2019;25(1):42–58. doi:10.3748/wjg.v25.i1.4230643357
  • Wonke B, Hoffbrand AV, Brown D, Dusheiko G. Antibody to hepatitis C virus in multiply transfused patients with thalassaemia major. J Clin Pathol. 1990;43(8):638–640. doi:10.1136/jcp.43.8.6382119395
  • Origa R, Ponti ML, Filosa A, et al. Treatment of hepatitis C virus infection with direct-acting antiviral drugs is safe and effective in patients with hemoglobinopathies. Am J Hematol. 2017;92(12):1349–1355. doi:10.1002/ajh.2491128929515
  • Mancuso A. Hepatocellular carcinoma in thalassemia: a critical review. World J Hepatol. 2010;2(5):171–174. doi:10.4254/wjh.v2.i5.17121160991
  • Schulze-Krebs A, Preimel D, Popov Y, et al. Hepatitis C virus-replicating hepatocytes induce fibrogenic activation of hepatic stellate cells. Gastroenterology. 2005;129(1):246–258. doi:10.1053/j.gastro.2005.03.08916012951
  • 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(1):17–21. doi:10.1182/blood.v100.1.1712070002
  • Zou DM, Sun WL. Relationship between hepatitis C virus infection and iron overload. Chin Med J (Engl). 2017;130(7):866–871. doi:10.4103/0366-6999.20273728345552
  • Girelli D, Pasino M, Goodnough JB, et al. Reduced serum hepcidin levels in patients with chronic hepatitis C. J Hepatol. 2009;51(5):845–852. doi:10.1016/j.jhep.2009.06.02719729219
  • 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(10):830–836. doi:10.1007/s00535-007-2095-z17940836
  • Kanda T, Goto T, Hirotsu Y, Moriyama M, Omata M. Molecular mechanisms driving progression of liver cirrhosis towards hepatocellular carcinoma in chronic hepatitis B and C infections: a review. Int J Mol Sci. 2019;20(6):1358. doi:10.3390/ijms20061358
  • Szmuness W. Hepatocellular carcinoma and the hepatitis B virus: evidence for a causal association. Prog Med Virol. 1978;24:40–69.212785
  • Petruzziello A. Epidemiology of hepatitis B virus (HBV) and hepatitis C virus (HCV) related hepatocellular carcinoma. Open Virol J. 2018;12(1):26–32. doi:10.2174/187435790181201002629541276
  • Takano S, Yokosuka O, Imazeki F, Tagawa M, Omata M. Incidence of hepatocellular carcinoma in chronic hepatitis B and C: a prospective study of 251 patients. Hepatology. 1995;21(3):650–655.7875662
  • Tawada A, Kanda T, Imazeki F, Yokosuka O. Prevention of hepatitis B virus-associated liver diseases by antiviral therapy. Hepatol Int. 2016;10(4):574–593. doi:10.1007/s12072-016-9720-y27026375
  • Kanda T, Wu S, Sasaki R, et al. HBV core protein enhances cytokine production. Dis (Basel, Switzerland). 2015;3(3):213–220.
  • Raimondo G, Navarra G, Mondello S, et al. Occult hepatitis B virus in liver tissue of individuals without hepatic disease. J Hepatol. 2008;48(5):743–746. doi:10.1016/j.jhep.2008.01.02318314221
  • Siagris D, Christofidou M, Triga K, et al. Occult hepatitis B virus infection in hemodialysis patients with chronic HCV infection. J Nephrol. 2006;19(3):327–333.16874693
  • Hollinger FB. Hepatitis B virus infection and transfusion medicine: science and the occult. Transfusion. 2008;48(5):1001–1026. doi:10.1111/j.1537-2995.2008.01701.x18454738
  • Spreafico M, Berzuini A, Foglieni B, et al. Poor efficacy of nucleic acid testing in identifying occult HBV infection and consequences for safety of blood supply in Italy. J Hepatol. 2015;63(5):1068–1076. doi:10.1016/j.jhep.2015.06.01626116791
  • Mortensen E, Kamali A, Schirmer PL, et al. Are current screening protocols for chronic hepatitis B virus infection adequate? Diagn Microbiol Infect Dis. 2016;85(2):159–167. doi:10.1016/j.diagmicrobio.2015.12.00527009896
  • Cholongitas E, Haidich A-B, Apostolidou-Kiouti F, Chalevas P, Papatheodoridis GV. Hepatitis B virus reactivation in HBsAg-negative, anti-HBc-positive patients receiving immunosuppressive therapy: a systematic review. Ann Gastroenterol. 2018;31(4):480–490. doi:10.20524/aog.2018.026629991894
  • Di Stefano M, Volpe A, Stallone G, et al. Occult HBV infection in hemodialysis setting is marked by presence of isolated antibodies to HBcAg and HCV. J Nephrol. 2009;22(3):381–386.19557715
  • Hassan ZK, Hafez MM, Mansor TM, Zekri ARN. Occult HBV infection among Egyptian hepatocellular carcinoma patients. Virol J. 2011;8:90. doi:10.1186/1743-422X-8-9021371325
  • Liu C-J, Chen D-S, Chen P-J. Epidemiology of HBV infection in Asian blood donors: emphasis on occult HBV infection and the role of NAT. J Clin Virol. 2006;36(Suppl 1):S33–S44.16831692
  • Candotti D, Grabarczyk P, Ghiazza P, et al. Characterization of occult hepatitis B virus from blood donors carrying genotype A2 or genotype D strains. J Hepatol. 2008;49(4):537–547. doi:10.1016/j.jhep.2008.04.01718602718
  • Raimondo G, Locarnini S, Pollicino T, Levrero M, Zoulim F, Lok AS. Update of the statements on biology and clinical impact of occult hepatitis b virus infection. J Hepatol. 2019. doi:10.1016/j.jhep.2019.03.034
  • Mandishona E, MacPhail AP, Gordeuk VR, et al. Dietary iron overload as a risk factor for hepatocellular carcinoma in Black Africans. Hepatology. 1998;27(6):1563–1566. doi:10.1002/hep.5102706149620327
  • Kowdley KV. Iron, hemochromatosis, and hepatocellular carcinoma. Gastroenterology. 2004;127(5 Suppl 1):S79–S86. doi:10.1016/j.gastro.2004.09.01915508107
  • Blanc JF, De Ledinghen V, Bernard PH, et al. Increased incidence of HFE C282Y mutations in patients with iron overload and hepatocellular carcinoma developed in non-cirrhotic liver. J Hepatol. 2000;32(5):805–811.10845668
  • Bralet MP, Regimbeau JM, Pineau P, et al. Hepatocellular carcinoma occurring in nonfibrotic liver: epidemiologic and histopathologic analysis of 80 French cases. Hepatology. 2000;32(2):200–204. doi:10.1053/jhep.2000.903310915724
  • Herrinton LJ, Friedman GD, Baer D, Selby JV. Transferrin saturation and risk of cancer. Am J Epidemiol. 1995;142(7):692–698.7572938
  • Ioannou GN, Kowdley KV. Iron, HFE mutations, and hepatocellular carcinoma: is hepatic iron a carcinogen? Clin Gastroenterol Hepatol. 2003;1(4):246–248. doi:10.1016/S1542-3565(03)00126-515017663
  • Moyo VM, Makunike R, Gangaidzo IT, et al. African iron overload and hepatocellular carcinoma (HA-7-0-080). Eur J Haematol. 1998;60(1):28–34.9451425
  • Turlin B, Juguet F, Moirand R, et al. Increased liver iron stores in patients with hepatocellular carcinoma developed on a noncirrhotic liver. Hepatology. 1995;22(2):446–450.7635411
  • Taher A, Hershko C, Cappellini MD. Iron overload in thalassaemia intermedia: reassessment of iron chelation strategies. Br J Haematol. 2009;147(5):634–640. doi:10.1111/j.1365-2141.2009.07848.x19681884
  • Nemeth E. Hepcidin in beta-thalassemia. Ann N Y Acad Sci. 2010;1202:31–35. doi:10.1111/j.1749-6632.2010.05585.x20712769
  • Matzner Y, Hershko C, Polliack A, Konijn AM, Izak G. Suppressive effect of ferritin on in vitro lymphocyte function. Br J Haematol. 1979;42(3):345–353. doi:10.1111/j.1365-2141.1979.tb01142.x157770
  • Walker EMJ, Walker SM. Effects of iron overload on the immune system. Ann Clin Lab Sci. 2000;30(4):354–365.11045759
  • Green R, Esparza I, Schreiber R. Iron inhibits the nonspecific tumoricidal activity of macrophages. A possible contributory mechanism for neoplasia in hemochromatosis. Ann N Y Acad Sci. 1988;526:301–309. doi:10.1111/j.1749-6632.1988.tb55514.x2455463
  • Djeha A, Brock JH. Effect of transferrin, lactoferrin and chelated iron on human T-lymphocytes. Br J Haematol. 1992;80(2):235–241. doi:10.1111/j.1365-2141.1992.tb08906.x1550782
  • Deugnier Y, Turlin B. Iron and hepatocellular carcinoma. J Gastroenterol Hepatol. 2001;16(5):491–494.11350542
  • Seligman PA, Kovar J, Gelfand EW. Lymphocyte proliferation is controlled by both iron availability and regulation of iron uptake pathways. Pathobiology. 1992;60(1):19–26. doi:10.1159/0001636921543546
  • Taher AT, Porter JB, Viprakasit V, et al. Defining serum ferritin thresholds to predict clinically relevant liver iron concentrations for guiding deferasirox therapy when MRI is unavailable in patients with non-transfusion-dependent thalassaemia. Br J Haematol. 2015;168(2):284–290.25212456
  • Ricchi P, Meloni A, Spasiano A, et al. The impact of liver steatosis on the ability of serum ferritin levels to be predictive of liver iron concentration in non-transfusion-dependent thalassaemia patients. Br J Haematol. 2018;180(5):721–726.29363758
  • Clouston AD, Powell EE. Interaction of non-alcoholic fatty liver disease with other liver diseases. Best Pract Res Clin Gastroenterol. 2002;16(5):767–781.12406444
  • Paradis V, Zalinski S, Chelbi E, et al. Hepatocellular carcinomas in patients with metabolic syndrome often develop without significant liver fibrosis: a pathological analysis. Hepatology. 2009;49(3):851–859.19115377
  • Divella R, Mazzocca A, Daniele A, Sabba C, Obesity PA. Nonalcoholic fatty liver disease and adipocytokines network in promotion of cancer. Int J Biol Sci. 2019;15(3):610–616.30745847
  • Younossi ZM, Otgonsuren M, Henry L, et al. Association of nonalcoholic fatty liver disease (NAFLD) with hepatocellular carcinoma (HCC) in the United States from 2004 to 2009. Hepatology. 2015;62(6):1723–1730.26274335
  • Puoti C. New insights on hepatocellular carcinoma: epidemiology and clinical aspects. Hepatoma Res. 2018;4(9):57.
  • Lin H, Fu C, Kannengiesser S, et al. Quantitative analysis of hepatic iron in patients suspected of coexisting iron overload and steatosis using multi-echo single-voxel magnetic resonance spectroscopy: Comparison with fat-saturated multi-echo gradient echo sequence. J Magn Reson Imaging. 2018;48(1):205–213. doi:10.1002/jmri.2596729513377
  • Gamberini MR, De Sanctis V, Gilli G. Hypogonadism, diabetes mellitus, hypothyroidism, hypoparathyroidism: incidence and prevalence related to iron overload and chelation therapy in patients with thalassaemia major followed from 1980 to 2007 in the Ferrara Centre. Pediatr Endocrinol Rev. 2008;6(Suppl 1):158–169.19337172
  • Multicentre study on prevalence of endocrine complications in thalassaemia major. Italian working group on endocrine complications in non-endocrine diseases. Clin Endocrinol (Oxf). 1995;42(6):581–586.7634497
  • De Sanctis V, Roos M, Gasser T, Fortini M, Raiola G, Galati MC. Impact of long-term iron chelation therapy on growth and endocrine functions in thalassaemia. J Pediatr Endocrinol Metab. 2006;19(4):471–480.16759032
  • Mancuso A, Perricone G. Time to define a new strategy for management of hepatocellular carcinoma in thalassaemia? Br J Haematol. 2015;168(2):304–305.25146379
  • Borgna-Pignatti C, Gamberini MR. Complications of thalassemia major and their treatment. Expert Rev Hematol. 2011;4(3):353–366.21668399
  • Koh W-P, Wang R, Jin A, Yu MC, J-M Y. Diabetes mellitus and risk of hepatocellular carcinoma: findings from the Singapore Chinese health study. Br J Cancer. 2013;108(5):1182–1188.23370206
  • Mantovani A, Targher G. Type 2 diabetes mellitus and risk of hepatocellular carcinoma: spotlight on nonalcoholic fatty liver disease. Ann Transl Med. 2017;5(13):270.28758096
  • Bruix J, Sherman M. Management of hepatocellular carcinoma. Hepatology. 2005;42(5):1208–1236.16250051
  • Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet. 2003;362(9399):1907–1917.14667750
  • Rampone B, Schiavone B, Martino A, Viviano C, Confuorto G. Current management strategy of hepatocellular carcinoma. World J Gastroenterol. 2009;15(26):3210–3216.19598295
  • Poon D, Anderson BO, Chen L-T, et al. Management of hepatocellular carcinoma in Asia: consensus statement from the Asian Oncology Summit 2009. Lancet Oncol. 2009;10(11):1111–1118.19880065
  • Verslype C, Van Cutsem E, Dicato M, et al. The management of hepatocellular carcinoma. Current expert opinion and recommendations derived from the 10th world congress on gastrointestinal cancer, Barcelona, 2008. Ann Oncol Off J Eur Soc Med Oncol. 2009;20(Suppl 7):vii1–vii6.
  • El-Serag HB, Marrero JA, Rudolph L, Reddy KR. Diagnosis and treatment of hepatocellular carcinoma. Gastroenterology. 2008;134(6):1752–1763.18471552
  • Tanwar S, Khan SA, Grover VPB, Gwilt C, Smith B, Brown A. Liver transplantation for hepatocellular carcinoma. World J Gastroenterol. 2009;15(44):5511–5516.19938188
  • Llovet JM, Bruix J. Novel advancements in the management of hepatocellular carcinoma in 2008. J Hepatol. 2008;48(Suppl 1):S20–S37.18304676
  • Bruix J, Sherman M, Llovet JM, et al. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL conference. European Association for the study of the liver. J Hepatol. 2001;35(3):421–430.11592607
  • Llovet JM, Ducreux M, Lencioni R, et al. EASL-EORTC Clinical practice guidelines: management of hepatocellular carcinoma. Eur J Cancer. 2012;48(5):599–641.22424278
  • Olivieri NF, Liu PP, Sher GD, et al. Brief report: combined liver and heart transplantation for end-stage iron-induced organ failure in an adult with homozygous beta-thalassemia. N Engl J Med. 1994;330(16):1125–1127.8133854
  • Mela M, Mancuso A, Burroughs AK. Review article: hepatocellular carcinoma: indications for liver transplantation. Aliment Pharmacol Ther. 2003;17(Suppl 2):130–137.12786624
  • Menon KV, Hakeem AR, Heaton ND. Review article: liver transplantation for hepatocellular carcinoma - a critical appraisal of the current worldwide listing criteria. Aliment Pharmacol Ther. 2014;40(8):893–902.25155143
  • Ricchi P, Meloni A, Di Matola T, et al. Galectin-3 and myocardial fibrosis by cardiac magnetic resonance in thalassaemia major. Blood. 2016;128(22):4832LP- 4832.
  • Trinchet JC, Chaffaut C, Bourcier V, et al. Ultrasonographic surveillance of hepatocellular carcinoma in cirrhosis: a randomized trial comparing 3- and 6-month periodicities. Hepatology. 2011;54(6):1987–1997. doi:10.1002/hep.2454522144108
  • Tzartzeva K, Obi J, Rich NE, et al. Surveillance imaging and alpha fetoprotein for early detection of hepatocellular carcinoma in patients with cirrhosis: a meta-analysis. Gastroenterology. 2018;154(6):1706–1718.e1. doi:10.1053/j.gastro.2018.01.06429425931
  • Musallam KM, Cappellini MD, Wood JC, et al. Elevated liver iron concentration is a marker of increased morbidity in patients with beta thalassemia intermedia. Haematologica. 2011;96(11):1605–1612. doi:10.3324/haematol.2011.04785221791471
  • Berdoukas V, Farmaki K, Wood JC, Coates T. Iron chelation in thalassemia: time to reconsider our comfort zones. Expert Rev Hematol. 2011;4(1):17–26. doi:10.1586/ehm.10.7421322775
  • Fibach E, Rachmilewitz EA. The role of antioxidants and iron chelators in the treatment of oxidative stress in thalassemia. Ann N Y Acad Sci. 2010;1202:10–16. doi:10.1111/j.1749-6632.2010.05577.x20712766
  • Di Marco V, Capra M, Angelucci E, et al. Management of chronic viral hepatitis in patients with thalassemia: recommendations from an international panel. Blood. 2010;116(16):2875–2883. doi:10.1182/blood-2009-11-24872420551378
  • 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(8):1243–1246. doi:10.3324/haematol.1255418556410
  • Hershko C. Pathogenesis and management of iron toxicity in thalassemia. Ann N Y Acad Sci. 2010;1202:1–9. doi:10.1111/j.1749-6632.2010.05544.x20712765
  • EASL. Recommendations on treatment of hepatitis C 2016. J Hepatol. 2017;66(1):153–194. doi:10.1016/j.jhep.2016.09.001.27667367
  • Harmatz P, Jonas MM, Kwiatkowski JL, et al. Safety and efficacy of pegylated interferon alpha-2a and ribavirin for the treatment of hepatitis C in patients with thalassemia. Haematologica. 2008;93(8):1247–1251. doi:10.3324/haematol.1235218556414
  • Inati A, Taher A, Ghorra S, et al. Efficacy and tolerability of peginterferon alpha-2a with or without ribavirin in thalassaemia major patients with chronic hepatitis C virus infection. Br J Haematol. 2005;130(4):644–646. doi:10.1111/j.1365-2141.2005.05645.x16098081
  • Di Marco V, D’Ambrosio R, Bronte F, et al. Dual therapy with peg-interferon and ribavirin in thalassemia major patients with chronic HCV infection: is there still an indication? Dig Liver Dis. 2016;48(6):650–655. doi:10.1016/j.dld.2016.02.00427012446
  • Kamal SM, Fouly AH, Mohamed MK, et al. Peginterferon alpha-2b therapy with and without ribavirin in patients with thalassemia: a randomized study. J Hepatol. 2006;44:S217. doi:10.1016/j.jhep.2005.09.019
  • Ricchi P, Ammirabile M, Costantini S, et al. The impact of previous or concomitant IFN therapy on deferiprone-induced agranulocytosis and neutropenia: a retrospective study. Expert Opin Drug Saf. 2010;9(6):875–881. doi:10.1517/14740338.2010.51083120945995
  • Ricchi P, Lanza AG, Ammirabile M, et al. Hepatitis C virus distribution and clearance following interferon-monotherapy among thalassaemia major and intermedia patients. Br J Haematol. 2011;155(4):524–527. doi:10.1111/j.1365-2141.2011.08717.x21564076
  • Kowdley KV, Gordon SC, Reddy KR, et al. Ledipasvir and sofosbuvir for 8 or 12 weeks for chronic HCV without cirrhosis. N Engl J Med. 2014;370(20):1879–1888. doi:10.1056/NEJMoa140235524720702
  • Prati D, Maggioni M, Milani S, et al. Clinical and histological characterization of liver disease in patients with transfusion-dependent beta-thalassemia. A multicenter study of 117 cases. Haematologica. 2004;89(10):1179–1186.15477201
  • Sulkowski MS, Jacobson IM, Nelson DR. Daclatasvir plus sofosbuvir for HCV infection. N Engl J Med. 2014;370(16):1560–1561. doi:10.1056/NEJMc1401726
  • Sharara AI, Rustom LBO, Marrache M, et al. Sofosbuvir/velpatasvir for chronic hepatitis C infection in patients with transfusion-dependent thalassemia. Am J Hematol. 2019;94(2):E43–E45. doi:10.1002/ajh.2533930394575
  • Ponti ML, Comitini F, Murgia D, et al. Impact of the direct-acting antiviral agents (DAAs) on chronic hepatitis C in Sardinian patients with transfusion-dependent Thalassemia major. Dig Liver Dis. 2019;51(4):561–567. doi:10.1016/j.dld.2018.12.01630658940
  • Aleman S, Rahbin N, Weiland O, et al. A risk for hepatocellular carcinoma persists long-term after sustained virologic response in patients with hepatitis C-associated liver cirrhosis. Clin Infect Dis. 2013;57(2):230–236.23616492
  • van der Meer AJ, Feld JJ, Hofer H, et al. Risk of cirrhosis-related complications in patients with advanced fibrosis following hepatitis C virus eradication. J Hepatol. 2017;66(3):485–493. doi:10.1016/j.jhep.2016.10.01727780714
  • El-Serag HB, Kanwal F, Richardson P, Kramer J. Risk of hepatocellular carcinoma after sustained virological response in veterans with hepatitis C virus infection. Hepatology. 2016;64(1):130–137. doi:10.1002/hep.2853526946190
  • Makiyama A, Itoh Y, Kasahara A, et al. Characteristics of patients with chronic hepatitis C who develop hepatocellular carcinoma after a sustained response to interferon therapy. Cancer. 2004;101(7):1616–1622. doi:10.1002/cncr.2053715378504
  • Su F, Ioannou GN. Hepatocellular carcinoma risk after direct‐acting antiviral therapy. Clin Liver Dis. 2019;13(1):6–12. doi:10.1002/cld.781
  • Li H, Huang M-H, Jiang J-D, Peng Z-G, Hepatitis C. From inflammatory pathogenesis to anti-inflammatory/hepatoprotective therapy. World J Gastroenterol. 2018;24(47):5297–5311. doi:10.3748/wjg.v24.i47.529730598575