Skip to main content
Gastrointestinal Tumors logoLink to Gastrointestinal Tumors
. 2015 Nov 18;2(4):188–194. doi: 10.1159/000441822

Nutrition and Hepatocellular Cancer

Kerstin Schütte 1, Christian Schulz 1, Peter Malfertheiner 1,*
PMCID: PMC4924450  PMID: 27403413

Abstract

Background

Hepatocellular carcinoma (HCC) significantly contributes to the global burden of cancer. Liver cancer is the third most frequent cause of cancer-related death with HCC representing more than 90% of primary liver cancers. The majority of patients are not only affected by the malignant disease but do also suffer from chronic liver disease. Therefore, several factors impact on the prognosis of patients with HCC, including tumor-related factors, liver function and patient-related factors such as performance status and other comorbidities. The nutritional status is of high significance for the patients' performance status, the tolerance of tumor-targeting therapy and the prognosis of cancer of any type and is specially referenced in HCC. This overview is on current concepts on the role of nutritional factors in hepatocarcinogenesis and the role of nutrition in patients affected by HCC.

Summary

Nutritional status and composition of diet are relevant factors related to the risk of HCC. They also have an important role concerning the prognosis of patients with HCC. Besides risk factors, several macro- and micronutrient components have been found to be inversely correlated with the risk of HCC. To prevent disease progression to liver cirrhosis or HCC in patients with nonalcoholic steatohepatitis, it is crucial to optimize the metabolic state

Key Message and Practical Implication

Evidence from well-designed prospective interventional trials with the aim to reduce the HCC incidence or to prolong survival in patients with HCC based on nutritional modification is still to be generated.

Key Words: Diet, Hepatocellular carcinoma, Malnutrition, Nutrition, Prognosis, Risk

Introduction

Hepatocellular carcinoma (HCC) significantly adds to the global burden of cancer. The most significant risk factor for HCC is liver cirrhosis, which is present in 70-90% of patients [1]. However, all risk factors that can lead to HCC in the presence of liver cirrhosis are also risk factors for the development of HCC in the absence of cirrhosis [2]. Worldwide, chronic viral hepatitis B (HBV) and C (HCV) are the most relevant factors associated with hepatocarcinogenesis. In industrialized countries, fatty liver disease, apart from viral hepatitis, is closely related to nutritional factors, and the prevalence of fatty liver disease is on the rise. High consumption of alcohol is one of the most significant risk factors for development of liver cirrhosis and HCC. However, in recent years, nonalcoholic fatty liver disease (NAFLD) has become more and more prevalent and nowadays has become the leading cause of HCC in some regions of Europe [3]. In patients with NAFLD the progression to HCC is frequent in the absence of cirrhosis. Metabolic risk factors such as obesity and diabetes mellitus have been identified as independent risk factors for HCC development [4,5,6,7].

The knowledge on the role of dietary habits which are protective or contribute to hepatocarcinogenesis, either associated with metabolic risk factors or independent of these, is limited.

In patients diagnosed with HCC, the nutritional status is of critical importance concerning the tolerability of tumor-directed therapy and survival.

Nutritional Factors in Hepatocarcinogenesis

Alcohol

Alcohol is a well-established cardinal risk factor for HCC development. There is little evidence of a direct carcinogenic effect of alcohol. The effect of alcohol on HCC development is driven by the alcohol-induced hepatic inflammation with progression to liver fibrosis or cirrhosis. However, alcohol is metabolized to acetaldehyde, which is capable of damaging DNA. A dose-effect relationship has been demonstrated for alcohol-related HCC [8]. Alcohol has a synergistic effect with other risk factors for HCC, in conjunction with HBV and HCV infections and, to a lesser extent, with diabetes [9].

Aflatoxin B1

Aflatoxin B1, a product of fungi of the Aspergillus species, is a common contaminant of grains, nuts and vegetables in regions in which climate and storage favor its development such as Asia and Africa, and is a direct carcinogen inducing specific point mutations in TP53 [10]. A simultaneous infection with the HBV potentiates the carcinogenic effect of aflatoxin B1 significantly [11]. It has been estimated that by reducing aflatoxin exposure in high-risk regions the number of HCC cases can be reduced by more than 20% [12].

Iron

Iron overload of the liver is a factor contributing to carcinogenesis. It can occur in hereditary hemochromatosis or as secondary iron overload because of chronic inflammation, especially in patients with alcohol abuse. Treatment with chelating agents and repeated phlebotomy in patients with hemochromatosis reduces the HCC risk [13]. In some regions of the world, e.g. Sub-Saharan Africa, high levels of ingested iron lead to hepatic iron overload [14] with levels comparable to those in hereditary hemochromatosis. The high amount of ingested iron results from large volumes of beer consumed which is home-brewed in cast iron drums or pots. It is estimated that this dietary iron overload, formerly referred to as Bantu siderosis, affects about 15% of black rural Africans [15,16].

Obesity and the Metabolic Syndrome

Obesity is one of the driving factors of NAFLD. Additionally, several studies have identified obesity as independent risk factor for HCC with a reported OR of 1.39-4.52 [4,5,6]. Weight loss is the cornerstone in the treatment of NAFLD. Changes in life style, pharmacological interventions and/or bariatric surgery can contribute to weight normalization. Whether persistent weight loss in obese patients also has the potential to positively influence the risk for HCC has not been adequately addressed in prospective clinical studies.

The options to positively influence NAFLD by pharmacotherapy are limited. Insulin resistance is an important factor in promoting NAFLD and may also be a causative factor in hepatocarcinogenesis. Drugs decreasing insulin resistance and lowering insulin levels, like the insulin sensitizer metformin, glitazones or GLP1 analogues like liraglutide, potentially have a positive impact on the risk for HCC in patients with NAFLD [14,17]. The same holds true for other interventions correcting aspects of the metabolic syndrome such as statins [18,19]. Obeticholic acid might expand the therapeutic options in nonalcoholic steatohepatitis, but its long-term beneficial effects in the prevention of HCC are not clear yet [20].

Composition of the Diet

There is sufficient evidence that NAFLD is associated with certain dietary patterns [21,22,23]. Several large cohort studies and few prospective randomized controlled trials have been conducted to analyze the role of diet and nutrition in liver cancer prevention, but the evidence for potential associations between nutrition and HCC is much weaker. While some of these studies evaluate the role of dietary patterns, others focus on distinct nutrients.

A prospective study from China analyzed food patterns in two large and prospectively followed cohorts. Using food frequency questionnaires at baseline, it was shown that a vegetable-based dietary pattern is associated with reduced liver cancer risk, while fruit- and meat-based diets did not show any association [24]. This observation was confirmed by a meta-analysis on published studies on the association between vegetable and fruit consumption and the risk of HCC: the risk of HCC decreases by 8% for every 100 g/day increase in vegetable intake [25]. In a large prospective evaluation in a large US cohort addressing the association of two dietary indices, the Healthy Eating Index-2010 (HEI-2010) and the alternate Mediterranean Diet Score (aMED), with HCC incidence and chronic liver disease mortality, it was shown that adherence to dietary recommendations has the potential to reduce the risk of both diseases. Higher HEI-2010 scores were significantly associated with a lower risk of HCC (HR 0.72; 95% CI 0.53-0.97), and the same holds true for high aMED scores (HR 0.62; 95% CI 0.47-0.84) [26]. In a European study on two cohorts from Greece and Italy, it was also demonstrated that the adherence to a Mediterranean diet pattern is associated with a lower risk for HCC [27]. In both cohorts, a high glycemic load of diet had a negative impact on the HCC risk [28,29].

EPIC is an ongoing multicenter prospective cohort study on the role of diet, lifestyle and environmental factors in the etiology of cancer and other diseases and has recruited more than 500,000 participants from all over Europe. From these studies several important data have been collected. An inverse association between total fat intake and risk of HCC (per 10 g/day HR 0.80; 95% CI 0.65-0.99) has been observed and appears to be mainly driven by monounsaturated fats [30]. Total fish intake was also inversely associated with HCC risk (per 20 g/day HR 0.83; 95% CI 0.74-0.95) while there was no association with meat consumption [31]. Higher consumption of dietary fiber (per 10 g/day HR 0.70; 95% CI 0.52-0.93), but not the intake of total carbohydrate, or the glycemic load or the glycemic index of diet, influenced the risk for HCC significantly [32]. A protective potential of fruits and vegetables high in flavonoids and antioxidants did not reach statistical significance [33]. These data obtained from populations from the European EPIC study are different from the results obtained from US cohorts. The NIH-AARP study that has prospectively enrolled almost 500,000 men and women reports an increased risk for HCC associated with the intake of red meat and saturated fat, and a stronger effect with the intake of saturated fat as compared to monounsaturated fat.

A recent meta-analysis on the risk of HCC and meat consumption identified seven cohort studies and ten case-control studies. It did not show an association of the intake of red meat, processed meat or total meat with HCC risk. High levels of white meat or fish consumption on the other hand can reduce the risk of HCC significantly [34].

Data from randomized controlled trials on nutrition and cancer prevention are scarce. A prospective randomized controlled trial in postmenopausal women could not show a beneficial effect of a low-fat diet on the incidence of liver cancer [35].

The nutritional status is a significant prognostic factor in patients with liver cirrhosis [36]. International guidelines recommend nutritional therapy for these patients whenever necessary. Optimal nutritional support prolongs survival, improves liver function and the nutritional status in patients with liver cirrhosis [37,38]. However, when focusing on the prevention of HCC, evidence-driven recommendations are lacking.

Coffee

Numerous studies have given evidence for an inverse association of coffee consumption and the risk for HCC [39,40,41,42,43]. An updated meta-analysis resulted in a risk reduction of 40% for any coffee consumption. The summary RR was 0.80 (95% CI 0.77-0.84) for an increment of 1 cup of coffee per day [44].

Branched-Chain Amino Acids

Oral branched-chain amino acid supplementation in patients with decompensated liver cirrhosis improves the nutritional status and prevents liver-related complications. In prospective clinical trials, this therapy was also shown to significantly lower HCC occurrence rates in patients with liver cirrhosis [45,46].

Vitamin D

Epidemiological studies show the inverse association between vitamin D status and the incidence of cancer, and biochemical studies suggest that vitamin D deficiency may play a role in the cause and progression of cancer [47]. Results of the EPIC trial revealed that higher baseline levels of 25(OH)D are associated with a lower risk of HCC (per 10 nmol/l increase IRR 0.80; 95% CI 0.68-0.94) [48]. In contrast, dietary calcium, vitamin D, fat and protein from dairy sources are associated with increased HCC risk while the same nutrients from nondairy sources show inverse or null associations [49].

Nutrition and Prognosis of HCC

Malnutrition is a frequent but underdiagnosed problem in cancer patients and is defined as ‘decline in lean body mass with the potential for functional impairment’ at multiple levels [50]. There is no commonly accepted gold standard for the diagnosis of malnutrition, but about one third of cancer patients are malnourished [51]. Patients with HCC are at a special increased risk for malnutrition. In patients with liver cirrhosis, malnutrition is a common finding and associated with mortality and reduced quality of life [52,53]. In HCC, the majority of cases are associated with liver function impairment because of liver cirrhosis [1], and tumor progression and tumor-directed therapies may further deteriorate liver function [54].

Prospective clinical studies have identified malnutrition as an independent negative prognostic factor in HCC, but interventional studies addressing the effect of nutritional therapy in HCC patients are few [55].

In patients undergoing resection of HCC in curative intent, perioperative nutritional management has been identified as key determinant of treatment success [56]. Modifications of the macronutrient and micronutrient composition of the diet as well as pharmacological interventions targeting nutritional and metabolic pathways are suggested to positively influence the patients' prognosis [14]. In patients on therapy for HCC, the supplementation of branched-chain amino acid improves liver function [57,58] and has the potential to prolong recurrence-free survival and overall survival [59]. These observations made from individual studies were only partially confirmed by a recent meta-analysis [60].

A prospective study of 200 patients with HCC revealed that 25(OH)D3 deficiency is associated with advanced stages of HCC and is a further prognostic indicator for poor outcome [61]. However, large prospective randomized controlled trials are still to be performed on the effect of nutritional interventions on prognosis of patients with HCC.

Conclusion

Nutritional status and composition of diet are relevant factors related to the risk of HCC. They also have an important role concerning the prognosis of patients with HCC. Several macro- and micronutrient components have been found to be inversely correlated with the risk for HCC. To prevent disease progression to liver cirrhosis or HCC in patients with nonalcoholic steatohepatitis, it is crucial to optimize the metabolic state. However, evidence from well-designed prospective interventional trials with the aim to reduce HCC incidence or to prolong survival in patients with HCC based on nutritional modification is still to be generated.

Disclosure Statement

The authors declare no conflicts of interest related to this paper.

References

  • 1.Schütte K, Bornschein J, Malfertheiner P. Hepatocellular carcinoma - epidemiological trends and risk factors. Dig Dis. 2009;27:80–92. doi: 10.1159/000218339. [DOI] [PubMed] [Google Scholar]
  • 2.Evert M, Dombrowski F. Hepatozelluläre Karzinome in der nichtzirrhotischen Leber. Pathologe. 2008;29:47–52. doi: 10.1007/s00292-007-0953-3. [DOI] [PubMed] [Google Scholar]
  • 3.Dyson J, Jaques B, Chattopadyhay D, Lochan R, Graham J, Das D, Aslam T, Patanwala I, Gaggar S, Cole M, Sumpter K, Stewart S, Rose J, Hudson M, Manas D, Reeves HL. Hepatocellular cancer: the impact of obesity, type 2 diabetes and a multidisciplinary team. J Hepatol. 2014;60:110–117. doi: 10.1016/j.jhep.2013.08.011. [DOI] [PubMed] [Google Scholar]
  • 4.Caldwell SH, Crespo DM, Kang HS, Al-Osaimi AMS. Obesity and hepatocellular carcinoma. Gastroenterology. 2004;127(5 suppl 1):S97–S103. doi: 10.1053/j.gastro.2004.09.021. [DOI] [PubMed] [Google Scholar]
  • 5.Borena W, Strohmaier S, Lukanova A, Bjørge T, Lindkvist B, Hallmans G, Edlinger M, Stocks T, Nagel G, Manjer J, Engeland A, Selmer R, Häggström C, Tretli S, Concin H, Jonsson H, Stattin P, Ulmer H. Metabolic risk factors and primary liver cancer in a prospective study of 578,700 adults. Int J Cancer. 2012;131:193–200. doi: 10.1002/ijc.26338. [DOI] [PubMed] [Google Scholar]
  • 6.Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348:1625–1638. doi: 10.1056/NEJMoa021423. [DOI] [PubMed] [Google Scholar]
  • 7.Welzel TM, Graubard BI, Zeuzem S, El-Serag HB, Davila JA, McGlynn KA. Metabolic syndrome increases the risk of primary liver cancer in the United States: a study in the SEER-Medicare database. Hepatology. 2011;54:463–471. doi: 10.1002/hep.24397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bagnardi V, Blangiardo M, La Vecchia C, Corrao G. A meta-analysis of alcohol drinking and cancer risk. Br J Cancer. 2001;85:1700–1705. doi: 10.1054/bjoc.2001.2140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.El-Serag HB, Kanwal F. Epidemiology of hepatocellular carcinoma in the United States: where are we? Where do we go? Hepatology. 2014;60:1767–1775. doi: 10.1002/hep.27222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nault J. Pathogenesis of hepatocellular carcinoma according to aetiology. Best Pract Res Clin Gastroenterol. 2014;28:937–947. doi: 10.1016/j.bpg.2014.08.006. [DOI] [PubMed] [Google Scholar]
  • 11.El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142:1264–1273. doi: 10.1053/j.gastro.2011.12.061. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Liu Y, Chang CH, Marsh GM, Wu F. Population attributable risk of aflatoxin-related liver cancer: systematic review and meta-analysis. Eur J Cancer. 2012;48:2125–2136. doi: 10.1016/j.ejca.2012.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Niederau C, Fischer R, Pürschel A, Stremmel W, Häussinger D, Strohmeyer G. Long-term survival in patients with hereditary hemochromatosis. Gastroenterology. 1996;110:1107–1119. doi: 10.1053/gast.1996.v110.pm8613000. [DOI] [PubMed] [Google Scholar]
  • 14.Smith RJ. Nutrition and metabolism in hepatocellular carcinoma. Hepatobiliary Surg Nutr. 2013;2:89–96. doi: 10.3978/j.issn.2304-3881.2012.11.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kew MC. Hepatic iron overload and hepatocellular carcinoma. Liver Cancer. 2014;3:31–40. doi: 10.1159/000343856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gordeuk VR, Boyd RD, Brittenham GM. Dietary iron overload persists in rural sub-Saharan Africa. Lancet. 1986;1(8493):1310–1313. doi: 10.1016/s0140-6736(86)91230-4. [DOI] [PubMed] [Google Scholar]
  • 17.Lai S, Chen P, Liao K, Muo C, Lin C, Sung F. Risk of hepatocellular carcinoma in diabetic patients and risk reduction associated with anti-diabetic therapy: a population-based cohort study. Am J Gastroenterol. 2012;107:46–52. doi: 10.1038/ajg.2011.384. [DOI] [PubMed] [Google Scholar]
  • 18.Bosch J, Forns X. Therapy. Statins and liver disease: from concern to ‘wonder’ drugs? Nat Rev Gastroenterol Hepatol. 2015;12:320–321. doi: 10.1038/nrgastro.2015.78. [DOI] [PubMed] [Google Scholar]
  • 19.McGlynn KA, Divine GW, Sahasrabuddhe VV, Engel LS, VanSlooten A, Wells K, Yood MU, Alford SH. Statin use and risk of hepatocellular carcinoma in a U.S. population. Cancer Epidemiol. 2014;38:523–527. doi: 10.1016/j.canep.2014.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Neuschwander-Tetri BA, Loomba R, Sanyal AJ, Lavine JE, van Natta ML, Abdelmalek MF, Chalasani N, Dasarathy S, Diehl AM, Hameed B, Kowdley KV, McCullough A, Terrault N, Clark JM, Tonascia J, Brunt EM, Kleiner DE, Doo E. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015;385:956–965. doi: 10.1016/S0140-6736(14)61933-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yang C, Shu L, Wang S, Wang J, Zhou Y, Xuan Y, Wang S. Dietary patterns modulate the risk of non-alcoholic fatty liver disease in Chinese adults. Nutrients. 2015;7:4778–4791. doi: 10.3390/nu7064778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Oddy WH, Herbison CE, Jacoby P, Ambrosini GL, O'Sullivan TA, Ayonrinde OT, Olynyk JK, Black LJ, Beilin LJ, Mori TA, Hands BP, Adams LA. The Western dietary pattern is prospectively associated with nonalcoholic fatty liver disease in adolescence. Am J Gastroenterol. 2013;108:778–785. doi: 10.1038/ajg.2013.95. [DOI] [PubMed] [Google Scholar]
  • 23.Yki-Järvinen H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome. Lancet Diabetes Endocrinol. 2014;2:901–910. doi: 10.1016/S2213-8587(14)70032-4. [DOI] [PubMed] [Google Scholar]
  • 24.Zhang W, Xiang Y, Li H, Yang G, Cai H, Ji B, Gao Y, Zheng W, Shu X. Vegetable-based dietary pattern and liver cancer risk: results from the Shanghai women's and men's health studies. Cancer Sci. 2013;104:1353–1361. doi: 10.1111/cas.12231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yang Y, Zhang D, Feng N, Chen G, Liu J, Chen G, Zhu Y. Increased intake of vegetables, but not fruit, reduces risk for hepatocellular carcinoma: a meta-analysis. Gastroenterology. 2014;147:1031–1042. doi: 10.1053/j.gastro.2014.08.005. [DOI] [PubMed] [Google Scholar]
  • 26.Li W, Park Y, McGlynn KA, Hollenbeck AR, Taylor PR, Goldstein AM, Freedman ND. Index-based dietary patterns and risk of incident hepatocellular carcinoma and mortality from chronic liver disease in a prospective study. Hepatology. 2014;60:588–597. doi: 10.1002/hep.27160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Turati F, Trichopoulos D, Polesel J, Bravi F, Rossi M, Talamini R, Franceschi S, Montella M, Trichopoulou A, La Vecchia C, Lagiou P. Mediterranean diet and hepatocellular carcinoma. J Hepatol. 2014;60:606–611. doi: 10.1016/j.jhep.2013.10.034. [DOI] [PubMed] [Google Scholar]
  • 28.Rossi M, Lipworth L, Maso LD, Talamini R, Montella M, Polesel J, McLaughlin JK, Parpinel M, Franceschi S, Lagiou P, La Vecchia C. Dietary glycemic load and hepatocellular carcinoma with or without chronic hepatitis infection. Ann Oncol. 2009;20:1736–1740. doi: 10.1093/annonc/mdp058. [DOI] [PubMed] [Google Scholar]
  • 29.Lagiou P, Rossi M, Tzonou A, Georgila C, Trichopoulos D, La Vecchia C. Glycemic load in relation to hepatocellular carcinoma among patients with chronic hepatitis infection. Ann Oncol. 2009;20:1741–1745. doi: 10.1093/annonc/mdp059. [DOI] [PubMed] [Google Scholar]
  • 30.Duarte-Salles T, Fedirko V, Stepien M, Aleksandrova K, Bamia C, Lagiou P, et al. Dietary fat, fat subtypes and hepatocellular carcinoma in a large European cohort. Int J Cancer. 2015;137:2715–2728. doi: 10.1002/ijc.29643. [DOI] [PubMed] [Google Scholar]
  • 31.Fedirko V, Trichopolou A, Bamia C, Duarte-Salles T, Trepo E, Aleksandrova K, et al. Consumption of fish and meats and risk of hepatocellular carcinoma: the European Prospective Investigation into Cancer and Nutrition (EPIC) Ann Oncol. 2013;24:2166–2173. doi: 10.1093/annonc/mdt168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fedirko V, Lukanova A, Bamia C, Trichopolou A, Trepo E, Nöthlings U, et al. Glycemic index, glycemic load, dietary carbohydrate, and dietary fiber intake and risk of liver and biliary tract cancers in Western Europeans. Ann Oncol. 2013;24:543–553. doi: 10.1093/annonc/mds434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zamora-Ros R, Fedirko V, Trichopoulou A, González CA, Bamia C, Trepo E, et al. Dietary flavonoid, lignan and antioxidant capacity and risk of hepatocellular carcinoma in the European prospective investigation into cancer and nutrition study. Int J Cancer. 2013;133:2429–2443. doi: 10.1002/ijc.28257. [DOI] [PubMed] [Google Scholar]
  • 34.Luo J, Yang Y, Liu J, Lu K, Tang Z, Liu P, Liu L, Zhu Y. Systematic review with meta-analysis: meat consumption and the risk of hepatocellular carcinoma. Aliment Pharmacol Ther. 2014;39:913–922. doi: 10.1111/apt.12678. [DOI] [PubMed] [Google Scholar]
  • 35.Prentice RL, Thomson CA, Caan B, Hubbell FA, Anderson GL, Beresford SAA, Pettinger M, Lane DS, Lessin L, Yasmeen S, Singh B, Khandekar J, Shikany JM, Satterfield S, Chlebowski RT. Low-fat dietary pattern and cancer incidence in the Women's Health Initiative Dietary Modification Randomized Controlled Trial. J Natl Cancer Inst. 2007;99:1534–1543. doi: 10.1093/jnci/djm159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Montano-Loza AJ. Clinical relevance of sarcopenia in patients with cirrhosis. World J Gastroenterol. 2014;20:8061–8071. doi: 10.3748/wjg.v20.i25.8061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hirsch S, Bunout D, La Maza P de, Iturriaga H, Petermann M, Icazar G, Gattas V, Ugarte G. Controlled trial on nutrition supplementation in outpatients with symptomatic alcoholic cirrhosis. JPEN J Parenter Enteral Nutr. 1993;17:119–124. doi: 10.1177/0148607193017002119. [DOI] [PubMed] [Google Scholar]
  • 38.Kondrup J, Müller MJ. Energy and protein requirements of patients with chronic liver disease. J Hepatol. 1997;27:239–247. doi: 10.1016/s0168-8278(97)80308-x. [DOI] [PubMed] [Google Scholar]
  • 39.Bamia C, Lagiou P, Jenab M, Trichopoulou A, Fedirko V, Aleksandrova K, et al. Coffee, tea and decaffeinated coffee in relation to hepatocellular carcinoma in a European population: multicentre, prospective cohort study. Int J Cancer. 2015;136:1899–1908. doi: 10.1002/ijc.29214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Setiawan VW, Wilkens LR, Lu SC, Hernandez BY, Le Marchand L, Henderson BE. Association of coffee intake with reduced incidence of liver cancer and death from chronic liver disease in the US multiethnic cohort. Gastroenterology. 2015;148:118–125. doi: 10.1053/j.gastro.2014.10.005. quiz e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lai GY, Weinstein SJ, Albanes D, Taylor PR, McGlynn KA, Virtamo J, Sinha R, Freedman ND. The association of coffee intake with liver cancer incidence and chronic liver disease mortality in male smokers. Br J Cancer. 2013;109:1344–1351. doi: 10.1038/bjc.2013.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Inoue M, Yoshimi I, Sobue T, Tsugane S. Influence of coffee drinking on subsequent risk of hepatocellular carcinoma: a prospective study in Japan. J Natl Cancer Inst. 2005;97:293–300. doi: 10.1093/jnci/dji040. [DOI] [PubMed] [Google Scholar]
  • 43.Petrick JL, Freedman ND, Graubard BI, Sahasrabuddhe VV, Lai GY, Alavanja MC, et al. Coffee consumption and risk of hepatocellular carcinoma and intrahepatic cholangiocarcinoma by sex: the Liver Cancer Pooling Project. Cancer Epidemiol Biomarkers Prev. 2015;24:1398–1406. doi: 10.1158/1055-9965.EPI-15-0137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bravi F, Bosetti C, Tavani A, Gallus S, La Vecchia C. Coffee reduces risk for hepatocellular carcinoma: an updated meta-analysis. Clin Gastroenterol Hepatol. 2013;11:1413–1421. doi: 10.1016/j.cgh.2013.04.039. e1. [DOI] [PubMed] [Google Scholar]
  • 45.Hayaishi S, Chung H, Kudo M, Ishikawa E, Takita M, Ueda T, Kitai S, Inoue T, Yada N, Hagiwara S, Minami Y, Ueshima K. Oral branched-chain amino acid granules reduce the incidence of hepatocellular carcinoma and improve event-free survival in patients with liver cirrhosis. Dig Dis. 2011;29:326–332. doi: 10.1159/000327571. [DOI] [PubMed] [Google Scholar]
  • 46.Kawaguchi T, Shiraishi K, Ito T, Suzuki K, Koreeda C, Ohtake T, et al. Branched-chain amino acids prevent hepatocarcinogenesis and prolong survival of patients with cirrhosis. Clin Gastroenterol Hepatol. 2014;12:1012–1018. doi: 10.1016/j.cgh.2013.08.050. e1. [DOI] [PubMed] [Google Scholar]
  • 47.Chiang K, Yeh C, Chen M, Chen TC. Hepatocellular carcinoma and vitamin D: a review. J Gastroenterol Hepatol. 2011;26:1597–1603. doi: 10.1111/j.1440-1746.2011.06892.x. [DOI] [PubMed] [Google Scholar]
  • 48.Fedirko V, Duarte-Salles T, Bamia C, Trichopoulou A, Aleksandrova K, Trichopoulos D, et al. Prediagnostic circulating vitamin D levels and risk of hepatocellular carcinoma in European populations: a nested case-control study. Hepatology. 2014;60:1222–1230. doi: 10.1002/hep.27079. [DOI] [PubMed] [Google Scholar]
  • 49.Duarte-Salles T, Fedirko V, Stepien M, Trichopoulou A, Bamia C, Lagiou P, et al. Dairy products and risk of hepatocellular carcinoma: the European Prospective Investigation into Cancer and Nutrition. Int J Cancer. 2014;135:1662–1672. doi: 10.1002/ijc.28812. [DOI] [PubMed] [Google Scholar]
  • 50.Jensen GL, Bistrian B, Roubenoff R, Heimburger DC. Malnutrition syndromes: a conundrum vs continuum. JPEN J Parenter Enteral Nutr. 2009;33:710–716. doi: 10.1177/0148607109344724. [DOI] [PubMed] [Google Scholar]
  • 51.Bozzetti F, Mariani L, Lo Vullo S, Amerio ML, Biffi R, Caccialanza G, Capuano G, Capuano G, Correja I, Cozzaglio L, Di Leo A, Di Cosmo L, Finocchiaro C, Gavazzi C, Giannoni A, Magnanini P, Mantovani G, Pellegrini M, Rovera L, Sandri G, Tinivella M, Vigevani E. The nutritional risk in oncology: a study of 1,453 cancer outpatients. Support Care Cancer. 2012;20:1919–1928. doi: 10.1007/s00520-012-1387-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Montano-Loza AJ, Duarte-Rojo A, Meza-Junco J, Baracos VE, Sawyer MB, Pang JXQ, Beaumont C, Esfandiari N, Myers RP. Inclusion of Sarcopenia within MELD (MELD-sarcopenia) and the prediction of mortality in patients with cirrhosis. Clin Transl Gastroenterol. 2015;6:e102. doi: 10.1038/ctg.2015.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Alberino F, Gatta A, Amodio P, Merkel C, Di Pascoli L, Boffo G, Caregaro L. Nutrition and survival in patients with liver cirrhosis. Nutrition. 2001;17:445–450. doi: 10.1016/s0899-9007(01)00521-4. [DOI] [PubMed] [Google Scholar]
  • 54.Hsu W, Tsai AC, Chan S, Wang P, Chung N. Mini-nutritional assessment predicts functional status and quality of life of patients with hepatocellular carcinoma in Taiwan. Nutr Cancer. 2012;64:543–549. doi: 10.1080/01635581.2012.675620. [DOI] [PubMed] [Google Scholar]
  • 55.Schütte K, Tippelt B, Schulz C, Röhl F, Feneberg A, Seidensticker R, Arend J, Malfertheiner P. Malnutrition is a prognostic factor in patients with hepatocellular carcinoma (HCC) Clin Nutr. 2014 doi: 10.1016/j.clnu.2014.11.007. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 56.Ciuni R, Biondi A, Grosso G, Nunnari G, Panascia E, Randisi L, Volpes R, Arcadipane A, Basile F, Gridelli B, Gruttadauria S. Nutritional aspects in patient undergoing liver resection. Updates Surg. 2011;63:249–252. doi: 10.1007/s13304-011-0121-4. [DOI] [PubMed] [Google Scholar]
  • 57.Morihara D, Iwata K, Hanano T, Kunimoto H, Kuno S, Fukunaga A, Yotsumoto K, Takata K, Tanaka T, Sakurai K, Iwashita H, Ueda S, Hirano G, Yokoyama K, Nakane H, Nishizawa S, Yoshikane M, Anan A, Takeyama Y, Kakumitsu S, Kitamura Y, Sakamoto M, Irie M, Shakado S, Sohda T, Watanabe H, Sakisaka S. Late-evening snack with branched-chain amino acids improves liver function after radiofrequency ablation for hepatocellular carcinoma. Hepatol Res. 2012;42:658–667. doi: 10.1111/j.1872-034X.2012.00969.x. [DOI] [PubMed] [Google Scholar]
  • 58.Nishikawa H, Osaki Y, Inuzuka T, Takeda H, Nakajima J, Matsuda F, Henmi S, Sakamoto A, Ishikawa T, Saito S, Kita R, Kimura T. Branched-chain amino acid treatment before transcatheter arterial chemoembolization for hepatocellular carcinoma. World J Gastroenterol. 2012;18:1379–1384. doi: 10.3748/wjg.v18.i12.1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ichikawa K, Okabayashi T, Maeda H, Namikawa T, Iiyama T, Sugimoto T, Kobayashi M, Mimura T, Hanazaki K. Oral supplementation of branched-chain amino acids reduces early recurrence after hepatic resection in patients with hepatocellular carcinoma: a prospective study. Surg Today. 2013;43:720–726. doi: 10.1007/s00595-012-0288-4. [DOI] [PubMed] [Google Scholar]
  • 60.Chen L, Chen Y, Wang X, Li H, Zhang H, Gong J, Shen S, Yin W, Hu H. Efficacy and safety of oral branched-chain amino acid supplementation in patients undergoing interventions for hepatocellular carcinoma: a meta-analysis. Nutr J. 2015;14:67. doi: 10.1186/s12937-015-0056-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Finkelmeier F, Kronenberger B, Köberle V, Bojunga J, Zeuzem S, Trojan J, Piiper A, Waidmann O. Severe 25-hydroxyvitamin D deficiency identifies a poor prognosis in patients with hepatocellular carcinoma - a prospective cohort study. Aliment Pharmacol Ther. 2014;39:1204–1212. doi: 10.1111/apt.12731. [DOI] [PubMed] [Google Scholar]

Articles from Gastrointestinal Tumors are provided here courtesy of Karger Publishers

RESOURCES