Hostname: page-component-76fb5796d-vfjqv Total loading time: 0 Render date: 2024-04-25T06:58:53.838Z Has data issue: false hasContentIssue false

Diet and liver cancer risk: a narrative review of epidemiological evidence

Published online by Cambridge University Press:  01 April 2020

Wan-Shui Yang
Affiliation:
Department of Nutrition, School of Public Health, Anhui Medical University, Hefei230032, People’s Republic of China
Xu-Fen Zeng
Affiliation:
Department of Nutrition, School of Public Health, Anhui Medical University, Hefei230032, People’s Republic of China
Zhi-Ning Liu
Affiliation:
Department of General Surgery, Second Affiliated Hospital of Anhui Medical University, Hefei230601, People’s Republic of China
Qi-Hong Zhao
Affiliation:
Department of Nutrition, School of Public Health, Anhui Medical University, Hefei230032, People’s Republic of China
Yu-Ting Tan
Affiliation:
State Key Laboratory of Oncogene and Related Genes and Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai200032, People’s Republic of China
Jing Gao
Affiliation:
State Key Laboratory of Oncogene and Related Genes and Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai200032, People’s Republic of China
Hong-Lan Li
Affiliation:
State Key Laboratory of Oncogene and Related Genes and Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai200032, People’s Republic of China
Yong-Bing Xiang*
Affiliation:
State Key Laboratory of Oncogene and Related Genes and Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai200032, People’s Republic of China
*
*Corresponding author: Professor Yong-Bing Xiang, email ybxiang@shsci.org
Rights & Permissions [Opens in a new window]

Abstract

Primary liver cancer is the third leading cause of cancer-related death worldwide. Most patients are diagnosed at late stages with poor prognosis; thus, identification of modifiable risk factors for primary prevention of liver cancer is urgently needed. The well-established risk factors of liver cancer include chronic infection with hepatitis B virus (HBV) or hepatitis C virus (HCV), heavy alcohol consumption, metabolic diseases such as obesity and diabetes, and aflatoxin exposure. However, a large proportion of cancer cases worldwide cannot be explained by current known risk factors. Dietary factors have been suspected as important, but dietary aetiology of liver cancer remains poorly understood. In this review, we summarised and evaluated the observational studies of diet including single nutrients, food and food groups, as well as dietary patterns with the risk of developing liver cancer. Although there are large knowledge gaps between diet and liver cancer risk, current epidemiological evidence supports an important role of diet in liver cancer development. For example, exposure to aflatoxin, heavy alcohol drinking and possibly dairy product (not including yogurt) intake increase, while intake of coffee, fish and tea, light-to-moderate alcohol drinking and several healthy dietary patterns (e.g. Alternative Healthy Eating Index) may decrease liver cancer risk. Future studies with large sample size and accurate diet measurement are warranted and need to consider issues such as the possible aetiological heterogeneity between liver cancer subtypes, the influence of chronic HBV or HCV infection, the high-risk populations (e.g. cirrhosis) and a potential interplay with host gut microbiota or genetic variations.

Type
Full Papers
Copyright
© The Authors 2020

Primary liver cancer is the third leading cause of cancer-related death worldwide(Reference Bray, Ferlay and Soerjomataram1). Hepatocellular carcinoma (HCC) is the most common (>80 %) histological type of liver cancer. There are large variations in geographical distribution of liver cancer worldwide. The disease burden is highest in areas with endemic hepatitis B virus (HBV) infection, such as in Asian countries, specifically in the East and South-East Asia, while North and South America have a relatively low incidence(Reference Petrick, Florio and Znaor2). About 72 % of all liver cancer occurs in Asia, with China accounting for 47 % of the global burden(Reference Petrick, Florio and Znaor2). Established risk factors of liver cancer include chronic infection with HBV or hepatitis C virus (HCV), excessive alcohol consumption, non-alcoholic fatty liver disease (NAFLD) and aflatoxin exposure. Beyond chronic hepatitis infections(Reference Mittal and El-Serag3Reference Yang, Gao and Li5), as shown in previous and our own studies, liver cancer risk consistently increased with obesity(Reference Yang, Petrick and Kelly6Reference Liu, Warren Andersen and Wen11) and type 2 diabetes (T2D)(Reference Campbell, Newton and Freedman10,Reference Petrick, Freedman and Demuth12,Reference Chen, Wu and Saito13) , suggesting an important role of insulinaemic and inflammatory pathways in hepatocarcinogenesis.

Currently, the population seroprevalence of HBV or HCV continues to decline, which was possibly due to large-scale HBV vaccination in newborns and common anti-HCV treatment worldwide(Reference Zhu, Shao and Chen14,Reference Thrift, El-Serag and Kanwal15) , and may contribute to the future declining rates of liver cancer in traditionally high-incident areas including East Asia(Reference Petrick, Florio and Znaor2). However, such gains in the control of liver cancer are at risk of being reversed by the rising rates of T2D, obesity and NAFLD, which were largely related to unhealthy diet(Reference Ng, Fleming and Robinson16,Reference Wang, Gao and Zhang17) . For example, in historically low-incident areas such as the USA, the liver cancer rates have tripled since the 1980s(Reference Petrick, Florio and Znaor2), and metabolic diseases are more prevalent than HBV or HCV infections. Therefore, HCC cases that are attributed to metabolic disorders (32 %) are higher than HBV (4 %) and HCV (21 %)(Reference Makarova-Rusher, Altekruse and McNeel18). Diet is a modifiable component of lifestyle that was suspected to influence liver cancer development, but dietary aetiology of liver cancer remains poorly understood. The World Cancer Research Foundation International/American Institute for Cancer Research (WCRF/AICR) concluded in their most recent Continuous Update Project that the established dietary factors for liver cancer only include aflatoxin-contaminated foods and heavy alcohol drinking, and possibly coffee consumption(19).

The liver plays a key role not only in the metabolism of carbohydrates, fats and proteins, but also in detoxification and hormone production. Thus, diet has measurable biological impacts on key pathways hypothesised to be involved in liver cancer risk. The proposed mechanisms linking diet and liver cancer development are shown in Fig. 1. For example, dietary carbohydrates are the primary determinants of the post-prandial glucose and insulin response(Reference Ludwig20), impacting circulating blood glucose, insulin demand and the bioavailability of insulin-like growth factor-1 (IGF-1)(Reference Malaguarnera and Belfiore21,Reference Shan, Shen and Liu22) . The essential branched-chain amino acids (BCAA), namely, leucine, isoleucine and valine, are essential amino acids that have known chemopreventive properties in patients with cirrhosis(Reference Muto, Sato and Watanabe23), via inhibition of hepatic IGF signalling and blockade of the phosphatidylinositol-3-kinase/protein kinase B (PI3k/Akt)/mammalian target of rapamycin (mTOR) pathway(Reference Iwasa, Shimizu and Shiraki24,Reference Alexia, Fallot and Lasfer25) . Recently, emerging evidence also showed that poor diet may cause prominent changes in gut microbiota which induce intestinal bacterial overgrowth, dysbiosis, intestinal permeability, bacterial translocation and endotoxaemia, leading to liver inflammation, chronic fibrosis, liver cirrhosis and hepatocarcinogenesis through the gut–liver axis(Reference Yu and Schwabe26,Reference Gupta, Youn and Shin27) .

Fig. 1. Proposed mechanisms linking diet and liver cancer risk. T2D, type 2 diabetes; NAFLD, non-alcoholic fatty liver disease; IGF-1, insulin-like growth factor-1.

In this review, we summarised the current epidemiological evidence of diet and the risk of incident liver cancer. Compared with a recent systematic literature review (SLR) in Continuous Update Project in 2013 by WCRF/AICR(19), we summarised and evaluated relevant studies published up to August 2019. In addition to evaluating nutrients and food or food groups(19), we further described the role of different dietary patterns in liver cancer development. We did not discuss the role of aflatoxin because the recent Continuous Update Project has determined exposure to aflatoxin as a convincing cause of liver cancer, and there are few studies(Reference Chu, Yang and Wu28,Reference Chu, Yang and Wu29) on this topic since the 2013 SLR(19).

Macro-/micronutrients and liver cancer risk

Carbohydrates

Over the past decades, sugar-sweetened beverages consumption has increased dramatically worldwide(Reference Bleich, Wang and Wang30). Although still inconclusive, this rising trend in sugar consumption, practically simple sugar (mainly fructose consumption), has been positively associated with weight gain and obesity(Reference Rebollo, Roglans and Alegret31), insulin resistance and T2D(Reference Vila, Roglans and Perna32), and NAFLD(Reference Roglans, Vila and Farre33Reference Rebollo, Roglans and Baena35). As mentioned above, insulin resistance, obesity and NAFLD may lead to the establishment of HCC. Thus, sugar-sweetened beverage consumption, mainly fructose, could be thereby linked to HCC development. This putative mechanism was supported by animal studies(Reference Laguna, Alegret and Roglans36). Likewise, food glycaemic index, glycaemic load, insulinaemic index and insulinaemic load, indicators of the glucose and insulin response to different dietary sugars, were positively associated with an increased risk of obesity(Reference Gaesser37), insulin resistance and T2D(Reference Lau, Faerch and Glumer38), liver steatosis(Reference Valtuena, Pellegrini and Ardigo39), and NAFLD(Reference Le and Bortolotti40).

However, only few observational studies have investigated the association between glycaemic index and glycaemic load (Reference Fedirko, Lukanova and Bamia41Reference Vogtmann, Li and Shu46) and dietary sugar(Reference Fedirko, Lukanova and Bamia41,Reference Vogtmann, Li and Shu46Reference Tasevska, Jiao and Cross48) in relation to liver cancer (online Supplementary Table S1). For glycaemic index and glycaemic load, the results among current epidemiological studies were inconclusive, varying from a positive(Reference George, Mayne and Leitzmann42,Reference Lagiou, Rossi and Tzonou44,Reference Rossi, Lipworth and Maso45) , null(Reference Fedirko, Lukanova and Bamia41,Reference Hu, La Vecchia and Augustin43,Reference Vogtmann, Li and Shu46) , to an inverse(Reference George, Mayne and Leitzmann42) association. Several studies(Reference George, Mayne and Leitzmann42,Reference Hu, La Vecchia and Augustin43,Reference Tasevska, Jiao and Cross48) have not considered the possible influence of chronic HBV and/or HCV infection in the analysis since they were primarily designed for cancer in general, and only two prospective studies(Reference Fedirko, Lukanova and Bamia41,Reference Rossi, Lipworth and Maso45) have adjusted for HBV and HCV infections in the analysis. In addition, the potential reverse causality and recall bias in retrospective case–control design(Reference Hu, La Vecchia and Augustin43,Reference Lagiou, Rossi and Tzonou44) may partially interpret such a discrepancy.

Similarly, there has been limited epidemiological evidence for the association between sugar-sweetened beverage intake and risk of HCC, with inconsistent results. Only two cohort studies(Reference Fedirko, Lukanova and Bamia41,Reference Vogtmann, Li and Shu46) have investigated the association between total carbohydrate consumption and liver cancer risk and yielded a null association, in which the chronic HBV and/or HCV infection have not been considered. Surprisingly, an inverse association of sucrose and fructose intake with the risk of HCC has been suggested in a case–control study nested within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort(Reference Fedirko, Lukanova and Bamia41) and in the National Institutes of Health-American Association of Retired Persons (NIH-AARP) cohort(Reference George, Mayne and Leitzmann42), respectively. However, the results could have been by chance given the limited number of liver cancer cases among the two studies. Overall, the relation has not been well defined based on current limited evidence. Given carbohydrates as a main energy source for human, future research should offer more insights on dietary carbohydrate quantity especially its quality with HCC risk, which may help individuals choose foods with ‘good’ carbohydrates. For example, in addition to glycaemic index, glycaemic load, insulinaemic index, and insulinaemic load, a carbohydrate:fibre ratio as a new measure of carbohydrate quality was shown to be more superior to other consumer-oriented methods for selecting more healthful foods(Reference Mozaffarian, Lee and Kennedy49), and a low carbohydrate:fibre ratio in diet was found to be associated with reduced risk of T2D(Reference AlEssa, Bhupathiraju and Malik50) and CHD(Reference AlEssa, Cohen and Malik51), though its association with liver cancer has not been studied.

Dietary fats/fatty acids

To date, the association between dietary fat intake and risk of HCC has not been well studied, and the existing epidemiological evidence is limited and inconclusive(Reference Kuper, Tzonou and Lagiou47,Reference Duarte-Salles, Fedirko and Stepien52Reference Yang, Sui and Ma57) . In current existing three case–control studies that were all conducted in European populations, two(Reference Kuper, Tzonou and Lagiou47,Reference Polesel, Talamini and Montella54) reported a null association between total fats and HCC risk, while another one(Reference Hadziyannis, Tabor and Kaklamani55) showed a significant positive association (OR 1·4). In a hospital-based case–control study from Italy, participants with higher intake of PUFA, but not MUFA or SFA, had a significant lower risk of HCC (OR 0·60, 95 % CI 0·40, 0·88)(Reference Polesel, Talamini and Montella54). Prospective studies include one conducted in the NIH-AARP Diet and Health cohort, which showed a statistically significant positive association (hazards ratios (HR) per 5 % increase in energy) for total (HR 1·10) and saturated fat (HR 1·38), and a null association for monounsaturated (HR 1·18) or polyunsaturated fats (HR 1·06)(Reference Freedman, Cross and McGlynn53). Likewise, a cohort study among Singapore Chinese showed similar patterns with HR (the highest v. lowest quartile) of 1·26 for total, 1·40 for saturated and 0·90 for monounsaturated fats (all P values for trend across quartiles were >0·05)(Reference Koh, Dan and Goh56). In contrast, a prospective study from the EPIC(Reference Duarte-Salles, Fedirko and Stepien52) showed a suggestive inverse HCC association for total fats (highest v. lowest quartile, HR 0·74, 95 % CI 0·45, 1·23), monounsaturated fats (HR 0·53, 95 % CI 0·27, 1·01) and polyunsaturated fats (HR 0·86, 95 % CI 0·55, 1·35).

The discrepancy among cohort studies of fat and HCC could be partly explained by different dietary sources and intake levels of fat. For example, for total dietary fat, the average intake levels for participants in the NIH-AARP cohort(Reference Freedman, Cross and McGlynn53) were lower than those in the EPIC(Reference Duarte-Salles, Fedirko and Stepien52) and higher than those from the Singapore cohort(Reference Koh, Dan and Goh56). Additionally, the main food sources of monounsaturated fat within the NIH-AARP are meat and meat products (>20 %). Unlike the NIH-AARP cohort, the main food sources of monounsaturated fat in the EPIC study were olive oil and nuts (about 20 %). The vegetable (plant) fat including fats from olive oil and nuts was shown to be associated with a lower risk of HCC.

A large number of laboratory studies indicate that n-3 PUFA possessed anti-inflammatory activity by inhibiting IL-1 and TNF synthesis(Reference Endres, Ghorbani and Kelley58,Reference James, Gibson and Cleland59) , which can contribute in HCC prevention, given that chronic inflammation plays a central role in HCC development. n-3 PUFA might exert anticancer effects also through their ability to induce apoptosis, to modulate cell cycle and eicosanoid production(Reference Fauser, Prisciandaro and Cummins60). In particular, n-3 PUFA have been shown to inhibit HCC growth in vitro through the blockage of β-catenin and cyclo-oxygenase-2(Reference Lim, Han and Dai61). Furthermore, it is observed that n-3 PUFA can improve hepatic steatosis and insulin sensitivity and reduce inflammation in patients with NAFLD(Reference Capanni, Calella and Biagini62Reference Di Minno, Russolillo and Lupoli64). Taken together, this evidence suggests a molecular mechanism of n-3 PUFA in HCC prevention. Conversely, metabolism of n-6 PUFA leads to increased levels of pro-inflammatory products, such as PG E2, thromboxane, and indirectly C-reactive protein, plasminogen activator inhibitor-1, TNF-a and IL-6(Reference Calder65,Reference Monteiro, Leslie and Moghadasian66) , which have been implicated in causing advanced fibrosis in non-alcoholic steatohepatitis, and subsequently to cirrhosis and ultimately to HCC as discussed above.

However, little is known about the relationship between dietary n-3 and n-6 PUFA and liver cancer in population studies. Only one study to date has investigated the association of n-6 PUFA with the risk of incident HCC in a cohort of 63 257 Chinese adults aged 45–74 years in Singapore and suggested a significantly positive association (HR 1·49)(Reference Koh, Dan and Goh56). This positive association was further confirmed among individuals who were free of chronic infection with HBV or HCV(Reference Koh, Dan and Goh56). Interestingly, they also found that higher n-6:n-3 PUFA ratio is associated with the increased risk of HCC, although with no association for the n-3 PUFA(Reference Koh, Dan and Goh56). Few studies have assessed the relation between n-3 PUFA and liver cancer risk, one in a Japanese cohort(Reference Sawada, Inoue and Iwasaki67) and one in a Italian hospital-based case–control study(Reference Polesel, Talamini and Montella54), both with an inverse association. Although there could be a potential beneficial role of plant fats and unsaturated fats especially n-3 PUFA in liver cancer development, no firm conclusion was reached based on current evidence. More epidemiological studies in this direction, together with considering the fat subtypes, fat food sources (animal, vegetable and dairy fat), and the possible substituting effect(Reference Song and Giovannucci68) of different fat subtypes such as substituting plant fat for animal or dairy fat, or substituting unsaturated fat for saturated fat in hepatocarcinogenesis are warranted, which may have both public health and aetiological importance.

Dietary proteins/amino acids

The three BCAA, leucine, isoleucine and valine, are among the nine essential amino acids for humans. They have been shown to affect gene expression, protein metabolism, apoptosis and regeneration of hepatocytes and insulin resistance(Reference Takami, Yamasaki and Saeki69). They have also been shown to inhibit the proliferation of liver cancer cells in vitro (Reference Sugiyama, Yu and Nagasue70). In addition, all three BCAA were found to accelerate insulin-induced vascular endothelial growth factor mRNA degradation at the post-transcriptional level, down-regulating vascular endothelial growth factor expression during the development of HCC(Reference Miuma, Ichikawa and Arima71). BCAA were also shown to induce apoptosis of liver cancer cell lines by inhibiting insulin-induced PI3K/Akt and NFκB pathways through mTORC1- and mTORC2-dependent mechanisms(Reference Hagiwara, Nishiyama and Ishizaki72). Moreover, BCAA may inhibit obesity-related hepatocarcinogenesis by suppressing the stimulatory effect of visfatin, an adipokine with a critical role in HCC proliferation(Reference Ninomiya, Shimizu and Imai73). In line with experimental studies, clinical studies showed that BCAA supplementation can help in the management of HCC(Reference Takami, Yamasaki and Saeki69), including reducing early recurrence of HCC(Reference Ichikawa, Okabayashi and Maeda74), improving quality of life(Reference Okabayashi, Iyoki and Sugimoto75,Reference Kuroda, Ushio and Miyamoto76) and liver functions(Reference Meng, Leung and Ho77) among patients undergoing liver resection for HCC, and increasing serum albumin levels during radiotherapy(Reference Lee, Seong and Bae78).

Interestingly, a recent meta-analysis of seven cohort and ten case–control studies indicated a potential protective association between BCAA and HCC risk(Reference Luo, Yang and Liu79). However, to date, there has been no epidemiological research to directly investigate the putative association between dietary BCAA intake and liver cancer risk to our knowledge.

Dietary trace elements and vitamins

In the two cohorts of Shanghai Men’s and Women’s Health Study with 132 765 Chinese adults and over 500 liver cancer cases, it suggested that dietary Mn intake was inversely associated with liver cancer risk (highest v. lowest quintile, HR 0·51, 95 % CI 0·35, 0·73; P trend = 0·001)(Reference Ma, Yang and Li80). This result was repeated in a nested case–control subset that adjusted for HBV infection(Reference Ma, Yang and Li80). However, we did not find any significant associations for folic acids or other trace elements including Se, Fe, Zn or Cu, although higher serum Cu and Cu:Zn ratio(Reference Fang, Chen and Wang81), and lower serum folate concentration(Reference Fang, Liu and Liao82) was suggested to be associated with poor prognosis among liver cancer patients. This is the first cohort design to investigate the association between trace elements and liver cancer risk, although two previous case–control studies have been conducted with limited cancer cases. One case–control study (ninety-seven cancer cases and 128 controls)(Reference Kuper, Tzonou and Lagiou47) showed a null association between trace elements and HCC, and another (185 cancer cases and 412 controls)(Reference Polesel, Talamini and Montella54) showed an inverse association for β-carotene intake and a suggestive inverse association with vitamin E intake. In the same cohorts, with 267 incident liver cancer cases, dietary vitamin E intake was inversely associated with liver cancer risk (highest v. lowest quintile, HR 0·60, 95 % CI 0·40, 0·89; P trend = 0·001)(Reference Zhang, Shu and Li83). Similar association was found for vitamin E supplement use (HR 0·52, 95 % CI 0·30, 0·90)(Reference Zhang, Shu and Li83).

In terms of vitamin A, interestingly, a case–control study in China showed a U-shaped association with HCC risk(Reference Lan, Zhang and Liao84). An intake level of 1000 μg retinol equivalent per d or more has shown beneficial association. Consistently, in another Shanghai male cohort(Reference Yuan, Gao and Ong85) and a Finnish cohort of middle-aged male smokers(Reference Lai, Weinstein and Albanes86), higher pre-diagnostic serum β-carotene and retinol levels were associated with lower risk of HCC. Although there is no observational studies investigating vitamin D intake in relation to HCC risk, a clinical cohort showed that only higher bioavailable (not bound to vitamin D-binding protein), rather than total circulating, 25-hydroxyvitamin D (25OHD) levels were associated with improved survival among HCC patients, indicating a potential utility of bioavailable 25OHD as a prognostic biomarker in HCC patients(Reference Fang, Long and Zhang87). Whether bioavailable but not total serum 25OHD is a useful biomarker in liver cancer risk prediction remains unknown.

The current experimental evidence supports the roles of several vitamins or trace elements (e.g. vitamins A, C and E, Se, Fe, Zn, Mn and Cu) as antioxidants in protection against oxidative stress (e.g. lipid peroxidation, DNA and protein damage)(Reference Wolonciej, Milewska and Roszkowska-Jakimiec88) which may generate reactive oxygen species and oxygen-free radicals, thereby relates to carcinogenesis(Reference Puertollano, Puertollano and de Cienfuegos89). Nonetheless, given the limited existing observational studies, all of which were conducted in China (high-risk country) except a cohort study in Finland(Reference Lai, Weinstein and Albanes86), and multiple comparisons among those studies(Reference Ma, Yang and Li80,Reference Zhang, Shu and Li83) , more related studies, especially studies in low-risk countries (e.g. European or Northern American countries) are warranted.

Foods or food groups and liver cancer risk

Dairy products

Previous studies suggested that high dairy product intake may increase the levels of plasma IGF-1(Reference Ma, Giovannucci and Pollak90Reference Heaney, McCarron and Dawson-Hughes93). The increased concentration of IGF-1, an important factor in the regulation of cell proliferation, differentiation, apoptosis and carcinogenesis, might contribute to the development of several cancers including HCC in experimental studies(Reference Malaguarnera and Belfiore21,Reference Hopfner, Huether and Sutter94) . In line with the evidence, two cohort studies(Reference Duarte-Salles, Fedirko and Stepien95,Reference Yang, Sui and Ma96) but not all(Reference Kuper, Tzonou and Lagiou47,Reference Fukuda, Shibata and Hirohata97Reference Park, Leitzmann and Subar101) showed that higher total dairy product intake (three serving per d or more) was associated with a statistically significant higher risk of HCC, while yogurt consumption was suggestively associated with lower HCC risk, and these associations remained within participants who were free of HBV and HCV infections (online Supplementary Table S2). Although dietary Ca, vitamin D, fat and protein from dairy sources were associated with increased HCC risk in EPIC cohort(Reference Duarte-Salles, Fedirko and Stepien95), we did not find such associations in the Nurses’ Health Study (NHS) and Health Professionals Follow-Up Study (HPFS)(Reference Yang, Sui and Ma96). Potential biologic mechanisms linking the positive associations between dairy products and HCC might include circulating IGF-1, but this needs further investigation in experimental and prospective settings. Future studies that comprehensively assess the association of dairy products according to different food groups (total dairy products, milk, cheese and yogurt) as well as their major micro- and macro-nutrient components (Ca, vitamin D, fats, lactose and protein) are needed to fully characterise the dairy products–liver cancer association.

Whole grains, fruit, vegetable and nuts

As mentioned above, we found that a vegetable-based dietary pattern was associated with a lower risk of liver cancer(Reference Zhang, Xiang and Li102). Consistently, total fibre and vegetable fibre, especially cereal fibre, were possibly associated with lower HCC risk, while fibre from fruit did not seem to be associated with HCC risk(Reference Fedirko, Lukanova and Bamia41,Reference Yang, Ma and Liu103) . These findings are less likely due to chance, since compared with fruit fibre, cereal fibre (possibly vegetable fibre) has been shown to be more consistently associated with lower risk of other health outcomes including total mortality(Reference Kim and Je104,Reference Aune, Keum and Giovannucci105) , CVD(Reference Aune, Keum and Giovannucci105,Reference Threapleton, Greenwood and Evans106) , T2D(Reference Aune, Keum and Giovannucci105,Reference Yao, Fang and Xu107) and colorectal cancer(Reference Aune, Chan and Lau108Reference Schatzkin, Mouw and Park112). The exact reasons for such a difference remain unknown. A potential explanation is that fruits, particularly fruit juice, contain sugar or added sugar (e.g. fructose and sucrose), which may cause hepatic damage and NAFLD(Reference DiNicolantonio, Subramonian and O’Keefe113), thereby masking the possible benefit of fibre intake.

Nuts are rich in unsaturated fatty acids, vegetable protein, vitamins, folate, fibre, minerals and other bioactive compounds(Reference Ros114,Reference Ros and Hu115) . Nut consumption was associated with reduced risk of NAFLD(Reference Han, Jo and Lee116). We did not find any protective association between total nut or walnut consumption and HCC risk in the two USA cohorts, although increasing intake of tree nuts was found to possibly reduce the HCC risk(Reference Sui, Yang and Ma117). These findings need to be validated due to limited HCC cases and relatively low intake levels of tree nuts in the study.

Meats

In 2015, the International Agency for Research on Cancer classified consumption of red meats as ‘probably carcinogenic to humans’ (Group 2A), while consumption of processed red meats was classified as ‘carcinogenic to humans’ (Group 1)(Reference Bouvard, Loomis and Guyton118). In addition to high levels of saturated fat and haem Fe found in meats, possible underlying mechanisms include the potentially carcinogenic chemicals N-nitroso compounds formed endogenously from nitrate or nitrite during meat processing or preservation, and heterocyclic amines (HCA) during meat cooking. By contrast, white meats (i.e. poultry and fish), particularly fish intake, have been shown to decrease cancer risk(Reference Luo, Yang and Liu79,Reference Huang, Duan and Hu119) , possibly due to long-chain n-3 PUFA present in fish, particularly fatty fish. Also, white meat including fish and poultry is a rich source of BCAA, which may play a protective role in hepatocarcinogenesis (see Section 1.3).

There have been few cohort studies of the association between red meat intake and HCC risk(Reference Freedman, Cross and McGlynn53,Reference Fedirko, Trichopolou and Bamia120Reference Ma, Yang and Li122) . For unprocessed red meat, two USA cohort studies, the NHS and HPFS(Reference Ma, Yang and Li122) as well as a European cohort study, the EPIC(Reference Fedirko, Trichopolou and Bamia120) did not observe an association between unprocessed red meat intake and HCC, which was consistent with findings from a recent meta-analysis(Reference Luo, Yang and Liu79), although another USA cohort, the NIH-AARP(Reference Freedman, Cross and McGlynn53), showed a suggestive positive association for unprocessed red meat. A non-significant association for unprocessed red meat with HCC was also seen in the Japanese Ministry of Education cohort(Reference Kurozawa, Ogimoto and Shibata121), showing no significant association between beef or pork intake and HCC mortality without adjustment for any risk factors(Reference Kurozawa, Ogimoto and Shibata121). Most current studies seemed to support a null association for processed meat(Reference Freedman, Cross and McGlynn53,Reference Luo, Yang and Liu79,Reference Fedirko, Trichopolou and Bamia120,Reference Kurozawa, Ogimoto and Shibata121) , or heterocyclic amines(Reference Freedman, Cross and McGlynn53), although a positive association between processed red meat and HCC risk was found in the NHS and HPFS(Reference Ma, Yang and Li122). Notably, cohort studies(Reference Freedman, Cross and McGlynn53,Reference Fedirko, Trichopolou and Bamia120,Reference Ma, Yang and Li122) and the meta-analyses(Reference Luo, Yang and Liu79,Reference Huang, Duan and Hu119) consistently suggested a protective association of white meat intake, particularly fish intake with HCC development, which was consistent with the findings form the most recent report by WCRF/AICR(19). Overall, intake of red meat particularly processed red meat may increase, while white meat possibly fish may decrease the risk of HCC. These findings need to be validated in more studies, ideally further considering the preparation or cooking methods of meat as well as the meat-derived mutagenicity or heterocyclic amines in liver cancer development.

Soya

Evidence showed that soya isoflavones may prevent liver cancer both in vitro and in vivo (Reference Su, Chow and Kung123). Compared with those in Europe and North America, Asia has one of the world’s highest intake levels of soya foods. There have been few observational studies of soya or soya isoflavones and liver cancer risk with inconclusive findings(Reference Zhang, Shu and Li83,Reference Sharp, Lagarde and Mizuno124Reference Zamora-Ros, Fedirko and Trichopoulou127) . In a Japanese nested case–control study (176 HCC cases and 560 controls among Japanese A-bomb survivors), consumption of miso soup and tofu was significantly inversely associated with HCC risk accounting for HBV and HCV infections(Reference Sharp, Lagarde and Mizuno124). This finding was consistent with our cohort study in China showing that increasing soya intake may decrease liver cancer risk(Reference Zhang, Shu and Li83). On the contrary, soya isoflavones (genistein or daidzein) showed a positive association with HCC risk in women and showed a null association in men in another Japanese cohort (101 HCC cases), which also carefully considered the HBV and HCV infections(Reference Kurahashi, Inoue and Iwasaki125). There have been two studies of dietary flavonoids and HCC risk in non-Asian countries with relatively low intake levels of soya(Reference Lagiou, Rossi and Lagiou126,Reference Zamora-Ros, Fedirko and Trichopoulou127) . One is a hospital-based case–control study in Greece(Reference Lagiou, Rossi and Lagiou126) showing a null association between dietary flavonoids from fresh beans and whole-grain bread and HCC risk; another is a prospective study in Europe(Reference Zamora-Ros, Fedirko and Trichopoulou127) showing a non-significant association between isoflavone intake (about 40 % from soya foods) and HCC risk, although an inverse association between total flavonoid intake and HCC risk was observed in the two studies(Reference Lagiou, Rossi and Lagiou126,Reference Zamora-Ros, Fedirko and Trichopoulou127) . These contradict results would be partly interpreted by challenge in assessing soya food intake.

Recently, in a case–control study nested within the Shanghai Men’s Health Study and Shanghai Women’s Health Study, we found that urinary excretion of genistein (a metabolite of soya flavonoids) was associated with lower risk of liver cancer in women, but not in men(Reference Zhang, Wang and Gao128). Future epidemiological study using ‘omics’ technology (e.g. metabolomics) to develop a metabolic signature reflecting soya intake (internal exposure) is needed. This approach using high-throughput metabolomics profiling to measure intermediate molecules and products of metabolism; metabolomic profiles reflects individuals’ dietary intakes and other sources of variability in metabolism (e.g. genetic variations(Reference Illig, Gieger and Zhai129) and diet–gut microbiome interplay(Reference Shoaie, Ghaffari and Kovatcheva-Datchary130)), thereby objectively assesses the diet and metabolic responses to diet intake(Reference Guertin, Moore and Sampson131Reference Guasch-Ferré, Bhupathiraju and Hu133).

Beverages

Tea

Tea is one of the most popular beverages consumed worldwide especially in China. Animal and in vitro studies showed a protective effect against cancer initiation and its subsequent development, which is largely owing to the polyphenolic antioxidants present in tea(Reference Yang, Wang and Lu134). We previously conducted a meta-analysis of six case–control and seven cohort studies and found a suggestive inverse association between total tea drinking and liver cancer risk (relative risk 0·77, 95 % CI 0·57, 1·03) and a significant inverse association for green tea (relative risk 0·79, 95 % CI 0·68, 0·93)(Reference Fon Sing, Yang and Gao135). After that, observational studies of tea drinking(Reference Bamia, Lagiou and Jenab136Reference Tamura, Wada and Konishi138) consistently showed a protective association with HCC risk. Although these findings were not supported by a nested case–control study (211 cases and 1067 matched controls in Shanghai cohort)(Reference Butler, Huang and Wang139) showing that higher urinary levels of catechins (partly reflect green tea drinking) may not decrease but increase risk of HCC development (online Supplementary Table S3). Future prospective studies that account for different characteristics of tea such as tea types, preparation methods and tea strength are needed to fully characterise such an association, ideally with useful biomarkers that can better reflect tea consumption in a subset of participants to confirm the consistency and to adjust for measurement error if possible.

Coffee and alcohol

The evidence that coffee protects against liver cancer has been determined as ‘probable’ but not ‘convincing’ by WCRF/AICR in their 2013 SLR(19). After 2013 SLR, a total of sixteen relevant studies including eight meta-analyses(Reference Bravi, Bosetti and Tavani140Reference Tamura, Hishida and Wakai147) and eight cohort studies(Reference Bamia, Lagiou and Jenab136,Reference Tamura, Wada and Konishi138,Reference Jang, Jeong and Lee148Reference Loftfield, Rothwell and Sinha153) were published. All above studies support an inverse association between coffee and liver cancer risk. Current meta-analyses(Reference Bravi, Bosetti and Tavani140,Reference Bravi, Tavani and Bosetti144,Reference Godos, Micek and Marranzano145) consistently showed that increasing coffee consumption by one cup/d was associated with a 15–20 % risk reduction of HCC. The protective association between regular coffee intake and liver cancer risk appeared stronger in men, in Asian populations, and in case–control studies compared with those in women, in non-Asian populations, and in cohort studies, respectively. Compared with caffeinated coffee, decaffeinated coffee showed a weaker non-significant inverse association with liver cancer risk(Reference Kennedy, Roderick and Buchanan146). For cancer subtypes, there seemed no association between coffee consumption and biliary tract cancers including intrahepatic cholangiocarcinoma (ICC)(Reference Godos, Micek and Marranzano145). The above differential associations according to sex, type of coffee consumed (caffeinated v. decaffeinated coffee) and liver cancer subtype (HCC v. ICC) were replicated in the Liver Cancer Pooling Project, a consortium of over ten US-based cohorts with 860 HCC cases and 260 ICC cases(Reference Petrick, Freedman and Graubard151). Further, the inverse association between coffee and HCC risk was consistent across strata of HBV and HCV infection status without significant heterogeneity(Reference Bamia, Lagiou and Jenab136,Reference Aleksandrova, Bamia and Drogan150) . Overall, these results strengthen the existing evidence regarding the inverse association between coffee and HCC risk. Future research in this direction needs to offer more mechanistic insights, although two recent large prospective studies suggested that the possible pathways linking coffee-HCC association include reduction of inflammation and hepatocellular injury(Reference Aleksandrova, Bamia and Drogan150) and regulation of metabolism of bile acids and glycerophospholipids(Reference Loftfield, Rothwell and Sinha153).

Heavy alcohol intake (i.e. >45 g/d, above about 3 drinks/d) has been determined as a convincing cause of liver cancer by WCRF/AICR(19), which was further confirmed by the following cohort studies(Reference Loomba, Yang and Su154,Reference Petrick, Campbell and Koshiol155) and meta-analyses(Reference Turati, Galeone and Rota156,Reference Chuang, Lee and Wu157) . However, whether light-to-moderate alcohol consumption (e.g. <3 drinks/d) is associated with liver cancer is unanswered in the 2013 SLR(19). Current epidemiological evidence including a meta-analysis of nineteen cohort studies(Reference Turati, Galeone and Rota156) and a US cohort consortium study(Reference Petrick, Campbell and Koshiol155) supports an inverse association of light-to-moderate alcohol drinking with HCC risk, but not with ICC. The moderate alcohol intake is associated with a decreased risk of T2D, possibly via increasing insulin sensitivity(Reference Schrieks, Heil and Hendriks158). Thus, light-to-moderate alcohol drinking may reduce the HCC risk through decreasing the risk of T2D. However, due to limited evidence, more studies are warranted.

Dietary pattern and liver cancer risk

Diets are complex combinations of nutrients and other compounds that act synergistically within individual foods and across food combinations(Reference Tapsell, Neale and Satija159). Thus, evaluating the association of dietary pattern with health outcomes may capture dietary effects on health more completely. Also, findings from such research might provide important data to the development of dietary guidelines. There are four studies(Reference Zhang, Xiang and Li102,Reference Chen, Fang and Wang160Reference Ma, Yang and Simon162) , two in China(Reference Zhang, Xiang and Li102,Reference Chen, Fang and Wang160) and two in the USA(Reference Li, Park and McGlynn161,Reference Ma, Yang and Simon162) , investigating the associations between certain dietary patterns and the risk of incident liver cancer to date (online Supplementary Table S4). Our study in the Shanghai Men’s Health Study and Shanghai Women’s Health Study(Reference Zhang, Xiang and Li102) showed that a vegetable-based dietary pattern was associated with a lower risk of liver cancer. Two cohort studies in the USA(Reference Li, Park and McGlynn161,Reference Ma, Yang and Simon162) suggested that participants with higher 2010 Alternative Healthy Eating Index (2010) and Alternate Mediterranean Diet scores had a lower risk of incident HCC(Reference Li, Park and McGlynn161). Consistently, one case–control study showed an association of higher adherence to Healthy Eating Index-2015 with lower HCC risk among Chinese adults(Reference Chen, Fang and Wang160). These healthy dietary patterns share several common dietary components (such as high intake of fruits and vegetables, nuts and legumes, and whole grains and low intake of red and processed meats) and showed moderate correlation between one another(Reference Li, Park and McGlynn161,Reference Ma, Yang and Simon162) , although they were not developed specifically for cancer prevention.

To strengthen the scientific evidence based on dietary patterns and to inform the Dietary Guidelines for Americans, the National Cancer Institute has initiated the Dietary Patterns Methods Project(163). This project aimed to compare key diet quality indices and their associations with cancer and CVD. These indices were selected based on a comprehensive review of literature, including the Alternative Healthy Eating Index, the Healthy Eating Index, the Dietary Approaches to Stop Hypertension and the Alternate Mediterranean Diet. However, little is known about the association between compliance to other country-specific dietary guidelines or recommendations and health outcomes including liver cancer development. Given large variations in diet and eating patterns across countries, this issue is of great important. In China, for example, only one study has evaluated the association of the 2016 Dietary Guidelines for Chinese with the risk of liver cancer(Reference Chen, Fang and Wang160). Future studies are needed to address this issue. In addition, given a potential mediated role of inflammation, insulin resistance, and gut microbiota (Fig. 1) in diet-liver cancer association, investigating the dietary pattern based on its inflammatory or insulinaemic potential (e.g. empirical dietary inflammatory index(Reference Tabung, Smith-Warner and Chavarro164) and empirical dietary index for hyperinsulinaemia(Reference Tabung, Wang and Fung165)) in relation to liver cancer risk would elucidate biological mechanisms relating diet to liver cancer.

Conclusion

In summary, current epidemiological evidence supports an important role of diet in liver cancer development, although there are large knowledge gaps between diet and liver cancer risk. Future nutritional epidemiological studies of diet and liver cancer with large sample size are warranted and need to consider: (1) how to best measure diet; (2) the possible aetiological heterogeneity between liver cancer subtypes (HCC v. ICC); (3) the potential influence of chronic HBV or HCV infection; (4) high-risk populations (e.g. patients with NAFLD or cirrhosis) and (5) a possible interplay with host gut microbiota or genetic variations. In addition, the underlying mechanisms linking diet and liver cancer remain to be further elucidated.

Acknowledgements

This work was supported by grants of the Scientific Research of BSKY (XJ201935) from Anhui Medical University, the National Key Project of Research and Development Program of China (2016YFC1302503) and the National Key Basic Research Program ‘973 Project’ (2015CB554000).

None of the funders had any influence on the study design; in the collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.

Y.-B. X. and W.-S. Y. obtained the funding. W.-S. Y. wrote the initial draft of the manuscript. All authors critically reviewed the manuscript.

The authors declare that there are no conflicts of interest.

Supplementary material

For supplementary material referred to in this article, please visit https://doi.org/10.1017/S0007114520001208

References

Bray, F, Ferlay, J, Soerjomataram, I, et al. (2018) Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68, 394424.10.3322/caac.21492CrossRefGoogle ScholarPubMed
Petrick, JL, Florio, AA, Znaor, A, et al. (2019) International trends in hepatocellular carcinoma incidence, 1978–2012. Int J Cancer (epublication ahead of print version 9 October 2019).Google ScholarPubMed
Mittal, S & El-Serag, HB (2013) Epidemiology of hepatocellular carcinoma: consider the population. J Clin Gastroenterol 47, S2S6.10.1097/MCG.0b013e3182872f29CrossRefGoogle ScholarPubMed
Davis, GL, Dempster, J, Meler, JD, et al. (2008) Hepatocellular carcinoma: management of an increasingly common problem. Proc (Bayl Univ Med Cent) 21, 266280.10.1080/08998280.2008.11928410CrossRefGoogle ScholarPubMed
Yang, Y, Gao, J, Li, HL, et al. (2016) Dose–response association between hepatitis B surface antigen levels and liver cancer risk in Chinese men and women. Int J Cancer 139, 355362.10.1002/ijc.30086CrossRefGoogle ScholarPubMed
Yang, B, Petrick, JL, Kelly, SP, et al. (2017) Adiposity across the adult life course and incidence of primary liver cancer: the NIH-AARP cohort. Int J Cancer 141, 271278.10.1002/ijc.30737CrossRefGoogle ScholarPubMed
Aleksandrova, K, Stelmach-Mardas, M & Schlesinger, S (2016) Obesity and liver cancer. Recent Results Cancer Res 208, 177198.10.1007/978-3-319-42542-9_10CrossRefGoogle ScholarPubMed
Sun, B & Karin, M (2012) Obesity, inflammation, and liver cancer. J Hepatol 56, 704713.10.1016/j.jhep.2011.09.020CrossRefGoogle ScholarPubMed
Qian, Y & Fan, JG (2005) Obesity, fatty liver and liver cancer. Hepatobiliary Pancreat Dis Int 4, 173177.Google ScholarPubMed
Campbell, PT, Newton, CC, Freedman, ND, et al. (2016) Body mass index, waist circumference, diabetes, and risk of liver cancer for U.S. adults. Cancer Res 76, 60766083.10.1158/0008-5472.CAN-16-0787CrossRefGoogle ScholarPubMed
Liu, Y, Warren Andersen, S, Wen, W, et al. (2016) Prospective cohort study of general and central obesity, weight change trajectory and risk of major cancers among Chinese women. Int J Cancer 139, 14611470.10.1002/ijc.30187CrossRefGoogle ScholarPubMed
Petrick, JL, Freedman, ND, Demuth, J, et al. (2016) Obesity, diabetes, serum glucose, and risk of primary liver cancer by birth cohort, race/ethnicity, and sex: multiphasic health checkup study. Cancer Epidemiol 42, 140146.10.1016/j.canep.2016.04.009CrossRefGoogle Scholar
Chen, Y, Wu, F, Saito, E, et al. (2017) Association between type 2 diabetes and risk of cancer mortality: a pooled analysis of over 771,000 individuals in the Asia Cohort Consortium. Diabetologia 60, 10221032.10.1007/s00125-017-4229-zCrossRefGoogle ScholarPubMed
Zhu, Q, Shao, X, Chen, S, et al. (2018) Epidemiological serosurvey of hepatitis B virus among children aged 1–14 years in Guangdong Province, China. Int J Infect Dis 71, 2529.10.1016/j.ijid.2018.01.027CrossRefGoogle ScholarPubMed
Thrift, AP, El-Serag, HB & Kanwal, F (2017) Global epidemiology and burden of HCV infection and HCV-related disease. Nat Rev Gastroenterol Hepatol 14, 122132.10.1038/nrgastro.2016.176CrossRefGoogle ScholarPubMed
Ng, M, Fleming, T, Robinson, M, et al. (2014) Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766781.10.1016/S0140-6736(14)60460-8CrossRefGoogle ScholarPubMed
Wang, L, Gao, P, Zhang, M, et al. (2017) Prevalence and ethnic pattern of diabetes and prediabetes in China in 2013. JAMA 317, 25152523.10.1001/jama.2017.7596CrossRefGoogle ScholarPubMed
Makarova-Rusher, OV, Altekruse, SF, McNeel, TS, et al. (2016) Population attributable fractions of risk factors for hepatocellular carcinoma in the United States. Cancer 122, 17571765.10.1002/cncr.29971CrossRefGoogle ScholarPubMed
World Cancer Research Fund International/American Institute for Cancer Research (2018) Diet, Nutrition, Physical Activity and Liver Cancer, Continuous Update Project Expert Report 2018. London: World Cancer Research Fund International/American Institute for Cancer Research.Google Scholar
Ludwig, DS (2002) The glycemic index: physiological mechanisms relating to obesity, diabetes, and cardiovascular disease. Jama 287, 24142423.10.1001/jama.287.18.2414CrossRefGoogle Scholar
Malaguarnera, R & Belfiore, A (2014) The emerging role of insulin and insulin-like growth factor signaling in cancer stem cells. Front Endocrinol 5, 10.CrossRefGoogle ScholarPubMed
Shan, J, Shen, J, Liu, L, et al. (2012) Nanog regulates self-renewal of cancer stem cells through the insulin-like growth factor pathway in human hepatocellular carcinoma. Hepatology 56, 10041014.CrossRefGoogle ScholarPubMed
Muto, Y, Sato, S, Watanabe, A, et al. (2006) Overweight and obesity increase the risk for liver cancer in patients with liver cirrhosis and long-term oral supplementation with branched-chain amino acid granules inhibits liver carcinogenesis in heavier patients with liver cirrhosis. Hepatol Res 35, 204214.Google ScholarPubMed
Iwasa, J, Shimizu, M, Shiraki, M, et al. (2010) Dietary supplementation with branched-chain amino acids suppresses diethylnitrosamine-induced liver tumorigenesis in obese and diabetic C57BL/KsJ-db/db mice. Cancer Sci 101, 460467.CrossRefGoogle ScholarPubMed
Alexia, C, Fallot, G, Lasfer, M, et al. (2004) An evaluation of the role of insulin-like growth factors (IGF) and of type-I IGF receptor signalling in hepatocarcinogenesis and in the resistance of hepatocarcinoma cells against drug-induced apoptosis. Biochem Pharmacol 68, 10031015.CrossRefGoogle ScholarPubMed
Yu, LX & Schwabe, RF (2017) The gut microbiome and liver cancer: mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol 14, 527539.10.1038/nrgastro.2017.72CrossRefGoogle ScholarPubMed
Gupta, H, Youn, GS, Shin, MJ, et al. (2019) Role of gut microbiota in hepatocarcinogenesis. Microorganisms 7, E121.CrossRefGoogle ScholarPubMed
Chu, YJ, Yang, HI, Wu, HC, et al. (2017) Aflatoxin B1 exposure increases the risk of cirrhosis and hepatocellular carcinoma in chronic hepatitis B virus carriers. Int J Cancer 141, 711720.CrossRefGoogle ScholarPubMed
Chu, YJ, Yang, HI, Wu, HC, et al. (2018) Aflatoxin B1 exposure increases the risk of hepatocellular carcinoma associated with hepatitis C virus infection or alcohol consumption. Eur J Cancer 94, 3746.10.1016/j.ejca.2018.02.010CrossRefGoogle ScholarPubMed
Bleich, SN, Wang, YC, Wang, Y, et al. (2009) Increasing consumption of sugar-sweetened beverages among US adults: 1988–1994 to 1999–2004. Am J Clin Nutr 89, 372381.10.3945/ajcn.2008.26883CrossRefGoogle ScholarPubMed
Rebollo, A, Roglans, N, Alegret, M, et al. (2012) Way back for fructose and liver metabolism: bench side to molecular insights. World J Gastroenterol 18, 65526559.CrossRefGoogle ScholarPubMed
Vila, L, Roglans, N, Perna, V, et al. (2011) Liver AMP/ATP ratio and fructokinase expression are related to gender differences in AMPK activity and glucose intolerance in rats ingesting liquid fructose. J Nutr Biochem 22, 741751.10.1016/j.jnutbio.2010.06.005CrossRefGoogle ScholarPubMed
Roglans, N, Vila, L, Farre, M, et al. (2007) Impairment of hepatic Stat-3 activation and reduction of PPARalpha activity in fructose-fed rats. Hepatology 45, 778788.10.1002/hep.21499CrossRefGoogle ScholarPubMed
Li, JM, Li, YC, Kong, LD, et al. (2010) Curcumin inhibits hepatic protein-tyrosine phosphatase 1B and prevents hypertriglyceridemia and hepatic steatosis in fructose-fed rats. Hepatology 51, 15551566.10.1002/hep.23524CrossRefGoogle ScholarPubMed
Rebollo, A, Roglans, N, Baena, M, et al. (2014) Liquid fructose downregulates Sirt1 expression and activity and impairs the oxidation of fatty acids in rat and human liver cells. Biochim Biophys Acta 1841, 514524.10.1016/j.bbalip.2014.01.002CrossRefGoogle ScholarPubMed
Laguna, JC, Alegret, M & Roglans, N (2014) Simple sugar intake and hepatocellular carcinoma: epidemiological and mechanistic insight. Nutrients 6, 59335954.10.3390/nu6125933CrossRefGoogle ScholarPubMed
Gaesser, GA (2007) Carbohydrate quantity and quality in relation to body mass index. J Am Diet Assoc 107, 17681780.10.1016/j.jada.2007.07.011CrossRefGoogle ScholarPubMed
Lau, C, Faerch, K, Glumer, C, et al. (2005) Dietary glycemic index, glycemic load, fiber, simple sugars, and insulin resistance: the Inter99 study. Diabet Care 28, 13971403.CrossRefGoogle ScholarPubMed
Valtuena, S, Pellegrini, N, Ardigo, D, et al. (2006) Dietary glycemic index and liver steatosis. Am J Clin Nutr 84, 136142; quiz 268–139.CrossRefGoogle ScholarPubMed
Le, KA & Bortolotti, M (2008) Role of dietary carbohydrates and macronutrients in the pathogenesis of nonalcoholic fatty liver disease. Curr Opin Clin Nutr Metab Care 11, 477482.10.1097/MCO.0b013e328302f3ecCrossRefGoogle ScholarPubMed
Fedirko, V, Lukanova, A, Bamia, C, et al. (2013) Glycemic index, glycemic load, dietary carbohydrate, and dietary fiber intake and risk of liver and biliary tract cancers in Western Europeans. Ann Oncol 24, 543553.CrossRefGoogle ScholarPubMed
George, SM, Mayne, ST, Leitzmann, MF, et al. (2009) Dietary glycemic index, glycemic load, and risk of cancer: a prospective cohort study. Am J Epidemiol 169, 462472.CrossRefGoogle ScholarPubMed
Hu, J, La Vecchia, C, Augustin, LS, et al. (2013) Glycemic index, glycemic load and cancer risk. Ann Oncol 24, 245251.10.1093/annonc/mds235CrossRefGoogle ScholarPubMed
Lagiou, P, Rossi, M, Tzonou, A, et al. (2009) Glycemic load in relation to hepatocellular carcinoma among patients with chronic hepatitis infection. Ann Oncol 20, 17411745.CrossRefGoogle ScholarPubMed
Rossi, M, Lipworth, L, Maso, LD, et al. (2009) Dietary glycemic load and hepatocellular carcinoma with or without chronic hepatitis infection. Ann Oncol 20, 17361740.CrossRefGoogle ScholarPubMed
Vogtmann, E, Li, HL, Shu, XO, et al. (2013) Dietary glycemic load, glycemic index, and carbohydrates on the risk of primary liver cancer among Chinese women and men. Ann Oncol 24, 238244.10.1093/annonc/mds287CrossRefGoogle ScholarPubMed
Kuper, H, Tzonou, A, Lagiou, P, et al. (2000) Diet and hepatocellular carcinoma: a case–control study in Greece. Nutr Cancer 38, 612.10.1207/S15327914NC381_2CrossRefGoogle Scholar
Tasevska, N, Jiao, L, Cross, AJ, et al. (2012) Sugars in diet and risk of cancer in the NIH-AARP diet and health study. Int J Cancer 130, 159169.10.1002/ijc.25990CrossRefGoogle ScholarPubMed
Mozaffarian, RS, Lee, RM, Kennedy, MA, et al. (2013) Identifying whole grain foods: a comparison of different approaches for selecting more healthful whole grain products. Public Health Nutr 16, 22552264.CrossRefGoogle ScholarPubMed
AlEssa, HB, Bhupathiraju, SN, Malik, VS, et al. (2015) Carbohydrate quality and quantity and risk of type 2 diabetes in US women. Am J Clin Nutr 102, 15431553.10.3945/ajcn.115.116558CrossRefGoogle Scholar
AlEssa, HB, Cohen, R, Malik, VS, et al. (2018) Carbohydrate quality and quantity and risk of coronary heart disease among US women and men. Am J Clin Nutr 107, 257267.10.1093/ajcn/nqx060CrossRefGoogle Scholar
Duarte-Salles, T, Fedirko, V, Stepien, M, et al. (2015) Dietary fat, fat subtypes and hepatocellular carcinoma in a large European cohort. Int J Cancer 137, 27152728.10.1002/ijc.29643CrossRefGoogle Scholar
Freedman, ND, Cross, AJ, McGlynn, KA, et al. (2010) Association of meat and fat intake with liver disease and hepatocellular carcinoma in the NIH-AARP cohort. J Natl Cancer Inst 102, 13541365.10.1093/jnci/djq301CrossRefGoogle ScholarPubMed
Polesel, J, Talamini, R, Montella, M, et al. (2007) Nutrients intake and the risk of hepatocellular carcinoma in Italy. Eur J Cancer 43, 23812387.10.1016/j.ejca.2007.07.012CrossRefGoogle ScholarPubMed
Hadziyannis, S, Tabor, E, Kaklamani, E, et al. (1995) A case–control study of hepatitis B and C virus infections in the etiology of hepatocellular carcinoma. Int J Cancer 60, 627631.CrossRefGoogle Scholar
Koh, WP, Dan, YY, Goh, GB, et al. (2016) Dietary fatty acids and risk of hepatocellular carcinoma in the Singapore Chinese health study. Liver Int 36, 893901.10.1111/liv.12978CrossRefGoogle ScholarPubMed
Yang, W, Sui, J, Ma, Y, et al. (2020) High dietary intake of vegetable or polyunsaturated fats is associated with reduced risk of hepatocellular carcinoma. Clin Gastroenterol Hepatol (epublication ahead of print version 9 January 2020).10.1016/j.cgh.2020.01.003CrossRefGoogle ScholarPubMed
Endres, S, Ghorbani, R, Kelley, VE, et al. (1989) The effect of dietary supplementation with n-3 polyunsaturated fatty acids on the synthesis of interleukin-1 and tumor necrosis factor by mononuclear cells. N Engl J Med 320, 265271.CrossRefGoogle ScholarPubMed
James, MJ, Gibson, RA & Cleland, LG (2000) Dietary polyunsaturated fatty acids and inflammatory mediator production. Am J Clin Nutr 71, 343S348S.CrossRefGoogle ScholarPubMed
Fauser, JK, Prisciandaro, LD, Cummins, AG, et al. (2011) Fatty acids as potential adjunctive colorectal chemotherapeutic agents. Cancer Biol Therapy 11, 724731.CrossRefGoogle ScholarPubMed
Lim, K, Han, C, Dai, Y, et al. (2009) Omega-3 polyunsaturated fatty acids inhibit hepatocellular carcinoma cell growth through blocking beta-catenin and cyclooxygenase-2. Mol Cancer Therapeut 8, 30463055.CrossRefGoogle ScholarPubMed
Capanni, M, Calella, F, Biagini, MR, et al. (2006) Prolonged n-3 polyunsaturated fatty acid supplementation ameliorates hepatic steatosis in patients with non-alcoholic fatty liver disease: a pilot study. Aliment Pharm Ther 23, 11431151.10.1111/j.1365-2036.2006.02885.xCrossRefGoogle ScholarPubMed
de Castro, GS & Calder, PC (2017) Non-alcoholic fatty liver disease and its treatment with n-3 polyunsaturated fatty acids. Clin Nutr 37, 3755.CrossRefGoogle ScholarPubMed
Di Minno, MN, Russolillo, A, Lupoli, R, et al. (2012) Omega-3 fatty acids for the treatment of non-alcoholic fatty liver disease. World J Gastroenterol 18, 58395847.CrossRefGoogle ScholarPubMed
Calder, PC (2006) Polyunsaturated fatty acids and inflammation. Prostagl Leukotr Ess Fatty Acids 75, 197202.CrossRefGoogle ScholarPubMed
Monteiro, J, Leslie, M, Moghadasian, MH, et al. (2014) The role of n-6 and n-3 polyunsaturated fatty acids in the manifestation of the metabolic syndrome in cardiovascular disease and non-alcoholic fatty liver disease. Food Funct 5, 426435.CrossRefGoogle ScholarPubMed
Sawada, N, Inoue, M, Iwasaki, M, et al. (2012) Consumption of n-3 fatty acids and fish reduces risk of hepatocellular carcinoma. Gastroenterology 142, 14681475.10.1053/j.gastro.2012.02.018CrossRefGoogle ScholarPubMed
Song, M & Giovannucci, E (2018) Substitution analysis in nutritional epidemiology: proceed with caution. Eur J Epidemiol 33, 137140.CrossRefGoogle Scholar
Takami, T, Yamasaki, T, Saeki, I, et al. (2016) Supportive therapies for prevention of hepatocellular carcinoma recurrence and preservation of liver function. World J Gastroenterol 22, 72527263.CrossRefGoogle ScholarPubMed
Sugiyama, K, Yu, L & Nagasue, N (1998) Direct effect of branched-chain amino acids on the growth and metabolism of cultured human hepatocellular carcinoma cells. Nutr Cancer 31, 6268.CrossRefGoogle ScholarPubMed
Miuma, S, Ichikawa, T, Arima, K, et al. (2012) Branched-chain amino acid deficiency stabilizes insulin-induced vascular endothelial growth factor mRNA in hepatocellular carcinoma cells. J Cell Biochem 113, 31133121.CrossRefGoogle ScholarPubMed
Hagiwara, A, Nishiyama, M & Ishizaki, S (2012) Branched-chain amino acids prevent insulin-induced hepatic tumor cell proliferation by inducing apoptosis through mTORC1 and mTORC2-dependent mechanisms. J Cell Physiol 227, 20972105.CrossRefGoogle ScholarPubMed
Ninomiya, S, Shimizu, M, Imai, K, et al. (2011) Possible role of visfatin in hepatoma progression and the effects of branched-chain amino acids on visfatin-induced proliferation in human hepatoma cells. Cancer Prevent Res 4, 20922100.10.1158/1940-6207.CAPR-11-0340CrossRefGoogle ScholarPubMed
Ichikawa, K, Okabayashi, T, Maeda, H, et al. (2013) Oral supplementation of branched-chain amino acids reduces early recurrence after hepatic resection in patients with hepatocellular carcinoma: a prospective study. Surg Today 43, 720726.CrossRefGoogle ScholarPubMed
Okabayashi, T, Iyoki, M, Sugimoto, T, et al. (2011) Oral supplementation with carbohydrate- and branched-chain amino acid-enriched nutrients improves postoperative quality of life in patients undergoing hepatic resection. Amino Acids 40, 12131220.CrossRefGoogle ScholarPubMed
Kuroda, H, Ushio, A, Miyamoto, Y, et al. (2010) Effects of branched-chain amino acid-enriched nutrient for patients with hepatocellular carcinoma following radiofrequency ablation: a one-year prospective trial. J Gastroenterol Hepatol 25, 15501555.CrossRefGoogle ScholarPubMed
Meng, WC, Leung, KL, Ho, RL, et al. (1999) Prospective randomized control study on the effect of branched-chain amino acids in patients with liver resection for hepatocellular carcinoma. Austr N Z J Surg 69, 811815.CrossRefGoogle Scholar
Lee, IJ, Seong, J, Bae, JI, et al. (2011) Effect of oral supplementation with branched-chain amino acid (BCAA) during radiotherapy in patients with hepatocellular carcinoma: a double-blind randomized study. Cancer Res Treat 43, 2431.CrossRefGoogle ScholarPubMed
Luo, J, Yang, Y, Liu, J, et al. (2014) Systematic review with meta-analysis: meat consumption and the risk of hepatocellular carcinoma. Aliment Pharmacol Ther 39, 913922.CrossRefGoogle ScholarPubMed
Ma, X, Yang, Y, Li, HL, et al. (2017) Dietary trace element intake and liver cancer risk: results from two population-based cohorts in China. Int J Cancer 140, 10501059.CrossRefGoogle ScholarPubMed
Fang, AP, Chen, PY, Wang, XY, et al. (2019) Serum copper and zinc levels at diagnosis and hepatocellular carcinoma survival in the Guangdong Liver Cancer Cohort. Int J Cancer 144, 28232832.CrossRefGoogle ScholarPubMed
Fang, AP, Liu, ZY, Liao, GC, et al. (2019) Serum folate concentrations at diagnosis are associated with hepatocellular carcinoma survival in the Guangdong Liver Cancer Cohort study. Br J Nutr 121, 13761388.10.1017/S0007114519000734CrossRefGoogle ScholarPubMed
Zhang, W, Shu, XO, Li, H, et al. (2012) Vitamin intake and liver cancer risk: a report from two cohort studies in China. J Natl Cancer Inst 104, 11731181.CrossRefGoogle ScholarPubMed
Lan, QY, Zhang, YJ, Liao, GC, et al. (2016) The association between dietary vitamin A and carotenes and the risk of primary liver cancer: a case–control study. Nutrients 8, 624.CrossRefGoogle ScholarPubMed
Yuan, JM, Gao, YT, Ong, CN, et al. (2006) Prediagnostic level of serum retinol in relation to reduced risk of hepatocellular carcinoma. J Natl Cancer Inst 98, 482490.10.1093/jnci/djj104CrossRefGoogle ScholarPubMed
Lai, GY, Weinstein, SJ, Albanes, D, et al. (2014) Association of serum alpha-tocopherol, beta-carotene, and retinol with liver cancer incidence and chronic liver disease mortality. Br J Cancer 111, 21632171.CrossRefGoogle ScholarPubMed
Fang, AP, Long, JA, Zhang, YJ, et al. (2019) Serum bioavailable, rather than total, 25-hydroxyvitamin D levels are associated with hepatocellular carcinoma survival. Hepatology (epublication ahead of print version 2 November 2019).10.1002/hep.31013CrossRefGoogle ScholarPubMed
Wolonciej, M, Milewska, E & Roszkowska-Jakimiec, W (2016) Trace elements as an activator of antioxidant enzymes. Postepy Hig Med Dosw 70, 14831498.CrossRefGoogle ScholarPubMed
Puertollano, MA, Puertollano, E, de Cienfuegos, GA, et al. (2011) Dietary antioxidants: immunity and host defense. Curr Top Med Chem 11, 17521766.10.2174/156802611796235107CrossRefGoogle ScholarPubMed
Ma, J, Giovannucci, E, Pollak, M, et al. (2001) Milk intake, circulating levels of insulin-like growth factor-I, and risk of colorectal cancer in men. J Natl Cancer Inst 93, 13301336.CrossRefGoogle ScholarPubMed
Holmes, MD, Pollak, MN, Willett, WC, et al. (2002) Dietary correlates of plasma insulin-like growth factor I and insulin-like growth factor binding protein 3 concentrations. Cancer Epidemiol Biomarkers Prevent 11, 852861.Google ScholarPubMed
Hoppe, C, Molgaard, C, Juul, A, et al. (2004) High intakes of skimmed milk, but not meat, increase serum IGF-I and IGFBP-3 in eight-year-old boys. Eur J Clin Nutr 58, 12111216.CrossRefGoogle Scholar
Heaney, RP, McCarron, DA, Dawson-Hughes, B, et al. (1999) Dietary changes favorably affect bone remodeling in older adults. J Am Dietetic Assoc 99, 12281233.CrossRefGoogle ScholarPubMed
Hopfner, M, Huether, A, Sutter, AP, et al. (2006) Blockade of IGF-1 receptor tyrosine kinase has antineoplastic effects in hepatocellular carcinoma cells. Biochem Pharmacol 71, 14351448.CrossRefGoogle ScholarPubMed
Duarte-Salles, T, Fedirko, V, Stepien, M, et al. (2014) Dairy products and risk of hepatocellular carcinoma: the European Prospective Investigation into Cancer and Nutrition. Int J Cancer 135, 16621672.CrossRefGoogle ScholarPubMed
Yang, W, Sui, J, Ma, Y, et al. (2020) A prospective study of dairy product intake and the risk of hepatocellular carcinoma in U.S. men and women. Int J Cancer 146, 12411249.CrossRefGoogle ScholarPubMed
Fukuda, K, Shibata, A, Hirohata, I, et al. (1993) A hospital-based case–control study on hepatocellular carcinoma in Fukuoka and Saga Prefectures, northern Kyushu, Japan. Jpn J Cancer Res 84, 708714.CrossRefGoogle ScholarPubMed
Talamini, R, Polesel, J, Montella, M, et al. (2006) Food groups and risk of hepatocellular carcinoma: a multicenter case–control study in Italy. Int J Cancer 119, 29162921.CrossRefGoogle ScholarPubMed
La Vecchia, C, Negri, E, Decarli, A, et al. (1988) Risk factors for hepatocellular carcinoma in northern Italy. Int J Cancer 42, 872876.CrossRefGoogle ScholarPubMed
Yu, SZ, Huang, XE, Koide, T, et al. (2002) Hepatitis B and C viruses infection, lifestyle and genetic polymorphisms as risk factors for hepatocellular carcinoma in Haimen, China. Jpn J Cancer Res 93, 12871292.CrossRefGoogle Scholar
Park, Y, Leitzmann, MF, Subar, AF, et al. (2009) Dairy food, calcium, and risk of cancer in the NIH-AARP Diet and Health Study. Arch Inter Med 169, 391401.CrossRefGoogle ScholarPubMed
Zhang, W, Xiang, YB, Li, HL, et al. (2013) Vegetable-based dietary pattern and liver cancer risk: results from the Shanghai women’s and men’s health studies. Cancer Sci 104, 13531361.CrossRefGoogle ScholarPubMed
Yang, W, Ma, Y, Liu, Y, et al. (2019) Association of intake of whole grains and dietary fiber with risk of hepatocellular carcinoma in US Adults. JAMA Oncol 5, 879886.CrossRefGoogle ScholarPubMed
Kim, Y & Je, Y (2014) Dietary fiber intake and total mortality: a meta-analysis of prospective cohort studies. Am J Epidemiol 180, 565573.10.1093/aje/kwu174CrossRefGoogle ScholarPubMed
Aune, D, Keum, N, Giovannucci, E, et al. (2016) Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: systematic review and dose–response meta-analysis of prospective studies. BMJ 353, i2716.CrossRefGoogle ScholarPubMed
Threapleton, DE, Greenwood, DC, Evans, CE, et al. (2013) Dietary fibre intake and risk of cardiovascular disease: systematic review and meta-analysis. BMJ 347, f6879.CrossRefGoogle ScholarPubMed
Yao, B, Fang, H, Xu, W, et al. (2014) Dietary fiber intake and risk of type 2 diabetes: a dose–response analysis of prospective studies. Eur J Epidemiol 29, 7988.CrossRefGoogle ScholarPubMed
Aune, D, Chan, DS, Lau, R, et al. (2011) Dietary fibre, whole grains, and risk of colorectal cancer: systematic review and dose–response meta-analysis of prospective studies. BMJ 343, d6617.CrossRefGoogle ScholarPubMed
Ben, Q, Sun, Y, Chai, R, et al. (2014) Dietary fiber intake reduces risk for colorectal adenoma: a meta-analysis. Gastroenterology 146, 689699.CrossRefGoogle ScholarPubMed
Murphy, N, Norat, T, Ferrari, P, et al. (2012) Dietary fibre intake and risks of cancers of the colon and rectum in the European Prospective Investigation into Cancer and Nutrition (EPIC). PLOS ONE 7, e39361.CrossRefGoogle Scholar
Hansen, L, Skeie, G, Landberg, R, et al. (2012) Intake of dietary fiber, especially from cereal foods, is associated with lower incidence of colon cancer in the HELGA cohort. Int J Cancer 131, 469478.CrossRefGoogle ScholarPubMed
Schatzkin, A, Mouw, T, Park, Y, et al. (2007) Dietary fiber and whole-grain consumption in relation to colorectal cancer in the NIH-AARP Diet and Health Study. Am J Clin Nutr 85, 13531360.CrossRefGoogle ScholarPubMed
DiNicolantonio, JJ, Subramonian, AM & O’Keefe, JH (2017) Added fructose as a principal driver of non-alcoholic fatty liver disease: a public health crisis. Open Heart 4, e000631.CrossRefGoogle ScholarPubMed
Ros, E (2010) Health benefits of nut consumption. Nutrients 2, 652682.CrossRefGoogle ScholarPubMed
Ros, E & Hu, FB (2013) Consumption of plant seeds and cardiovascular health: epidemiological and clinical trial evidence. Circulation 128, 553565.CrossRefGoogle ScholarPubMed
Han, JM, Jo, AN, Lee, SM, et al. (2014) Associations between intakes of individual nutrients or whole food groups and non-alcoholic fatty liver disease among Korean adults. J Gastroenterol Hepatol 29, 12651272.CrossRefGoogle ScholarPubMed
Sui, J, Yang, W, Ma, Y, et al. (2019) A prospective study of nut consumption and risk of primary hepatocellular carcinoma in the U.S. women and men. Cancer Prevent Res 12, 367374.CrossRefGoogle ScholarPubMed
Bouvard, V, Loomis, D, Guyton, KZ, et al. (2015) Carcinogenicity of consumption of red and processed meat. Lancet Oncol 16, 15991600.CrossRefGoogle ScholarPubMed
Huang, RX, Duan, YY & Hu, JA (2015) Fish intake and risk of liver cancer: a meta-analysis. PLOS ONE 10, e0096102.CrossRefGoogle ScholarPubMed
Fedirko, V, Trichopolou, A, Bamia, C, et al. (2013) Consumption of fish and meats and risk of hepatocellular carcinoma: the European Prospective Investigation into Cancer and Nutrition (EPIC). Ann Oncol 24, 21662173.CrossRefGoogle Scholar
Kurozawa, Y, Ogimoto, I, Shibata, A, et al. (2004) Dietary habits and risk of death due to hepatocellular carcinoma in a large scale cohort study in Japan. Univariate analysis of JACC study data. Kurume Med J 51, 141149.CrossRefGoogle Scholar
Ma, Y, Yang, W, Li, T, et al. (2019) Meat intake and risk of hepatocellular carcinoma in two large US prospective cohorts of women and men. Int J Epidemiol 48, 18631871.Google ScholarPubMed
Su, SJ, Chow, NH, Kung, ML, et al. (2003) Effects of soy isoflavones on apoptosis induction and G2-M arrest in human hepatoma cells involvement of caspase-3 activation, Bcl-2 and Bcl-XL downregulation, and Cdc2 kinase activity. Nutr Cancer 45, 113123.CrossRefGoogle ScholarPubMed
Sharp, GB, Lagarde, F, Mizuno, T, et al. (2005) Relationship of hepatocellular carcinoma to soya food consumption: a cohort-based, case–control study in Japan. Int J Cancer 115, 290295.CrossRefGoogle ScholarPubMed
Kurahashi, N, Inoue, M, Iwasaki, M, et al. (2009) Isoflavone consumption and subsequent risk of hepatocellular carcinoma in a population-based prospective cohort of Japanese men and women. Int J Cancer 124, 16441649.CrossRefGoogle Scholar
Lagiou, P, Rossi, M, Lagiou, A, et al. (2008) Flavonoid intake and liver cancer: a case–control study in Greece. Cancer Causes Control 19, 813818.CrossRefGoogle Scholar
Zamora-Ros, R, Fedirko, V, Trichopoulou, A, et al. (2013) Dietary flavonoid, lignan and antioxidant capacity and risk of hepatocellular carcinoma in the European Prospective Investigation into Cancer and Nutrition study. Int J Cancer 133, 24292443.CrossRefGoogle ScholarPubMed
Zhang, W, Wang, J, Gao, J, et al. (2019) Prediagnostic level of dietary and urinary isoflavonoids in relation to risk of liver cancer in Shanghai, China. Cancer Epidemiol Biomarkers Prev 28, 17121719.CrossRefGoogle ScholarPubMed
Illig, T, Gieger, C, Zhai, G, et al. (2009) A genome-wide perspective of genetic variation in human metabolism. Nat Genet 42, 137.CrossRefGoogle ScholarPubMed
Shoaie, S, Ghaffari, P, Kovatcheva-Datchary, P, et al. (2015) Quantifying diet-induced metabolic changes of the human gut microbiome. Cell Metab 22, 320331.CrossRefGoogle ScholarPubMed
Guertin, KA, Moore, SC, Sampson, JN, et al. (2014) Metabolomics in nutritional epidemiology: identifying metabolites associated with diet and quantifying their potential to uncover diet–disease relations in populations. Am J Clin Nutr 100, 208217.CrossRefGoogle ScholarPubMed
Bhupathiraju, SN & Hu, FB (2017) One (small) step towards precision nutrition by use of metabolomics. Lancet Diabetes Endocrinol 5, 154155.CrossRefGoogle ScholarPubMed
Guasch-Ferré, M, Bhupathiraju, SN & Hu, FB (2018) Use of metabolomics in improving assessment of dietary intake. Clin Chem 64, 8298.CrossRefGoogle ScholarPubMed
Yang, CS, Wang, X, Lu, G, et al. (2009) Cancer prevention by tea: animal studies, molecular mechanisms and human relevance. Nat Rev Cancer 9, 429439.CrossRefGoogle ScholarPubMed
Fon Sing, M, Yang, WS, Gao, S, et al. (2011) Epidemiological studies of the association between tea drinking and primary liver cancer: a meta-analysis. Eur J Cancer Prev 20, 157165.CrossRefGoogle ScholarPubMed
Bamia, C, Lagiou, P, Jenab, M, et al. (2015) Coffee, tea and decaffeinated coffee in relation to hepatocellular carcinoma in a European population: multicentre, prospective cohort study. Int J Cancer 136, 18991908.CrossRefGoogle Scholar
Huang, YQ, Lu, X, Min, H, et al. (2016) Green tea and liver cancer risk: a meta-analysis of prospective cohort studies in Asian populations. Nutrition 32, 38.CrossRefGoogle ScholarPubMed
Tamura, T, Wada, K, Konishi, K, et al. (2018) Coffee, green tea, and caffeine intake and liver cancer risk: a prospective cohort study. Nutr Cancer 70, 12101216.CrossRefGoogle ScholarPubMed
Butler, LM, Huang, JY, Wang, R, et al. (2015) Urinary biomarkers of catechins and risk of hepatocellular carcinoma in the Shanghai Cohort Study. Am J Epidemiol 181, 397405.CrossRefGoogle ScholarPubMed
Bravi, F, Bosetti, C, Tavani, A, et al. (2013) Coffee reduces risk for hepatocellular carcinoma: an updated meta-analysis. Clin Gastroenterol Hepatol 11, 14131421.CrossRefGoogle ScholarPubMed
Sang, LX, Chang, B, Li, XH, et al. (2013) Consumption of coffee associated with reduced risk of liver cancer: a meta-analysis. BMC Gastroenterol 13, 34.CrossRefGoogle ScholarPubMed
Bai, K, Cai, Q, Jiang, Y, et al. (2016) Coffee consumption and risk of hepatocellular carcinoma: a meta-analysis of eleven epidemiological studies. Oncol Targets Ther 9, 43694375.Google ScholarPubMed
Yu, C, Cao, Q, Chen, P, et al. (2016) An updated dose–response meta-analysis of coffee consumption and liver cancer risk. Sci Rep 6, 37488.CrossRefGoogle ScholarPubMed
Bravi, F, Tavani, A, Bosetti, C, et al. (2017) Coffee and the risk of hepatocellular carcinoma and chronic liver disease: a systematic review and meta-analysis of prospective studies. Eur J Cancer Prev 26, 368377.CrossRefGoogle ScholarPubMed
Godos, J, Micek, A, Marranzano, M, et al. (2017) Coffee consumption and risk of biliary tract cancers and liver cancer: a dose–response meta-analysis of prospective cohort studies. Nutrients 9, E950.CrossRefGoogle ScholarPubMed
Kennedy, OJ, Roderick, P, Buchanan, R, et al. (2017) Coffee, including caffeinated and decaffeinated coffee, and the risk of hepatocellular carcinoma: a systematic review and dose–response meta-analysis. BMJ Open 7, e013739.CrossRefGoogle ScholarPubMed
Tamura, T, Hishida, A & Wakai, K (2019) Coffee consumption and liver cancer risk in Japan: a meta-analysis of six prospective cohort studies. Nagoya J Med Sci 81, 143150.Google ScholarPubMed
Jang, ES, Jeong, SH, Lee, SH, et al. (2013) The effect of coffee consumption on the development of hepatocellular carcinoma in hepatitis B virus endemic area. Liver Int 33, 10921099.CrossRefGoogle ScholarPubMed
Lai, GY, Weinstein, SJ, Albanes, D, et al. (2013) The association of coffee intake with liver cancer incidence and chronic liver disease mortality in male smokers. Br J Cancer 109, 13441351.CrossRefGoogle ScholarPubMed
Aleksandrova, K, Bamia, C, Drogan, D, et al. (2015) The association of coffee intake with liver cancer risk is mediated by biomarkers of inflammation and hepatocellular injury: data from the European Prospective Investigation into Cancer and Nutrition. Am J Clin Nutr 102, 14981508.CrossRefGoogle ScholarPubMed
Petrick, JL, Freedman, ND, Graubard, BI, et al. (2015) Coffee consumption and risk of hepatocellular carcinoma and intrahepatic cholangiocarcinoma by sex: the liver cancer pooling project. Cancer Epidemiol Biomarkers Prev 24, 13981406.CrossRefGoogle ScholarPubMed
Setiawan, VW, Wilkens, LR, Lu, SC, et al. (2015) Association of coffee intake with reduced incidence of liver cancer and death from chronic liver disease in the US multiethnic cohort. Gastroenterology 148, 118125; quiz e115.CrossRefGoogle ScholarPubMed
Loftfield, E, Rothwell, JA, Sinha, R, et al. (2019) Prospective investigation of serum metabolites, coffee drinking, liver cancer incidence, and liver disease mortality. J Natl Cancer Inst 112, 286294.CrossRefGoogle Scholar
Loomba, R, Yang, HI, Su, J, et al. (2013) Synergism between obesity and alcohol in increasing the risk of hepatocellular carcinoma: a prospective cohort study. Am J Epidemiol 177, 333342.CrossRefGoogle ScholarPubMed
Petrick, JL, Campbell, PT, Koshiol, J, et al. (2018) Tobacco, alcohol use and risk of hepatocellular carcinoma and intrahepatic cholangiocarcinoma: the liver cancer pooling project. Br J Cancer 118, 10051012.CrossRefGoogle ScholarPubMed
Turati, F, Galeone, C, Rota, M, et al. (2014) Alcohol and liver cancer: a systematic review and meta-analysis of prospective studies. Ann Oncol 25, 15261535.CrossRefGoogle ScholarPubMed
Chuang, SC, Lee, YC, Wu, GJ, et al. (2015) Alcohol consumption and liver cancer risk: a meta-analysis. Cancer Causes Control 26, 12051231.CrossRefGoogle ScholarPubMed
Schrieks, IC, Heil, AL, Hendriks, HF, et al. (2015) The effect of alcohol consumption on insulin sensitivity and glycemic status: a systematic review and meta-analysis of intervention studies. Diabetes Care 38, 723732.Google ScholarPubMed
Tapsell, LC, Neale, EP, Satija, A, et al. (2016) Foods, Nutrients, and Dietary Patterns: interconnectionsd and implications for dietary guidelines. Adv Nutr 7, 445454.CrossRefGoogle ScholarPubMed
Chen, PY, Fang, AP, Wang, XY, et al. (2018) Adherence to the Chinese or American dietary guidelines is associated with a lower risk of primary liver cancer in China: a case–control study. Nutrients 10.CrossRefGoogle ScholarPubMed
Li, WQ, Park, Y, McGlynn, KA, et al. (2014) Index-based dietary patterns and risk of incident hepatocellular carcinoma and mortality from chronic liver disease in a prospective study. Hepatology 60, 588597.CrossRefGoogle ScholarPubMed
Ma, Y, Yang, W, Simon, TG, et al. (2018) Dietary patterns and risk of hepatocellular carcinoma among U.S. men and women. Hepatology 70, 577586.CrossRefGoogle Scholar
National Cancer Institute (2018) Applied Research: Cancer Control and Population Sciences. Dietary patterns methods project. https://epi.grants.cancer.gov/dietary-patterns/ (accessed September 2018).Google Scholar
Tabung, FK, Smith-Warner, SA, Chavarro, JE, et al. (2016) Development and validation of an empirical dietary inflammatory index. J Nutr 146, 15601570.CrossRefGoogle ScholarPubMed
Tabung, FK, Wang, W, Fung, TT, et al. (2016) Development and validation of empirical indices to assess the insulinaemic potential of diet and lifestyle. Br J Nutr 116, 17871798.CrossRefGoogle Scholar
Figure 0

Fig. 1. Proposed mechanisms linking diet and liver cancer risk. T2D, type 2 diabetes; NAFLD, non-alcoholic fatty liver disease; IGF-1, insulin-like growth factor-1.

Supplementary material: File

Yang et al. supplementary material

Tables S1-S4

Download Yang et al. supplementary material(File)
File 138.8 KB