Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Sep 28, 2016; 8(27): 1119-1127
Published online Sep 28, 2016. doi: 10.4254/wjh.v8.i27.1119
Molecular pathological epidemiology in diabetes mellitus and risk of hepatocellular carcinoma
Chun Gao, Department of Gastroenterology, China-Japan Friendship Hospital, Ministry of Health, Beijing 100029, China
Author contributions: Gao C conceived the topic, performed research, retrieved concerned literatures and wrote the paper.
Supported by Beijing NOVA Programme of Beijing Municipal Science and Technology Commission, No. Z13110.7000413067.
Conflict-of-interest statement: No conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Chun Gao, MD, Department of Gastroenterology, China-Japan Friendship Hospital, Ministry of Health, No. 2 Yinghua East Road, Beijing 100029, China. gaochun@bjmu.edu.cn
Telephone: +86-10-84205313 Fax: +86-10-64481924
Received: April 2, 2016
Peer-review started: April 7, 2016
First decision: June 6, 2016
Revised: June 28, 2016
Accepted: August 6, 2016
Article in press: August 8, 2016
Published online: September 28, 2016

Abstract

Molecular pathological epidemiology (MPE) is a multidisciplinary and transdisciplinary study field, which has emerged as an integrated approach of molecular pathology and epidemiology, and investigates the relationship between exogenous and endogenous exposure factors, tumor molecular signatures, and tumor initiation, progression, and response to treatment. Molecular epidemiology broadly encompasses MPE and conventional-type molecular epidemiology. Hepatocellular carcinoma (HCC) is the third most common cause of cancer-associated death worldwide and remains as a major public health challenge. Over the past few decades, a number of epidemiological studies have demonstrated that diabetes mellitus (DM) is an established independent risk factor for HCC. However, how DM affects the occurrence and development of HCC remains as yet unclearly understood. MPE may be a promising approach to investigate the molecular mechanisms of carcinogenesis of DM in HCC, and provide some useful insights for this pathological process, although a few challenges must be overcome. This review highlights the recent advances in this field, including: (1) introduction of MPE; (2) HCC, risk factors, and DM as an established independent risk factor for HCC; (3) molecular pathology, molecular epidemiology, and MPE in DM and HCC; and (4) MPE studies in DM and risk of HCC. More MPE studies are expected to be performed in future and I believe that this field can provide some very important insights on the molecular mechanisms, diagnosis, personalized prevention and treatment for DM and risk of HCC.

Key Words: Diabetes mellitus, Molecular pathological epidemiology, Hepatocellular carcinoma, Risk factor, Molecular mechanism

Core tip: Diabetes mellitus (DM) is an established independent risk factor for hepatocellular carcinoma (HCC); however, how DM affects the occurrence and development of HCC remains as yet unclearly understood. Molecular pathological epidemiology (MPE) may be a promising approach to investigate the molecular mechanisms of carcinogenesis of DM in HCC, and provide some useful insights for this pathological process. This review highlights the recent advances in this field and more MPE studies are expected to be performed for this question in future.



INTRODUCTION

Molecular pathology examines the expression of molecular markers within bodily fluids, tissues or organs, and focuses on the diagnosis and studies of diseases, such as tumors[1,2]. Epidemiology is focused upon the studies of distributions and determinants of diseases and health conditions in specific populations[3,4]. Molecular pathological epidemiology (MPE) is a multidisciplinary and transdisciplinary study field, which has emerged as an integrated approach of molecular pathology and epidemiology, and investigates the relationship between tumour molecular markers, exposure of endogenous and exogenous factors, and development, progression and prognosis of tumors[5-8]. Molecular epidemiology broadly encompasses MPE and conventional-type molecular epidemiology.

In MPE, researchers investigate the relationships between: (1) changes of extracellular or cellular molecules (disease molecular signatures); (2) genetic, dietary, environmental and lifestyle factors; and (3) development and progression of diseases, such as tumors[6]. In 2010, Professor Shuji Ogino and Professor Meir Stampfer[5] were the first to introduce the concept of MPE. They consolidated this concept mainly based on the researches of colorectal cancer (CRC), particularly the prototypical study in the evolving field of MPE, which was conducted by Professor Peter T Campbell and others[9].

This case-control study of Campbell et al[9] was conducted to determine the relationships between CRC microsatellite instability (MSI) status, risk of CRC, and human body mass index (BMI). The results showed that an increased CRC risk was found in those patients with a high BMI; however, this risk of CRC was associated with the MSI status. For patients with MS-stable, the adjusted odds ratio (OR) was 1.38 and 95%CI was 1.24-1.54, for an increment of 5 kg/m2 of BMI; for patients with MSI-low, the OR was 1.33 and 95%CI was 1.04-1.72; however, for patients with MSI-high tumours, the value of OR and 95%CI were 1.05 and 0.84-1.31, respectively[9]. The authors concluded that the relationship between the high BMI and increased CRC risk was related to the tumor MSI status[9]. According to the concept and principle of MPE, this prototypical study addressed the relationship between exposure factor (high BMI), molecular change (CRC MSI status) and tumor initiation (risk of CRC)[5,10].

MPE addresses two questions: (1) the association of particular exposure factors with specific molecular changes; and (2) the interaction of particular exposure factors with specific molecular changes to affect development, progression and prognosis of tumors. The typical research of cancer MPE is used to examine the relationship between exposure factors and risk of tumors according to the status of tumor signatures[9,10]. Cancer MPE techniques and studies can help us understand the carcinogenesis of certain exposure factors, through the examination of molecular pathological signatures associated with initiation and progression of tumors, and the exposures[5,6].

HEPATOCELLULAR CARCINOMA AND RISK FACTORS

Hepatocellular carcinoma (HCC) has been confirmed as the third leading cause globally, among all the cancer-related deaths[10-12]. For primary liver cancers, more than 80% are HCC and the incidence rate annually of HCC is 4.9 per 100000 persons. Although some advances have been gained in the diagnosis and treatment of HCC, the prognosis remains very poor. Similarly, the annual mortality rate remains very high and HCC has also been ranked as one of the most lethal cancers[13].

With the using and popularization of hepatitis B virus (HBV) vaccination, the improvement of people’s living standard and life style, and advancement of early diagnosis and treatment of premalignant lesions, the incidence of HCC had been anticipated to be decreased. However, the incidence rate of HCC has already been found to be increased significantly in the past thirty years in some countries, including the United States, China and Japan[14,15]. For example, during the period of 1981-1983 in the United States, the age adjusted incidence rate was 1.3 per 100000; however, this rate increased to 3.0 per 100000 during the period of 1996-1998[14,15]. Although more than fifty percent of this increase has been attributed to hepatitis virus C (HCV), other hepatitis viruses and alcoholic liver disease[16], the reason remains as unclear.

The identified risk factors of HCC include liver cirrhosis, HBV, HCV, heavy alcoholic consumption, alfatoxin exposure, non-alcoholic steatohepatitis, positive family history, male sex, and increasing age[17-19]. Over the past few decades, a number of epidemiological studies have demonstrated that diabetes mellitus (DM) is an established independent risk factor for HCC[12,20-23].

DM AS AN ESTABLISHED INDEPENDENT RISK FACTOR FOR HCC

In the year of 1986, for the first time, Lawson et al[24] proposed accidentally the positive association of DM with HCC. The authors observed that, in Western Europe, the incidence rate of primary liver cancers was increased, and deduced that this increase might in part be associated with the induction of hepatic microsomal enzyme caused by long-term usage of some drugs. On the basis of this assumption, the authors designed and performed an observational case-control study, which included 105 patients with HCC and long-term drug use, and 105 age and sex-matched patients with colorectal tumors and with fractures of femur[24]. Surprisingly, the results demonstrated that compared to the control group, the HCC group patients had four-fold excess of diabetic cases, and this association was independent of those pre-existing diseases, for example viral hepatitis, alcoholic cirrhosis and haemochromatosis[24]. The relationship between DM and HCC was proposed clearly although some limitations could not be avoided.

Following the publication of this study, only a few researches attempted to elucidate the association of diabetes with HCC in the next more than ten years; however, over the past more than one decade, more and more researches have been designed and performed to address this relationship[21,25-27]. Earlier epidemiologic studies showed inconsistent findings relating to the association of DM with HCC[21,28-30] whereas more and more recent studies have identified DM as an established independent risk factor for HCC, especially two prospectively large-scale population-based cohort studies[31,32]. In 2008, a review published in the journal of LANCET ranked diabetes as the fourth risk factor for HCC, following cirrhosis, viral hepatitis B and C, and non-alcoholic steatohepatitis[17].

Of the two prospectively large-scale population-based cohort studies[31,32], one was performed in the Sweden, which used the Swedish In-patient Register and included 153852 patients diagnosed with diabetes during the period between 1965 and 1983[31]. The patients were followed up through December 31, 1989. The authors identified those incident cases of cancer using the database and excluded those patients who were diagnosed with liver cancers during the first year of follow-up. The results showed that an increased risk of developing primary liver cancers was found in the diabetic patients (standardized incidence ratio, SIR = 4.1; 95%CI: 3.8-4.5). After exclusion of those concomitant diseases which have been associated with HCC, for example hepatitis, cirrhosis, and alcoholism, the persistence of an approximately threefold excess risk was observed[31].

The conclusion from the Swedish study was supported by another followed cohort study conducted in the United States[32], which was performed by doctors in the Department of Veterans Affairs. In this study, the authors also used the computerized records to identify all the patients with a hospital discharge diagnosis of DM in the period from 1985 to 1990, and matched randomly three patients without DM for every diabetic patient. Follow-up of these patients was taken through December 31, 2000. The major strength of this study was the strict inclusion and exclusion criteria and they were pre-determined perfectly on the basis of our current knowledge. The authors decided and used three periods, including: (1) the period dating back to 1980; (2) the period of index hospitalization; and (3) the period of the first year of follow-up. During these three above-mentioned periods, those patients with all kinds of liver diseases, abnormal liver function tests, alcoholism, or other identified risk factors for HCC, such as HBV and HCV, had been excluded from the study population[32]. The authors concluded that among men with diabetes, the risk of HCC was increased, which was not associated with demographic features, viral hepatitis, cirrhosis, and alcoholic liver disease.

The recently published systematic review in this field was designed to evaluate the impact of DM on the risk of HCC among patients with HCV infection[33]. This research included seven articles and all of them were conducted in Asian cohorts, including three studies from Taiwan, China, and four from Japan[34-40]. Among these studies, six were observational cohorts and six studies were of good quality. The results showed that a significantly increased risk of HCC was associated with DM in five of these seven studies and the effect sizes ranged from HR = 1.73 (95%CI: 1.30-2.30) to RR = 3.52 (95%CI: 1.29-9.24)[33].

MOLECULAR PATHOLOGY, MOLECULAR EPIDEMIOLOGY AND MPE IN DIABETES
Molecular pathology in diabetes

Pathology is an important constituent part of diagnostics, modern medicine and causal studies of diseases, which focuses upon four research fields of diseases: Etiology (causes), pathogenesis (mechanisms of development and progression), morphologic alterations (structural changes of cells, tissues and organs), and clinical manifestations (consequences of alterations)[41,42]. Molecular pathology (MP), whose focus is the examination of molecular signatures, has some similar aspects of practice to other disciplines, such as anatomic pathology, genetics, biochemistry, proteomics, molecular biology, and clinical pathology. Application of modern MP often encompasses three components: (1) exploration and confirmation of predictive molecular biomarkers for development, progression and treatment of diseases; (2) development of genetic and molecular approaches for diagnosis and classification of human diseases; and (3) susceptibility of individuals of different genetic constitution to particular disorders.

Molecular pathological studies in diabetes provide better insight into the etiology. For example type 1 or insulin-dependent diabetes, at least 20 genes have been identified and the dominant susceptibility locus maps to the major histocompatibility complex[43,44]. Major areas of MP research include environmental trigger factors, modification of the beta cells, infiltration of the islets by immuno-inflammatory cells, and autoimmune-mediated destruction of the beta cells. For T2DM, since the early genome-wide association studies (GWAS) in 2007, hundreds of genetic loci have been identified. Elucidating the pathology of DM at the molecular level is very important for developing innovative, personalized, and evidence-based treatments[45,46].

From the viewpoint of MP of DM in cancers, disruption of homeostatic glucose metabolism has been significantly associated with the malignant cellular transformation and tumor progression. In addition, the pathophysiology of disrupted glucose-insulin axis pathways of DM has been understood deeply at the subcellular level, thanking for the recent advances in biochemical and molecular technology. They may be useful for better understanding of the malignant cellular transformation, such as HCC.

Molecular epidemiology in diabetes

In the late 20th century, with great advancement of biomedical sciences, a number of molecular signatures or biomarkers were identified as predictors of disease initiation, progression, and response to treatment, including diabetes and tumors. Since the identification of these molecular signatures, molecular epidemiology has evolved and been broadly named, which refers to the branch of epidemiology, where investigators examine these signatures in special study populations and its interaction with environmental, lifestyle or dietary factors, to perform the causal studies of diseases with aetiological factors[6,10]. Since the 2000s, GWAS has been commonly performed to identify genetic risk factors for diseases and health conditions[47,48].

Molecular epidemiology in diabetes is focused upon the contribution of possible environmental and genetic risk factors, to the distributions and determinants of DM within families and across populations, at the molecular level. For example, a number of molecular epidemiological studies demonstrate that some growth factors, including insulin, growth hormone, insulin-like growth factors and their binding proteins, may be important in the pathophysiological processes of T2DM[49]. In addition, many physiological changes have been associated with T2DM, including insulin resistance and hyperinsulinemia, increased estrogen levels, increased inflammatory cytokines such as tumor necrosis factor (TNF)-α, and interleukin (IL)-6, as well as altered levels of circulating adipokines[50]. It is well known that some of these molecular signatures and physiological changes may contribute to the development of cancers. Therefore, the relationship between DM and cancers, such as HCC, may be built via these molecular signatures or biomarkers.

MPE in diabetes

MPE emerges as an integrated approach of molecular pathology and epidemiology, and investigates the relationship between risk factors, molecular signatures, and development and progression of diseases[10]. According to the concept and principle of MPE, the MPE approaches can also be used in non-neoplastic diseases, such as DM[51]. Although great advancements have been made in molecular pathology and molecular epidemiology, and a lot of molecular signatures have been associated with DM, no MEP studies in DM have been performed and the reason may be deduced that no identified risk factors are found for DM, such as HBV or HCV for HCC.

However, a few MPE studies had been performed when DM was treated as the risk factor for other diseases, such as cancers and coronary artery lesions, before the proposal and/or use of the concept of MPE, and they were conducted usually under the umbrella of molecular epidemiology. For example, one MPE study was designed to determine the relationship between 8-oxoguanine glycosylase (hOGG1) Ser326Cys gene polymorphism and coronary artery lesions in patients with DM[52]. In this study, 323 diabetic patients were included and the results showed that hOGG1 Ser326Cys polymorphism was correlated with coronary artery lesions in patients with DM, and Cys/Cys genotype may be associated with the more severity of lesions[52].

MOLECULAR PATHOLOGY, MOLECULAR EPIDEMIOLOGY AND MPE IN HCC
Molecular pathology in HCC

For human cancers, including CRC and HCC[53-55], molecular pathology is commonly used in the diagnosis and classification. Traditional molecular pathology studies are focused upon the molecular characteristics in cancer cells to improve our understanding of tumor cell behavior and carcinogenic processes[1,6,10]. However, human cancers are complex multifactorial diseases. Recent studies suggest that cancers should be classified based on salient clinical and pathologic features as well as on molecular fingerprints, which has been named “molecular classification”, because of the premise that tumors with similar characteristics share common pathogenic mechanisms and progression patterns, despite each tumor undergoing its own unique neoplastic transformation[5,6,56]. Molecular classification is helpful to better understand the pathogenesis of tumors, predict the development and progression of each tumor, and for personalized cancer medicine, optimize the preventive and treatment strategies[5,6,56]. For cancer molecular classification, informative biomarkers are needed to be identified to stratify tumors or patients[57-62].

Examples of well-established informative biomarkers include ESR1 (ER-α), PGR and ERBB2 (HER2) expression in breast cancer[63-65], EGFR mutations in lung cancer[66,67], MSI in colorectal cancer[68-70], TMPRSS2-ERG translocation in prostate cancer[71], and TP53, PIK3CA, BRAF and KRAS mutations, and CpG island methylation in multiple cancers[72-74]. Some molecular changes or biomarkers in HCC have also been previously identified. Ojanguren et al[75] showed that the positive expression of mutant p53 was related to alcohol abuse (42%) and HBV infection (21%). Park et al[76] found that TNF and IL-6 signaling was correlated with obesity-associated HCC development. In the obese patients, insulin and insulin-like growth factors, TNF-α, IL-1 and IL-6, leptin, adipokines, adiponectin, and plasminogen activator inhibitor-1 are significantly associated with the occurrence and development of some cancers, including HCC[77].

Molecular epidemiology in HCC

HCC is also very complex, for example it occurs in about 1%-7% of cirrhotic patients annually, whereas most of the cirrhotic patients do not progress to HCC during their lifetimes[78]. Molecular biomarkers are expected to satisfy this need and resolve the question at the molecular level. To date, molecular epidemiology studies show that a number of molecular risk factors of HCC have been identified, such as numerous genetic polymorphisms reported as host genetic factors[79]. Most of HCC-associated single-nucleotide polymorphisms are identified in genes involved in biological pathways, including oxidative stress (GSTT1, GSTM1), cell cycle (MDM2), immune response (IL10, TNF), DNA damage repair (XPC), growth signaling (EGF), and iron metabolism (HFE) in viral hepatitis- or alcohol-related HCC[80-84]. Recent GWAS identifies the DEPDC5 locus as the risk loci in viral hepatitis-related HCC[85].

Molecular factors associated with etiological agents, for example HBV and HCV could also influence the risk of HCC. It is well known that a high level of serum HBV DNA is indicative of increased risk of HCC. Some studies have demonstrated that HBV genotype is related to the HCC risk[86]. Genomics technology has revealed that HCC should be regarded as a heterogenous group of diseases, not one single disease entity, because each sub-group HCC has different sets of epigenetic and genetic alterations[87]. The heterogeneous molecular features of HCC tumors are associated with the biological behavior, clinical outcome and prognosis[87-91]. Molecular classification is recommended to HCC, and previous studies have identified subsets of HCC tumors characterized by TP53 and CTNNB1 activation mutations, progenitor cell–like features, Met activation, Myc activation, and transforming growth factor-β activation[92-94]. These molecular risk factors of HCC would play important roles in the design and implementation of MPE studies.

MPE in HCC

Epidemiological studies have showed that DM is an established independent risk factor for HCC[12,20-23]; however, how DM affects the development and progression of HCC has not been explained clearly. MPE approaches and studies may be helpful to improve our understanding of the molecular mechanisms of carcinogenesis of HCC. MPE can be used to investigate the relationship between DM and risk of HCC by molecular subtypes. A few MPE studies have been performed for this question, although they were usually under the umbrella of molecular epidemiology. They would be described in the next section in detail. MPE can provide some useful insights for the pathological processes of DM in HCC, although a few challenges must be overcome.

MPE IN DM AND RISK OF HCC

Currently, based on our knowledge, very few MPE researches are available for DM and risk of HCC[95-97]. For these studies, the original design are not for MPE, and the term of “molecular pathological epidemiology” have not appeared in their articles, but they can be treated as MPE researches, according to the objectives and methods.

One MPE research which was performed in the Japan was designed to determine the relationship between PNPLA3 and JAZF1, and risk of HCC, in patients with non-viral hepatitis and type 2 DM[95]. The objective of this research was to identify genetic determinants associated with T2DM patients who have a high risk of developing HCC by genotyping T2DM susceptibility loci and PNPLA3. This study included 389 T2DM patients, including 59 patients with HCC (DM-HCC) and 330 patients without HCC (DM-non-HCC). Those patients who followed these criteria were included: (1) history of T2DM > 10 years; (2) alcohol intake < 60 g/d; and (3) negative for anti-HCV Ab and HBs-Ag. The authors found that the SNP rs738409 located in PNPLA3 was the greatest risk factor associated with HCC in these diabetic patients. Compared to DM-non-HCC patients, DM-HCC patients had the significantly higher frequency of the PNPLA3 G allele (OR = 2.53, P = 1.05 × 10-5). Moreover, among the 115 DM patients homozygous for the PNPLA3 G allele, HCC patients had the significantly higher frequency of the JAZF1 rs864745 G allele (OR = 3.44, P = 0.0002)[95]. They concluded that PNPLA3 and JAZF1 were associated with an increased risk of developing HCC among T2DM patients without viral hepatitis[95].

Another study was designed to evaluate the cytokinome profile, including the serum levels of growth factors, chemokines, cytokines, as well as of other diabetes and cancer biomarkers, in a cohort of patients, including 17 patients with T2DM, 20 patients with chronic hepatitis C infection, 34 patients with HCC, 10 patients with T2DM-HCC, and 20 healthy controls[96]. The results demonstrated that: (1) T2DM-HCC patients had the higher levels of IL-2R, sIL-6Ra, IL-16, IL-18, HGF, β-NGF, CXCL1, CXCL12, ADIPOQ, and IFN-α than those with T2DM or HCC; (2) T2DM-HCC patients had the lower level of LEP than those with T2DM or HCC; (3) T2DM-HCC and only HCC patients had the similar levels of CXCL9, PECAM-1, Prolactin, glucagon, sVEGFR-1 and sVEGFR-2; (4) T2DM-HCC patients had the higher levels of CXCL9, PECAM-1, Prolactin, and glucagon than those with only T2DM; and (5) T2DM-HCC patients had the lower levels of sVEGFR-1 and sVEGFR-2 than those with only T2DM[96]. The major limitation of this study was the very limited number of included patients; however, these molecular changes could be used to design and perform the MPE researches in DM and risk of HCC in future.

Some molecular pathology researches can also be regarded as MPE studies, for example one study which was conducted in the Second Military Medical University, Shanghai, China[97]. The objectives of this study were to determine the effect of p-Ser9-GSK-3β (glycogen synthase kinase-3β) on the prognosis in HCC patients and to explore the interaction between GSK-3β, T2DM and prognosis of HCC. This research included 178 HCC patients after curative partial hepatectomy and showed that expression of P-Ser9-GSK-3β was significantly higher in tumor tissues than that in their normal counterparts[97]. Moreover, the authors also found that: (1) over-expression of p-Ser9-GSK-3β was associated with T2DM; (2) T2DM and over-expression of p-Ser9-GSK-3β were closely related with each other; and (3) these two variables were independently associated with poor prognosis of HCC[97]. Therefore, p-Ser9-GSK-3β may be regarded as the mediator between T2DM and HCC.

One case report which was published in 2015 was also considered to be related to this field[98]. This report describes a 23-year-old woman with HCC and type 2 DM; and results of histological and immunohistochemical examination showed that this HCC arose in the background of hepatocyte nuclear factor-1α mutated hepatocellular adenomas (H-HCA). However, traditionally, we consider that H-HCA have no minimal malignant potential. For the molecular changes and tumor biomarkers of HCC, the authors found that by immunohistochemical tests, CD34 expression in sinusoidal endothelial cells and expression of glutamine synthetase in tumor cells were increased, whereas exon 3 of CTNNB1 and TERT promoter mutations, and nuclear expression of β-catenin were absent in this patients with HCC and DM. Although such cases are rare, they reinforce the potential of H-HCA for HCC, which may be related to DM[98].

Considering that DM is an independent risk factor for HCC, some efforts have been focused on understanding of the molecular mechanisms of DM in the development and progression of HCC, which may be useful for the design and implementation of MPE studies. For example, one mini-review focused on the impact of TNF-α and IL-6 along with epigenetic regulations[99]. Two approaches are suggested as followed: (1) the first is about the role of TNF-α and IL-6 as inflammatory mediators, from the point of role of apoptosis and inflammation in HCC; and apoptotic regulators can be used for this purpose, such as Bax (bcl-2-like protein 4 encoded by the BAX gene) and Bcl-2 (B-cell lymphoma 2 protein encoded by BCL2 gene); and (2) the second is about the possible epigenomic reprogramming, from the point of role of epigenetic modification of DNA in HCC. According to these two app–roaches, apoptotic and inflammatory markers (Bcl2 and Bax), and DNA methylation, hypomethylation or histone modifications can be used as the candidated molecular biomarkers for the understanding of role of DM in HCC[99].

Another review focused on the influence of insulin resistance and hyperinsulinemia of DM in the pathogenesis of hepatocarcinogenesis, and the author summarized that some molecular pathways were involved, for example phosphatase and tensin homolog/P13K/Akt and MAPK kinase[100]. It is well known that different anti-diabetic medications have different influences on the risk of HCC in diabetic patients[23,100]. Metformin has been associated with the decreased risk of HCC in patients diagnosed with DM[23]. The molecular mechanism is deduced that metformin can activate 5-adenosine monophosphate-activated protein kinase (AMPK) and decrease the expression of protein Livin[100]. AMPK can inhibit its downstream target mammalian target of rapamycin, and then inhibit the growth of human cancer cell lines. Livin has been involved in both cell proliferation and survival. Thiazolidinediones seem to inhibit peroxisome proliferator-activated receptor gamma-independent regulation of nucleophosmin and prevent tumor formation[100].

Although these studies are not enough for understanding of molecular mechanisms of DM in the increased risk of HCC, they and the involved molecular biomarkers can be very useful for future MPE researches. I hope that more and more MPE researches are performed exploring the molecular mechanisms as well as novel biomarkers.

CONCLUSION

DM is an established independent risk factor for HCC; however, how DM affects the occurrence and development of HCC remains as yet unclearly understood. “MPE” is the branch of epidemiology and pathology, and its basis is the molecular classification of tumors. MPE is a multidisciplinary, interdisciplinary and transdisciplinary study field, and molecular pathology plays a central role in this relatively new field. In MPE, investigators examine the relationship between tumor molecular signatures, endogenous and exogenous factors, and development, progression and prognosis of tumors. I believe that this research field can be very helpful to improve our understanding of the pathogenesis, molecular mechanisms, diagnosis, personalized prevention and treatment for DM and risk of HCC in future.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Haidara MAA S- Editor: Qi Y L- Editor: A E- Editor: Li D

References
1.  Harris TJ, McCormick F. The molecular pathology of cancer. Nat Rev Clin Oncol. 2010;7:251-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 170]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
2.  Menendez KR, García M, Spatz S, Tablante NL. Molecular epidemiology of infectious laryngotracheitis: a review. Avian Pathol. 2014;43:108-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 46]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
3.  Izzotti A, Neri M, Vecchio D, Puntoni R. Molecular epidemiology in cancer research (review). Int J Oncol. 1997;11:1053-1069.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Lloyd C, Cullinan P. Year in review 2014: basic science and epidemiology. Thorax. 2015;70:581-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
5.  Ogino S, Stampfer M. Lifestyle factors and microsatellite instability in colorectal cancer: the evolving field of molecular pathological epidemiology. J Natl Cancer Inst. 2010;102:365-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 153]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
6.  Ogino S, Chan AT, Fuchs CS, Giovannucci E. Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut. 2011;60:397-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 436]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
7.  Ogino S, Nosho K, Meyerhardt JA, Kirkner GJ, Chan AT, Kawasaki T, Giovannucci EL, Loda M, Fuchs CS. Cohort study of fatty acid synthase expression and patient survival in colon cancer. J Clin Oncol. 2008;26:5713-5720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 143]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
8.  Morikawa T, Kuchiba A, Yamauchi M, Meyerhardt JA, Shima K, Nosho K, Chan AT, Giovannucci E, Fuchs CS, Ogino S. Association of CTNNB1 (beta-catenin) alterations, body mass index, and physical activity with survival in patients with colorectal cancer. JAMA. 2011;305:1685-1694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 153]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
9.  Campbell PT, Jacobs ET, Ulrich CM, Figueiredo JC, Poynter JN, McLaughlin JR, Haile RW, Jacobs EJ, Newcomb PA, Potter JD. Case-control study of overweight, obesity, and colorectal cancer risk, overall and by tumor microsatellite instability status. J Natl Cancer Inst. 2010;102:391-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 144]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
10.  Gao C. Molecular pathological epidemiology: an interdisciplinary field for study of hepatocellular carcinoma. Austin J Gastroenterol. 2015;2:1040.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012;62:10-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8406]  [Cited by in F6Publishing: 8901]  [Article Influence: 741.8]  [Reference Citation Analysis (0)]
12.  Zhang H, Gao C, Fang L, Yao SK. Increased international normalized ratio level in hepatocellular carcinoma patients with diabetes mellitus. World J Gastroenterol. 2013;19:2395-2403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
13.  Madkhali AA, Fadel ZT, Aljiffry MM, Hassanain MM. Surgical treatment for hepatocellular carcinoma. Saudi J Gastroenterol. 2015;21:11-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
14.  El-Serag HB, Davila JA, Petersen NJ, McGlynn KA. The continuing increase in the incidence of hepatocellular carcinoma in the United States: an update. Ann Intern Med. 2003;139:817-823.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Yuen MF, Hou JL, Chutaputti A. Hepatocellular carcinoma in the Asia pacific region. J Gastroenterol Hepatol. 2009;24:346-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 330]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
16.  Davila JA, Morgan RO, Shaib Y, McGlynn KA, El-Serag HB. Diabetes increases the risk of hepatocellular carcinoma in the United States: a population based case control study. Gut. 2005;54:533-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 480]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
17.  Schuppan D, Afdhal NH. Liver cirrhosis. Lancet. 2008;371:838-851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1446]  [Cited by in F6Publishing: 1428]  [Article Influence: 89.3]  [Reference Citation Analysis (0)]
18.  Hirokawa F, Hayashi M, Asakuma M, Shimizu T, Inoue Y, Uchiyama K. Risk factors and patterns of early recurrence after curative hepatectomy for hepatocellular carcinoma. Surg Oncol. 2016;25:24-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
19.  Yang WT, Wu LW, Tseng TC, Chen CL, Yang HC, Su TH, Wang CC, Kuo SF, Liu CH, Chen PJ. Hepatitis B Surface Antigen Loss and Hepatocellular Carcinoma Development in Patients With Dual Hepatitis B and C Infection. Medicine (Baltimore). 2016;95:e2995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
20.  Gao C, Fang L, Zhao HC, Li JT, Yao SK. Potential role of diabetes mellitus in the progression of cirrhosis to hepatocellular carcinoma: a cross-sectional case-control study from Chinese patients with HBV infection. Hepatobiliary Pancreat Dis Int. 2013;12:385-393.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Gao C, Yao SK. Diabetes mellitus: a “true” independent risk factor for hepatocellular carcinoma? Hepatobiliary Pancreat Dis Int. 2009;8:465-473.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Gao C, Zhao HC, Li JT, Yao SK. Diabetes mellitus and hepatocellular carcinoma: comparison of Chinese patients with and without HBV-related cirrhosis. World J Gastroenterol. 2010;16:4467-4475.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Zhang H, Gao C, Fang L, Zhao HC, Yao SK. Metformin and reduced risk of hepatocellular carcinoma in diabetic patients: a meta-analysis. Scand J Gastroenterol. 2013;48:78-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 89]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
24.  Lawson DH, Gray JM, McKillop C, Clarke J, Lee FD, Patrick RS. Diabetes mellitus and primary hepatocellular carcinoma. Q J Med. 1986;61:945-955.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Fujino Y, Mizoue T, Tokui N, Yoshimura T. Prospective study of diabetes mellitus and liver cancer in Japan. Diabetes Metab Res Rev. 2001;17:374-379.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  El-Serag HB, Richardson PA, Everhart JE. The role of diabetes in hepatocellular carcinoma: a case-control study among United States Veterans. Am J Gastroenterol. 2001;96:2462-2467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 182]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
27.  Yu L, Sloane DA, Guo C, Howell CD. Risk factors for primary hepatocellular carcinoma in black and white Americans in 2000. Clin Gastroenterol Hepatol. 2006;4:355-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
28.  Kessler II. Cancer mortality among diabetics. J Natl Cancer Inst. 1970;44:673-686.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Ragozzino M, Melton LJ, Chu CP, Palumbo PJ. Subsequent cancer risk in the incidence cohort of Rochester, Minnesota, residents with diabetes mellitus. J Chronic Dis. 1982;35:13-19.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Lu SN, Lin TM, Chen CJ, Chen JS, Liaw YF, Chang WY, Hsu ST. A case-control study of primary hepatocellular carcinoma in Taiwan. Cancer. 1988;62:2051-2055.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Adami HO, Chow WH, Nyrén O, Berne C, Linet MS, Ekbom A, Wolk A, McLaughlin JK, Fraumeni JF. Excess risk of primary liver cancer in patients with diabetes mellitus. J Natl Cancer Inst. 1996;88:1472-1477.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  El-Serag HB, Tran T, Everhart JE. Diabetes increases the risk of chronic liver disease and hepatocellular carcinoma. Gastroenterology. 2004;126:460-468.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Dyal HK, Aguilar M, Bartos G, Holt EW, Bhuket T, Liu B, Cheung R, Wong RJ. Diabetes Mellitus Increases Risk of Hepatocellular Carcinoma in Chronic Hepatitis C Virus Patients: A Systematic Review. Dig Dis Sci. 2016;61:636-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
34.  Arase Y, Kobayashi M, Suzuki F, Suzuki Y, Kawamura Y, Akuta N, Kobayashi M, Sezaki H, Saito S, Hosaka T. Effect of type 2 diabetes on risk for malignancies includes hepatocellular carcinoma in chronic hepatitis C. Hepatology. 2013;57:964-973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 138]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
35.  Wang CS, Yao WJ, Chang TT, Wang ST, Chou P. The impact of type 2 diabetes on the development of hepatocellular carcinoma in different viral hepatitis statuses. Cancer Epidemiol Biomarkers Prev. 2009;18:2054-2060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 85]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
36.  Chen CL, Yang HI, Yang WS, Liu CJ, Chen PJ, You SL, Wang LY, Sun CA, Lu SN, Chen DS. Metabolic factors and risk of hepatocellular carcinoma by chronic hepatitis B/C infection: a follow-up study in Taiwan. Gastroenterology. 2008;135:111-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 386]  [Cited by in F6Publishing: 406]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
37.  Kawamura Y, Arase Y, Ikeda K, Hirakawa M, Hosaka T, Kobayashi M, Saitoh S, Yatsuji H, Sezaki H, Akuta N. Diabetes enhances hepatocarcinogenesis in noncirrhotic, interferon-treated hepatitis C patients. Am J Med. 2010;123:951-956.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
38.  Ko WH, Chiu SY, Yang KC, Chen HH. Diabetes, hepatitis virus infection and hepatocellular carcinoma: A case-control study in hepatitis endemic area. Hepatol Res. 2012;42:774-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
39.  Konishi I, Hiasa Y, Shigematsu S, Hirooka M, Furukawa S, Abe M, Matsuura B, Michitaka K, Horiike N, Onji M. Diabetes pattern on the 75 g oral glucose tolerance test is a risk factor for hepatocellular carcinoma in patients with hepatitis C virus. Liver Int. 2009;29:1194-1201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
40.  Ohata K, Hamasaki K, Toriyama K, Matsumoto K, Saeki A, Yanagi K, Abiru S, Nakagawa Y, Shigeno M, Miyazoe S. Hepatic steatosis is a risk factor for hepatocellular carcinoma in patients with chronic hepatitis C virus infection. Cancer. 2003;97:3036-3043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 239]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
41.  van den Tweel JG, Taylor CR. A brief history of pathology: Preface to a forthcoming series that highlights milestones in the evolution of pathology as a discipline. Virchows Arch. 2010;457:3-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 63]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
42.  Jiang C, Gu J. History and current state of pathology in China. Virchows Arch. 2013;463:599-608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
43.  Campbell IL, Harrison LC. Molecular pathology of type 1 diabetes. Mol Biol Med. 1990;7:299-309.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Adorini L, Gregori S, Harrison LC. Understanding autoimmune diabetes: insights from mouse models. Trends Mol Med. 2002;8:31-38.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Wiltshire S, Bell JT, Groves CJ, Dina C, Hattersley AT, Frayling TM, Walker M, Hitman GA, Vaxillaire M, Farrall M. Epistasis between type 2 diabetes susceptibility Loci on chromosomes 1q21-25 and 10q23-26 in northern Europeans. Ann Hum Genet. 2006;70:726-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
46.  Lima SM, Grisi DC, Kogawa EM, Franco OL, Peixoto VC, Gonçalves-Júnior JF, Arruda MP, Rezende TM. Diabetes mellitus and inflammatory pulpal and periapical disease: a review. Int Endod J. 2013;46:700-709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
47.  Tenesa A, Dunlop MG. New insights into the aetiology of colorectal cancer from genome-wide association studies. Nat Rev Genet. 2009;10:353-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 320]  [Article Influence: 22.9]  [Reference Citation Analysis (0)]
48.  Fletcher O, Houlston RS. Architecture of inherited susceptibility to common cancer. Nat Rev Cancer. 2010;10:353-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 148]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
49.  Sandhu MS. Insulin-like growth factor-I and risk of type 2 diabetes and coronary heart disease: molecular epidemiology. Endocr Dev. 2005;9:44-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
50.  Gallagher EJ, LeRoith D. Epidemiology and molecular mechanisms tying obesity, diabetes, and the metabolic syndrome with cancer. Diabetes Care. 2013;36 Suppl 2:S233-S239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 100]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
51.  Field AE, Camargo CA, Ogino S. The merits of subtyping obesity: one size does not fit all. JAMA. 2013;310:2147-2148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 75]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
52.  Wu ZY, Wang MH, Qi HM, Wu MH, Ge YZ, Li HT. Relationship between hOGG1 Ser326Cys gene polymorphism and coronary artery lesions in patients with diabetes mellitus. Int J Clin Exp Med. 2015;8:18629-18637.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Remo A, Pancione M, Zanella C, Vendraminelli R. Molecular pathology of colorectal carcinoma. A systematic review centred on the new role of the pathologist. Pathologica. 2012;104:432-441.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Paral J, Slaninka I, Kalabova H, Hadzi-Nikolov D. Gastrointestinal stromal tumors: review on morphology, molecular pathology, diagnostics, prognosis and treatment options. Acta Gastroenterol Belg. 2010;73:349-359.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Ray A, Manjila S, Hdeib AM, Radhakrishnan A, Nock CJ, Cohen ML, Sloan AE. Extracranial metastasis of gliobastoma: Three illustrative cases and current review of the molecular pathology and management strategies. Mol Clin Oncol. 2015;3:479-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
56.  Ogino S, Goel A. Molecular classification and correlates in colorectal cancer. J Mol Diagn. 2008;10:13-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 341]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
57.  Baudhuin LM, Donato LJ, Uphoff TS. How novel molecular diagnostic technologies and biomarkers are revolutionizing genetic testing and patient care. Expert Rev Mol Diagn. 2012;12:25-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
58.  Roukos DH. Novel clinico-genome network modeling for revolutionizing genotype-phenotype-based personalized cancer care. Expert Rev Mol Diagn. 2010;10:33-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 94]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
59.  Metodiev MV. Biomarkers research in Europe: focus on personalized medicine. Expert Rev Mol Diagn. 2011;11:689-690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
60.  Hamilton SR. Targeted therapy of cancer: new roles for pathologists in colorectal cancer. Mod Pathol. 2008;21 Suppl 2:S23-S30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
61.  Gulley ML, Braziel RM, Halling KC, Hsi ED, Kant JA, Nikiforova MN, Nowak JA, Ogino S, Oliveira A, Polesky HF. Clinical laboratory reports in molecular pathology. Arch Pathol Lab Med. 2007;131:852-863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
62.  Tonellato PJ, Crawford JM, Boguski MS, Saffitz JE. A national agenda for the future of pathology in personalized medicine: report of the proceedings of a meeting at the Banbury Conference Center on genome-era pathology, precision diagnostics, and preemptive care: a stakeholder summit. Am J Clin Pathol. 2011;135:668-672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 36]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
63.  Dolle JM, Daling JR, White E, Brinton LA, Doody DR, Porter PL, Malone KE. Risk factors for triple-negative breast cancer in women under the age of 45 years. Cancer Epidemiol Biomarkers Prev. 2009;18:1157-1166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 175]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
64.  Trivers KF, Lund MJ, Porter PL, Liff JM, Flagg EW, Coates RJ, Eley JW. The epidemiology of triple-negative breast cancer, including race. Cancer Causes Control. 2009;20:1071-1082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 238]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
65.  Pervaiz F, Rehmani S, Majid S, Anwar H. Evaluation of Hormone Receptor Status (ER/PR/HER2-neu) in Breast Cancer in Pakistan. J Pak Med Assoc. 2015;65:747-752.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Ichihara E, Lovly CM. Shades of T790M: Intratumor Heterogeneity in EGFR-Mutant Lung Cancer. Cancer Discov. 2015;5:694-696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
67.  Li W, Qu J, Xu Z. Clinical features and mutation status of EGFR, KRAS, BRAF, EML4-ALK and ROS1 between surgical resection samples and non surgical resection samples in lung cancer. J Thorac Dis. 2015;7:875-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
68.  Webber EM, Kauffman TL, O’Connor E, Goddard KA. Systematic review of the predictive effect of MSI status in colorectal cancer patients undergoing 5FU-based chemotherapy. BMC Cancer. 2015;15:156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 77]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
69.  Yamane LS, Scapulatempo-Neto C, Alvarenga L, Oliveira CZ, Berardinelli GN, Almodova E, Cunha TR, Fava G, Colaiacovo W, Melani A. KRAS and BRAF mutations and MSI status in precursor lesions of colorectal cancer detected by colonoscopy. Oncol Rep. 2014;32:1419-1426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
70.  Genther Williams SM, Kuznicki AM, Andrade P, Dolinski BM, Elbi C, O’Hagan RC, Toniatti C. Treatment with the PARP inhibitor, niraparib, sensitizes colorectal cancer cell lines to irinotecan regardless of MSI/MSS status. Cancer Cell Int. 2015;15:14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
71.  Rastogi A, Tan SH, Mohamed AA, Chen Y, Hu Y, Petrovics G, Sreenath T, Kagan J, Srivastava S, McLeod DG. Functional antagonism of TMPRSS2-ERG splice variants in prostate cancer. Genes Cancer. 2014;5:273-284.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Esteller M. Epigenetics in cancer. N Engl J Med. 2008;358:1148-1159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2672]  [Cited by in F6Publishing: 2493]  [Article Influence: 155.8]  [Reference Citation Analysis (0)]
73.  Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2599]  [Cited by in F6Publishing: 2647]  [Article Influence: 132.4]  [Reference Citation Analysis (0)]
74.  Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318:1108-1113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2283]  [Cited by in F6Publishing: 2213]  [Article Influence: 130.2]  [Reference Citation Analysis (0)]
75.  Ojanguren I, Castellà E, Llatjós M, Ariza A, Navas Palacios JJ. p53 immunoreaction in hepatocellular carcinoma and its relationship to etiologic factors. A fine needle aspiration study. Acta Cytol. 1996;40:1148-1153.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, Osterreicher CH, Takahashi H, Karin M. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 2010;140:197-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1169]  [Cited by in F6Publishing: 1283]  [Article Influence: 91.6]  [Reference Citation Analysis (1)]
77.  Jiang N, Sun R, Sun Q. Leptin signaling molecular actions and drug target in hepatocellular carcinoma. Drug Des Devel Ther. 2014;8:2295-2302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
78.  Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet. 2012;379:1245-1255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3249]  [Cited by in F6Publishing: 3477]  [Article Influence: 289.8]  [Reference Citation Analysis (3)]
79.  Yu XJ, Fang F, Tang CL, Yao L, Yu L, Yu L. dbHCCvar: a comprehensive database of human genetic variations in hepatocellular carcinoma. Hum Mutat. 2011;32:E2308-E2316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
80.  Wei Y, Liu F, Li B, Chen X, Ma Y, Yan L, Wen T, Xu M, Wang W, Yang J. Polymorphisms of tumor necrosis factor-alpha and hepatocellular carcinoma risk: a HuGE systematic review and meta-analysis. Dig Dis Sci. 2011;56:2227-2236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 34]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
81.  Wei YG, Liu F, Li B, Chen X, Ma Y, Yan LN, Wen TF, Xu MQ, Wang WT, Yang JY. Interleukin-10 gene polymorphisms and hepatocellular carcinoma susceptibility: a meta-analysis. World J Gastroenterol. 2011;17:3941-3947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 27]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
82.  Wang B, Huang G, Wang D, Li A, Xu Z, Dong R, Zhang D, Zhou W. Null genotypes of GSTM1 and GSTT1 contribute to hepatocellular carcinoma risk: evidence from an updated meta-analysis. J Hepatol. 2010;53:508-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 101]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
83.  Tanabe KK, Lemoine A, Finkelstein DM, Kawasaki H, Fujii T, Chung RT, Lauwers GY, Kulu Y, Muzikansky A, Kuruppu D. Epidermal growth factor gene functional polymorphism and the risk of hepatocellular carcinoma in patients with cirrhosis. JAMA. 2008;299:53-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 160]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
84.  Jin F, Qu LS, Shen XZ. Association between C282Y and H63D mutations of the HFE gene with hepatocellular carcinoma in European populations: a meta-analysis. J Exp Clin Cancer Res. 2010;29:18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
85.  Miki D, Ochi H, Hayes CN, Abe H, Yoshima T, Aikata H, Ikeda K, Kumada H, Toyota J, Morizono T. Variation in the DEPDC5 locus is associated with progression to hepatocellular carcinoma in chronic hepatitis C virus carriers. Nat Genet. 2011;43:797-800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 124]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
86.  Yang HI, Yeh SH, Chen PJ, Iloeje UH, Jen CL, Su J, Wang LY, Lu SN, You SL, Chen DS. Associations between hepatitis B virus genotype and mutants and the risk of hepatocellular carcinoma. J Natl Cancer Inst. 2008;100:1134-1143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 447]  [Cited by in F6Publishing: 458]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
87.  Kim SM, Leem SH, Chu IS, Park YY, Kim SC, Kim SB, Park ES, Lim JY, Heo J, Kim YJ. Sixty-five gene-based risk score classifier predicts overall survival in hepatocellular carcinoma. Hepatology. 2012;55:1443-1452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 100]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
88.  Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, Calderaro J, Bioulac-Sage P, Letexier M, Degos F. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012;44:694-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 962]  [Cited by in F6Publishing: 1059]  [Article Influence: 88.3]  [Reference Citation Analysis (0)]
89.  Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, Aoki M, Hosono N, Kubo M, Miya F. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 2012;44:760-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 631]  [Cited by in F6Publishing: 681]  [Article Influence: 56.8]  [Reference Citation Analysis (0)]
90.  Hoshida Y, Nijman SM, Kobayashi M, Chan JA, Brunet JP, Chiang DY, Villanueva A, Newell P, Ikeda K, Hashimoto M. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res. 2009;69:7385-7392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 753]  [Cited by in F6Publishing: 851]  [Article Influence: 56.7]  [Reference Citation Analysis (0)]
91.  Yamashita T, Forgues M, Wang W, Kim JW, Ye Q, Jia H, Budhu A, Zanetti KA, Chen Y, Qin LX. EpCAM and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma. Cancer Res. 2008;68:1451-1461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 545]  [Cited by in F6Publishing: 561]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
92.  Hoshida Y, Toffanin S, Lachenmayer A, Villanueva A, Minguez B, Llovet JM. Molecular classification and novel targets in hepatocellular carcinoma: recent advancements. Semin Liver Dis. 2010;30:35-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 243]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
93.  Villanueva A, Newell P, Chiang DY, Friedman SL, Llovet JM. Genomics and signaling pathways in hepatocellular carcinoma. Semin Liver Dis. 2007;27:55-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 406]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
94.  Hoshida Y, Moeini A, Alsinet C, Kojima K, Villanueva A. Gene signatures in the management of hepatocellular carcinoma. Semin Oncol. 2012;39:473-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
95.  Ueyama M, Nishida N, Korenaga M, Korenaga K, Kumagai E, Yanai H, Adachi H, Katsuyama H, Moriyama S, Hamasaki H. The impact of PNPLA3 and JAZF1 on hepatocellular carcinoma in non-viral hepatitis patients with type 2 diabetes mellitus. J Gastroenterol. 2016;51:370-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
96.  Capone F, Guerriero E, Colonna G, Maio P, Mangia A, Marfella R, Paolisso G, Izzo F, Potenza N, Tomeo L. The Cytokinome Profile in Patients with Hepatocellular Carcinoma and Type 2 Diabetes. PLoS One. 2015;10:e0134594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
97.  Qiao G, Le Y, Li J, Wang L, Shen F. Glycogen synthase kinase-3β is associated with the prognosis of hepatocellular carcinoma and may mediate the influence of type 2 diabetes mellitus on hepatocellular carcinoma. PLoS One. 2014;9:e105624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
98.  Stueck AE, Qu Z, Huang MA, Campreciós G, Ferrell LD, Thung SN. Hepatocellular Carcinoma Arising in an HNF-1α-Mutated Adenoma in a 23-Year-Old Woman with Maturity-Onset Diabetes of the Young: A Case Report. Semin Liver Dis. 2015;35:444-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
99.  Ali Kamkar MM, Ahmad R, Alsmadi O, Behbehani K. Insight into the impact of diabetes mellitus on the increased risk of hepatocellular carcinoma: mini-review. J Diabetes Metab Disord. 2014;13:57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
100.  Facciorusso A. The influence of diabetes in the pathogenesis and the clinical course of hepatocellular carcinoma: recent findings and new perspectives. Curr Diabetes Rev. 2013;9:382-386.  [PubMed]  [DOI]  [Cited in This Article: ]