Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 21, 2016; 22(27): 6100-6113
Published online Jul 21, 2016. doi: 10.3748/wjg.v22.i27.6100
Diabetes mellitus and metformin in hepatocellular carcinoma
Koji Fujita, Hisaaki Miyoshi, Joji Tani, Kyoko Oura, Tomoko Tadokoro, Teppei Sakamoto, Takako Nomura, Asahiro Morishita, Hirohito Yoneyama, Tsutomu Masaki, Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa 760-8521, Japan
Hisakazu Iwama, Life Science Research Center, Kagawa University, Kagawa 760-8521, Japan
Author contributions: All authors made contributions to this manuscript.
Conflict-of-interest statement: The authors declare no conflicts 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: Tsutomu Masaki, Professor, Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Ikenobe 1750-1, Miki, Kita, Kagawa 760-8521, Japan. tmasaki@med.kagawa-u.ac.jp
Telephone: +81-87-8912156 Fax: +81-87-8912158
Received: March 28, 2016
Peer-review started: March 29, 2016
First decision: May 12, 2016
Revised: May 25, 2016
Accepted: June 15, 2016
Article in press: June 15, 2016
Published online: July 21, 2016
Processing time: 108 Days and 19.7 Hours

Abstract

Hepatocellular carcinoma (HCC) is the leading cause of cancer-related death worldwide. Diabetes mellitus, a risk factor for cancer, is also globally endemic. The clinical link between these two diseases has been the subject of investigation for a century, and diabetes mellitus has been established as a risk factor for HCC. Accordingly, metformin, a first-line oral anti-diabetic, was first proposed as a candidate anti-cancer agent in 2005 in a cohort study in Scotland. Several subsequent large cohort studies and randomized controlled trials have not demonstrated significant efficacy for metformin in suppressing HCC incidence and mortality in diabetic patients; however, two recent randomized controlled trials have reported positive data for the tumor-preventive potential of metformin in non-diabetic subjects. The search for biological links between cancer and diabetes has revealed intracellular pathways that are shared by cancer and diabetes. The signal transduction mechanisms by which metformin suppresses carcinogenesis in cell lines or xenograft tissues and improves chemoresistance in cancer stem cells have also been elucidated. This review addresses the clinical and biological links between HCC and diabetes mellitus and the anti-cancer activity of metformin in clinical studies and basic experiments.

Key Words: Hepatocellular carcinoma, Diabetes mellitus, Metformin, Risk

Core tip: Diabetes mellitus, an increasing risk factor for hepatocellular carcinoma (HCC), shares pathological mechanisms with HCC. Thus, the first-line anti-diabetic metformin was anticipated to reduce cancer risk. Though basic research has provided evidence of its anti-cancer effect, clinical studies of diabetic patients have not provided conclusive data that metformin reduces HCC risk. Clinical studies have suggested that metformin may suppress cancer in non-diabetic subjects. Basic research on cancer stem cell-targeting therapies has also examined the potential of metformin.



INTRODUCTION

Hepatocellular carcinoma (HCC) is the sixth most frequent cancer worldwide and the third most common cause of cancer death[1,2]. This primary liver cancer originating from hepatocytes is characterized by poor prognosis, and patients with HCC incident risks must be monitored closely for HCC occurrence[3]. Patients in the early stage of malignancy onset can be cured by surgery[4-6] or radiofrequency ablation therapy[7,8] However, HCC recurs in 60%-80% of patients within 5 years of curative treatments[9-12] due to intrahepatic metastasis or multicentric occurrence facilitated by the long-term exposure of the liver to chronic viral hepatitis, fibrotic changes and hyperinsulinemia. Patients who do not meet the criteria for surgical resection or radiofrequency ablation therapy can be treated with either transcatheter arterial chemoembolization therapy[13,14], an anti-cancer drug and lipiodol emulsion[15], or drug-eluting beads[16] with the intention of downstaging the tumor to enable hepatic resection or await liver transplantation. Liver transplantation, a radical curative surgery with a 3-year overall survival rate of approximately 40%, has been established as a standard treatment for HCC using the Milan criteria[17]. However, patients who require liver transplantation must confront a shortage of donors and progression of their cancer stage while awaiting a donor, particularly in Asia[18]. Patients who fail or are excluded from any of the above treatments might receive sorafenib, a multi-kinase inhibitor that has exhibited effectiveness for improving HCC patient prognosis in randomized control trials[19,20] but improves survival by only 3 mo.

Therefore, the prevention of HCC and the intense screening of high-risk individuals are of great importance. The risk factors for HCC are now well-established, and great efforts have been made to decrease HCC prevalence, recurrence and death. The medical histories of HCC patients range from liver-specific viral infections, such as HBV and HCV, and alcohol consumption to metabolic disorders including diabetes mellitus and obesity[21]. Effective anti-viral agents for HBV and HCV significantly suppressed HCC prevalence in clinical trials[22-25]. However, type 2 diabetes mellitus, characterized by hyperinsulinemia in its early stage and usually linked to obesity, is increasing worldwide.

Metformin, an oral anti-diabetic drug that is less expensive than any other anti-cancer agent in use, first attracted attention in 2005 for its potential to suppress not only serum glucose levels but also the incidence of various cancers in an observational study[26]. Metformin has subsequently been investigated as an anti-cancer medicine for malignancies including HCC in diabetic and non-diabetic subjects. This review describes the links between HCC and type 2 diabetes mellitus in terms of their epidemiology and pathology and addresses the benefits and limitations of metformin in the prevention and treatment of HCC.

DIABETES MELLITUS IN HCC EPIDEMIOLOGICAL TRENDS

Some trends of HCC epidemiology have recently attracted attention. First, the increase in HCC incidence rates has ceased in two nations, Japan and the United States[27-29], though the prevalence of HCC and HCC-related deaths will continue to increase in the future[2,30]. The restriction of these phenomena to these two developed countries reflects the diversity of HCC incidence with geography, socioeconomic condition, race, generation and gender in these two countries[31]. In Japan, liver cancer incidence rates rapidly increased between the mid-1970s and 1990s but then leveled off and began to decrease by 2003[27,32]. This trend in a Japanese city has been attributed to a decline in HCV infection, previously the major cause of HCC in Japan, partly by avoiding HCV-contaminated blood transfusion and suppression and by restraining injections of drugs of abuse after the Second World War. In the United States, the absence of a significant increase in HCC incidence rates is due in part to the decreased prevalence of HCC among the largest group with an HCC risk caused by HBV, Asians/Pacific islanders, particularly men aged 35-49 years[28].

Second, a frequent risk factor for HCC, the hepatitis C virus, can now be more easily and thoroughly eradicated by direct anti-viral agents[24,25], which are newly developed oral medications that might affect HCC prevalence more strongly than interferon[33,34]. HBV is another hepatocarcinogenic virus that infects hundreds of millions of people globally. The replication and inflammatory activity of HBV in a HBV-infected liver can be inhibited by the newly developed nucleotide analogues entecavir[22,23] and tenofovir[35]. These two retrograde transcriptional viral inhibitors have largely overcome the disadvantages of the previous nucleotide analogues, lamivudine and adefovir, which usually caused viral mutations and drug resistance in 70% of patients who took lamivudine for 4 years[36] and in 20% of patients who were prescribed adefovir for 5 years[37,38].

Third, the prevalence of another HCC risk factor, type 2 diabetes mellitus, continues to increase[39,40] with the increase in the incidence of obesity[41]. The number of diabetic patients is estimated to increase to 300-400 million worldwide by 2030[39,40,42]. Such predictions are based on increases in populations living in urban areas in developing countries, in senior diabetic patients and in obesity.

DIABETES MELLITUS AS A RISK FACTOR FOR HCC

Diabetes mellitus was first investigated as a risk factor for cancer death at the beginning of the 20th century, when the etiologies of these two major deadly diseases were unknown. An observational study addressing cancer deaths in United States cities in 1910 concluded that the correlation between cancer and diabetes mellitus was not fortuitous nor due merely to errors of observation[43], although cancer prevalence at that time was biased by the availability of medical schools at which post-mortem examinations and cancer diagnoses could be performed, as discussed by Greenwood[44]. Greenwood himself analyzed the correlation between death rates due to diabetes and cancer. He concluded that cancer mortality was significantly associated with diabetes in the United States and not in Europe, but he did not assess the correlation between organ-specific cancer deaths and diabetes[44].

Among various organ-specific cancers, pancreatic cancer was initially determined to have coincidence with diabetic conditions in an observation of 10000 diabetic patients in 1934, although that conclusion may have been due to reverse causation[45]. In 1970, Kessler reported an association between pancreatic cancer mortality and diabetes mellitus prevalence in human males, although no excess deaths via any other type of cancer were observed among diabetic patients; this phenomenon occurred in part because diabetic patients died from diabetes itself or from cardiovascular diseases before cancers other than those of pancreatic origin became fatal[46].

The strength of the association between cancers and diabetes depends on the cancer species; however, a hospital-based case control study in 1986 demonstrated that more than 4 times as many HCC patients suffered from diabetes mellitus than colorectal tumor patients and femoral bone fracture patients[47]. A large population-based cohort study in Uppsala, Sweden, confirmed the significantly increased risk of HCC as well as pancreatic cancer in diabetic patients with a relative ratio of approximately 1.5, which was higher in males than in females[48]. The link between diabetes mellitus and the larger HCC population is supported by two major prospective cohort studies, one in Sweden[49] and the other in Denmark[50], followed by another case-control study in Italy[51]. Diabetes mellitus has subsequently been investigated as a risk factor for the prevalence[52,53], recurrence[54-57] and mortality[58,59] of HCC.

Diabetes mellitus is now considered an independent risk factor for HCC[60,61] and has been proven to increase the risk of HCC even in those not infected with HBV or HCV[57,62,63]. Increased incidence and mortality of several malignancies other than HCC, including pancreatic cancer, endometrial cancer and colon cancer, have been observed among diabetic patients in a series of studies[48,58,64], partly based on obesity, which has also been identified as a risk factor for cancers including HCC[65,66]. An association between post-load plasma glucose in a non-diabetic individual who has the potential to develop diabetes mellitus and cancer mortality due to HCC has also been suggested[67,68].

These observational studies assessing the potential role of diabetes mellitus as a risk factor for cancers were not free from detection bias and reverse causation; the cancer risk was highest immediately after the diabetes cases were registered or diagnosed in each study and then decreased gradually with time[69]. However, the risks of HCC, pancreatic cancer and endometrial cancer remained significant after adjusting for detection bias and reverse causation[70]. Major clinical studies of the relationships between diabetes mellitus and HCC and other cancers are illustrated in Table 1.

Table 1 The influence of diabetes mellitus on the incidence, recurrence, and mortality of hepatocellular carcinoma.
Ref.YearStudy designType of diabetesResults
Maynard[43]1910Case-controlNot differentiatedCancer mortality increased
Greenwood and Wood[44]1914Case-controlNot differentiatedCancer mortality increased in American cities; no significant correlation was observed in European cities
Marble[45]1934Case-controlNot differentiatedPancreatic cancer incidence increased
Kessler[46]1970Case-controlNot differentiatedPancreatic cancer deaths increased
Lawson et al[47]1986Case-controlNot differentiatedHCC incidence increased (HR = 3.9)
Levine et al[67]1990CohortIGTHCC deaths increased in men; post-load plasma glucose increased
Adami et al[48]1991CohortNot differentiatedIncidences of primary liver (RR = 1.5), pancreatic (RR = 1.4) and endometrial (RR = 1.5) cancers increased
Smith et al[164]1992CohortIGTPancreatic cancer increased (RR = 2.25); post-load plasma glucose increased in IGT men. HCC was not analyzed in organ-specific statistics
La Vecchia et al[51]1994Case-controlNot differentiatedLiver cancer incidence remained elevated 10 yr after the diagnosis of diabetes (RR = 2.6)
Adami et al[49]1996CohortNot differentiatedPrimary liver cancer incidence increased (SIR = 4.7 in men and 3.4 in women)
Wideroff et al[50]1997CohortNot differentiatedPrimary liver cancer incidence increased (SIR = 4.0 in men and 2.1 in women)
La Vecchia et al[165]1997Case-controlNot differentiatedLiver cancer incidence increased (OR = 2.2) for at least 10 yr after the diagnosis of diabetes
Ikeda et al[54]1998CohortNot differentiatedRecurrence-free survival after hepatic resection decreased in diabetic cases
Balkau et al[52]2001CohortNot differentiatedHCC incidence increased with fasting hyperinsulinemia (HR = 2.72) and 2-h hyperinsulinemia (HR = 3.41)
Huo et al[55]2003CohortNot differentiatedHCC recurrence increased in HBV-seropositive cases
Coughlin et al[58]2004CohortNot differentiatedLiver cancer mortality increased in men (RR = 2.19)
Batty et al[68]2004CohortIGTHCC (HR = 2.47) and pancreatic cancer (HR = 1.35) increased; post-load plasma glucose increased in IGT men
El-Serag et al[60]2006Meta-analysisNot differentiatedHCC incidence increased in 9 case-control studies (OR = 2.5) and 7 cohort studies (OR = 2.5)
Inoue et al[64]2006CohortNot differentiatedHCC incidence increased (HR = 2.24 in men and 1.94 in women)
Komura et al[56]2007CohortNot differentiatedPostoperative recurrence-free survival decreased in diabetic cases
Kawamura et al[57]2008CohortNot differentiatedHCC recurrence increased (HR = 4.61)
Landman et al[59]2010CohortType 2HCC death increased (SMR = 1.47)
Lee et al[53]2011CohortType 2Incidences of total cancer, HCC and pancreatic cancer increased
Hense et al[166]2011CohortType 2HCC incidence increased (SIR = 1.94)
Johnson et al[70]2011CohortType 2After detection biases were excluded, incidences of HCC (HR = 2.53), pancreatic (HR = 1.65) and endometrial (HR = 1.58) cancers increased
Wang et al[167]2012Meta-analysisType 1 and type 2HCC incidence (RR = 2.23) and mortality (RR = 2.43) increased in cohort studies
Wang et al[168]2012Meta-analysisNot differentiatedHCC incidence (RR = 2.01) and mortality (RR = 1.56) increased
Lai et al[101]2012CohortNot differentiatedHCC incidence increased (HR = 1.73)
Schlesinger et al[62]2013CohortNot differentiatedHCC incidence increased (RR = 2.17) in HBV/HCV-negative individuals
Koh et al[63]2013CohortNot differentiatedHCC incidence increased (HR = 2.14), particularly in non-viral cases (HR = 5.15)
Wang et al[169]2014Meta-analysisNot differentiatedHCC in diabetic cases was related to overall survival (RR = 1.46) and disease-free survival (RR = 1.57)
Harding et al[69]2015Case-controlType 1 and type 2Incidences of total, liver, pancreatic and endometrial cancer increased in cases involving type 2 diabetes mellitus
BIOLOGICAL LINKAGE BETWEEN DIABETES MELLITUS AND CANCER

In 1910, Maynard hypothesized that cancer occurrence might be due to meteorological conditions, such as hours of sunshine, mean temperature, rainfall and other indicators, but observed no significant correlations[43]. He subsequently focused on diabetes mellitus as a possible cause of cancer because the two diseases occurred at similar ages, were increasing in prevalence and had no known etiologies at that time, as discussed by Greenwood[44]. The subsequent body of research has since established that the etiologies of diabetes mellitus and cancer share a number of biological pathways[61], some of which are based on central obesity and insulin resistance, common risk factors for both diseases[71].

The insulin/IGF-1 axis involves over-activation of mTOR

The classical pathways shared by diabetes mellitus and cancer are the Insulin/IGF- axis, including over-activation of mTOR. Type 2 diabetes mellitus is characterized by hyperinsulinemia. Insulin exerts its proliferative effects directly through the insulin receptor (IR) and indirectly by increasing circulating levels of IGF-1[72]. Insulin increases circulating IGF-1 by decreasing hepatic production of IGF-binding protein 1, a ligand of IGF-1, thus increasing levels of free IGF-1[73,74]. In hyperinsulinemia, the activity of insulin becomes less metabolic and more mitogenic. Insulin decreases its metabolic activity by over-activation of mTOR, which phosphorylates IR-substrate-1 and attenuates metabolic pathways downstream of insulin signals. Simultaneously, insulin up-regulates IR-substrate-2 and induces the mitogen-activated kinase pathway, thereby enhancing cell survival[75].

Hyperinsulinemia is regarded as an independent risk factor for HCC, and major dysregulations of insulin-dependent pathways have been reported in HCC[76]. The effect of HCC on development also depends on excess signals from IGF-I but more strongly on signals from IGF-II[77]. Aberrant mTOR signaling in HCC has been confirmed in human tumor samples[78].

Chronic inflammation caused by adipokines

Central obesity, which usually accompanies type 2 diabetes mellitus, may be a trigger for carcinogenesis via pro-inflammatory cytokines secreted from visceral adipose tissues. Adipokines such as tumor necrosis factor-α and interleukin-6 are produced in the excess visceral fatty compartment and perpetuate chronic low-grade inflammation in peripheral tissues, which provides microenvironments suitable for tumorigenesis[79]. Another adipose tissue-derived hormone, leptin, promotes or suppresses cell proliferation[80]. Adiponectin, which is produced most highly by adipokines, presents both anti-inflammatory and anti-tumor activities[81].

However, in the case of HCC, the carcinogenic or anti-tumor effects of the two hormones, leptin and adiponectin, have been the subject of contradictory reports, and more conclusive data are needed[82].

Hyperglycemia

Hyperglycemia is a feature of diabetes mellitus, and Warburg first hypothesized that hyperglycemia itself might have carcinogenic potential[83]. In general, cancer cells produce ATP by anaerophilic glycolysis. Under aerophilic conditions, cytoplasmic ATP production via glycolysis is less efficient than synthesis via oxidative phosphorylation in mitochondria. Cancer cells are consequently assumed to require more glucose than normal cells, and several types of cancers have been detected by positron emission tomography based on this theory[84].

Direct carcinogenic effects of hyperglycemia combined with the Wnt signaling pathway were recently proposed to promote carcinogenesis, resulting in nuclear beta-catenin accumulation[85] via aberrant acetylation of beta-catenins. A national cohort study in Taiwan revealed a linear relationship between HCC occurrence and HbA1c in hyperglycemia[86]. In addition, a case-control study in Japan demonstrated that post-challenge hyperglycemia was an independent risk factor for HCC[87].

However, hyperglycemia has been considered subordinate to hyperinsulinemia as a carcinogen, and a meta-analysis of large randomized controlled trials did not indicate definitive cancer risk reduction by intensive glycemic controls in patients with type 2 diabetes mellitus[88,89].

Estrogen

Estrogen is produced primarily in the body fat of postmenopausal women and obesity, a background metabolic disorder of diabetes mellitus, is linked to elevated serum estrogen levels. Therefore, estrogen is recognized as a carcinogenic risk factor for breast, endometrial and ovarian cancers in post-menopausal women[90].

In HCC, however, primary liver malignancies occur predominantly in males, and male HCC patients usually present with a poorer prognosis than female HCC patients[91]. Among women, post-menopausal women suffer from an elevated incidence of HCC, which is epidemiologically suppressed by estrogen therapy[92]. The relatively lower estrogen levels in males compared to females and in post-menopausal women compared to estrogen-supplemented women suggests that low estrogen might contribute to the more frequent cancer prevalence in men, particularly for HCC, because estrogen appears to suppress HCC development by inactivating chronic low-grade inflammation in the liver[93].

METFORMIN, A DRUG TO POTENTIALLY PREVENT HCC OCCURRENCE, RECURRENCE AND DEATH

Metformin, a first-line oral anti-diabetic[94], was associated with reduced prevalence of cancers in type 2 diabetic patients by Evans in 2005[26]. This pilot case-control study, which did not include site-specific cancer data, provided the foundation for epidemiological studies on the anti-tumor effects of metformin. As a matter of fact, an herb called Galega Officinalis, which contains large amounts of guanidine, the original molecule of metformin, was prescribed as long ago as the 17th century to relieve diabetic symptoms[95].

A population-based cohort study by Bowker et al[96] in 2006 demonstrated that cancer mortality in the type 2 diabetic group decreased when metformin was prescribed compared to insulin injection. A Scottish cohort study also demonstrated a protective effect of metformin against total cancers[97].

In the case of HCC, a case-control study suggested that HCC risk was reduced in male type 2 diabetic patients prescribed metformin[98] and a subsequent cohort study including male and female subjects[99]. A hospital-based case-control study in the United States also indicated that metformin reduces the incidence of HCC in type 2 diabetic patients[100]. A prospective cohort study in Taiwan performed by Lee et al[53] identified benefits of metformin for HCC prevention compared to other anti-diabetics, with a reduced risk of other tumors, pancreatic and colorectal cancer as well. Another cohort study in Taiwan also demonstrated that the development of HCC was suppressed by metformin administration[101].

However, observational studies of cancer incidence and mortality are subject to analyses of time-related biases, which have led to debate and controversy[102,103]. Two retrospective cohort studies observed no influence of metformin on cancer risk[104,105]. A meta-analysis excluding studies with time-related biases stated that metformin did not significantly reduce the risk of HCC. However, colon cancer was the only type of cancer that remained significant in a site-specific cancer risk analysis, and a 10% risk reduction for total cancers remained[106]. A randomized controlled trial comparing metformin to rosiglitazone failed to support a significant difference in cancer occurrence between the two oral anti-diabetics[107]. A meta-analysis of randomized controlled trials concluded that metformin provided patients with little benefit with respect to overall mortality compared to other anti-diabetics or insulin therapy and a 10% reduction in mortality compared to placebo or usual care that did not reach statistical significance[108].

In summary, the limited epidemiological research on the anti-cancer activity of metformin in diabetic patients indicates that this drug definitely exhibited an association with deceased cancer prevalence in case-control studies and cohort studies but has failed all randomized controlled trials in diabetic subjects[109,110]. No conclusive data from clinical trials regarding the prevention of cancers, including HCC in diabetic subjects, by metformin are available.

Clinical studies of metformin as an adjuvant to conventional chemotherapy and radiotherapy have reported promising data in case-control studies intended for patients with pancreatic cancers[111], breast cancers[112], lung cancers[113] and colorectal cancers[114]. However, a randomized controlled trial investigating the adjuvant use of metformin with conventional chemotherapy intended for pancreatic cancer failed to demonstrate a significant improvement of overall survival[115]. As an adjuvant to an estrogen-synthesis inhibitor, metformin is being prescribed to estrogen receptor-positive post-menopausal breast cancer patients without diabetes mellitus in a phase II randomized controlled trial[116]. No randomized controlled trials are available to demonstrate the feasibility of metformin as an adjuvant to non-surgical therapy.

For HCC, a combination of metformin and radiation therapy yielded prolonged overall survival compared to controls[117], although an adjuvant to sorafenib resulted in shorter progression-free survival and poorer overall survival[118]. As for primary cancer prevention, results on the adjuvant use of metformin in clinical trials are not conclusive, and further investigations are needed.

The application of metformin as an agent against premalignant tumor activity is being explored in non-diabetic cases to prevent tumor incidence. Metformin suppressed the incidence of colorectal aberrant crypt foci, surrogate markers of colon cancer, in non-diabetic subjects in a small randomized controlled trial[119]. A double-blind randomized controlled trial in Japan demonstrated a significant reduction in colorectal polyp formation in non-diabetic patients after 1 year of administration of low-dose metformin[120]. Similar trials on HCC have not been performed, and the anti-tumor activity of metformin in non-diabetic cases requires further elucidation. The major clinical studies of the anti-cancer effect of metformin against HCC and other cancer types are illustrated in Table 2.

Table 2 Efficacy of metformin on the incidence, recurrence and mortality of hepatocellular carcinoma and other tumors.
Ref.YearStudy designType of diabetesResults
Evans et al[26]2005Case-controlType 2HCC incidence decreased (OR = 0.79)
Bowker et al[96]2006CohortType 2Mortality was lower among metformin users than among insulin or sulfonylurea users (HR = 0.77)
Libby et al[97]2009CohortType 2Total cancer incidence decreased (HR = 0.63)
Donadon et al[98]2009Case-controlType 2HCC incidence was lower among metformin users (OR = 0.33) than among insulin users (OR = 2.99)
Donadon et al[99]2010CohortType 2HCC incidence was lower among metformin users (OR = 0.15) than among insulin or sulfonylurea users
Hassan et al[100]2010Case-controlNot differentiatedHCC incidence decreased (OR = 0.30)
Home et al[107]2010Randomized controlled trialType 2Total cancer incidence did not decrease compared with rosiglitazone users
Landman et al[59]2010CohortType 2HCC deaths decreased (HR = 0.43)
Hosono et al[119]2010Randomized controlled trialNon-diabeticA surrogate marker of colorectal cancer incidence decreased
Ferrara et al[104]2011CohortNot differentiatedNo decreases in the incidence of any cancer; no data on HCC were available
Lee et al[53]2011CohortType 2Incidences of total cancer (HR = 0.12), HCC (HR = 0.06) and colorectal cancer (HR = 0.36) decreased
Hense et al[166]2011CohortType 2HCC incidence did not decrease
Lai et al[101]2012CohortNot differentiatedHCC incidence was decreased by metformin (HR = 0.49) and thiazolidinedione (HR = 0.56)
Ruiter et al[170]2012CohortNot differentiatedIncidences of total cancer (HR = 0.90) and HCC (HR = 0.67) were lower among metformin users than among sulfonylurea users
Stevens et al[108]2012Meta-analysisType 2 and at-risk for diabetesThe summary RR for cancer outcomes was 1.02 across all trials
Thakkar et al[109]2013Meta-analysisType 2Total cancer incidence decreased in case-control studies (RR = 0.90) and cohort studies (RR = 0.70) but did not significantly decrease in randomized controlled trials
Yin et al[110]2013Meta-analysisType 2Overall survival (HR = 0.65) and cancer-specific survival (HR = 0.62) for total cancers were better for metformin than for other glucose-lowering medications
Tsilidis et al[105]2014CohortType 2Incidences of total cancer and HCC were not significantly lower among metformin users than among sulfonylurea users
Gandini et al[106]2014Meta-analysisNot differentiatedAfter adjusting for time-related biases, total cancer incidence decreased (RR = 0.90), but this decrease became insignificant after adjusting for BMI in addition to time-related biases. Total cancer mortality and HCC incidence did not decrease after adjusting for time-related biases
Higurashi et al[120]2016Randomized controlled trialNon-diabeticIncidences of metachronous colorectal adenomas (HR = 0.60) and total polyps (HR = 0.67) decreased
ANTI-CANCER MECHANISM OF METFORMIN

Studies of the anti-cancer mechanism of metformin have followed basic research on the intracellular signaling downstream of metformin to improve hyperglycemia, hyperlipidemia and hyperinsulinemia. An anti-cancer role of metformin had not been proposed when AMPK was identified as a major target molecule of metformin[121]. The identification of LKB1, a tumor suppressor gene, upstream of AMPK highlighted the biguanides as candidate anti-cancer drugs[122,123]. Downstream of AMPK, mTOR, an energy sensor and a gene that plays multiple roles in cell proliferation, was identified[124,125]. An in vivo study employing LKB1 knockout mice clarified that metformin signals in the liver via the LKB1/AMPK axis in the context of glucose homeostasis[126]. A role of the LKB1/AMPK/mTOR axis in carcinogenesis and mediating the anti-cancer signaling of metformin was subsequently identified. Another study in LKB1-AMPK double knockout mice identified an AMPK-independent pathway that improves the diabetic state[127]. AMPK-independent anti-carcinogenic pathways of metformin have also been investigated.

LKB1/AMPK/mTOR axis

Metformin halts the respiratory chain in mitochondria and increases cell energy stress, which activates LKB1 and AMPK. AMPK activation inhibits mTOR and suppresses cell proliferation[124,125]. Furthermore, LKB1/AMPK disturbs insulin signals by degrading IR-substrate-1, resulting in the suppression of insulin/IGF-1 signaling[128], a pathway shared by diabetes and cancer. In lipid metabolism, which is indispensable for tumor growth[129], metformin directly inhibits fatty acid synthase[130]. Metformin arrests the cell cycle in malignant cells via activated AMPK, which is correlated with the downregulation of cyclin D1 and the upregulation of p21CIP and p27KIP[131,132].

AMPK-independent pathways

AMPK-independent pathways downstream of metformin vary. Metformin is thought to protect against DNA damage from reactive oxygen species (ROS) by inhibiting ROS production when metformin inhibits the mitochondrial respiratory chain[133]. Metformin can also bypass AMPK and inhibit mTOR signaling[134] and induce cell cycle arrest by down-regulating cyclin D1 via p53[135,136]. An AMPK knockdown study demonstrated that metformin up-regulates apoptosis and autophagy via a Stat3/Bcl2 pathway[137]. Metformin decreases glucose uptake into cancer cell decreases via a direct allosteric effect on hexokinase II[138].

MicroRNAs as mediators of the anti-cancer activity of metformin

Metformin exerts its anti-carcinogenic activity by regulating microRNA (miRNA) expression to down-regulate target messenger RNAs. miRNAs are small non-coding RNAs with a length of 20-25 nucleotides. miRNAs can bind to messenger RNAs at their 3’-UTR and inhibit their translation, that is, they regulate gene expression at the post-transcriptional level and modulate biological processes, such as intracellular metabolism, cell proliferation, differentiation, apoptosis and angiogenesis[139].

Metformin inhibits the cell cycle of various gastrointestinal tumors, including HCC, by up-regulating the let-7 family in vitro and in vivo[140-145]. For HCC in particular, metformin’s anti-cancer activities are mediated through let-7c, which targets RAS[146]; miR-140-5p, which targets TGFR1, FGF9[147] and DNMT1[148]; and miR-222, which targets PTEN and p57[149]. In pancreatic cancer cell lines, metformin suppresses HMGA1, a pseudogene gene highly expressed in cancer cells, by up-regulating miR-26a, which binds to and degrades the HMGA1 messenger RNA[150]. MiRNAs and their target messenger RNAs in cancers originating from other organs have been well summarized by Pulito[151].

CANCER STEM CELLS AS A TARGET OF METFORMIN IN ADJUVANT THERAPY

Cancer stem cells (CSCs) or tumor-initiating stem cells are a minor subset of the cancer cell population and have been hypothesized to exist among cancer cells. These cells should self-renew indefinitely to generate cancer clones hierarchically and resist chemotherapy and radiotherapy more strongly than any other cancerous daughter cells[152,153]. CSCs do not express definitive cell surface markers and have not been well defined due to extensive heterogeneity[154]. However, using surface markers and the enhanced ALDH1 activity of normal stem cells[155,156], research on CSCs has developed, and a small subset of cells that might include CSCs has been isolated and subjected to further analysis. In the case of HCC, several cell surface markers, such as CD 133, CD90, CD44, EpCAM, OV6 and SP, have been employed to focus on specific cells, including hepatic CSCs[157,158].

Tumors have the potential to be resistant to chemotherapy and radiotherapy, and this potential has been attributed to CSCs[159]. Targeting CSCs in cancer that is refractory to non-surgical treatments may provide a cure. Metformin as an adjuvant to conventional chemotherapy was determined to be effective against CSCs in vitro and in vivo[159]. For hepatic CSCs, metformin administration reduced EpCAM-positive cells, partly depending on the AMPK/mTOR pathway in cell lines and xenograft tumors[160].

CONCLUSION

Diabetes mellitus is globally endemic and has been established as a risk factor for HCC incidence in a large number of observational studies in which researchers critically analyzed study data and adjusted for as many biases as possible. Thus, future increases in diabetes mellitus will likely result in increases in the incidence of HCC. Metformin, a first-line oral anti-diabetic, has been shown to prevent cancer and reduce cancer mortality among diabetic patients in observational studies. Further investigations, particularly randomized controlled trials involving diabetic and non-diabetic subjects, remain necessary. In vitro and in vivo experiments have already provided evidence of the anti-tumor activity of metformin. Newly developed topics that are being investigated further include the AMPK-independent pathway represented by the LKB1/AMPK/mTOR axis; miRNAs downstream of this biguanide and their messenger RNAs that are pivotal to cell survival and proliferation; and cancer stem cells in HCC that are nearly completely identified using cell surface markers.

In daily clinical practice, the administration of metformin to cancer patients, including those with HCC, is associated with few complications. The biguanides exhibit good tolerance in diabetic patients, even those suffering from cirrhosis[161]. Lactic acidosis is not significantly associated with metformin[162]. Although a recent study proposed that metformin might impair cognitive function[163], causality between metformin prescription and cognitive impairment, including Alzheimer’s dementia, has not been confirmed. In summary, metformin is a safe drug for cancer patients as well as diabetic patients. Further clinical evidence of the anti-cancer activity of metformin would have implications for many patients suffering from cancer with or without diabetes mellitus.

Footnotes

Manuscript source: Invited manuscript

P- Reviewer: Guerrieri F, Li YX S- Editor: Yu J L- Editor: A E- Editor: Wang CH

References
1.  El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142:1264-1273.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1196]  [Cited by in F6Publishing: 1296]  [Article Influence: 86.4]  [Reference Citation Analysis (0)]
2.  Altekruse SF, McGlynn KA, Reichman ME. Hepatocellular carcinoma incidence, mortality, and survival trends in the United States from 1975 to 2005. J Clin Oncol. 2009;27:1485-1491.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Bruix J, Sherman M, American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53:1020-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5972]  [Cited by in F6Publishing: 6372]  [Article Influence: 490.2]  [Reference Citation Analysis (1)]
4.  Regimbeau JM, Kianmanesh R, Farges O, Dondero F, Sauvanet A, Belghiti J. Extent of liver resection influences the outcome in patients with cirrhosis and small hepatocellular carcinoma. Surgery. 2002;131:311-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 214]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
5.  Hasegawa K, Kokudo N, Imamura H, Matsuyama Y, Aoki T, Minagawa M, Sano K, Sugawara Y, Takayama T, Makuuchi M. Prognostic impact of anatomic resection for hepatocellular carcinoma. Ann Surg. 2005;242:252-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 488]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
6.  Hasegawa K, Kokudo N, Makuuchi M, Izumi N, Ichida T, Kudo M, Ku Y, Sakamoto M, Nakashima O, Matsui O. Comparison of resection and ablation for hepatocellular carcinoma: a cohort study based on a Japanese nationwide survey. J Hepatol. 2013;58:724-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 274]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
7.  Shiina S, Teratani T, Obi S, Sato S, Tateishi R, Fujishima T, Ishikawa T, Koike Y, Yoshida H, Kawabe T. A randomized controlled trial of radiofrequency ablation with ethanol injection for small hepatocellular carcinoma. Gastroenterology. 2005;129:122-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 664]  [Cited by in F6Publishing: 613]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
8.  Lin SM, Lin CJ, Lin CC, Hsu CW, Chen YC. Randomised controlled trial comparing percutaneous radiofrequency thermal ablation, percutaneous ethanol injection, and percutaneous acetic acid injection to treat hepatocellular carcinoma of 3 cm or less. Gut. 2005;54:1151-1156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 415]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
9.  Poon RT, Fan ST, Lo CM, Liu CL, Wong J. Long-term survival and pattern of recurrence after resection of small hepatocellular carcinoma in patients with preserved liver function: implications for a strategy of salvage transplantation. Ann Surg. 2002;235:373-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 647]  [Cited by in F6Publishing: 666]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
10.  Zhou L, Rui JA, Wang SB, Chen SG, Qu Q, Chi TY, Wei X, Han K, Zhang N, Zhao HT. Factors predictive for long-term survival of male patients with hepatocellular carcinoma after curative resection. J Surg Oncol. 2007;95:298-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
11.  Naito S, Imamura H, Tukada A, Matsuyama Y, Yoshimoto J, Sugo H, Ishizaki Y, Kawasaki S. Postoperative recurrence pattern and prognosis of patients with hepatocellular carcinoma, with particular reference to the hepatitis viral infection status. Liver Int. 2014;34:802-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
12.  Guo R, Feng X, Xiao S, Yan J, Xia F, Ma K, Li X. Short- and long-term outcomes of hepatectomy with or without radiofrequency-assist for the treatment of hepatocellular carcinomas: a retrospective comparative cohort study. Biosci Trends. 2015;9:65-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
13.  Llovet JM, Real MI, Montaña X, Planas R, Coll S, Aponte J, Ayuso C, Sala M, Muchart J, Solà R. Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet. 2002;359:1734-1739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2502]  [Cited by in F6Publishing: 2507]  [Article Influence: 114.0]  [Reference Citation Analysis (0)]
14.  Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, Poon RT, Fan ST, Wong J. Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology. 2002;35:1164-1171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1904]  [Cited by in F6Publishing: 1912]  [Article Influence: 86.9]  [Reference Citation Analysis (0)]
15.  Matsui O, Kadoya M, Yoshikawa J, Gabata T, Arai K, Demachi H, Miyayama S, Takashima T, Unoura M, Kogayashi K. Small hepatocellular carcinoma: treatment with subsegmental transcatheter arterial embolization. Radiology. 1993;188:79-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 312]  [Cited by in F6Publishing: 305]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
16.  Lammer J, Malagari K, Vogl T, Pilleul F, Denys A, Watkinson A, Pitton M, Sergent G, Pfammatter T, Terraz S. Prospective randomized study of doxorubicin-eluting-bead embolization in the treatment of hepatocellular carcinoma: results of the PRECISION V study. Cardiovasc Intervent Radiol. 2010;33:41-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1063]  [Cited by in F6Publishing: 1133]  [Article Influence: 75.5]  [Reference Citation Analysis (0)]
17.  Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, Montalto F, Ammatuna M, Morabito A, Gennari L. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996;334:693-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5110]  [Cited by in F6Publishing: 5052]  [Article Influence: 180.4]  [Reference Citation Analysis (0)]
18.  de Villa V, Lo CM. Liver transplantation for hepatocellular carcinoma in Asia. Oncologist. 2007;12:1321-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
19.  Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9016]  [Cited by in F6Publishing: 9673]  [Article Influence: 604.6]  [Reference Citation Analysis (1)]
20.  Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, Luo R, Feng J, Ye S, Yang TS. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3854]  [Cited by in F6Publishing: 4397]  [Article Influence: 274.8]  [Reference Citation Analysis (0)]
21.  de Lope CR, Tremosini S, Forner A, Reig M, Bruix J. Management of HCC. J Hepatol. 2012;56 Suppl 1:S75-S87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in F6Publishing: 463]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
22.  Hosaka T, Suzuki F, Kobayashi M, Seko Y, Kawamura Y, Sezaki H, Akuta N, Suzuki Y, Saitoh S, Arase Y. Long-term entecavir treatment reduces hepatocellular carcinoma incidence in patients with hepatitis B virus infection. Hepatology. 2013;58:98-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 519]  [Cited by in F6Publishing: 505]  [Article Influence: 45.9]  [Reference Citation Analysis (0)]
23.  Wong GL, Chan HL, Mak CW, Lee SK, Ip ZM, Lam AT, Iu HW, Leung JM, Lai JW, Lo AO. Entecavir treatment reduces hepatic events and deaths in chronic hepatitis B patients with liver cirrhosis. Hepatology. 2013;58:1537-1547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 347]  [Cited by in F6Publishing: 369]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
24.  Lawitz E, Poordad FF, Pang PS, Hyland RH, Ding X, Mo H, Symonds WT, McHutchison JG, Membreno FE. Sofosbuvir and ledipasvir fixed-dose combination with and without ribavirin in treatment-naive and previously treated patients with genotype 1 hepatitis C virus infection (LONESTAR): an open-label, randomised, phase 2 trial. Lancet. 2014;383:515-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 429]  [Cited by in F6Publishing: 456]  [Article Influence: 45.6]  [Reference Citation Analysis (0)]
25.  Afdhal N, Reddy KR, Nelson DR, Lawitz E, Gordon SC, Schiff E, Nahass R, Ghalib R, Gitlin N, Herring R. Ledipasvir and sofosbuvir for previously treated HCV genotype 1 infection. N Engl J Med. 2014;370:1483-1493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1065]  [Cited by in F6Publishing: 1042]  [Article Influence: 104.2]  [Reference Citation Analysis (0)]
26.  Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005;330:1304-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1692]  [Cited by in F6Publishing: 1757]  [Article Influence: 92.5]  [Reference Citation Analysis (0)]
27.  Tanaka H, Imai Y, Hiramatsu N, Ito Y, Imanaka K, Oshita M, Hijioka T, Katayama K, Yabuuchi I, Yoshihara H. Declining incidence of hepatocellular carcinoma in Osaka, Japan, from 1990 to 2003. Ann Intern Med. 2008;148:820-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 100]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
28.  Altekruse SF, Henley SJ, Cucinelli JE, McGlynn KA. Changing hepatocellular carcinoma incidence and liver cancer mortality rates in the United States. Am J Gastroenterol. 2014;109:542-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 323]  [Article Influence: 32.3]  [Reference Citation Analysis (1)]
29.  Njei B, Rotman Y, Ditah I, Lim JK. Emerging trends in hepatocellular carcinoma incidence and mortality. Hepatology. 2015;61:191-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 408]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
30.  Venook AP, Papandreou C, Furuse J, de Guevara LL. The incidence and epidemiology of hepatocellular carcinoma: a global and regional perspective. Oncologist. 2010;15 Suppl 4:5-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 626]  [Cited by in F6Publishing: 711]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
31.  Major JM, Sargent JD, Graubard BI, Carlos HA, Hollenbeck AR, Altekruse SF, Freedman ND, McGlynn KA. Local geographic variation in chronic liver disease and hepatocellular carcinoma: contributions of socioeconomic deprivation, alcohol retail outlets, and lifestyle. Ann Epidemiol. 2014;24:104-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
32.  Tanaka H, Uera F, Tsukuma H, Ioka A, Oshima A. Distinctive change in male liver cancer incidence rate between the 1970s and 1990s in Japan: comparison with Japanese-Americans and US whites. Jpn J Clin Oncol. 2007;37:193-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
33.  Kasahara A, Hayashi N, Mochizuki K, Takayanagi M, Yoshioka K, Kakumu S, Iijima A, Urushihara A, Kiyosawa K, Okuda M. Risk factors for hepatocellular carcinoma and its incidence after interferon treatment in patients with chronic hepatitis C. Osaka Liver Disease Study Group. Hepatology. 1998;27:1394-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 357]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
34.  Lok AS, Everhart JE, Wright EC, Di Bisceglie AM, Kim HY, Sterling RK, Everson GT, Lindsay KL, Lee WM, Bonkovsky HL. Maintenance peginterferon therapy and other factors associated with hepatocellular carcinoma in patients with advanced hepatitis C. Gastroenterology. 2011;140:840-849; quiz e12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 151]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
35.  Snow-Lampart A, Chappell B, Curtis M, Zhu Y, Myrick F, Schawalder J, Kitrinos K, Svarovskaia ES, Miller MD, Sorbel J. No resistance to tenofovir disoproxil fumarate detected after up to 144 weeks of therapy in patients monoinfected with chronic hepatitis B virus. Hepatology. 2011;53:763-773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 141]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
36.  Liaw YF, Leung NW, Chang TT, Guan R, Tai DI, Ng KY, Chien RN, Dent J, Roman L, Edmundson S. Effects of extended lamivudine therapy in Asian patients with chronic hepatitis B. Asia Hepatitis Lamivudine Study Group. Gastroenterology. 2000;119:172-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 521]  [Cited by in F6Publishing: 500]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
37.  Hadziyannis SJ, Tassopoulos NC, Heathcote EJ, Chang TT, Kitis G, Rizzetto M, Marcellin P, Lim SG, Goodman Z, Ma J. Long-term therapy with adefovir dipivoxil for HBeAg-negative chronic hepatitis B for up to 5 years. Gastroenterology. 2006;131:1743-1751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 674]  [Cited by in F6Publishing: 639]  [Article Influence: 35.5]  [Reference Citation Analysis (0)]
38.  Marcellin P, Chang TT, Lim SG, Sievert W, Tong M, Arterburn S, Borroto-Esoda K, Frederick D, Rousseau F. Long-term efficacy and safety of adefovir dipivoxil for the treatment of hepatitis B e antigen-positive chronic hepatitis B. Hepatology. 2008;48:750-758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 259]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
39.  King H, Aubert RE, Herman WH. Global burden of diabetes, 1995-2025: prevalence, numerical estimates, and projections. Diabetes Care. 1998;21:1414-1431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3616]  [Cited by in F6Publishing: 3312]  [Article Influence: 127.4]  [Reference Citation Analysis (0)]
40.  Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract. 2010;87:4-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4438]  [Cited by in F6Publishing: 4250]  [Article Influence: 303.6]  [Reference Citation Analysis (3)]
41.  Kelly T, Yang W, Chen CS, Reynolds K, He J. Global burden of obesity in 2005 and projections to 2030. Int J Obes (Lond). 2008;32:1431-1437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1843]  [Cited by in F6Publishing: 1938]  [Article Influence: 121.1]  [Reference Citation Analysis (2)]
42.  Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004;27:1047-1053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9344]  [Cited by in F6Publishing: 8778]  [Article Influence: 438.9]  [Reference Citation Analysis (1)]
43.  Maynard GD. A statistical study in cancer death-rates. Biometrik. 1910;7: 276-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
44.  Greenwood M, Wood F. The Relation between the Cancer and Diabetes Death-rates. J Hyg (Lond). 1914;14:83-118.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Marble A. Diabetes and cancer. N Engl J Med. 1934;211:339-349.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Kessler II. Cancer mortality among diabetics. J Natl Cancer Inst. 1970;44:673-686.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  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: ]
48.  Adami HO, McLaughlin J, Ekbom A, Berne C, Silverman D, Hacker D, Persson I. Cancer risk in patients with diabetes mellitus. Cancer Causes Control. 1991;2:307-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 228]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
49.  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: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 274]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
50.  Wideroff L, Gridley G, Mellemkjaer L, Chow WH, Linet M, Keehn S, Borch-Johnsen K, Olsen JH. Cancer incidence in a population-based cohort of patients hospitalized with diabetes mellitus in Denmark. J Natl Cancer Inst. 1997;89:1360-1365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 453]  [Cited by in F6Publishing: 455]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
51.  La Vecchia C, Negri E, Franceschi S, D’Avanzo B, Boyle P. A case-control study of diabetes mellitus and cancer risk. Br J Cancer. 1994;70:950-953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 221]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
52.  Balkau B, Kahn HS, Courbon D, Eschwège E, Ducimetière P. Hyperinsulinemia predicts fatal liver cancer but is inversely associated with fatal cancer at some other sites: the Paris Prospective Study. Diabetes Care. 2001;24:843-849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 79]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
53.  Lee MS, Hsu CC, Wahlqvist ML, Tsai HN, Chang YH, Huang YC. Type 2 diabetes increases and metformin reduces total, colorectal, liver and pancreatic cancer incidences in Taiwanese: a representative population prospective cohort study of 800,000 individuals. BMC Cancer. 2011;11:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 340]  [Article Influence: 26.2]  [Reference Citation Analysis (0)]
54.  Ikeda Y, Shimada M, Hasegawa H, Gion T, Kajiyama K, Shirabe K, Yanaga K, Takenaka K, Sugimachi K. Prognosis of hepatocellular carcinoma with diabetes mellitus after hepatic resection. Hepatology. 1998;27:1567-1571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 77]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
55.  Huo TI, Wu JC, Lui WY, Lee PC, Huang YH, Chau GY, Tsay SH, Chang FY, Lee SD. Diabetes mellitus is a recurrence-independent risk factor in patients with hepatitis B virus-related hepatocellular carcinoma undergoing resection. Eur J Gastroenterol Hepatol. 2003;15:1203-1208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
56.  Komura T, Mizukoshi E, Kita Y, Sakurai M, Takata Y, Arai K, Yamashita T, Ohta T, Shimizu K, Nakamoto Y. Impact of diabetes on recurrence of hepatocellular carcinoma after surgical treatment in patients with viral hepatitis. Am J Gastroenterol. 2007;102:1939-1946.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
57.  Kawamura Y, Ikeda K, Arase Y, Yatsuji H, Sezaki H, Hosaka T, Akuta N, Kobayashi M, Saitoh S, Suzuki F. Diabetes mellitus worsens the recurrence rate after potentially curative therapy in patients with hepatocellular carcinoma associated with nonviral hepatitis. J Gastroenterol Hepatol. 2008;23:1739-1746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 27]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
58.  Coughlin SS, Calle EE, Teras LR, Petrelli J, Thun MJ. Diabetes mellitus as a predictor of cancer mortality in a large cohort of US adults. Am J Epidemiol. 2004;159:1160-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 565]  [Cited by in F6Publishing: 600]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
59.  Landman GW, van Hateren KJ, Kleefstra N, Groenier KH, Gans RO, Bilo HJ. The relationship between glycaemic control and mortality in patients with type 2 diabetes in general practice (ZODIAC-11). Br J Gen Pract. 2010;60:172-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 36]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
60.  El-Serag HB, Hampel H, Javadi F. The association between diabetes and hepatocellular carcinoma: a systematic review of epidemiologic evidence. Clin Gastroenterol Hepatol. 2006;4:369-380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 581]  [Cited by in F6Publishing: 559]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
61.  Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG, Yee D. Diabetes and cancer: a consensus report. CA Cancer J Clin. 2010;60:207-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 585]  [Cited by in F6Publishing: 630]  [Article Influence: 45.0]  [Reference Citation Analysis (0)]
62.  Schlesinger S, Aleksandrova K, Pischon T, Jenab M, Fedirko V, Trepo E, Overvad K, Roswall N, Tjønneland A, Boutron-Ruault MC. Diabetes mellitus, insulin treatment, diabetes duration, and risk of biliary tract cancer and hepatocellular carcinoma in a European cohort. Ann Oncol. 2013;24:2449-2455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 100]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
63.  Koh WP, Wang R, Jin A, Yu MC, Yuan JM. Diabetes mellitus and risk of hepatocellular carcinoma: findings from the Singapore Chinese Health Study. Br J Cancer. 2013;108:1182-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 65]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
64.  Inoue M, Iwasaki M, Otani T, Sasazuki S, Noda M, Tsugane S. Diabetes mellitus and the risk of cancer: results from a large-scale population-based cohort study in Japan. Arch Intern Med. 2006;166:1871-1877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 384]  [Cited by in F6Publishing: 398]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
65.  Møller H, Mellemgaard A, Lindvig K, Olsen JH. Obesity and cancer risk: a Danish record-linkage study. Eur J Cancer. 1994;30A:344-350.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Wolk A, Gridley G, Svensson M, Nyrén O, McLaughlin JK, Fraumeni JF, Adam HO. A prospective study of obesity and cancer risk (Sweden). Cancer Causes Control. 2001;12:13-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 455]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
67.  Levine W, Dyer AR, Shekelle RB, Schoenberger JA, Stamler J. Post-load plasma glucose and cancer mortality in middle-aged men and women. 12-year follow-up findings of the Chicago Heart Association Detection Project in Industry. Am J Epidemiol. 1990;131:254-262.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Batty GD, Shipley MJ, Marmot M, Smith GD. Diabetes status and post-load plasma glucose concentration in relation to site-specific cancer mortality: findings from the original Whitehall study. Cancer Causes Control. 2004;15:873-881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 101]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
69.  Harding JL, Shaw JE, Peeters A, Cartensen B, Magliano DJ. Cancer risk among people with type 1 and type 2 diabetes: disentangling true associations, detection bias, and reverse causation. Diabetes Care. 2015;38:264-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 173]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
70.  Johnson JA, Bowker SL, Richardson K, Marra CA. Time-varying incidence of cancer after the onset of type 2 diabetes: evidence of potential detection bias. Diabetologia. 2011;54:2263-2271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 101]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
71.  Hjartåker A, Langseth H, Weiderpass E. Obesity and diabetes epidemics: cancer repercussions. Adv Exp Med Biol. 2008;630:72-93.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Pollak M. Insulin, insulin-like growth factors and neoplasia. Best Pract Res Clin Endocrinol Metab. 2008;22:625-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
73.  Powell DR, Suwanichkul A, Cubbage ML, DePaolis LA, Snuggs MB, Lee PD. Insulin inhibits transcription of the human gene for insulin-like growth factor-binding protein-1. J Biol Chem. 1991;266:18868-18876.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Ooi GT, Tseng LY, Tran MQ, Rechler MM. Insulin rapidly decreases insulin-like growth factor-binding protein-1 gene transcription in streptozotocin-diabetic rats. Mol Endocrinol. 1992;6:2219-2228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 29]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
75.  Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149:274-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5794]  [Cited by in F6Publishing: 6205]  [Article Influence: 517.1]  [Reference Citation Analysis (1)]
76.  Chettouh H, Lequoy M, Fartoux L, Vigouroux C, Desbois-Mouthon C. Hyperinsulinaemia and insulin signalling in the pathogenesis and the clinical course of hepatocellular carcinoma. Liver Int. 2015;35:2203-2217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 76]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
77.  Enguita-Germán M, Fortes P. Targeting the insulin-like growth factor pathway in hepatocellular carcinoma. World J Hepatol. 2014;6:716-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
78.  Villanueva A, Chiang DY, Newell P, Peix J, Thung S, Alsinet C, Tovar V, Roayaie S, Minguez B, Sole M. Pivotal role of mTOR signaling in hepatocellular carcinoma. Gastroenterology. 2008;135:1972-1983, 1983.e1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 520]  [Cited by in F6Publishing: 562]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
79.  Garg SK, Maurer H, Reed K, Selagamsetty R. Diabetes and cancer: two diseases with obesity as a common risk factor. Diabetes Obes Metab. 2014;16:97-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 176]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
80.  Guo S, Liu M, Wang G, Torroella-Kouri M, Gonzalez-Perez RR. Oncogenic role and therapeutic target of leptin signaling in breast cancer and cancer stem cells. Biochim Biophys Acta. 2012;1825:207-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 77]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
81.  Becker S, Dossus L, Kaaks R. Obesity related hyperinsulinaemia and hyperglycaemia and cancer development. Arch Physiol Biochem. 2009;115:86-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 108]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
82.  Duan XF, Tang P, Li Q, Yu ZT. Obesity, adipokines and hepatocellular carcinoma. Int J Cancer. 2013;133:1776-1783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
83.  Warburg O. On the origin of cancer cells. Science. 1956;123:309-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9117]  [Cited by in F6Publishing: 9383]  [Article Influence: 138.0]  [Reference Citation Analysis (0)]
84.  Miles KA, Williams RE. Warburg revisited: imaging tumour blood flow and metabolism. Cancer Imaging. 2008;8:81-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 104]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
85.  Chocarro-Calvo A, García-Martínez JM, Ardila-González S, De la Vieja A, García-Jiménez C. Glucose-induced β-catenin acetylation enhances Wnt signaling in cancer. Mol Cell. 2013;49:474-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 105]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
86.  Li CI, Chen HJ, Lai HC, Liu CS, Lin WY, Li TC, Lin CC. Hyperglycemia and chronic liver diseases on risk of hepatocellular carcinoma in Chinese patients with type 2 diabetes--National cohort of Taiwan Diabetes Study. Int J Cancer. 2015;136:2668-2679.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
87.  Takahashi H, Mizuta T, Eguchi Y, Kawaguchi Y, Kuwashiro T, Oeda S, Isoda H, Oza N, Iwane S, Izumi K. Post-challenge hyperglycemia is a significant risk factor for the development of hepatocellular carcinoma in patients with chronic hepatitis C. J Gastroenterol. 2011;46:790-798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
88.  Johnson JA, Bowker SL. Intensive glycaemic control and cancer risk in type 2 diabetes: a meta-analysis of major trials. Diabetologia. 2011;54:25-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 99]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
89.  Yang XL, Ma RC, Chan JC. Meta-analysis of trial data may support a causal role of hyperglycaemia in cancer. Diabetologia. 2011;54:709-710; author reply 711-712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
90.  Brown SB, Hankinson SE. Endogenous estrogens and the risk of breast, endometrial, and ovarian cancers. Steroids. 2015;99:8-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 102]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
91.  El-Serag HB, Mason AC, Key C. Trends in survival of patients with hepatocellular carcinoma between 1977 and 1996 in the United States. Hepatology. 2001;33:62-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 262]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
92.  Yu MW, Chang HC, Chang SC, Liaw YF, Lin SM, Liu CJ, Lee SD, Lin CL, Chen PJ, Lin SC. Role of reproductive factors in hepatocellular carcinoma: Impact on hepatitis B- and C-related risk. Hepatology. 2003;38:1393-1400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 55]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
93.  Shi L, Feng Y, Lin H, Ma R, Cai X. Role of estrogen in hepatocellular carcinoma: is inflammation the key? J Transl Med. 2014;12:93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 83]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
94.  Nathan DM, Buse JB, Davidson MB, Ferrannini E, Holman RR, Sherwin R, Zinman B. Medical management of hyperglycaemia in type 2 diabetes mellitus: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetologia. 2009;52:17-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 519]  [Cited by in F6Publishing: 537]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
95.  Bailey CJ, Day C. Traditional plant medicines as treatments for diabetes. Diabetes Care. 1989;12:553-564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 455]  [Cited by in F6Publishing: 403]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
96.  Bowker SL, Majumdar SR, Veugelers P, Johnson JA. Increased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin. Diabetes Care. 2006;29:254-258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 685]  [Cited by in F6Publishing: 685]  [Article Influence: 38.1]  [Reference Citation Analysis (0)]
97.  Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JM. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care. 2009;32:1620-1625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 778]  [Cited by in F6Publishing: 782]  [Article Influence: 52.1]  [Reference Citation Analysis (0)]
98.  Donadon V, Balbi M, Ghersetti M, Grazioli S, Perciaccante A, Della Valentina G, Gardenal R, Dal Mas M, Casarin P, Zanette G. Antidiabetic therapy and increased risk of hepatocellular carcinoma in chronic liver disease. World J Gastroenterol. 2009;15:2506-2511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 109]  [Cited by in F6Publishing: 109]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
99.  Donadon V, Balbi M, Mas MD, Casarin P, Zanette G. Metformin and reduced risk of hepatocellular carcinoma in diabetic patients with chronic liver disease. Liver Int. 2010;30:750-758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 206]  [Article Influence: 14.7]  [Reference Citation Analysis (1)]
100.  Hassan MM, Curley SA, Li D, Kaseb A, Davila M, Abdalla EK, Javle M, Moghazy DM, Lozano RD, Abbruzzese JL. Association of diabetes duration and diabetes treatment with the risk of hepatocellular carcinoma. Cancer. 2010;116:1938-1946.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 232]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
101.  Lai SW, Chen PC, Liao KF, Muo CH, Lin CC, Sung FC. Risk of hepatocellular carcinoma in diabetic patients and risk reduction associated with anti-diabetic therapy: a population-based cohort study. Am J Gastroenterol. 2012;107:46-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 232]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
102.  Suissa S, Azoulay L. Metformin and the risk of cancer: time-related biases in observational studies. Diabetes Care. 2012;35:2665-2673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 377]  [Cited by in F6Publishing: 390]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
103.  Johnson JA, Carstensen B, Witte D, Bowker SL, Lipscombe L, Renehan AG. Diabetes and cancer (1): evaluating the temporal relationship between type 2 diabetes and cancer incidence. Diabetologia. 2012;55:1607-1618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 162]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
104.  Ferrara A, Lewis JD, Quesenberry CP, Peng T, Strom BL, Van Den Eeden SK, Ehrlich SF, Habel LA. Cohort study of pioglitazone and cancer incidence in patients with diabetes. Diabetes Care. 2011;34:923-929.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 144]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
105.  Tsilidis KK, Capothanassi D, Allen NE, Rizos EC, Lopez DS, van Veldhoven K, Sacerdote C, Ashby D, Vineis P, Tzoulaki I. Metformin does not affect cancer risk: a cohort study in the U.K. Clinical Practice Research Datalink analyzed like an intention-to-treat trial. Diabetes Care. 2014;37:2522-2532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 132]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
106.  Gandini S, Puntoni M, Heckman-Stoddard BM, Dunn BK, Ford L, DeCensi A, Szabo E. Metformin and cancer risk and mortality: a systematic review and meta-analysis taking into account biases and confounders. Cancer Prev Res (Phila). 2014;7:867-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 294]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
107.  Home PD, Kahn SE, Jones NP, Noronha D, Beck-Nielsen H, Viberti G. Experience of malignancies with oral glucose-lowering drugs in the randomised controlled ADOPT (A Diabetes Outcome Progression Trial) and RECORD (Rosiglitazone Evaluated for Cardiovascular Outcomes and Regulation of Glycaemia in Diabetes) clinical trials. Diabetologia. 2010;53:1838-1845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 131]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
108.  Stevens RJ, Ali R, Bankhead CR, Bethel MA, Cairns BJ, Camisasca RP, Crowe FL, Farmer AJ, Harrison S, Hirst JA. Cancer outcomes and all-cause mortality in adults allocated to metformin: systematic review and collaborative meta-analysis of randomised clinical trials. Diabetologia. 2012;55:2593-2603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 133]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
109.  Thakkar B, Aronis KN, Vamvini MT, Shields K, Mantzoros CS. Metformin and sulfonylureas in relation to cancer risk in type II diabetes patients: a meta-analysis using primary data of published studies. Metabolism. 2013;62:922-934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 104]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
110.  Yin M, Zhou J, Gorak EJ, Quddus F. Metformin is associated with survival benefit in cancer patients with concurrent type 2 diabetes: a systematic review and meta-analysis. Oncologist. 2013;18:1248-1255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 117]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
111.  Choi Y, Kim TY, Oh DY, Lee KH, Han SW, Im SA, Kim TY, Bang YJ. The Impact of Diabetes Mellitus and Metformin Treatment on Survival of Patients with Advanced Pancreatic Cancer Undergoing Chemotherapy. Cancer Res Treat. 2016;48:171-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
112.  Jiralerspong S, Palla SL, Giordano SH, Meric-Bernstam F, Liedtke C, Barnett CM, Hsu L, Hung MC, Hortobagyi GN, Gonzalez-Angulo AM. Metformin and pathologic complete responses to neoadjuvant chemotherapy in diabetic patients with breast cancer. J Clin Oncol. 2009;27:3297-3302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 627]  [Cited by in F6Publishing: 653]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
113.  Tan BX, Yao WX, Ge J, Peng XC, Du XB, Zhang R, Yao B, Xie K, Li LH, Dong H. Prognostic influence of metformin as first-line chemotherapy for advanced nonsmall cell lung cancer in patients with type 2 diabetes. Cancer. 2011;117:5103-5111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 124]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
114.  Lee JH, Kim TI, Jeon SM, Hong SP, Cheon JH, Kim WH. The effects of metformin on the survival of colorectal cancer patients with diabetes mellitus. Int J Cancer. 2012;131:752-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 157]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
115.  Kordes S, Pollak MN, Zwinderman AH, Mathôt RA, Weterman MJ, Beeker A, Punt CJ, Richel DJ, Wilmink JW. Metformin in patients with advanced pancreatic cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol. 2015;16:839-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 267]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
116.  Kim J, Lim W, Kim EK, Kim MK, Paik NS, Jeong SS, Yoon JH, Park CH, Ahn SH, Kim LS. Phase II randomized trial of neoadjuvant metformin plus letrozole versus placebo plus letrozole for estrogen receptor positive postmenopausal breast cancer (METEOR). BMC Cancer. 2014;14:170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
117.  Jang WI, Kim MS, Lim JS, Yoo HJ, Seo YS, Han CJ, Park SC, Kay CS, Kim M, Jang HS. Survival Advantage Associated with Metformin Usage in Hepatocellular Carcinoma Patients Receiving Radiotherapy: A Propensity Score Matching Analysis. Anticancer Res. 2015;35:5047-5054.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Casadei Gardini A, Marisi G, Scarpi E, Scartozzi M, Faloppi L, Silvestris N, Masi G, Vivaldi C, Brunetti O, Tamberi S. Effects of metformin on clinical outcome in diabetic patients with advanced HCC receiving sorafenib. Expert Opin Pharmacother. 2015;16:2719-2725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 57]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
119.  Hosono K, Endo H, Takahashi H, Sugiyama M, Sakai E, Uchiyama T, Suzuki K, Iida H, Sakamoto Y, Yoneda K. Metformin suppresses colorectal aberrant crypt foci in a short-term clinical trial. Cancer Prev Res (Phila). 2010;3:1077-1083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 235]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
120.  Higurashi T, Hosono K, Takahashi H, Komiya Y, Umezawa S, Sakai E, Uchiyama T, Taniguchi L, Hata Y, Uchiyama S. Metformin for chemoprevention of metachronous colorectal adenoma or polyps in post-polypectomy patients without diabetes: a multicentre double-blind, placebo-controlled, randomised phase 3 trial. Lancet Oncol. 2016;17:475-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 214]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
121.  Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3802]  [Cited by in F6Publishing: 4034]  [Article Influence: 175.4]  [Reference Citation Analysis (0)]
122.  Hawley SA, Boudeau J, Reid JL, Mustard KJ, Udd L, Mäkelä TP, Alessi DR, Hardie DG. Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J Biol. 2003;2:28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1203]  [Cited by in F6Publishing: 1280]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
123.  Lizcano JM, Göransson O, Toth R, Deak M, Morrice NA, Boudeau J, Hawley SA, Udd L, Mäkelä TP, Hardie DG. LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily, including MARK/PAR-1. EMBO J. 2004;23:833-843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1048]  [Cited by in F6Publishing: 1075]  [Article Influence: 53.8]  [Reference Citation Analysis (0)]
124.  Kimura N, Tokunaga C, Dalal S, Richardson C, Yoshino K, Hara K, Kemp BE, Witters LA, Mimura O, Yonezawa K. A possible linkage between AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) signalling pathway. Genes Cells. 2003;8:65-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 279]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
125.  Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell. 2003;115:577-590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2835]  [Cited by in F6Publishing: 2893]  [Article Influence: 137.8]  [Reference Citation Analysis (0)]
126.  Shaw RJ, Lamia KA, Vasquez D, Koo SH, Bardeesy N, Depinho RA, Montminy M, Cantley LC. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science. 2005;310:1642-1646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1494]  [Cited by in F6Publishing: 1453]  [Article Influence: 76.5]  [Reference Citation Analysis (0)]
127.  Foretz M, Hébrard S, Leclerc J, Zarrinpashneh E, Soty M, Mithieux G, Sakamoto K, Andreelli F, Viollet B. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest. 2010;120:2355-2369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 858]  [Cited by in F6Publishing: 917]  [Article Influence: 65.5]  [Reference Citation Analysis (0)]
128.  Takano A, Usui I, Haruta T, Kawahara J, Uno T, Iwata M, Kobayashi M. Mammalian target of rapamycin pathway regulates insulin signaling via subcellular redistribution of insulin receptor substrate 1 and integrates nutritional signals and metabolic signals of insulin. Mol Cell Biol. 2001;21:5050-5062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 198]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
129.  Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer. 2007;7:763-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1925]  [Cited by in F6Publishing: 2045]  [Article Influence: 120.3]  [Reference Citation Analysis (0)]
130.  Cantoria MJ, Boros LG, Meuillet EJ. Contextual inhibition of fatty acid synthesis by metformin involves glucose-derived acetyl-CoA and cholesterol in pancreatic tumor cells. Metabolomics. 2014;10:91-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
131.  Cai X, Hu X, Cai B, Wang Q, Li Y, Tan X, Hu H, Chen X, Huang J, Cheng J. Metformin suppresses hepatocellular carcinoma cell growth through induction of cell cycle G1/G0 phase arrest and p21CIP and p27KIP expression and downregulation of cyclin D1 in vitro and in vivo. Oncol Rep. 2013;30:2449-2457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 53]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
132.  Cai X, Hu X, Tan X, Cheng W, Wang Q, Chen X, Guan Y, Chen C, Jing X. Metformin Induced AMPK Activation, G0/G1 Phase Cell Cycle Arrest and the Inhibition of Growth of Esophageal Squamous Cell Carcinomas In Vitro and In Vivo. PLoS One. 2015;10:e0133349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
133.  Algire C, Moiseeva O, Deschênes-Simard X, Amrein L, Petruccelli L, Birman E, Viollet B, Ferbeyre G, Pollak MN. Metformin reduces endogenous reactive oxygen species and associated DNA damage. Cancer Prev Res (Phila). 2012;5:536-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 245]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
134.  Kalender A, Selvaraj A, Kim SY, Gulati P, Brûlé S, Viollet B, Kemp BE, Bardeesy N, Dennis P, Schlager JJ. Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner. Cell Metab. 2010;11:390-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 629]  [Cited by in F6Publishing: 653]  [Article Influence: 46.6]  [Reference Citation Analysis (0)]
135.  Ben Sahra I, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger P, Tanti JF, Le Marchand-Brustel Y, Bost F. The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene. 2008;27:3576-3586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 633]  [Cited by in F6Publishing: 658]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
136.  Ben Sahra I, Regazzetti C, Robert G, Laurent K, Le Marchand-Brustel Y, Auberger P, Tanti JF, Giorgetti-Peraldi S, Bost F. Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res. 2011;71:4366-4372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 471]  [Article Influence: 36.2]  [Reference Citation Analysis (0)]
137.  Feng Y, Ke C, Tang Q, Dong H, Zheng X, Lin W, Ke J, Huang J, Yeung SC, Zhang H. Metformin promotes autophagy and apoptosis in esophageal squamous cell carcinoma by downregulating Stat3 signaling. Cell Death Dis. 2014;5:e1088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 195]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
138.  Salani B, Marini C, Rio AD, Ravera S, Massollo M, Orengo AM, Amaro A, Passalacqua M, Maffioli S, Pfeffer U. Metformin impairs glucose consumption and survival in Calu-1 cells by direct inhibition of hexokinase-II. Sci Rep. 2013;3:2070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 89]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
139.  Morishita A, Masaki T. miRNA in hepatocellular carcinoma. Hepatol Res. 2015;45:128-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 81]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
140.  Kato K, Gong J, Iwama H, Kitanaka A, Tani J, Miyoshi H, Nomura K, Mimura S, Kobayashi M, Aritomo Y. The antidiabetic drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Mol Cancer Ther. 2012;11:549-560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 174]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
141.  Miyoshi H, Kato K, Iwama H, Maeda E, Sakamoto T, Fujita K, Toyota Y, Tani J, Nomura T, Mimura S. Effect of the anti-diabetic drug metformin in hepatocellular carcinoma in vitro and in vivo. Int J Oncol. 2014;45:322-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 66]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
142.  Kobayashi M, Kato K, Iwama H, Fujihara S, Nishiyama N, Mimura S, Toyota Y, Nomura T, Nomura K, Tani J. Antitumor effect of metformin in esophageal cancer: in vitro study. Int J Oncol. 2013;42:517-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
143.  Fujimori T, Kato K, Fujihara S, Iwama H, Yamashita T, Kobayashi K, Kamada H, Morishita A, Kobara H, Mori H. Antitumor effect of metformin on cholangiocarcinoma: In vitro and in vivo studies. Oncol Rep. 2015;34:2987-2996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
144.  Fujihara S, Kato K, Morishita A, Iwama H, Nishioka T, Chiyo T, Nishiyama N, Miyoshi H, Kobayashi M, Kobara H. Antidiabetic drug metformin inhibits esophageal adenocarcinoma cell proliferation in vitro and in vivo. Int J Oncol. 2015;46:2172-2180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
145.  Kato K, Iwama H, Yamashita T, Kobayashi K, Fujihara S, Fujimori T, Kamada H, Kobara H, Masaki T. The anti-diabetic drug metformin inhibits pancreatic cancer cell proliferation in vitro and in vivo: Study of the microRNAs associated with the antitumor effect of metformin. Oncol Rep. 2016;35:1582-1592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
146.  Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ. RAS is regulated by the let-7 microRNA family. Cell. 2005;120:635-647.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Yang H, Fang F, Chang R, Yang L. MicroRNA-140-5p suppresses tumor growth and metastasis by targeting transforming growth factor β receptor 1 and fibroblast growth factor 9 in hepatocellular carcinoma. Hepatology. 2013;58:205-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 217]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
148.  Takata A, Otsuka M, Yoshikawa T, Kishikawa T, Hikiba Y, Obi S, Goto T, Kang YJ, Maeda S, Yoshida H. MicroRNA-140 acts as a liver tumor suppressor by controlling NF-κB activity by directly targeting DNA methyltransferase 1 (Dnmt1) expression. Hepatology. 2013;57:162-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 86]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
149.  Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, Ngankeu A, Taccioli C, Pichiorri F, Alder H, Secchiero P. miR-221& amp; 222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell. 2009;16:498-509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 617]  [Cited by in F6Publishing: 642]  [Article Influence: 42.8]  [Reference Citation Analysis (0)]
150.  Li W, Yuan Y, Huang L, Qiao M, Zhang Y. Metformin alters the expression profiles of microRNAs in human pancreatic cancer cells. Diabetes Res Clin Pract. 2012;96:187-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
151.  Pulito C, Donzelli S, Muti P, Puzzo L, Strano S, Blandino G. microRNAs and cancer metabolism reprogramming: the paradigm of metformin. Ann Transl Med. 2014;2:58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 22]  [Reference Citation Analysis (0)]
152.  Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6844]  [Cited by in F6Publishing: 6752]  [Article Influence: 293.6]  [Reference Citation Analysis (0)]
153.  Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8:755-768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2552]  [Cited by in F6Publishing: 2557]  [Article Influence: 159.8]  [Reference Citation Analysis (0)]
154.  Sell S, Leffert HL. Liver cancer stem cells. J Clin Oncol. 2008;26:2800-2805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 162]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
155.  Boman BM, Wicha MS. Cancer stem cells: a step toward the cure. J Clin Oncol. 2008;26:2795-2799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 199]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
156.  Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu S. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2860]  [Cited by in F6Publishing: 2972]  [Article Influence: 185.8]  [Reference Citation Analysis (0)]
157.  Zhu CP, Wang AQ, Zhang HH, Wan XS, Yang XB, Chen SG, Zhao HT. Research progress and prospects of markers for liver cancer stem cells. World J Gastroenterol. 2015;21:12190-12196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
158.  Chiba T, Iwama A, Yokosuka O. Cancer stem cells in hepatocellular carcinoma: Therapeutic implications based on stem cell biology. Hepatol Res. 2016;46:50-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 49]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
159.  Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res. 2009;69:7507-7511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 803]  [Cited by in F6Publishing: 855]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
160.  Saito T, Chiba T, Yuki K, Zen Y, Oshima M, Koide S, Motoyama T, Ogasawara S, Suzuki E, Ooka Y. Metformin, a diabetes drug, eliminates tumor-initiating hepatocellular carcinoma cells. PLoS One. 2013;8:e70010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
161.  Zhang X, Harmsen WS, Mettler TA, Kim WR, Roberts RO, Therneau TM, Roberts LR, Chaiteerakij R. Continuation of metformin use after a diagnosis of cirrhosis significantly improves survival of patients with diabetes. Hepatology. 2014;60:2008-2016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 121]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
162.  Salpeter SR, Greyber E, Pasternak GA, Salpeter EE. Risk of fatal and nonfatal lactic acidosis with metformin use in type 2 diabetes mellitus. Cochrane Database Syst Rev. 2010;CD002967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 175]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
163.  Moore EM, Mander AG, Ames D, Kotowicz MA, Carne RP, Brodaty H, Woodward M, Boundy K, Ellis KA, Bush AI. Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care. 2013;36:2981-2987.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 257]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
164.  Smith GD, Egger M, Shipley MJ, Marmot MG. Post-challenge glucose concentration, impaired glucose tolerance, diabetes, and cancer mortality in men. Am J Epidemiol. 1992;136:1110-1114.  [PubMed]  [DOI]  [Cited in This Article: ]
165.  La Vecchia C, Negri E, Decarli A, Franceschi S. Diabetes mellitus and the risk of primary liver cancer. Int J Cancer. 1997;73:204-207.  [PubMed]  [DOI]  [Cited in This Article: ]
166.  Hense HW, Kajüter H, Wellmann J, Batzler WU. Cancer incidence in type 2 diabetes patients - first results from a feasibility study of the D2C cohort. Diabetol Metab Syndr. 2011;3:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
167.  Wang P, Kang D, Cao W, Wang Y, Liu Z. Diabetes mellitus and risk of hepatocellular carcinoma: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2012;28:109-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 201]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
168.  Wang C, Wang X, Gong G, Ben Q, Qiu W, Chen Y, Li G, Wang L. Increased risk of hepatocellular carcinoma in patients with diabetes mellitus: a systematic review and meta-analysis of cohort studies. Int J Cancer. 2012;130:1639-1648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 288]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
169.  Wang YG, Wang P, Wang B, Fu ZJ, Zhao WJ, Yan SL. Diabetes mellitus and poorer prognosis in hepatocellular carcinoma: a systematic review and meta-analysis. PLoS One. 2014;9:e95485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 64]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
170.  Ruiter R, Visser LE, van Herk-Sukel MP, Coebergh JW, Haak HR, Geelhoed-Duijvestijn PH, Straus SM, Herings RM, Stricker BH. Lower risk of cancer in patients on metformin in comparison with those on sulfonylurea derivatives: results from a large population-based follow-up study. Diabetes Care. 2012;35:119-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 181]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]