Abstract
A high increase witnessed in type II diabetes mellitus (T2DM) globally has increasingly posed a serious threat to global increases in liver cancer with the association between diabetes mellitus type II and the survival rate in liver cancer patients showing unstable findings. An increase in the development and progression of chronic liver disease from diabetes mellitus patients may be connected to cancer of the liver with several links such as Hepatitis B and C virus and heavy consumption of alcohol. The link between T2DM patients and liver cancer is centered on non-alcoholic fatty liver disease (NAFLD) which could be a serious threat globally if not clinically addressed. Several reports identified metformin treatment as linked to a lower risk of liver cancer prognosis while insulin treatment or sulphonylureas posed a serious threat. Mechanistically, the biological linkage between diabetes type II mellitus and liver cancer are still complex to understand with only the existence of a relationship between NAFLD and high level of energy intake and diabetes mellitus induces hepatic damage, increased liver weight thereby causes multiple pro-inflammatory cytokines that lead to the development of liver cancer. Therefore, this review gives an account of the pathophysiological importance of liver cancer position with T2DM, with the role of NAFLD as an important factor that bridges them.
Keywords: liver cancer, diabetes mellitus, prognosis, cytokines, steatohepatitis, cirrhosis
Introduction
Type II diabetes mellitus (T2DM) is a chronic metabolic disorder of global burden that is associated with impaired insulin secretion/function and glucose metabolism.1 Oxidative stress has been implicated in the etiology of T2DM with consequent secondary complications including neuronal impairment, kidney and liver damage, and liver cancer.2–4 Individuals with T2DM are at greater risk of developing cancer and dying from it.5
An increase in the death rate globally from chronic liver disease had been linked to T2DM patients and liver cancer.4 The serious threat posed by cancer and this metabolic disease to people's health is alarming in both developed and developing nations. Although accumulating evidence has implicated genetic alterations in cancer development, progression, and worse clinical outcome,6–12 the question of whether T2DM influences cancer incidence or influences cancer's natural history and vice versa remains an integral part for investigating the relationship between T2DM and liver cancer pathophysiology.13–17 Moreover, a higher level of energy intake against energy expenditure, which results in insulin sensitivity impairment, increase in liver weight, fat mass and non-alcoholic fatty liver disease (NAFLD) has been linked to evidence between T2DM and liver cancer.16–21
The attributed fact related to sporadic increase in T2DM patients and obesity with prevalence in NAFLD and increase in liver weight and other related symptoms posed a threat to the increase in liver cancer cases. The progressive hepatic damage from steatosis to steatohepatitis and cirrhosis surfaced in the larger percent of the patients, which have also been observed in T2DM conditions.22–25
Given the sharp increase in T2DM globally and its risk factors linked to NAFLD and liver cancer, scientists like diabetologists and oncologists considered managing diabetes patients who had been diagnosed with liver cancer because of the strong link between T2DM and liver cancer. The link between T2DM and HCC is mediated by a chronic inflammatory state.26–29 Indeed, the eradication of HCV infection with direct antiviral agents (DAAs), leading to a reduction of the chronic inflammatory state, determines the reduction both of the onset of type 2 diabetes,30 and the risk of HCC.31,32
In this review, we carefully examine the temporality of the relationship between T2DM and liver cancer with the aim to potentially evaluate the modification role of NAFLD and other potential risk factors in the context of the relationship between TDM2 and liver cancer.
Global Studies Show Linkage of T2DM to Liver Cancer Risk
Global data on T2DM has increased greatly in recent years with the progressive increase of T2DM worldwide.13,33,34 The estimated world prevalence of T2DM is approximated to be 51% higher by 2045 with 700 million people projected to be affected if not properly controlled.35 Similarly, around 10 million deaths were recorded in 2020 cancer cases with uncontrollable abnormal cell growth in the organ or tissue placing liver cancer as one of the leading causes of cancer death.17,36 An evidence-based study shows that T2DM is increasing by 2 to 5-fold in liver cancer patients after changing the biasing of other factors.37–40 A progressive increase of T2DM in diabetes conditions leads to an increase in liver cancer by 10-fold in the presence of viral hepatitis condition and alcohol intake.41 A Competing Risks Analysis study by Baena-Díez et al42 concluded that diabetes is associated with premature death from cardiovascular disease, cancer, and noncardiovascular, noncancer causes,42 and individuals with T2DM are at greater risk of developing cancer and of dying from it.5 An evidenced-based report of an Italian cohort over an 11-year period, in which death certificates were reviewed, showed that death from site-specific cancer of the liver showed a higher rate of patients dying with T2DM compared with other.16,43 Based on the same findings, which are similar to this evidence based on site-specific cancer in relation to T2DM, the American Diabetes Association and American Cancer Association agreed that the incidence rate of liver cancer is increasingly higher in patients suffering from T2DM.44–47 Thirty-four years ago, a link was reported among patients with T2DM and cancer of the liver where a case-control study involving 105 liver cancer patients and other related cancer cases were matched by age and sex.48 Between 1984 and 1989, La Vecchia et al revealed that out of 242 cases reported, patients with T2DM had an increase of 2.5-fold in liver cancer development and other independent metabolic factors and potential confounding variables.49 The result from epidemiologists revealed a high risk of liver cirrhosis connected with NAFLD patients leading to liver cancer.19,50 Therefore, more evidence is needed for the establishment of epidemiological relations and cause-effect association between T2DM and liver cancer disease.
Mitochondria compose a dynamic population of organelles, existing partly both as units and as an interconnected network viewed under the microscope as constantly moving.51 It takes the microtubule track through dynein motors for mitochondria to travel within the cell to regions of high-energy demand via the uptake of calcium by mitochondria regulating ATP production.52,53 Given the role of mitochondria in β-cells stimulus-secretion coupling, a few genetic studies in humans implicate mitochondrial dysfunction in the pathogenesis of diabetes mellitus.54–56 Nevertheless, research revealed that mutation in the mitochondrial tRNA synthase tRNALeu results in inherited diabetes while the variant in the mitochondrial transcription factor TFB1M has been implicated worldwide by GWAS (genome-wide association studies).56 The mitochondria and ER were larger in size, close to each other with mitochondria changing formation in diabetes conditions thereby reflecting the stress of ER and dysfunction of mitochondria.57
In recent years, it was reported that liver cancer not only developed in patients suffering from cirrhotic NAFLD but developed increasingly in non-cirrhotic patients with NASH.50,58–60 Other reports described the frequent consumption of alcohol and age-old condition as risk factors for the development of liver disease. In all the research studies carried out, the survival rate of patients with cirrhotic NAFLD and with liver cancer disease was significantly shorter compared with patients with liver cancer secondary to HCV cirrhosis due to their tenacity in older age and the possibility of larger tumor diameters with less surveillance to patients on liver cancer secondary to HCV cirrhosis.50,61,62 (Figure 1).
Figure 1.
Non-alcoholic fatty liver disease (NAFLD), which can proceed to cirrhosis and hepatocellular cancer, can be caused by type 2 diabetes mellitus (HCC).
Pathophysiological Mechanism Between T2DM and NAFLD, a Potential Risk Factor for Liver Cancer
The details of pathophysiological links between T2DM, NAFLD, and liver cancer are unclear but the liver cancer mechanistic approach in relation to this context is increasingly being understood with more research in finding the pathophysiological link between them. Elevated levels of hepatic insulin resistance, oxidative stress, chronic low pro-inflammation level, and lipotoxicity are the strongest indicators that exist between T2DM and NAFLD. This is because an increase in interleukin-1, interleukin-6, tumor necrosis factor-alpha, and tumor growth factor-beta occurred as a result of the development of insulin resistance and lipotoxicity.21,63,64 Moreover, there is an elevated level of vasoactive factors and pro-oxidant molecules in the blood bloodstream that results in hepatic cellular growth and multiplication with inhibition in cellular apoptosis, which eventually results in liver cancer.60,65–69 Observed concentration increases of insulin in the blood increases insulin-like growth factor-1 (IGF-1) thereby stimulating insulin receptor substrate-1 (IRS-1), an activator of some intracellular signaling pathways.67,69–72 (Figure 2).
Figure 2.
The details of pathophysiological links between T2DM, NAFLD, and liver cancer are unclear but the liver cancer mechanistic approach in relation to this context is increasingly improved recently with more research to find the pathophysiological link. Elevated levels of hepatic insulin resistance, oxidative stress, chronic low pro-inflammation level, and lipotoxicity are the strongest indicative stages that exist between T2DM and NAFLD.
Previous evidence revealed that T2DM was a bad prognostic factor for the long-term survival of cirrhotic patients.73 This showed similar evidence in NAFLD because of their relationship in activating oxidative stress thereby release of reactive oxygen species (ROS).70,74 Several studies revealed ROS is produced when hepatocytes are steatotic thereby promoting the development of liver cancer and other cancers.64,65,75,76 The increase caused by oxidative stress production results in DNA damage, cytotoxicity as well as activation and suppression of multiple genes that are potentially implicated in cellular proliferation and growth thereby producing hepatic carcinogenesis.76 Therefore, several reports have shown a closed relationship between T2DM and NAFLD due to their disrupted mitochondria as a result of ROS production.64,76–78
The mechanistic process of NAFLD production from T2DM is complicated, and this has been explored in isolated biological systems. Fatty liver, obesity, and insulin resistance have all been shown to be co-factors in liver disease. Because of increased absorption of free fatty acids and de-novo liponeogenesis in hepatocytes, fatty liver results in an intracellular build-up of triglycerides.79,80 At the same time, the hepatic secretion of extremely low-density lipoproteins is reduced. The liver damage includes cellular necrosis and inflammation, which are caused by an increase in mitochondria oxidative stress on triglycerides, resulting in the formation of free radicals and peroxisomes.81,82 Adipokines (cytokines generated by adipocytes), such as leptin and tumor necrosis factor (TNF), are produced in excess, and worsen mitochondria oxidative stress. The regulating adipokine adiponectin is reduced, thereby encouraging the action of inflammatory adipokines. These chemical mediators are produced as a result of inflammation, cell necrosis, and adipokines that stimulate liver stellate cells, causing them to produce more collagen, connective tissue growth factor, and extracellular matrix, promoting fibrosis.83–85 (Figure 3).
Figure 3.
Result of diabetes type II in the damaged liver. Insulin resistance causes adipose tissue to emit free fatty acids (FFA). FFAs accumulate in liver cells, and de novo liponeogenesis (DNL) plays a role as well. Hepatic cells’ decreased release of very low-density lipoprotein (VLDL) saturates hepatocytes, resulting in steatosis. Excess intracellular FFAs and the impact of adipokines (leptin and tumor necrosis factor-alpha (TNF-) exacerbate mitochondrial oxidative stress. Excessive oxidative stress results in the production of free radicals, which causes inflammation and cellular necrosis. The stellate cells are stimulated to make collagen by tissue inflammation.
A recent report has also linked the gut microbiota with obesity, thereby alluding to the importance of gut microbiota, a key factor in energy production. Most metabolites generated from human mammalian blood come from gut microbiota with BA (bile acids) as the most important metabolite produced in the liver.86,87 Therefore, an alteration of gut microbiota results in T2DM, NAFLD, and obesity pathogenesis with several pieces of evidence proving the possible implication of gut microbiota in hepatic carcinogenesis.88–90
In addition, cell cycle regulators in the gene encoding for albumin (TP53 and CDKN2A) and (CTNNB1 and AXIN1), genes of the β-catenin/WNT signaling pathway were revealed in wide studies of the pathophysiology of the cancer liver using the next generation sequencing and other omics techniques.64,91,92 Other experimental reports revealed that patients suffering from NASH pose a risk of a high level of genomic instability, which results in the development of liver cancer.93,94 Furthermore, there is increased evidence of the presence of the patatin-like phospholipase domain-containing protein-3 (PNPLA3) gene. This gene encodes for adiponutrin (protein) found in intra-hepatic lipid droplets, promoting lipogenesis and lipolysis. The gene has also been reported to be linked with an increased risk of NAFLD progression.95–97 Therefore, there is a mechanism of PNPLA3 I148M polymorphism of genotyping revealing the damaged hepatic failure and carcinogenesis.
Some Glucose-Lowering Medications and Their Relationship to Liver Cancer Risk
The liver as one of the most important organs that play a central role in the metabolism of drugs, xenobiotic, and nutrients are highly susceptible to drug-induced damage.98 Apart from the study carried out by Evans et al in 2005 which suggested giving metformin, a glucose attenuating medication, for liver cancer in patients suffering from T2DM.99 A group of scientists has demonstrated the evidence-based research on a population-based cohort of 10,309 T2DM patients followed up for nearly 5 years with the outcome revealing that cancer mortality significantly differed among the various treatments for diabetes at baseline: 3.5% for metformin users, 4.9% for sulphonylureas users and 5.8% for insulin users.100,101
Several other scientists have shown proven evidence in their research findings relating to those treated with metformin had a lower incidence of total cancer than those treated with sulphonylureas or insulin, independent of age, sex, BMI, hemoglobin A1c, smoking, and use of other medications including the hospital-based findings where sulfonylureas or insulin conferred the highest HCC risk, while the metformin or glitazones usage associated with a larger percent risk reduction in liver cancer in patients with T2DM.102,103 Contrary to the above finding, Tsilidis et al revealed that users of sulphonylureas or metformin had similar incidence rates of total cancer over 5 years of follow-up in a retrospective cohort study of nearly 96,000 individuals with T2DM who make use of metformin or other oral glucose-lowering agents within 12 months of the diagnosis.104
Studies have also reported the beneficial effects of metformin as an anti-aging agent.105 Metformin also acts as an endothelial protector5 that inhibit tumor growth, and metastasis via an AMPK-dependent signaling network.106 Interestingly, metformin has also been reported to synergize with and improved the activities and safety of clinical drugs for the treatment of lung cancer.107 Altogether, it's evident that metformin does not only alleviate hyperglycemia but also protects against the development of cancer and aging
Thus, the idea that a medication designed for the use of diabetes treatment may either increase or decrease the risk of liver cancer, or even influence cancer prognosis is still unclear because most observational studies predict that metformin, one of the drugs for diabetes patients, might have chemopreventive potential against liver cancer and a biologically plausible mechanism also exists (metformin drug activates AMP-activated protein kinase (AMPK) and inhibits the PI3K/AKT/mTOR signaling pathway that is important in regulating the cell cycle). Furthermore, it is unclear whether an insulin-related increase in liver cancer risk is related to toxicity associated with the medication, or if it is simply reflective of increased HCC risk in patients with more severe T2DM.108,109
Conclusion
The high increase witnessed in T2DM globally has increasingly posed a serious threat to the global increase in liver cancer with an association between T2DM and liver cancer. However, the involvement of several underlying pathophysiologies of this cancer is linked to several diseases like NAFLD, increased hepatic insulin resistance hyperinsulinemia, activated level of pro-inflammatory mediators, oxidative stress, JNK-1 activation, increased IGF-1 activity, altered gut microbiota, and immunomodulation. Therefore, in-depth knowledge of the underlying pathophysiology could provide treatment breakthroughs for patients being treated when confronted with both T2DM and liver cancer.
Footnotes
Author Contributions: OSA designed and conducted the study and B L., B.O.A, B.O., A.F., O.A.O., S.K., G.E., and A.J oversaw the study. All author read and approved the final version of the manuscript.
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: The authors are thankful for the financial support provided by the Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) Brazil — grant number [E-26/200.891/2021], and the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) - grant number [313119/2020-1].
ORCID iDs: Sunday Amos Onikannihttps://orcid.org/0000-0003-0491-3301
Bashir Lawal https://orcid.org/0000-0003-0676-5875
Saeed M Kabrah https://orcid.org/0000-0003-2992-8497
Carlos Adam Conte-Junior https://orcid.org/0000-0001-6133-5080
References
- 1.Abubakar AN, Badmos FO, Saidu AN, Yunus IO, Hamzah RU, Lawal B. Phytochemical compositions, and hypoglycemic effect of methanol leaf extract of Telfairia Occidentalis in alloxan-induced diabetic rats. AROC Nat Prod Res. 2021;1(1):052-060. [Google Scholar]
- 2.Onikanni AS, Lawal B, Oyinloye BEet al. Therapeutic efficacy of clompanus pubescent leaves fractions via downregulation of neuronal cholinesterases/Na + -K + ATPase/IL-1 β, and improving the neurocognitive and antioxidants status of streptozotocin-induced diabetic rats. Biomed Pharmacother. 2022;148: 112730. 10.1016/j.biopha.2022.112730. [DOI] [PubMed] [Google Scholar]
- 3.Onikanni AS, Lawal B, Olusola AO, et al. Sterculia tragacantha lindl leaf extract ameliorates STZ-induced diabetes, oxidative stress, inflammation, and neuronal impairment. J Inflamm Res. 2021;14:6749-6764. doi: 10.2147/jir.s319673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kim WR, Brown RS, Jr, Terrault NA, El-Serag H. Burden of liver disease in the United States: summary of a workshop. Hepatology. 2002;36(1):227-242. [DOI] [PubMed] [Google Scholar]
- 5.Rey-Reñones C, Baena-Díez JM, Aguilar-Palacio I, Miquel C, Grau M. Type 2 diabetes Mellitus and cancer: epidemiology, physiopathology and prevention. Biomedicines. 2021;9:1429. doi: 10.3390/biomedicines9101429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Yeh YC, Lawal B, Hsiao M, Huang TH, Huang CYF. Identification of NSP3 (SH2D3C) as a prognostic biomarker of tumor progression and immune evasion for lung cancer and evaluation of organosulfur compounds from Allium sativum L. As therapeutic candidates. Biomedicines. 2021;9(11):1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lawal B, Kuo YC, Tang SLet al. et al. Transcriptomic-Based identification of the immuno-oncogenic signature of cholangiocarcinoma for HLC-018 multi-target therapy exploration. Cells. 2021;10(11):2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wu ATH, Yeh YC, Huang YJ, Mokgautsi N, Lawal B, Huang TH. Gamma-Mangostin isolated from Garcinia mangostana suppresses colon carcinogenesis and stemness by downregulating the GSK3β/β-catenin/CDK6 cancer stem pathway. Phytomedicine. 2021;95:153797. doi: 10.1016/j.phymed.2021.153797. [DOI] [PubMed] [Google Scholar]
- 9.Wu SY, Lin KC, Lawal B, Wu ATH, Wu CZ. MXD3 As an onco-immunological biomarker encompassing the tumor microenvironment, disease staging, prognoses, and therapeutic responses in multiple cancer types. Comput Struct Biotechnol J. 2021;19:4970-4983. doi: 10.1016/j.csbj.2021.08.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lawal B, Wang YC, Wu ATH, Huang HS. Pro-Oncogenic c-met/EGFR, biomarker signatures of the tumor microenvironment are clinical and therapy response prognosticators in colorectal cancer, and therapeutic targets of 3-phenyl-2H-benzo[e][1,3]-oxazine-2,4(3H)-dione derivatives. Front Pharmacol. 2021;12:1–24. doi: 10.3389/fphar.2021.691234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lawal B, Tseng SH, Olugbodi JO, et al. Pan-Cancer analysis of immune complement signature C3/C5/C3AR1/C5AR1 in association with tumor immune evasion and therapy resistance. Cancers (Basel). 2021;13:4124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chen JH, Wu ATH, Lawal Bet al. et al. Identification of cancer hub gene signatures associated with immune-suppressive tumor microenvironment and ovatodiolide as a potential cancer immunotherapeutic agent. Cancers (Basel). 2021;13(15):3847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Whiting DR, Guariguata L, Weil C, Shaw J. IDF Diabetes atlas: global estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Res Clin Pract. 2011;94(3):311-321. [DOI] [PubMed] [Google Scholar]
- 14.Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U, Shaw JE. Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract. 2014;103(2):137-149. [DOI] [PubMed] [Google Scholar]
- 15.McGlynn KA, London WT. The global epidemiology of hepatocellular carcinoma: present and future. Clin Liver Dis. 2011;15(2):223-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mantovani A, Targher G. Type 2 diabetes mellitus and risk of hepatocellular carcinoma: spotlight on nonalcoholic fatty liver disease. Ann Transl Med. 2017;5(13):270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87-108. [DOI] [PubMed] [Google Scholar]
- 18.Sun B, Karin M. Obesity, inflammation, and liver cancer. J Hepatol. 2012;56(3):704-713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Margini C, Dufour JF. The story of HCC in NAFLD: from epidemiology, across pathogenesis, to prevention and treatment. Liver Int. 2016;36(3):317-324. [DOI] [PubMed] [Google Scholar]
- 20.Streba LAM, Vere CC, Rogoveanu I, Streba CT. Nonalcoholic fatty liver disease, metabolic risk factors, and hepatocellular carcinoma: an open question. World J Gastroenterol. 2015;21(14):4103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Noureddin M, Rinella ME. Nonalcoholic fatty liver disease, diabetes, obesity, and hepatocellular carcinoma. Clin Liver Dis. 2015;19(2):361-379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73-84. [DOI] [PubMed] [Google Scholar]
- 23.Lonardo A, Bellentani S, Argo CK, et al. Epidemiological modifiers of non-alcoholic fatty liver disease: focus on high-risk groups. Dig Liver Dis. 2015;47(12):997-1006. [DOI] [PubMed] [Google Scholar]
- 24.Portillo-Sanchez P, Bril F, Maximos M, et al. High prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus and normal plasma aminotransferase levels. J Clin Endocrinol Metab. 2015;100(6):2231-2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol. 2015;62(1):S47-S64. [DOI] [PubMed] [Google Scholar]
- 26.Matteoni CA, Younossi ZM, Gramlich T, Boparai N, Liu YC, McCullough AJ. Nonalcoholic fatty liver disease: a spectrum of clinical and pathological severity. Gastroenterology. 1999;116(6):1413-1419. [DOI] [PubMed] [Google Scholar]
- 27.Falck-Ytter Y, Younossi ZM, Marchesini G, McCullough AJ. Clinical features and natural history of nonalcoholic steatosis syndromes. In Proceedings of Seminars in liver disease; pp. 017-026. [DOI] [PubMed] [Google Scholar]
- 28.Belfiore F, Iannello S. Insulin resistance in obesity: metabolic mechanisms and measurement methods. Mol Genet Metab. 1998;65(2):121-128. [DOI] [PubMed] [Google Scholar]
- 29.El-Serag HB, Tran T, Everhart JE. Diabetes increases the risk of chronic liver disease and hepatocellular carcinoma. Gastroenterology. 2004;126(2):460-468. [DOI] [PubMed] [Google Scholar]
- 30.Adinolfi LE, Petta S, Fracanzani AL, et al. Reduced incidence of type 2 diabetes in patients with chronic hepatitis C virus infection cleared by direct-acting antiviral therapy: a prospective study. Diabetes, Obe Metab. 2020;22(12):2408-2416. 10.1111/dom.14168. [DOI] [PubMed] [Google Scholar]
- 31.Rinaldi L, Perrella A, Guarino M, et al. Incidence and risk factors of early HCC occurrence in HCV patients treated with direct acting antivirals: a prospective multicentre study. J Transl Med. 2019;17(1):292. doi: 10.1186/s12967-019-2033-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rinaldi L, Nevola R, Franci G, et al. Risk of hepatocellular carcinoma after HCV clearance by direct-acting antivirals treatment predictive factors and role of epigenetics. Cancers (Basel). 2020;12(6):1351. doi: 10.3390/cancers12061351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sudharsanan N, Ali MK, Mehta NK, Narayan KV. Population aging, macroeconomic changes, and global diabetes prevalence, 1990–2008. Popul Health Metr. 2015;13(2):1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Leahy S, O’Halloran A, O’Leary Net al. et al. Prevalence and correlates of diagnosed and undiagnosed type 2 diabetes mellitus and pre-diabetes in older adults: findings from the Irish longitudinal study on ageing (TILDA). Diabetes Res Clin Pract. 2015;110(3):241-249. [DOI] [PubMed] [Google Scholar]
- 35.Federation ID. IDF Diabetes atlas ninth. 9th ed. Dunia: IDF. 2019:978-2-930229-87-4. [Google Scholar]
- 36.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394-424. [DOI] [PubMed] [Google Scholar]
- 37.El-Serag HB, Richardson PA, Everhart JE. The role of diabetes in hepatocellular carcinoma: a case-control study among United States veterans. Am J Gastroenterol. 2001;96(8):2462-2467. [DOI] [PubMed] [Google Scholar]
- 38.Beasley RP. Diabetes and hepatocellular carcinoma editorial. Hepatology. 2006;44(6):1408-1410. [DOI] [PubMed] [Google Scholar]
- 39.Davila J, Morgan R, Shaib Y, McGlynn K, El-Serag H. Diabetes increases the risk of hepatocellular carcinoma in the United States: a population based case control study. Gut. 2005;54:533-539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Amarapurkar DN, Patel ND, Kamani PM. Impact of diabetes mellitus on outcome of HCC. Ann Hepatol. 2008;7(4):148-151. [PubMed] [Google Scholar]
- 41.Hassan MM, Hwang LY, Hatten CJet al. et al. Risk factors for hepatocellular carcinoma: synergism of alcohol with viral hepatitis and diabetes mellitus. Hepatology. 2002;36(5):1206-1213. [DOI] [PubMed] [Google Scholar]
- 42.Baena-Díez JM, Peñafiel J, Subirana I, et al. Risk of cause-specific death in individuals with diabetes: a competing risks analysis. Diabetes Care. 2016;39(11):1987-1995. doi: 10.2337/dc16-0614. [DOI] [PubMed] [Google Scholar]
- 43.Verlato G, Zoppini G, Bonora E, Muggeo M. Mortality from site-specific malignancies in type 2 diabetic patients from verona. Diabetes Care. 2003;26(4):1047-1051. [DOI] [PubMed] [Google Scholar]
- 44.Carstensen B, Witte D, Friis S. Cancer occurrence in danish diabetic patients: duration and insulin effects. Diabetologia. 2012;55(4):948-958. [DOI] [PubMed] [Google Scholar]
- 45.Geier A, Wellmann J, Wellmann Iet al. et al. Cancer detection rates following enrolment in a disease management programme for type 2 diabetes. Diabetologia. 2013;56(9):1944-1948. [DOI] [PubMed] [Google Scholar]
- 46.Tseng CH. Diabetes and risk of prostate cancer: a study using the national health insurance. Diabetes Care. 2011;34(3):616-621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Giovannucci E, Harlan DM, Archer MCet al. Diabetes and cancer: a consensus report. CA Cancer J Clin. 2010;60(4):207-221. [DOI] [PubMed] [Google Scholar]
- 48.Lawson D, Gray J, McKillop C, Clarke J, Lee F, Patrick R. Diabetes mellitus and primary hepatocellular carcinoma. QJM: Int J Med 1986;61(234):945-955. [PubMed] [Google Scholar]
- 49.Prizment AE, Folsom AR, Cerhan JR, Flood A, Ross JA, Anderson KE. History of allergy and reduced incidence of colorectal cancer, iowa women’s health study. Cancer Epidemiol Prev Biomark. 2007;16(11):2357-2362. [DOI] [PubMed] [Google Scholar]
- 50.Calzadilla Bertot L, Adams LA. The natural course of non-alcoholic fatty liver disease. Int J Mol Sci. 2016;17(5):774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Rutter GA, Rizzuto R. Regulation of mitochondrial metabolism by ER Ca2 + release: an intimate connection. Trends Biochem Sci. 2000;25(5):215-221. [DOI] [PubMed] [Google Scholar]
- 52.Denton RM. Regulation of mitochondrial dehydrogenases by calcium ions. Biochim Biophys Acta (BBA)-Bioenerget. 2009;1787(11):1309-1316. [DOI] [PubMed] [Google Scholar]
- 53.Mishra P, Chan DC. Metabolic regulation of mitochondrial dynamics. J Cell Biol. 2016;212(4):379-387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Rutter GA, Pullen TJ, Hodson DJ, Martinez-Sanchez A. Pancreatic β-cell identity, glucose sensing and the control of insulin secretion. Biochem J. 2015;466(2):203-218. [DOI] [PubMed] [Google Scholar]
- 55.Maechler P, Wollheim CB. Mitochondrial function in normal and diabetic β-cells. Nature. 2001;414(6865):807-812. [DOI] [PubMed] [Google Scholar]
- 56.Mulder H. Transcribing β-cell mitochondria in health and disease. Mol Metab. 2017;6(9):1040-1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fex M, Nitert MD, Wierup N, Sundler F, Ling C, Mulder H. Enhanced mitochondrial metabolism may account for the adaptation to insulin resistance in islets from C57BL/6J mice fed a high-fat diet. Diabetologia. 2007;50(1):74-83. [DOI] [PubMed] [Google Scholar]
- 58.Kawada N, Imanaka K, Kawaguchi Tet al. Hepatocellular carcinoma arising from non-cirrhotic nonalcoholic steatohepatitis. J Gastroenterol. 2009;44(12):1190. [DOI] [PubMed] [Google Scholar]
- 59.Rahman RN, Ibdah JA. Nonalcoholic fatty liver disease without cirrhosis is an emergent and independent risk factor of hepatocellular carcinoma: a population based study. In Proceedings of Hepatology; pp. 241A-241A. [Google Scholar]
- 60.Dongiovanni P, Romeo S, Valenti L. Hepatocellular carcinoma in nonalcoholic fatty liver: role of environmental and genetic factors. World J Gastroenterol. 2014;20(36):12945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Guzman G, Brunt EM, Petrovic LM, Chejfec G, Layden TJ, Cotler SJ. Does nonalcoholic fatty liver disease predispose patients to hepatocellular carcinoma in the absence of cirrhosis? Arch Pathol Lab Med. 2008;132(8):1761-1766. [DOI] [PubMed] [Google Scholar]
- 62.Reddy SK, Steel JL, Chen HW, et al. Outcomes of curative treatment for hepatocellular cancer in nonalcoholic steatohepatitis versus hepatitis C and alcoholic liver disease. Hepatology. 2012;55(6):1809-1819. [DOI] [PubMed] [Google Scholar]
- 63.Weiskirchen R, Tacke F. Immune surveillance of liver cancer in non-alcoholic fatty liver disease: excess lipids cause CD4 T-cells loss and promote hepatocellular carcinoma development. Hepatobiliary Surg Nutr. 2016;5(5):433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wainwright P, Scorletti E, Byrne C. Type 2 diabetes and hepatocellular carcinoma: risk factors and pathogenesis. Curr Diab Rep. 2017;17(4):20. [DOI] [PubMed] [Google Scholar]
- 65.Sakurai T, He G, Matsuzawa Aet al. et al. Hepatocyte necrosis induced by oxidative stress and IL-1α release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell. 2008;14(2):156-165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hoesel B, Schmid JA. The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer. 2013;12(86):1-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Tunissiolli NM, Castanhole-Nunes MMU, Biselli-Chicote PM, Pavarino ÉC, da Silva RF. Hepatocellular carcinoma: a comprehensive review of biomarkers, clinical aspects, and therapy. Asian Pac J Cancer Prev. 2017;18(4):863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Huan L, Liang LH, He XH. Role of microRNAs in inflammation-associated liver cancer. Cancer Biol Med. 2016;13(4):407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Michelotti GA, Machado MV, Diehl AM. NAFLD, NASH And liver cancer. Nat Rev Gastroenterol Hepatol. 2013;10(11):656-665. [DOI] [PubMed] [Google Scholar]
- 70.Elia G, Fallahi P. Hepatocellular carcinoma and CXCR3 chemokines: a narrative review. Clin Ter. 2017;168(1):e37-e41. [DOI] [PubMed] [Google Scholar]
- 71.Zhang Q, Su J, Wang Zet al. et al. MicroRNA-149* suppresses hepatic inflammatory response through antagonizing STAT3 signaling pathway. Oncotarget. 2017;8(39):65397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Krishna-Subramanian S, Singer S, Armaka M, et al. RIPK1 And death receptor signaling drive biliary damage and early liver tumorigenesis in mice with chronic hepatobiliary injury. Cell Death Differ. 2019;26(12):2710-2726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bianchi G, Marzocchi R, Lorusso C, Ridolfi V, Marchesini G. Nutritional treatment of chronic liver failure. Hepatol Res. 2008;38(1):S93-S101. [DOI] [PubMed] [Google Scholar]
- 74.Schlesinger S, Aleksandrova K, Pischon T, et al. Abdominal obesity, weight gain during adulthood and risk of liver and biliary tract cancer in a European cohort. Int J Cancer. 2013;132(3):645-657. [DOI] [PubMed] [Google Scholar]
- 75.Spickett CM. The lipid peroxidation product 4-hydroxy-2-nonenal: advances in chemistry and analysis. Redox Biol. 2013;1(1):145-152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hu W, Feng Z, Eveleigh Jet al. et al. The major lipid peroxidation product, trans-4-hydroxy-2-nonenal, preferentially forms DNA adducts at codon 249 of human p53 gene, a unique mutational hotspot in hepatocellular carcinoma. Carcinogenesis. 2002;23(11):1781-1789. [DOI] [PubMed] [Google Scholar]
- 77.Rhee SG, Bae YS, Lee SR, Kwon J. Hydrogen peroxide: a key messenger that modulates protein phosphorylation through cysteine oxidation. Sci Signal. 2000;2000(53):pe1-pe1. [DOI] [PubMed] [Google Scholar]
- 78.Yang S, Zhu H, Li Yet al. et al. Mitochondrial adaptations to obesity-related oxidant stress. Arch Biochem Biophys. 2000;378(2):259-268. [DOI] [PubMed] [Google Scholar]
- 79.Hickman IJ, Macdonald GA. Impact of diabetes on the severity of liver disease. Am J Med. 2007;120(10):829-834. [DOI] [PubMed] [Google Scholar]
- 80.Tolman KG, Fonseca V, Dalpiaz A, Tan MH. Spectrum of liver disease in type 2 diabetes and management of patients with diabetes and liver disease. Diabetes Care. 2007;30(3):734-743. [DOI] [PubMed] [Google Scholar]
- 81.Chalasani N, Gorski JC, Asghar MSet al. et al. Hepatic cytochrome P450 2E1 activity in nondiabetic patients with nonalcoholic steatohepatitis. Hepatology. 2003;37(3):544-550. [DOI] [PubMed] [Google Scholar]
- 82.Pessayre D, Fromenty B, Mansouri A. Mitochondrial injury in steatohepatitis. Eur J Gastroenterol Hepatol. 2004;16(11):1095-1105. [DOI] [PubMed] [Google Scholar]
- 83.Bertolani C, Marra F. The role of adipokines in liver fibrosis. Pathophysiology. 2008;15(2):91-101. [DOI] [PubMed] [Google Scholar]
- 84.Sanyal AJ. AGA Technical review on nonalcoholic fatty liver disease. Gastroenterology. 2002;123(5):1705-1725. [DOI] [PubMed] [Google Scholar]
- 85.Sanyal AJ, Campbell–Sargent C, Mirshahi F, et al. Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Gastroenterology. 2001;120(5):1183-1192. [DOI] [PubMed] [Google Scholar]
- 86.Zhao L. The gut microbiota and obesity: from correlation to causality. Nat Rev Microbiol. 2013;11(9):639-647. [DOI] [PubMed] [Google Scholar]
- 87.Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341(6150):1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Adams LA, Morrison M. The microbiome in obesity, diabetes, and NAFLD: what is your gut telling us? Curr Hepatol Rep. 2016;15(5):96-102. [Google Scholar]
- 89.Darnaud M, Faivre J, Moniaux N. Targeting gut flora to prevent progression of hepatocellular carcinoma. J Hepatol. 2013;58:385-387. [DOI] [PubMed] [Google Scholar]
- 90.Yoshimoto S, Loo TM, Atarashi K, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499(7456):97-101. [DOI] [PubMed] [Google Scholar]
- 91.Schulze K, Imbeaud S, Letouzé E, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 2015;47(5):505-511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Liu YL, Patman G, Leathart J, et al. Carriage of the PNPLA3 rs738409 C > G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma. J Hepatol. 2014;61(1):75-81. [DOI] [PubMed] [Google Scholar]
- 93.Herath NI, Leggett BA, MacDonald GA. Review of genetic and epigenetic alterations in hepatocarcinogenesis. J Gastroenterol Hepatol. 2006;21(1):15-21. [DOI] [PubMed] [Google Scholar]
- 94.Dongiovanni P, Donati B, Fares Ret al. et al. PNPLA3 I148m polymorphism and progressive liver disease. World J Gastroenterol. 2013;19(41):6969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Chamoun Z, Vacca F, Parton RG, Gruenberg J. PNPLA3/adiponutrin Functions in lipid droplet formation. Biol Cell. 2013;105(5):219-233. [DOI] [PubMed] [Google Scholar]
- 96.Romeo S, Kozlitina J, Xing C, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2008;40(12):1461-1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Falleti E, Fabris C, Cmet S, et al. PNPLA3 Rs738409c/G polymorphism in cirrhosis: relationship with the aetiology of liver disease and hepatocellular carcinoma occurrence. Liver Int. 2011;31(1):1137-1143. [DOI] [PubMed] [Google Scholar]
- 98.Alorabi M, Mohammed DS, Mostafa-Hedeab G, et al. Combination treatment of Omega-3 fatty acids and vitamin C exhibited promising therapeutic effect against oxidative impairment of the liver in methotrexate-intoxicated mice. BioMed Res Int. 2022;2022(7):4122166. doi: 10.1155/2022/4122166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. Br Med J. 2005;330(7503):1304-1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Dulskas A, Patasius A, Linkeviciute-Ulinskiene D, Zabuliene L, Smailyte G. A cohort study of antihyperglycemic medication exposure and survival in patients with gastric cancer. Aging (Albany NY). 2019;11(17):7197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Horibe Y, Adachi S, Ohno T, et al. Alpha-glucosidase inhibitor use is associated with decreased colorectal neoplasia risk in patients with type 2 diabetes mellitus receiving colonoscopy: a retrospective study. Oncotarget. 2017;8(58):97862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.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(5):750-758. [DOI] [PubMed] [Google Scholar]
- 103.Hassan MM, Curley SA, Li D, et al. Association of diabetes duration and diabetes treatment with the risk of hepatocellular carcinoma. Cancer. 2010;116(8):1938-1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Tsilidis KK, Capothanassi D, Allen NE, et al. Metformin does not affect cancer risk: a cohort study in the UK clinical practice research datalink analyzed like an intention-to-treat trial. Diabetes Care. 2014;37(10):2522-2532. [DOI] [PubMed] [Google Scholar]
- 105.Salvatore T, Pafundi PC, Morgillo F, et al. Metformin: an old drug against old age and associated morbidities. Diabetes Res Clin Pract. 2020;160:108025. doi: 10.1016/j.diabres.2020.108025. [DOI] [PubMed] [Google Scholar]
- 106.Choi YK, Park KG. Metabolic roles of AMPK and metformin in cancer cells. Mol Cells. 2013;36(4):279-287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Morgillo F, Fasano M, Della Corte CM, et al. Results of the safety run-in part of the METAL (METformin in advanced lung cancer) study: a multicentre, open-label phase I-II study of metformin with erlotinib in second-line therapy of patients with stage IV non-small-cell lung cancer. ESMO Open. 2017;2(2):e000132. doi: 10.1136/esmoopen-2016-000132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Klil-Drori AJ, Azoulay L, Pollak MN. Cancer, obesity, diabetes, and antidiabetic drugs: is the fog clearing? Nat Rev Clin Oncol. 2017;14(2):85-99. [DOI] [PubMed] [Google Scholar]
- 109.Singh S, Singh PP, Singh AG, Murad MH, Sanchez W. Anti-diabetic medications and the risk of hepatocellular cancer: a systematic review and meta-analysis. J Am College Gastroenterol ACG 2013;108(6):881-891. [DOI] [PubMed] [Google Scholar]



