• Open Access
    Editorial

    Extra-hepatic cancers in metabolic fatty liver syndromes

    Amedeo Lonardo *

    Explor Dig Dis. 2023;2:11–17 DOI: https://doi.org/10.37349/edd.2023.00014

    Received: December 29, 2022 Accepted: February 01, 2023 Published: February 24, 2023

    Academic Editor: Agustín Albillos, University of Alcalá, Spain

    The liver operates an array of vital digestive, metabolic, immunological, and purificatory physiological functions including synthesis of albumin, regulation of glucose and lipid homeostasis, detoxification of ammonia and xenobiotics, choleresis and production of hepatokines and hormones [1]. However, in healthy states, it is not involved in the storage of fatty substrates and, whenever the normal liver threshold is exceeded, accumulated triglycerides will impair whole-body insulin sensitivity, trigger sterile low-grade hepatic metaflammation, and promote the progression of the fibrosing process that will eventually conduce to cirrhosis in a subset of individuals [2, 3].

    While the histopathological cascade described above was first recognized in the 1800s by distinguished liver pathologists, such as Addison, Rokitansky, Pepper, and Bartolow, until the years 1958–1971 the debate, with the contributions by Zelman and Thaler, mainly focused on such hepato-histopathological changes being indistinguishable irrespective of the inciting triggers: alcohol, obesity, and diabetes [4]. From the perspective of clinical nosography, this finding eventually led to coining the innovative definitions of “nonalcoholic steatohepatitis (NASH)” and “nonalcoholic fatty liver disease (NAFLD)” in the 1980s. “Nonalcoholic” is a composite word that, at least in the spelling used by Ludwig et al., and by Schaffner and Thaler; does not require any hyphens [4] at variance with the use that is sometimes encountered in contemporary literature (“non-alcoholic”).

    To convert a “negative” definition (i.e., nonalcoholic) into a positive diagnosis highlighting the pathogenic (dysmetabolic) origin, while also avoiding stigmatizing alcohol consumption, a panel of international experts have proposed renaming NAFLD to metabolic dysfunction-associated fatty liver disease (MAFLD). Although endorsed to an unprecedented extent [5], MAFLD retains some elements of ambiguity and amplifies NAFLD’s heterogeneity, therefore creating major implications for research arena and clinical practice [6, 7].

    Compared to the history of NAFLD briefly summarized above, the notions that NAFLD and NAFLD-related metabolic conditions could also be a risk factor for the development of primary liver cancer (PLC) in a limited proportion of individuals are more recent (Table 1) [813].

    Earliest case reports and systematic published studies supporting the notions that “cryptogenic cirrhosis” equals “NAFLD-cirrhosis” in most cases; and that PLCs, hepatocellular carcinoma (HCC), and cholangiocarcinoma (CC) may indeed occur as a complication of NAFLD and related metabolic disorders

    Author, year [Ref]MethodFindingsConclusion
    Caldwell et al. [8], 1999Findings from 70 consecutive CryptoCir probands reassessed for alcohol consumption, evaluated by the IAH score and for risks of viral hepatitis and NASH were compared to 50 consecutive NASH patients, 39 nonalcoholic patients with HCV cirrhosis, and 33 with cirrhosis owing to PBCThe CryptoCir group, that comprised 70% of women in their 60s, had a prevalence of T2D and obesity significantly higher than those with cirrhosis owing to either PBC or HCV. Conversely, the prevalence of obesity and T2D was similar to the NASH patients who were, on average, 10 years youngerData suggest that NASH is an under-recognized cause in many CryptoCir patients, most of whom are older ladies with T2D and obesity
    Zen et al. [9], 2001A 58-year-old lady who did not drink alcohol and was negative for all serological markers of HBV and HCV infection received a diagnosis of T2D, treated with insulin therapy. Four years later, liver biopsy (performed to investigate altered liver tests) was compatible with NASHIn the follow-up, the patient was re-biopsied for multifocal hepatopathy and 3 out of the 4 liver nodules were moderately differentiated HCC (10 years after the diagnosis of NASH), well-differentiated HCC (11 years later) and dysplastic nodule (11 years later)This case study is the first proof-of-concept published anecdotal evidence that HCC may develop as a late NASH complication
    Bugianesi et al. [10], 2002Twenty-three out of forty-four CryptoCir patients retrospectively identified among 641 cirrhosis-associated HCCs were actively followed up and compared to viral- and alcohol-associated HCCThe prevalence of obesity and T2D was significantly higher in patients with CC, who also had higher glucose, cholesterol, triglyceridemia, and IR; aminotransferase levels were lower. Iron status and prevalence of mutations in the HFE gene did not differ. At LRA hypertriglyceridemia, T2D, and normal aminotransferases were independently associated with HCC arising in CryptoCirCharacteristics compatible with NASH are more common in HCC arising in patients with CryptoCir than in age- and sex-matched HCC cases owing to viral or alcoholic etiology suggesting that HCC may occur as a late complication of NASH-cirrhosis
    Marrero et al. [11], 2002Among 105 consecutive HCC patients, the most common etiologies of CLD were HCV and CC (51% and 29%, respectively). Half of the CryptoCir patients exhibited either histologic or clinical features associated with NAFLD. In 50% of cases, HCC was diagnosed during surveillance (group I); in the remaining patients, HCC was symptomatic (group II)Group I patients had smaller cancers (P = 0.01), were more likely to be eligible for surgical treatment (P = 0.005) and had higher survival rates than group II patients (P = 0.001). CC patients were less likely to have been submitted to surveillance for HCC and, accordingly, were diagnosed larger tumor burdensHCV and CryptoCir were the most common etiologies of HCC. NAFLD accounted for at least 13% of the cases
    Michelini et al. [12], 2007The authors speculated that IR might be a risk factor also for CC, like other cancer typesTo illustrate their speculation, these authors reported on 3 ICC cases that exhibited clinical manifestations of IR such as obesity, dyslipidemia, and NAFLD as the common grounds predisposing to CCThis case study is the first proof-of-concept published anecdotal evidence that conditions belonging to the domain of MetS may be the only biologically plausible risk factor in a fraction of CC cases
    Welzel et al. [13], 2007The SEERM database was utilized to evaluate comorbid conditions of 535 ICC patients, 549 ECC patients, and 102,782 cancer-free controls. Data were analyzed with LRAFurther to established risk factors, several endocrine and metabolic comorbidities were strongly associated with both ECC and ICC: cholelithiasis (P < 0.001), diabetes (P < 0.001), thyrotoxicosis (ECC, P = 0.006; ICC, P = 0.04). Conditions associated with ICC alone included obesity (P = 0.01) and NAFLD (P = 0.02)This pioneering study identified several novel metabolic risk factors for ECC and ICC
    Display full size

    CLD: chronic liver disease; CryptoCir: cryptogenic cirrhosis; ECC: extrahepatic cholangiocarcinoma; HBV: hepatitis B virus; HCV: hepatitis C virus; IAH: International Autoimmune Hepatitis; ICC: intrahepatic cholangiocarcinoma; IR: insulin resistance; LRA: logistic regression analysis; MetS: metabolic syndrome; PBC: primary biliary cholangitis; SEERM: Surveillance, Epidemiology and End Results-Medicare; T2D: type 2 diabetes

    While it is logical and somewhat foreseeable that PLC may eventually develop in some individuals with NAFLD, more recent observations substantiate the probably unexpected theory that, further to PLCs, NAFLD might also be a precursor of some extra-hepatic cancer types. This invited editorial will shortly summarize history, mechanisms, and implications of this intriguing association.

    In 2014, evidence was published that NAFLD patients (particularly those with NASH) were exposed to a strong risk of developing colorectal neoplasms, in addition to other extra-hepatic outcomes (such as cardiovascular disease, T2D, and chronic kidney disease) [14, 15]. Five years later, a seminal study by Allen et al. [16], pointed out that this risk was NAFLD-related and independent of obesity. This is noteworthy given that obesity had historically been deemed responsible for increased mortality rates owing to a gamut of cancers of potentially metabolic origin, notably including cancers of the colon and rectum, esophagus, liver, gallbladder, pancreas, kidney; non-Hodgkin’s lymphoma and multiple myeloma (in either sex); stomach and prostate (in men); genital tract (in women) [17].

    Over time, a consistent body of accumulating epidemiological studies have fully supported the notion that, collectively, the “metabolic fatty liver syndromes (MFLS)”, namely both NAFLD and MAFLD, are indeed associated with a variety of extra-hepatic cancers. For example, in 2021, Mantovani et al. [18] conducted a meta-analytical review of 10 published observational studies globally comprising 182,202 adults, approximately one quarter of whom had NAFLD, diagnosed with either imaging techniques or International Classification of Diseases codes. During a 5.8-year median follow-up, 8,485 incident extrahepatic cancers were registered, conferring to NAFLD an excess risk ranging from 1.5-fold to 2-fold of developing gastro-esophageal, pancreatic, and colorectal cancers and a slightly lower excess risk (ranging from 1.2-fold to 1.5-fold) of incident cancers of lung, breast, female genital tract and the urinary system. Interestingly, all the estimates of risks were independent of potential confounding factors (such as age, sex, smoking, obesity, and diabetes). Among the strengths of this study, the overall heterogeneity of pooled analyses was low. Moreover, findings were unaffected by sensitivity analyses and no significant publication bias was revealed by funnel plot analysis. Among the weaknesses of the published studies included in this metanalysis, there was no biopsy-proven diagnosis of NAFLD in any of them. A subsequent meta-analytic study published by Thomas et al. [19] by comparing 64 studies for analysis of incident HCC (625,984 patients) and extrahepatic cancer (41,027), found that extra-hepatic cancers were over 8-fold more common than HCC in NAFLD and were not associated with the stage of advanced hepatic fibrosis or cirrhosis. This is in striking contrast with HCC where the most advanced stages of liver fibrosis are associated with an increasing risk of disease [20, 21]. Finally, an umbrella meta-analysis by Yi et al. [22] based on the scrutiny of 39 published meta-analyses results found that individuals with NAFLD exhibited an increased risk of the following extra-hepatic cancers: thyroid, extra-hepatic and intra-hepatic cholangiocarcinoma, pancreatic, esophago-gastric, urinary tract, breast, and lung. The entity of this risk, however, ranged, based on various cancer types, from hazard ratio (HR) 1.25 for lung cancer to HR 2.63 for thyroid cancer. No significant association was found linking NAFLD with the following cancer types: blood, female genital tract, and prostate. Compared to the well-consolidated body of published studies demonstrating the association of NAFLD with extra-hepatic cancer, the association of MAFLD with extra-hepatic cancer appears to be at its dawn. However, as awaited, some recent studies have documented an increased risk of colon cancer [23] and excess mortality of MAFLD owing to malignancy [24].

    While it is readily intuitive to associate those profound cellular, vascular, histological, and immunological perturbations as well as the metabolic remodeling that collectively occur in NAFLD, and particularly in fibrosing NASH and NASH-cirrhosis, with the development of NASH-HCC also in non-cirrhotic livers [2527] how can NAFLD be postulated to mechanistically predispose to incident extra-hepatic cancers? In this regard, it is important to remember that NAFLD is a systemic disorder [28] and even more so is, by definition, MAFLD [24]. On these grounds, one can hypothesize that either the MFLS and extra-hepatic cancer share a common ancestor, for example, a pro-inflammatory systemic milieu associated with metabolic derangements; or that the MFLS are directly involved in the various phases of initiation, development, and progression of cancer at various anatomic sites. The anatomical location of such cancer types, the gastrointestinal tract being more exposed than the genito-urinary tract, may probably offer a clue to understanding these poorly defined pathomechanisms. Excellent reviews have specifically been devoted to exploring this fascinating topic [29, 30]. Additionally, it is conceivable that NAFLD heterogeneity accounts for seemingly conflicting findings published by different investigators. For example, “lean NAFLD” compared to “obese NAFLD” and MAFLD probably exhibits different patho-mechanisms given that expanded adipose tissue per se acts as an endocrine organ favoring the development and progression of hormone-sensitive cancers. Other major modifiers of extra-hepatic cancer risk likely include the stage of hepatic fibrosis [31] and the estimated cardiovascular risk [32]. The Figure 1 illustrates the best-characterized modulators of the risk of extra-hepatic cancers in those with the MFLS.

    Principal modifiers and putative pathomechanisms involved in the association of NAFLD and MAFLD with extra-hepatic cancers. This illustration, based on published studies [18, 19, 2224, 2932], highlights the knowns of this topic. However, our understanding of this complex scenario is far from complete

    Supplementary disease cofactors [33] and particularly lifestyle habits may also participate in fine-tuning the odds of extra-hepatic cancers among those with MFLS. These risk modifiers include epigenetics, alcohol consumption, eating habits (scarcity of fiber and an excess of saturated fats), and sedentary behavior which closely interact with viral infections, drugs, and immunological factors in predisposing to/protecting from extra-hepatic cancers.

    A detailed analysis of molecular mechanisms that can lead to extra-hepatic cancers is out of the scope of this editorial and has been covered elsewhere [29, 30]. In short, while the chief mechanisms may vary according to the variable extra-hepatic cancer site, they can generally be categorized into four effects. A) Insulin resistance that will induce proliferative and anti-apoptotic effects via increased insulin growth factor-1 factor axis; B) dysfunctional adipose tissue which, via nuclear factor kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α), mitogen-activated protein kinase (MAPK) decreased adiponectin, increased leptin and resistin, carries out anti-apoptotic effects, enhanced proliferation and angiogenesis, invasiveness, and increased motility; C) inflammation, which facilitates cell proliferation, neo-angiogenesis, de-differentiation, and metastasis development via interleukin-6 (IL-6)/Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) and IL-6/MAPK TNF-α/Wnt/β-catenin; and D) gut dysbiosis which further triggers inflammation and exerts anti-apoptotic effects via microorganism-associated molecular patterns (MAMPs)/ toll-like receptors (TLRs) inflammasome-derived IL-18 [34, 35].

    However, given that the picture is incomplete, additional prospective studies will have to better define the pathomechanics of this scenario. Such as extensively discussed elsewhere [3638], an improved understanding of this will lead to personalized medicine approaches in prevention, diagnosis, management, prognostication, and individualized follow-up protocols.

    Abbreviations

    HCC:

    hepatocellular carcinoma

    IL-6:

    interleukin-6

    MAFLD:

    metabolic dysfunction-associated fatty liver disease

    MFLS:

    metabolic fatty liver syndromes

    NAFLD:

    nonalcoholic fatty liver disease

    NASH:

    nonalcoholic steatohepatitis

    PLC:

    primary liver cancer

    T2D:

    type 2 diabetes

    Declarations

    Author contributions

    The author contributed solely to the work.

    Conflicts of interest

    The author declares that there are no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Gille C, Bölling C, Hoppe A, Bulik S, Hoffmann S, Hübner K, et al. HepatoNet1: a comprehensive metabolic reconstruction of the human hepatocyte for the analysis of liver physiology. Mol Syst Biol. 2010;6:411. [DOI] [PubMed] [PMC]
    Lonardo A, Ballestri S, Marchesini G, Angulo P, Loria P. Nonalcoholic fatty liver disease: a precursor of the metabolic syndrome. Dig Liver Dis. 2015;47:18190. [DOI] [PubMed]
    Lonardo A, Mantovani A, Targher G, Baffy G. Nonalcoholic fatty liver disease and chronic kidney disease: epidemiology, pathogenesis, and clinical and research implications. Int J Mol Sci. 2022;23:13320. [DOI] [PubMed] [PMC]
    Lonardo A, Leoni S, Alswat KA, Fouad Y. History of nonalcoholic fatty liver disease. Int J Mol Sci. 2020;21:5888. [DOI] [PubMed] [PMC]
    Méndez-Sánchez N, Bugianesi E, Gish RG, Lammert F, Tilg H, Nguyen MH, et al.; Global multi-stakeholder consensus on the redefinition of fatty liver disease. Global multi-stakeholder endorsement of the MAFLD definition. Lancet Gastroenterol Hepatol. 2022;7:38890. [DOI] [PubMed]
    Polyzos SA, Kang ES, Tsochatzis EA, Kechagias S, Ekstedt M, Xanthakos S, et al. Commentary: nonalcoholic or metabolic dysfunction-associated fatty liver disease? The epidemic of the 21st century in search of the most appropriate name. Metabolism. 2020;113:154413. [DOI] [PubMed]
    Chan KE, Ng CH, Fu CE, Quek J, Kong G, Goh YJ, et al. The spectrum and impact of metabolic dysfunction in MAFLD: a longitudinal cohort analysis of 32,683 overweight and obese individuals. Clin Gastroenterol Hepatol. 2022;[Epub ahead of print]. [DOI] [PubMed]
    Caldwell SH, Oelsner DH, Iezzoni JC, Hespenheide EE, Battle EH, Driscoll CJ. Cryptogenic cirrhosis: clinical characterization and risk factors for underlying disease. Hepatology. 1999;29:6649. [DOI] [PubMed]
    Zen Y, Katayanagi K, Tsuneyama K, Harada K, Araki I, Nakanuma Y. Hepatocellular carcinoma arising in non-alcoholic steatohepatitis. Pathol Int. 2001;51:12731. [DOI] [PubMed]
    Bugianesi E, Leone N, Vanni E, Marchesini G, Brunello F, Carucci P, et al. Expanding the natural history of nonalcoholic steatohepatitis: from cryptogenic cirrhosis to hepatocellular carcinoma. Gastroenterology. 2002;123:13440. [DOI] [PubMed]
    Marrero JA, Fontana RJ, Su GL, Conjeevaram HS, Emick DM, Lok AS. NAFLD may be a common underlying liver disease in patients with hepatocellular carcinoma in the United States. Hepatology. 2002;36:134954. [DOI] [PubMed]
    Michelini E, Lonardo A, Ballestri S, Costantini M, Caporali C, Bonati ME, et al. Is cholangiocarcinoma another complication of insulin resistance: a report of three cases. Metab Syndr Relat Disord. 2007;5:194202. [DOI] [PubMed]
    Welzel TM, Graubard BI, El-Serag HB, Shaib YH, Hsing AW, Davila JA, et al. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma in the United States: a population-based case-control study. Clin Gastroenterol Hepatol. 2007;5:12218. [DOI] [PubMed] [PMC]
    Shen H, Lipka S, Kumar A, Mustacchia P. Association between nonalcoholic fatty liver disease and colorectal adenoma: a systemic review and meta-analysis. J Gastrointest Oncol. 2014;5:4406. [DOI] [PubMed] [PMC]
    Armstrong MJ, Adams LA, Canbay A, Syn WK. Extrahepatic complications of nonalcoholic fatty liver disease. Hepatology. 2014;59:117497. [DOI] [PubMed]
    Allen AM, Hicks SB, Mara KC, Larson JJ, Therneau TM. The risk of incident extrahepatic cancers is higher in non-alcoholic fatty liver disease than obesity - a longitudinal cohort study. J Hepatol. 2019;71:122936. [DOI] [PubMed] [PMC]
    Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348:162538. [DOI] [PubMed]
    Mantovani A, Petracca G, Beatrice G, Csermely A, Tilg H, Byrne CD, et al. Non-alcoholic fatty liver disease and increased risk of incident extrahepatic cancers: a meta-analysis of observational cohort studies. Gut. 2022;71:77888. [DOI] [PubMed]
    Thomas JA, Kendall BJ, Dalais C, Macdonald GA, Thrift AP. Hepatocellular and extrahepatic cancers in non-alcoholic fatty liver disease: a systematic review and meta-analysis. Eur J Cancer. 2022;173:25062. [DOI] [PubMed]
    Zhang DY, Friedman SL. Fibrosis-dependent mechanisms of hepatocarcinogenesis. Hepatology. 2012;56:76975. [DOI] [PubMed] [PMC]
    Ahmad MI, Khan MU, Kodali S, Shetty A, Bell SM, Victor D. Hepatocellular carcinoma due to nonalcoholic fatty liver disease: current concepts and future challenges. J Hepatocell Carcinoma. 2022;9:47796. [DOI] [PubMed] [PMC]
    Yi M, Peng W, Feng X, Teng F, Tang Y, Kong Q, et al. Extrahepatic morbidities and mortality of NAFLD: an umbrella review of meta-analyses. Aliment Pharmacol Ther. 2022;56:111930. [DOI] [PubMed]
    Lee H, Lee HW, Kim SU, Chang Kim H. Metabolic dysfunction-associated fatty liver disease increases colon cancer risk: a nationwide cohort study. Clin Transl Gastroenterol. 2022;13:e00435. [DOI] [PubMed] [PMC]
    Quek J, Ng CH, Tang ASP, Chew N, Chan M, Khoo CM, et al. Metabolic associated fatty liver disease increases the risk of systemic complications and mortality. A meta-analysis and systematic review of 12 620 736 individuals. Endocr Pract. 2022;28:66772. [DOI] [PubMed]
    Zoller H, Tilg H. Nonalcoholic fatty liver disease and hepatocellular carcinoma. Metabolism. 2016;65:115160. [DOI] [PubMed]
    Rui L, Lin JD. Reprogramming of hepatic metabolism and microenvironment in nonalcoholic steatohepatitis. Annu Rev Nutr. 2022;42:91113. [DOI] [PubMed]
    Peiseler M, Schwabe R, Hampe J, Kubes P, Heikenwälder M, Tacke F. Immune mechanisms linking metabolic injury to inflammation and fibrosis in fatty liver disease - novel insights into cellular communication circuits. J Hepatol. 2022;77:113660. [DOI] [PubMed]
    Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol. 2015;62:S4764. [DOI] [PubMed]
    Dang CV. Links between metabolism and cancer. Genes Dev. 2012;26:87790. [DOI] [PubMed] [PMC]
    Lonardo F, Ballouk C. Metabolism and cancer-select topics. Metab Target Organ Damage. 2022;2:8. [DOI]
    Kim MC, Park JG, Jang BI, Lee HJ, Lee WK. Liver fibrosis is associated with risk for colorectal adenoma in patients with nonalcoholic fatty liver disease. Medicine (Baltimore). 2019;98:e14139. [DOI] [PubMed] [PMC]
    Chen X, Chen Z, Jiang L, Huang J, Zhu Y, Lin S. MAFLD is associated with increased all-cause mortality in low cardiovascular-risk individuals but not in intermediate to high-risk individuals. Nutr Metab Cardiovasc Dis. 2022. [DOI] [PubMed]
    Lonardo A, Singal AK, Osna N, Kharbanda KK. Effect of cofactors on NAFLD/NASH and MAFLD. A paradigm illustrating the pathomechanics of organ dysfunction. Metab Target Organ Damage. 2022;2:12. [DOI] [PubMed] [PMC]
    Sanna C, Rosso C, Marietti M, Bugianesi E. Non-alcoholic fatty liver disease and extra-hepatic cancers. Int J Mol Sci. 2016;17:717. [DOI] [PubMed] [PMC]
    Veracruz N, Hameed B, Saab S, Wong RJ. The association between nonalcoholic fatty liver disease and risk of cardiovascular disease, stroke, and extrahepatic cancers. J Clin Exp Hepatol. 2021;11:4581. [DOI] [PubMed] [PMC]
    Lonardo A, Byrne CD, Targher G. Precision medicine approaches in metabolic disorders and target organ damage: where are we now, and where are we going? Metab Target Organ Damage. 2021;1:3. [DOI]
    Lonardo A, Arab JP, Arrese M. Perspectives on precision medicine approaches to NAFLD diagnosis and management. Adv Ther. 2021;38:213058. [DOI] [PubMed] [PMC]
    Lonardo A. Precision medicine in nonalcoholic fatty liver disease. J Gastroenterol Hepatol. 2022;37:11758. [DOI] [PubMed]
    • Citation
      Cite this Article