Download PDF
Review  |  Open Access  |  28 Sep 2024

MASLD: an emerging factor in the pathophysiology and clinical management of ICC

Views: 56 |  Downloads: 26 |  Cited:  0
Hepatoma Res 2024;10:41.
10.20517/2394-5079.2024.63 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

This article explores the emerging role of metabolic dysfunction-associated steatotic liver disease (MASLD) in the pathophysiology of intrahepatic cholangiocarcinoma (ICC). Despite advances in public health, ICC incidence has risen, suggesting overlooked risk factors. Recent studies indicate a significant association between MASLD and ICC, though causality remains unproven. The article reviews current research, highlighting limitations such as retrospective designs and small sample sizes. It emphasizes the need for comprehensive investigations into MASLD’s role in ICC pathogenesis, prognosis, and management. Future research should focus on integrating advanced methodologies to identify novel biomarkers and therapeutic strategies, aiming to improve patient outcomes and develop tailored prevention and treatment approaches.

Keywords

Metabolic dysfunction-associated steatotic liver disease, intrahepatic cholangiocarcinoma, risk factors, pathogenesis, progression, prognosis, treatment

INTRODUCTION

Previous studies have identified chronic inflammation caused by viruses, parasites, biliary obstruction, or stones as the most significant risk factor for intrahepatic cholangiocarcinoma (ICC). Despite advances in public health, living standards, medical technology, and vaccine distribution, the incidence of ICC has not only stabilized but has increased sharply by 140% over the past 40 years. This trend suggests that current research does not entirely elucidate the mechanisms underlying ICC and that some potential risk factors may be overlooked.

Recent findings indicate that diabetes and obesity significantly elevate the risk of ICC, illustrating the critical role of metabolic diseases in its pathophysiology. Metabolic dysfunction-associated steatotic liver disease (MASLD), which is associated not only with hepatitis and liver fibrosis but also with various metabolic disorders, has seen a dramatic rise in prevalence, affecting over 30% of the global population. This suggests that MASLD may be an emerging factor in the pathophysiology of ICC.

There is currently a significant gap in research exploring the association between MASLD and ICC. Given MASLD’s high prevalence and severity, overlooking its role in the pathogenesis of ICC could hinder early intervention efforts and worsen both ICC incidence and the global burden of liver disease. Therefore, thorough investigations into the relationship between MASLD and ICC are essential.

This study aims to provide a concise review of current research on MASLD and ICC while laying the groundwork for further exploration in this important area.

MASLD IS A LATENT RISK FACTOR

Research overview

The relevant research began in 2007. Welzel et al. identified an increasing incidence of ICC in the United States, contrasting with stable rates of extrahepatic cholangiocarcinoma (eCCA)[1]. This observation led to the hypothesis that unknown risk factors, potentially including MASLD, might contribute to ICC pathogenesis. Their subsequent investigations utilizing the SEER-Medicare database indicated a significant association, with an odds ratio (OR) of 3.0 (95%CI: 1.2-7.3). However, it is crucial to note that these findings, while suggestive, do not establish causation.

In 2016, Kinoshita et al. further explored this potential link, noting a higher prevalence of metabolic dysfunction-associated steatohepatitis (MASH) in ICC patients without known risk factors compared to those with liver metastatic carcinoma[2]. They formally proposed MASLD as a risk factor associated with ICC.

Meta-analyses and cohort studies, such as those by Wongjarupong et al. in 2017, have reinforced the hypothesis of an increased risk, reporting an OR of 2.22 (95%CI: 1.52-3.24) for cholangiocarcinoma (CCA)[3]. Similarly, Petrick et al.’s randomized controlled study suggested a more than threefold increase in risk (OR = 3.52, 95%CI: 2.87-4.32) for ICC[4]. Despite these findings, the absence of prospective data means that the role of MASLD as a risk factor remains speculative.

In addition to these studies, numerous scholars have investigated the relationship between MASLD and ICC in depth [Table 1]. In conclusion, while the current evidence indicates a potential link between MASLD and ICC, it is premature to draw definitive conclusions about causality.

Table 1

Research on MASLD and the risk of ICC

AuthorsYear of publicationCountryRelated conclusions
Zhou et al.[5]2009ChinaMASLD may be a risk factor for ICC
Reddy et al.[6]2013USAMASH may affect up to 20% of patients with resectable ICC
Nkontchou et al.[7]2013FrenchThe incidence of MASH was higher (19%) in ICC patients with no other known risk factors
Chang et al.[8]2013Taiwan, ChinaMASLD increases the risk of CCA
Choi et al.[9]2016USAMASLD is not a risk factor for CCA (OR = 1.40, 95%CI: 0.94-2.09)
Xiong et al.[10]2018ChinaMetabolic syndrome was significantly associated with a 1.86-fold increased risk of CCA
Sirica et al.[11]2019USAMASLD and MASH are risk factors for HCC and ICC
De Lorenzo et al.[12]2020ItalyMASH is a risk factor for ICC, MASLD is not
Corrao et al.[13]2021ItalyMASLD is significantly correlated with the progress of ICC, not eCCA
Ghidini et al.[14]2021ItalyMASLD and MASH are associated with a high risk of CCA, especially ICC

Critical re-examination

Current research on the association between MASLD and the risk of ICC is constrained by several limitations, including retrospective designs, small sample sizes, and single-center cohort studies. These limitations contribute to weak evidence, potential biases, and significant heterogeneity in research outcomes, making it challenging to draw consistent conclusions. Several specific issues warrant further attention:

(1) Temporal Relevance: The incidence of MASLD has increased dramatically over the past decade[15], yet many studies rely on data from the late 20th century. This temporal disconnect, coupled with inadequate control of confounding factors, compromises the accuracy of research findings. Future studies should utilize up-to-date, comprehensive datasets and account for time effects and potential confounders.

(2) Geographic and Ethnic Considerations: The influence of regional and ethnic factors cannot be overlooked. While MASLD was traditionally considered a Western disease[16], its prevalence in Asia, the Middle East, and North Africa has surged over the past three decades, with related mortality rates surpassing those in Europe and the United States[17]. The predominance of studies based on European and American populations may introduce bias due to genetic, environmental, and lifestyle differences across regions and ethnicities. Moreover, the current data inadequately represent the baseline characteristics and disease patterns of Asian populations, potentially leading to misinterpretation of the MASLD-ICC association in these groups.

(3) Metabolic Syndrome Features: A critical oversight in current research is the insufficient focus on the individual components of metabolic syndrome, namely obesity and diabetes, as these factors may play pivotal roles in the observed association between MASLD and ICC. Obesity, a proinflammatory state, can exacerbate liver pathology, possibly accelerating the progression from MASLD to ICC. Similarly, diabetes, through its systemic metabolic effects, might influence carcinogenic pathways, thereby increasing ICC risk. Future studies must employ a multifaceted approach to address this issue: (a) Comprehensive patient profiling: Researchers should conduct detailed metabolic assessments, including body mass index (BMI), waist circumference, lipid profiles, fasting glucose levels, and insulin resistance markers; (b) Stratified analysis: Studies should stratify patients based on the presence and severity of individual metabolic syndrome components to isolate their effects on ICC risk; (c) Longitudinal designs: Long-term follow-up studies are needed to track the progression of metabolic syndrome features and their temporal relationship with ICC development; (d) Molecular investigations: Research should explore potential molecular mechanisms linking specific metabolic abnormalities to cholangiocarcinogenesis, such as examining the role of adipokines, proinflammatory cytokines, and insulin signaling pathways; (e) Dose-response relationships: Investigations into whether the severity or duration of metabolic syndrome features correlates with ICC risk could provide valuable insights.

By meticulously accounting for these factors, future research can more accurately evaluate the independent contribution of MASLD to ICC risk, potentially uncovering complex interactions between metabolic syndrome components and liver pathology in cholangiocarcinogenesis.

PATHOGENESIS OF MASLD-RELATED ICC

Research overview

Research on the pathogenesis of MASLD and ICC remains limited. To date, only two articles address this topic [Figure 1]. The first article[18] focuses on the genetic factors, offering an in-depth examination of the role of MASLD in the initiation and proliferation of ICC. The second article[19] analyzes the impacts of metabolic syndrome (MetS) and fibrosis on inflammation and the mechanism that promotes cholangiocyte cancer, framed within the context of systemic metabolism. Together, these studies not only provide valuable insights for future research but also underscore the pressing need for comprehensive investigations and expansions within this domain.

MASLD: an emerging factor in the pathophysiology and clinical management of ICC

Figure 1. Potential pathogenesis of MASLD-related ICC. MASH: Metabolic dysfunction-associated steatohepatitis; MetS: metabolic syndrome; HC: hepatic cells; YAP 1: yes-associated protein 1; SOX 9: SRY-box transcription factor 9; OPN: osteopontin; TnC: tenascin-C; POSTN: periostin; ICC: intrahepatic cholangiocarcinoma.

High expression of YAP1 and SOX9 promotes ICC progression

In 2020, Hu et al. reported that over 90% of ICC patients exhibited alterations in YAP1 and/or SOX9[18]. Utilizing dual tissue microarray chips and a preclinical ICC mouse model, their findings included: (a) Nuclear levels of SOX9 and YAP1 were elevated in over 90% of human CCA samples (n = 108). Additionally, the expression of p-AKT, SOX9, and YAP1 was significantly upregulated in the hepatic cells of mice with MASH or primary sclerosing cholangitis (PSC); (b) The deletion of either YAP1 or SOX9 alone led to ICC pathogenesis, which was characterized by high expression levels of the remaining gene. In contrast, concurrent deletion of both genes completely inhibited ICC pathogenesis; (c) YAP1 deficiency impeded the reprogramming of hepatic cells to biliary epithelial cells (BECs) and tumor cell proliferation. In contrast, SOX9 deficiency solely inhibited tumor cell proliferation. Based on these observations, the authors propose a model of MASLD-related ICC pathogenesis. This model suggests that steatohepatitis in MASLD patients promotes elevated expression of YAP1 and SOX9 in liver cells, facilitating the transdifferentiation of hepatic cells to BECs and subsequent tumor cell proliferation.

The biological heterogeneity of ICC significantly impacts the efficacy of surgical and chemotherapeutic interventions. This is primarily due to the diverse origins of ICC cells[20]. This study not only underscores the complex interplay between MASLD, HCC, and ICC but also advocates for a comprehensive approach to hepatobiliary diseases, avoiding isolated disease analyses. Furthermore, the study highlights common features of ICC heterogeneity, offering novel insights for the development of broad-spectrum therapeutic strategies for advanced ICC and improving prognosis.

Osteopontin overexpression is a prominent feature of ICC patients with MetS

In 2023, Cadamuro et al. highlighted recent epidemiological evidence suggesting that MetS is frequently associated with MASLD or MASH, positioning it as a potential risk factor for ICC[3,14,19]. Concurrently, studies have demonstrated that ICC typically involves the deposition and remodeling of a dense extracellular matrix (ECM)[21-23], with ECM remodeling identified as a critical mechanism underlying cardiovascular mortality in patients with MetS and MASLD[19]. Thus, the authors hypothesize that ECM remodeling driven by MetS is a key pathological mechanism in MASLD that may precipitate ICC.

In their study, Cadamuro et al. analyzed 44 ICC patients with MetS and 22 without, focusing on the levels of osteopontin (OPN), tenascin-C (TnC), and periostin (POSTN) within the ECM to assess changes in its composition and impact on ICC cell biology[19].

Their findings revealed that: (a) Enhanced deposition of OPN, TnC, and POSTN was observed in the tumors of the 44 ICC patients with MetS compared to adjacent non-tumorous tissue; (b) Significantly greater OPN accumulation was observed in MetS-afflicted ICC patients relative to those without MetS; (c) OPN, TnC, and POSTN markedly promoted cell viability and a cancer stem cell-like phenotype in human ICC cell lines; (d) Differences in the distribution and composition of fibrosis between MetS and non-MetS ICC patients are likely due to OPN-mediated recruitment of cancer-associated fibroblasts.

The study concluded that de novo expression of OPN, TnC, and POSTN was significantly associated with the promotion of motility of malignant cholangiocytes. At the same time, fibrosis can promote tumor progression. The overexpression of OPN is a significant feature of ICC with MetS and a key effector of fibrosis.

This investigation provides a foundational analysis of the pathophysiological links between MetS, ECM remodeling, and ICC, offering crucial insights for monitoring MetS and MASLD patients’ risk of progressing to ICC. This work also establishes a theoretical basis for early prevention, precise diagnosis, and the development of novel ICC treatment strategies. However, further investigation is required to elucidate the specific roles and mechanisms of various OPN isoforms (including OPNa, OPNb, OPNc, OPN4 and OPN5) in ICC progression, suggesting substantial opportunities for future research in this domain.

Future perspective

MASLD-related ICC represents a complex disease trajectory originating from metabolic disorders and culminating in malignancy. Current research into its pathogenesis remains sparse, and existing studies fall short of providing a comprehensive understanding of the disease’s overall mechanistic blueprint. However, revisiting the literature from the perspectives of origin and outcome offers valuable insights.

Origin perspective

From the origin standpoint, existing studies have delved into the mechanisms by which metabolic disorders such as diabetes and obesity contribute to CCA. These studies highlight key features such as hepatic steatosis, insulin resistance, and the subsequent upregulation of genes promoting cell turnover in the pathogenesis of CCA [Table 2]. Given that MASLD shares commonalities with these metabolic disorders, it is anticipated that MASLD-related ICC will exhibit similar characteristics. Future research can build upon these findings by investigating:

Table 2

Metabolic factors affect the pathophysiology of ICC

Metabolic factorProposed mechanismReference
LeptinOverexpression enhances cholangiocarcinoma cell growth[24]
Adipose tissue inflammationChronic hepatic inflammation and fibrosis due to IL-6 and TNFα release[25]
Insulin resistanceCompensatory hyperinsulinemia and increased IGF-1 production, promoting cell proliferation and survival[4,26-28]
HyperinsulinemiaDirect stimulation of cholestatic cell proliferation; increased IGF-1 and insulin receptor expression in tumor cells[29-33]
HyperglycemiaSTAT3 activation leading to aggressive CCA phenotypes; NF-κB glycosylation and nuclear localization promoting aggressive phenotype[31,34,35]
Low APN levelsReduced AdipoR expression leading to APN insensitivity; impaired activation of anti-neoplastic pathways (MAPK, mTOR, STAT-3, etc.)[36-40]

(1) The specific molecular pathways linking MASLD to hepatic steatosis, insulin resistance, and altered gene expression in cholangiocytes.

(2) The potential synergistic effects of various metabolic derangements commonly observed in MASLD (e.g., dyslipidemia, hyperglycemia, and altered adipokine profiles) on ICC development.

(3) The role of genetic predisposition and epigenetic modifications in modulating the susceptibility of individuals with MASLD to ICC.

Outcome perspective

From the outcome perspective, the pathogenesis of ICC, including MASLD-related ICC, involves intricate physiological systems and metabolic processes. The convergence of pathological progression in ICC, regardless of the initial causative factors, suggests shared mechanisms that warrant further exploration. In general, the investigation of ICC pathogenesis can be divided into three main categories: (a) Studies of metabolic-related pathways: for instance, the association between the overexpression of ErbB family receptor tyrosine kinases in the bile of ICC patients and the origin of ICC[41]; (b) Investigations at the cellular and molecular levels, including mutations in PTEN, TP53, KRAS, CXCL12, and other genes in ICC cell lines and tissue[42-44], as well as the promoting effects of multiple uncontrolled miRNA expressions on the progression of ICC[45,46]; (c) Analysis of related signaling pathways: the potential roles of C-JNK, Wnt/β-catenin, and JAK-STAT signaling pathways in the proliferation and metastasis of ICC. Building on these established research foundations, we can delve deeper into the key genes and signaling pathways upstream or downstream of MASLD-related ICC to uncover its unique pathological processes and provide new insights for developing treatment strategies.

Inflammation as a central element

Inflammation is a critical factor linking MASLD to ICC, given its close association with hepatic steatosis. Extensive research has demonstrated the role of inflammation in ICC pathogenesis [Figure 2][47,48], with inflammatory mediators like IL-6 and TNFα activating pathways such as JAK-STAT, p38 MAPK, and Akt, which enhance cell growth, survival, and proliferation. The upregulation of COX-2 and Mcl-1 due to inflammation confers resistance to apoptosis, further promoting oncogenesis. Understanding the pivotal role of inflammation in the pathophysiological process of MASLD-related ICC is essential for unraveling its pathogenesis. Future studies should focus on delineating the inflammatory pathways and mediators that serve as critical links between MASLD and ICC, providing a foundation for targeted therapeutic strategies.

MASLD: an emerging factor in the pathophysiology and clinical management of ICC

Figure 2. The molecular pathogenesis of ICC induced by inflammation. IL-6: Interleukin-6; TNF-α: tumor necrosis factor alpha; Wnt: wingless/integrated; EGFR: epidermal growth factor receptor; ErbB2: Erb-B2 receptor tyrosine kinase 2; IL-6R: interleukin-6 receptor; TNFR: tumor necrosis factor receptor; LPR: low-density lipoprotein receptor-related protein; FZD: frizzled; iNOS: inducible nitric oxide synthase; COX2: cyclooxygenase-2; miR: microRNA; IncRNA: long non-coding RNA; NO: nitric oxide; Notch: notch signaling pathway; p38: p38 mitogen-activated protein kinase; MAPK: mitogen-activated protein kinase; MAP3K8: mitogen-activated protein kinase kinase kinase 8; DNMT1: DNA methyltransferase 1; JAK-STAT3: janus kinase-signal transducer and activator of transcription 3; p44/42 MAPK: p44/p42 mitogen-activated protein kinase (also known as ERK1/ERK2); P21: cyclin-dependent kinase inhibitor 1; SOCS3: suppressor of cytokine signaling 3; Mid-1: midline 1; P16: cyclin-dependent kinase inhibitor 2A; RASSF1A: RAS-association domain family 1, isoform A; NF2: neurofibromatosis type 2; AJID: atypical junctional intercellular domain; TCF: T-cell factor; CEF: cell-free extract.

Additional insights and revelations

Recent studies have indicated that certain ICCs share a common progenitor cell origin as HCC[49,50]. This finding underscores the importance of adopting a holistic approach to hepatobiliary diseases and highlights the necessity of a unified metabolic interpretation. It also reveals a significant correlation between MASLD-related ICC and the broader spectrum of existing MASLD diseases. As research progresses, the MASLD disease spectrum may be refined, with eventual outcomes potentially extending beyond HCC to include ICC.

Furthermore, it is important that MASLD without cirrhosis has been associated with HCC or ICC[51,52]. This association suggests that certain elements of the MASLD disease progression may not always manifest as traditionally expected. Therefore, in investigating the pathogenesis of MASLD-related ICC, it is crucial to elucidate this phenomenon and transcend the limitations of conventional progression models in order to expand the scope of research perspectives.

PROGNOSIS OF MASLD-RELATED ICC

Research overview

The prognosis of patients is a critical factor in predicting the course of progression and provides an essential basis for treatment decisions by healthcare providers. Recent studies on the prognosis of MASLD-related ICC have primarily focused on differences between ICC patients with and without MASLD, yielding varied conclusions that warrant further investigation.

Reddy et al. reported no significant differences in tumor-free survival and overall survival between ICC patients with or without MASLD[6]. Conversely, De Lorenzo et al. found that the median survival of patients with MASLD (38.5 months) was significantly shorter than that of patients without traditional risk factors including HBV, HCV, alcohol, and environmental toxins (48.1 months, P = 0.003), and was comparable to that of patients with traditional risk factors (31.9 months, P = 0.948)[12]. Yu et al. observed that, in comparison to HBV-associated ICC, MASLD-related ICC patients exhibited a lower 5-year overall survival rate (24.0% vs. 48.9%), a higher 5-year recurrence rate (80.9% vs. 55.0%), and a higher early recurrence rate (58.5% vs. 30.0%) (all P < 0.01)[53].

In conclusion, the results of current cohort studies on the prognosis of MASLD-related ICC patients are inconclusive. Meta-analyses are challenging due to the limited number of studies and diverse research objectives. Further investigation is needed to elucidate the role of MASLD in the onset and progression of ICC and its impact on quality of life. A clear understanding of the prognosis of MASLD-related ICC patients can facilitate more accurate disease progression assessment and subsequent treatment adjustments, such as the introduction of new adjuvant therapies or postoperative radio-chemotherapy, with the aim of enhancing patient prognosis.

Critical re-examination

The prognosis of MASLD-related ICC patients is closely related to the treatment. Currently, the treatment strategy is mostly referred to the general ICC patients, but the surgical procedure has been updated in recent years. New adjuvant chemotherapies have been recommended as first-line treatments for locally advanced ICC[54-56]. Additionally, the role of lymph node dissection in surgery has gained focus[57,58], with the latest treatment guidelines advocating for its widespread application during surgical intervention[59].

However, due to the extensive time span of samples included in relevant studies, the underutilization of novel adjuvant chemotherapies and lymph node dissection in earlier cases may lead to a pessimistic prognosis assessment for MASLD-related ICC. Therefore, further studies are necessary to determine the prognosis and optimal treatment strategies for MASLD-related ICC patients.

CLINICAL MANAGEMENT

Early intervention

Malignant tumors, both intrahepatic and extrahepatic, rank as the second most common cause of death among patients with MASLD, following cardiovascular events. Large cohort studies[60] have identified a significant link between MASLD and an elevated cancer risk. Further investigations[61] have revealed that all histological stages of MASLD are significantly associated with increased overall mortality. This risk intensifies with the histological progression of MAFLD (HR = 1.71, 95%CI: 1.64-1.79 for simple steatosis; HR = 2.14, 95%CI: 1.93-2.38 for MASH; HR = 2.44, 95%CI: 2.22-2.69 for non-cirrhotic fibrosis; HR = 3.79, 95%CI: 3.34-4.30 for cirrhosis). Consequently, the early identification of MAFLD patients at high risk of malignancy and timely intervention to obstruct disease progression is crucial for optimizing the management of MAFLD-related ICC.

Key intervention[62] targets connecting MASLD and ICC include metabolic function, inflammation, insulin resistance, and liver fibrosis, making them essential focal points for early intervention strategies. Current research primarily focuses on evaluating the effects of pharmacological interventions on these aspects of MASLD. Two main approaches have emerged: one investigates the repurposing of existing drugs for other metabolic disorders (such as hyperlipidemia and diabetes) for use in MASLD[62,63] [Table 3]; the other concentrates on the development of new drugs specifically targeting MASLD. Recently, phase 3 clinical trial data for the new drug Rezdiffra demonstrated a significant improvement in liver fibrosis among MASH patients with moderate to severe non-cirrhotic fibrosis[64]. As a result, Rezdiffra was approved, marking a milestone as the first drug sanctioned for MASH treatment, effectively addressing a long-standing therapeutic gap.

Table 3

Effects of drugs on hepatic steatosis, inflammation and fibrosis, and cardiovascular events

DrugLiverCardiovascular system
Vitamin EImproves non-T2DM fatty liver and hepatocyte ballooning/inflammation, does not improve fibrosisIncreases the risk of heart failure in T2DM patients
Obeticholic acidImproves liver fibrosis, prevents the progression of NASHNo effect
StatinsPrevent the progression of MASLD, reduce the risk of HCCReduce cardiovascular risk
MetforminPrevents the progression of MASLD, reduce the risk of HCCNo effect
PioglitazoneReduces liver fat content in DM patients, alleviates NASHReduces the risk of cardiovascular events in T2DM and NASH patients
GLP-1RAThe effect on improving fatty liver, liver enzyme levels, and liver fibrosis in NASH patients needs further researchReduces the risk of cardiovascular events in T2DM patients
SGLT-2iThe effect on improving fatty liver in T2DM patients, and liver fibrosis in NASH patients needs further researchReduces the risk of cardiovascular and kidney events in T2DM patients
DPP-4iCannot alleviate fatty liver and fibrosis in overweight T2DM patientsReduces the risk of cardiovascular events in T2DM patients

Additionally, some scholars have argued[65] that new drug development should focus on the broader “metabolic dysfunction” rather than solely aiming to ameliorate inflammation and fibrosis pre- and post-liver biopsy. Reducing the risk of malignancy and improving patient quality of life should be key objectives in pharmacotherapy.

Looking ahead, the future of MASLD management is promising, with innovative approaches under exploration. These include targeting the gut-liver axis via microbiome modulation, leveraging incretin-based therapies, and investigating epigenetic modifications in disease progression[66]. Furthermore, precision medicine approaches utilizing genetic and metabolomic profiling may enable more personalized and effective interventions.

Treatment and challenges

Currently, the U.S. National Comprehensive Cancer Network’s 2023 CCA Guidelines[59] do not specify distinct treatment protocols for patients with MASLD-related ICC. Consequently, these patients typically receive standard ICC treatments, which encompass a range of modalities depending on disease stage and patient factors.

For resectable tumors, surgical resection remains the gold standard, offering the best chance for long-term survival. However, only 30%-40% of patients are candidates for this approach at diagnosis[67]. Additionally, the presence of MASLD can complicate surgical outcomes due to impaired liver function and regenerative capacity.

For unresectable cases, systemic therapy is the mainstay of treatment. The current first-line regimen consists of gemcitabine plus cisplatin, which has shown improved overall survival compared to gemcitabine alone[68]. In addition, recent studies have proposed new regimens such as TOPAZ-1[69] and KEYNOTE-966[70], which can benefit patients more than traditional first-line regimens. Recent advancements in targeted therapies, such as FGFR2 inhibitors for FGFR2 fusion-positive ICC, have opened new avenues for personalized treatment[71]. However, the efficacy of these regimens in MASLD-related ICC is not well-documented, and the potential for increased toxicity due to underlying liver dysfunction warrants careful consideration.

Locoregional therapies play a crucial role in managing ICC, particularly for patients with limited hepatic disease[71,72]. These include radiofrequency ablation (RFA), microwave ablation (MWA), and transarterial chemoembolization (TACE). Stereotactic body radiation therapy (SBRT) has also shown promise in local tumor control. However, the radiosensitivity of MASLD-affected liver tissue and the risk of radiation-induced liver disease necessitate precise planning and dose management. In addition, the altered vascular architecture and regenerative limitations in MASLD may impact their effectiveness and safety.

Liver transplantation, while a potential curative option for select patients with MASH-related end-stage liver disease and early-stage ICC[66], poses significant challenges in the context of MASLD. The increased risk of postoperative complications, including the risk of developing recurrent or de novo MASLD, graft dysfunction, and cardiovascular events[73], highlights the need for stringent candidate selection and comprehensive perioperative management.

Despite these options, the unique pathophysiology of MASLD-related ICC presents distinct challenges. The altered hepatic microenvironment, chronic inflammation, and metabolic dysregulation associated with MASLD may influence treatment efficacy and tolerability. This underscores a significant gap in current oncological practice, highlighting the need for tailored approaches that consider the specific biological context of MASLD-related ICC.

Perioperative management

Recent studies[74] have demonstrated that patients with MetS, MASLD, or MASH face significantly increased risks of postoperative complications such as cardiovascular events, infections, and renal insufficiency following liver resection or transplantation. A meta-analysis by Meijer et al. indicated that both MASLD and MASH are independent risk factors for postoperative complications after hepatectomy, with risks escalating alongside disease severity[75]. Neal et al. suggested that only MASH was associated with increased complication risks, but the majority of studies highlight the need for intensive perioperative management due to elevated surgical risks in MASLD patients[76]. Mechanistic studies by Veteläinen et al. and McCuskey et al. have demonstrated that MASH can exacerbate postoperative inflammatory responses, impair hepatocyte function, diminish liver regenerative capacity, and lead to microvascular dysfunction[77,78]. Collectively, these factors reduce surgical tolerance and increase the risk of postoperative mortality in these patients.

Therefore, for patients with MASLD, irrespective of disease stage, enhanced perioperative management is crucial. Poon et al. recommend comprehensive preoperative liver function assessments, preoperative interventions such as portal vein embolization, and meticulous surgical techniques to minimize liver injury[79]. Cauchy et al. and Reddy et al. noted that even patients with latent or mild MASLD are at increased risk of postoperative complications[80,81]. Cauchy et al. further emphasized that the reduced liver regenerative capacity associated with MASH necessitates combining tumor treatment with minimally invasive techniques during surgery[82].

In conclusion, detailed and comprehensive treatment and management strategies should be emphasized for MASLD-related ICC patients to mitigate surgical risks and reduce the incidence of complications regardless of their stage of MASLD. Additionally, specific treatment options for this subgroup have not received adequate attention, underscoring the need for further research to fundamentally improve prognosis for these patients.

DEFINITION UPDATE

MASLD represents a novel conceptualization of non-alcoholic fatty liver disease (NAFLD), establishing more precise diagnostic criteria. It is crucial to recognize that MASLD should not be viewed exclusively as the hepatic manifestation of metabolic syndrome. Instead, it encompasses a broader spectrum of intra- and extrahepatic complications, including insulin resistance and alcoholic liver disease[15]. The recent consensus to further rename NAFLD to MASLD underscores the pivotal role of metabolic cardiovascular risk factors in its pathogenesis[83]. This refinement in terminology allows for a more nuanced diagnosis: patients with fatty liver disease and a normal BMI without diabetes can be diagnosed with MASLD based on a single metabolic cardiovascular risk factor, while those with multiple risk factors require at least two for diagnosis.

These terminological shifts emphasize the need for a more comprehensive understanding of the disease, particularly highlighting the central role of metabolic dysfunction in its initiation and progression. This evolving conceptualization is crucial for developing more targeted therapeutic interventions. Moreover, it underscores the importance of categorizing MASLD alongside other metabolic disorders such as diabetes, chronic kidney disease, and cardiovascular disease, facilitating a more integrated approach to treatment and management.

The relationship between MASLD and NAFLD has been a subject of debate in recent literature. Some researchers argue that these should be considered distinct disease entities. In a meta-analysis involving 980,867 patients, Ayada et al. compared the definitions of the two diseases[84]. The study found that 79.9% of patients with fatty liver met the criteria for both conditions, 4.0% met only the NAFLD criteria, and 15.1% met the MASLD criteria. Notably, patients diagnosed with MASLD exhibited a higher prevalence of liver fibrosis progression than those with NAFLD or overlapping conditions. Alharthi et al. considered MASLD a superior name compared with NAFLD, and the transformation in name and definition from NAFLD to MAFLD represents an important milestone, which indicates significant tangible progress toward a more inclusive, equitable, and patient-centered approach to addressing the profound challenges of this disease[85].

However, other studies suggest a high degree of overlap between MASLD and NAFLD, indicating that they may be more similar than different. Younossi et al. found that only 5.29% of patients with NAFLD do not fulfill MASLD criteria in their database and the National Health and Nutrition Examination Survey of 6,429 patients[86]. Similarly, a study by Suzuki et al. reported that 96.7% of MASLD patients in their database met NAFLD criteria[87]. Zou et al. further corroborated these findings, suggesting that insights from NAFLD research remain largely applicable to MASLD[88]. These studies collectively argue that the clinical characteristics, diagnostic criteria, and research findings for NAFLD can be largely applied to MASLD, indicating a high degree of continuity between the two concepts.

In conclusion, while the shift from NAFLD to MASLD represents a significant advancement in our understanding of fatty liver disease, the high degree of overlap between these concepts suggests that much of our existing knowledge remains relevant. However, the more precise and inclusive nature of MASLD criteria may lead to better identification and management of patients at risk for metabolic liver disease. As we move forward, a balanced approach that acknowledges both the continuity and the refinements brought by the MASLD concept will be crucial for advancing our understanding and treatment of this increasingly prevalent condition.

LEAN MASLD

Obesity is a well-established risk factor and diagnostic criterion for MASLD. The increasing prevalence of MASLD over recent decades can largely be attributable to an obesogenic environment characterized by high-calorie food intake and physical inactivity[89,90]. However, it is important to note that obesity is not present in all MASLD patients. Global studies have identified that individuals with a normal BMI (18.5-25 kg/m²) who present with at least two associated risk factors - such as increased waist circumference, hypertension, low HDL-C levels, hypertriglyceridemia, impaired fasting glucose, insulin resistance, and chronic subclinical inflammation - can also develop MASLD. These cases, often termed “lean MASLD,” account for 15% to 50% of all MASLD cases[91].

While some studies suggest that lean MASLD patients may share similar morphological characteristics with their non-lean counterparts[92] or even exhibit better histological and metabolic profiles[93], long-term prognostic data often indicate worse outcomes. Meta-analyses have demonstrated that lean MASLD is associated with a higher risk of various extrahepatic complications[94] and increased all-cause mortality compared to non-lean MASLD patients[95-97]. Nonetheless, some studies report no significant survival differences between these groups. Despite the limited and somewhat conflicting prognostic data, outcomes for lean MASLD patients tend to be poorer than those of healthy individuals and may be similar to or worse than those of non-lean MASLD patients. Therefore, there is a critical need for targeted management strategies aimed at slowing disease progression and improving prognosis in lean MASLD patients.

Recent research has highlighted the role of metabolic adaptation in the pathogenesis of lean MASLD, which may explain why lean MASLD patients have similar or worse long-term outcomes compared to their non-lean counterparts. Initially, lean MASLD patients exhibit adaptive metabolic responses characterized by increased serum bile acids and elevated Farnesoid X Receptor (FXR) activity[98,99]. This adaptation appears to mitigate hepatic inflammation in the early stages of the disease. However, as the disease progresses, this adaptive response wanes, leading to a failure to attenuate hepatic inflammatory responses and contributing to disease progression. The impaired bile acid signaling, driven by alterations in the macrophage epigenome and increased expression of proinflammatory cytokines, underscores the complexity of lean MASLD pathophysiology.

Currently, no specific guidelines exist for managing lean MASLD due to a paucity of research evidence. Nevertheless, some studies indicate that weight loss can effectively mitigate fatty liver disease, with a dose-dependent relationship observed[100]. Additionally, poor diet quality is a principal modifiable risk factor for worsening metabolic health[101]. Therefore, it is important to provide all MASLD patients, particularly those with metabolically unhealthy profiles, with personalized dietary guidance and encourage physical activity (while reducing sedentary behavior) to optimize metabolic function and maintain cardiovascular health.

FUTURE INSIGHTS

The intricate interplay between metabolic dysregulation, chronic inflammation, and biliary carcinogenesis warrants further investigation. Our preliminary search on ClinicalTrials.gov using the terms “MASLD” and “intrahepatic cholangiocarcinoma” did not yield any ongoing or completed trials. Expanding the search to “metabolism” and “primary liver cancer” identified 13 relevant clinical trials, providing insights into the current research landscape on metabolic dysfunction and liver cancer.

These trials [Table 4] predominantly investigate the metabolic reprogramming of cancer cells, the impact of lifestyle factors such as diet and obesity on liver cancer, and the exploration of metabolic markers for early diagnosis and prognosis of HCC. For instance, trial NCT06278701 examines dietary interventions on liver function recovery post-hepatectomy, while NCT05794048 aims to identify metabolic markers for optimizing patient care. Trials such as NCT03663062 and NCT03356535 explore links between obesity, metabolic syndrome, and liver cancer risk, emphasizing the complex relationship between metabolic health and liver carcinogenesis.

Table 4

Clinical trials of metabolism and PLC

NCT numberCountryFocusObjective
NCT06278701ChinaDietary interventionEvaluate the effects of late-evening snacks on nutritional status, liver function recovery, complication rate, quality of life, and metabolic patterns in HCC patients post-hepatectomy
NCT05794048FranceMetabolic profilingCharacterize metabolic signatures of HCC and pancreatic tumors to identify prognostic and theranostic markers
NCT06315361ItalyDiabetes and NAFLDInvestigate the clinical impact of NAFLD in diabetic patients and assess the applicability of non-invasive fibrosis scores in this high-risk population
NCT05623150FranceAlcohol-related liver disease and metabolic syndromeIdentify metabolic, immune, and imaging factors associated with HCC development in patients with alcohol-related liver disease or non-alcoholic steatohepatitis
NCT04371042ItalyNAFLD and metabolic syndromeEstablish an observational cohort of NAFLD patients at different stages and a comparative cohort without NAFLD to characterize clinical phenotypes and collect biological samples for future omics-based studies
NCT03663062UKObesity and liver diseaseAssess the link between body weight, fatty liver disease, and colorectal adenomas in a UK population undergoing colonoscopy
NCT02730611USAMetabolic syndrome and bile acid metabolismInvestigate the relationship between bile acid profiles, metabolic syndrome, and the efficacy of vertical sleeve gastrectomy in obese patients, with a focus on HCC and cholangiocarcinoma risk
NCT03356535FranceLifestyle and metabolic signaturesIdentify metabolic signatures associated with healthy lifestyle behaviors and assess their relationship with HCC risk using data from a large European cohort
NCT01798173FranceMetabolic syndrome and HCC in cirrhosisIdentify metabolic signatures associated with healthy lifestyle behaviors and assess their relationship with HCC risk using data from a large European cohort
NCT01342705FranceIron overload and HCC riskAssess the effect of phlebotomy on HCC risk in patients with compensated alcoholic cirrhosis and hepatic iron overload
NCT04649671ChinaExercise and insulin resistanceInvestigate the impact of a mobile health-based exercise program on insulin resistance in HCC patients
NCT02561546ChinaGene therapy and diabetes in HCCEvaluate the preliminary efficacy of p53 gene therapy in treating diabetes in patients with HCC
NCT06072287UKPsychological distress in chronic diseasesDevelop and validate a new measure of illness-related distress for individuals living with long-term health conditions, including liver diseases

Given that ICC and HCC both fall under PLC and share common progenitor cell origins, future clinical research on MASLD-related ICC should leverage insights from these existing studies. Looking ahead, we propose several key directions for MASLD-related ICC:

(1) Prospective Cohort Studies: Large-scale studies are needed to delineate the natural history and specific risk factors of MASLD-related ICC. Incorporating advanced imaging techniques, such as those in NCT05794048, may help identify early radiological signatures of ICC in MASLD patients.

(2) Metabolomic and Proteomic Approaches: Methods used in trials like NCT04371042 and NCT03356535 should be applied to ICC research to discover novel biomarkers for early detection and prognosis.

(3) Interventional Studies on Lifestyle Modifications: Adapt strategies from trials such as NCT06278701 (late-evening snack strategy) and NCT04649671 (mobile health-based exercise program) for MASLD patients at high risk of ICC.

(4) Innovative Therapeutic Approaches: Explore novel treatments like p53 gene therapy investigated in NCT02561546 in the context of MASLD-related ICC to open new avenues for targeted therapy.

By addressing these priorities, we anticipate significant advancements in our understanding of MASLD-related ICC. Development of tailored screening protocols, identification of novel therapeutic targets, and ultimately improved outcomes for at-risk or diagnosed patients are expected. Interdisciplinary collaboration and integration of cutting-edge technologies will be pivotal in unraveling the complex interplay between metabolic dysfunction and cholangiocarcinogenesis, paving the way for personalized prevention and treatment strategies in the precision medicine era.

CONCLUSION

In general, it is evident that MASLD is an emerging factor in the pathophysiology of ICC. Despite the increasing prevalence of MASLD and its potential role in ICC progression, significant research gaps remain, particularly concerning the pathogenesis, prognosis, and optimal management strategies for MASLD-related ICC. Current studies are limited by retrospective designs, small sample sizes, and regional biases, necessitating large-scale, multicenter prospective cohort studies to elucidate the natural history and specific risk factors of MASLD-related ICC. Additionally, the intricate interplay between metabolic disorders, inflammation, and genetic factors in ICC pathogenesis warrants further exploration. Future research should focus on integrating advanced imaging, metabolomic, and proteomic approaches to identify novel biomarkers for early detection and prognosis. Moreover, interventional studies on lifestyle modifications and targeted therapeutic strategies are crucial for improving patient outcomes. Addressing these gaps will not only enhance our understanding of MASLD-related ICC but also pave the way for more effective prevention and treatment paradigms.

DECLARATIONS

Authors’ contributions

Conceptualized the review, conducted the literature review, and contributed to the writing and editing of the manuscript: Li ZL, Tang ZH

Assisted in the literature review: Chen JL, Tang Y, Qin DL

Availability of data and materials

Not applicable.

Financial support and sponsorship

National Natural Science Foundation of China (81772521); Hospital-level Clinical Research Cultivation Fund of Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (17CSK06); Multicenter Clinical Study of Shanghai Jiao Tong University School of Medicine (DLY201807).

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Welzel TM, Graubard BI, El-Serag HB, et al. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma in the United States: a population-based case-control study. Clin Gastroenterol Hepatol 2007;5:1221-8.

2. Kinoshita M, Kubo S, Tanaka S, et al. The association between non-alcoholic steatohepatitis and intrahepatic cholangiocarcinoma: a hospital based case-control study. J Surg Oncol 2016;113:779-83.

3. Wongjarupong N, Assavapongpaiboon B, Susantitaphong P, et al. Non-alcoholic fatty liver disease as a risk factor for cholangiocarcinoma: a systematic review and meta-analysis. BMC Gastroenterol 2017;17:149.

4. Petrick JL, Yang B, Altekruse SF, et al. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma in the United States: a population-based study in SEER-Medicare. PLoS One 2017;12:e0186643.

5. Zhou HB, Xu QR, Wang H, et al. Risk factors of intrahepatic cholangiocarcinoma: a case-control study. Zhonghua Gan Zang Bing Za Zhi 2009;17:935-9.

6. Reddy SK, Hyder O, Marsh JW et al. Prevalence of nonalcoholic steatohepatitis among patients with resectable intrahepatic cholangiocarcinoma. J Gastrointest Surg 2013;17:748-55.

7. Nkontchou G, Tran Van Nhieu J, Ziol M, et al. Peripheral intrahepatic cholangiocarcinoma occurring in patients without cirrhosis or chronic bile duct diseases: epidemiology and histopathology of distant nontumoral liver in 57 White patients. Eur J Gastroenterol Hepatol 2013;25:94-8.

8. Chang JS, Tsai CR, Chen LT. Medical risk factors associated with cholangiocarcinoma in Taiwan: a population-based case-control study. PLoS One 2013;8:e69981.

9. Choi J, Ghoz HM, Peeraphatdit T, et al. Aspirin use and the risk of cholangiocarcinoma. Hepatology 2016;64:785-96.

10. Xiong J, Lu X, Xu W, et al. Metabolic syndrome and the risk of cholangiocarcinoma: a hospital-based case-control study in China. Cancer Manag Res 2018;10:3849-55.

11. Sirica AE, Gores GJ, Groopman JD, et al. Intrahepatic cholangiocarcinoma: continuing challenges and translational advances. Hepatology 2019;69:1803-15.

12. De Lorenzo S, Tovoli F, Mazzotta A, et al. Non-alcoholic steatohepatitis as a risk factor for intrahepatic cholangiocarcinoma and its prognostic role. Cancers (Basel) 2020;12:3182.

13. Corrao S, Natoli G, Argano C. Nonalcoholic fatty liver disease is associated with intrahepatic cholangiocarcinoma and not with extrahepatic form: definitive evidence from meta-analysis and trial sequential analysis. Eur J Gastroenterol Hepatol 2021;33:62-8.

14. Ghidini M, Ramai D, Facciorusso A, et al. Metabolic disorders and the risk of cholangiocarcinoma. Expert Rev Gastroenterol Hepatol 2021;15:999-1007.

15. Eslam M, Newsome PN, Sarin SK, et al. A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J Hepatol 2020;73:202-9.

16. Crane H, Gofton C, Sharma A, George J. MAFLD: an optimal framework for understanding liver cancer phenotypes. J Gastroenterol 2023;58:947-64.

17. Golabi P, Paik JM, AlQahtani S, Younossi Y, Tuncer G, Younossi ZM. Burden of non-alcoholic fatty liver disease in Asia, the Middle East and North Africa: data from Global Burden of Disease 2009-2019. J Hepatol 2021;75:795-809.

18. Hu S, Molina L, Tao J, et al. Therapeutic implication of SOX9 and YAP1 co-repression in advanced intrahepatic cholangiocarcinoma. The FASEB Journal 2022:36.

19. Cadamuro M, Sarcognato S, Camerotto R, et al. Intrahepatic cholangiocarcinoma developing in patients with metabolic syndrome is characterized by osteopontin overexpression in the tumor stroma. Int J Mol Sci 2023;24:4748.

20. Ko S, Molina L, Tao J, et al. Simultaneous Yap and Sox9 repression eliminates Notch-dependent hepatocyte-driven cholangiocarcinogenesis. The FASEB Journal 2020;34:1-1.

21. Cadamuro M, Stecca T, Brivio S, et al. The deleterious interplay between tumor epithelia and stroma in cholangiocarcinoma. Biochim Biophys Acta Mol Basis Dis 2018;1864:1435-43.

22. Sirica AE, Strazzabosco M, Cadamuro M. Intrahepatic cholangiocarcinoma: morpho-molecular pathology, tumor reactive microenvironment, and malignant progression. Adv Cancer Res 2021;149:321-87.

23. Fabris L, Cadamuro M, Cagnin S, Strazzabosco M, Gores GJ. Liver matrix in benign and malignant biliary tract disease. Semin Liver Dis 2020;40:282-97.

24. Fava G, Alpini G, Rychlicki C, et al. Leptin enhances cholangiocarcinoma cell growth. Cancer Res 2008;68:6752-61.

25. Alzahrani B, Iseli TJ, Hebbard LW. Non-viral causes of liver cancer: does obesity led inflammation play a role? Cancer Lett 2014;345:223-9.

26. Lewitt MS, Dent MS, Hall K. The insulin-like growth factor system in obesity, insulin resistance and type 2 diabetes mellitus. J Clin Med 2014;3:1561-74.

27. Gallagher EJ, LeRoith D. Minireview: IGF, insulin, and cancer. Endocrinology 2011;152:2546-51.

28. Weroha SJ, Haluska P. The insulin-like growth factor system in cancer. Endocrinol Metab Clin North Am 2012;41:335-50.

29. Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer 2012;12:159-69.

30. Marzioni M, Fava G, Alvaro D, Alpini G, Benedetti A. Control of cholangiocyte adaptive responses by visceral hormones and neuropeptides. Clin Rev Allergy Immunol 2009;36:13-22.

31. Alvaro D, Barbaro B, Franchitto A, et al. Estrogens and insulin-like growth factor 1 modulate neoplastic cell growth in human cholangiocarcinoma. Am J Pathol 2006;169:877-88.

32. Shimizu Y, Demetris AJ, Gollin SM, et al. Two new human cholangiocarcinoma cell lines and their cytogenetics and responses to growth factors, hormones, cytokines or immunologic effector cells. Int J Cancer 1992;52:252-60.

33. Xu L, Hausmann M, Dietmaier W, et al. Expression of growth factor receptors and targeting of EGFR in cholangiocarcinoma cell lines. BMC Cancer 2010;10:302.

34. Phoomak C, Vaeteewoottacharn K, Silsirivanit A, et al. High glucose levels boost the aggressiveness of highly metastatic cholangiocarcinoma cells via O-GlcNAcylation. Sci Rep 2017;7:43842.

35. Saengboonmee C, Seubwai W, Pairojkul C, Wongkham S. High glucose enhances progression of cholangiocarcinoma cells via STAT3 activation. Sci Rep 2016;6:18995.

36. Hebbard LW, Garlatti M, Young LJ, Cardiff RD, Oshima RG, Ranscht B. T-cadherin supports angiogenesis and adiponectin association with the vasculature in a mouse mammary tumor model. Cancer Res 2008;68:1407-16.

37. Wendel HG, De Stanchina E, Fridman JS, et al. Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature 2004;428:332-7.

38. Clements O, Eliahoo J, Kim JU, Taylor-Robinson SD, Khan SA. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma: a systematic review and meta-analysis. J Hepatol 2020;72:95-103.

39. Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature 2001;410:37-40.

40. Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 2003;423:762-9.

41. Sirica AE. Role of ErbB family receptor tyrosine kinases in intrahepatic cholangiocarcinoma. World J Gastroenterol 2008;14:7033-58.

42. Wang L, Zhu H, Zhao Y, et al. Comprehensive molecular profiling of intrahepatic cholangiocarcinoma in the Chinese population and therapeutic experience. J Transl Med 2020;18:273.

43. Miyata T, Yamashita YI, Yoshizumi T, et al. CXCL12 expression in intrahepatic cholangiocarcinoma is associated with metastasis and poor prognosis. Cancer Sci 2019;110:3197-203.

44. Cen W, Li J, Tong C, et al. Intrahepatic cholangiocarcinoma cells promote epithelial-mesenchymal transition of hepatocellular carcinoma cells by secreting LAMC2. J Cancer 2021;12:3448-57.

45. Shi T, Morishita A, Kobara H, Masaki T. The role of microRNAs in cholangiocarcinoma. Int J Mol Sci 2021;22:7627.

46. Pu T, Fang Q, Chen Z, et al. Advances in molecular pathogenesis and treatment of intrahepatic cholangiocarcinoma. Available from: https://pesquisa.bvsalud.org/portal/resource/pt/wpr-865095 [Last accessed on 25 Sep 2024].

47. Chen C, Qiu Y. Advances in research, diagnosis and treatment of intrahepatic cholangiocarcinoma based on molecular mechanisms. Journal of Abdominal Surgery 2022;35:245-51.

48. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021;18:151-66.

49. Alison MR, Lovell MJ. Liver cancer: the role of stem cells. Cell Prolif 2005;38:407-21.

50. Roskams T. Liver stem cells and their implication in hepatocellular and cholangiocarcinoma. Oncogene 2006;25:3818-22.

51. Foerster F, Gairing SJ, Müller L, Galle PR. NAFLD-driven HCC: safety and efficacy of current and emerging treatment options. J Hepatol 2022;76:446-57.

52. Takahashi Y, Dungubat E, Kusano H, Fukusato T. Pathology and pathogenesis of metabolic dysfunction-associated steatotic liver disease-associated hepatic tumors. Biomedicines 2023;11:2761.

53. Yu Q, Lei Z, Ma W, et al. Postoperative prognosis of non-alcoholic fatty liver disease-associated intrahepatic cholangiocarcinoma: a multi-center propensity score matching analysis. J Gastrointest Surg 2023;27:2403-13.

54. Le Roy B, Gelli M, Pittau G, et al. Neoadjuvant chemotherapy for initially unresectable intrahepatic cholangiocarcinoma. Br J Surg 2018;105:839-47.

55. Toyoda J, Sahara K, Takahashi T, et al. Neoadjuvant therapy for extrahepatic biliary tract cancer: a propensity score-matched survival analysis. J Clin Med 2023;12:2654.

56. Kato A, Shimizu H, Ohtsuka M, et al. Surgical resection after downsizing chemotherapy for initially unresectable locally advanced biliary tract cancer: a retrospective single-center study. Ann Surg Oncol 2013;20:318-24.

57. Chang ME, Lei HJ, Chen MH, et al. Evaluation of prognostic factors and implication of lymph node dissection in intrahepatic cholangiocarcinoma: 10-year experience at a tertiary referral center. J Chin Med Assoc 2017;80:140-6.

58. Weber SM, Ribero D, O'Reilly EM, Kokudo N, Miyazaki M, Pawlik TM. Intrahepatic cholangiocarcinoma: expert consensus statement. HPB (Oxford) 2015;17:669-80.

59. Benson AB, D'Angelica MI, Abrams T, et al. NCCN guidelines® insights: biliary tract cancers, version 2.2023. J Natl Compr Canc Netw 2023;21:694-704.

60. 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;71:1229-36.

61. Simon TG, Roelstraete B, Khalili H, Hagström H, Ludvigsson JF. Mortality in biopsy-confirmed nonalcoholic fatty liver disease: results from a nationwide cohort. Gut 2021;70:1375-82.

62. Zhou XD, Targher G, Byrne CD, et al. An international multidisciplinary consensus statement on MAFLD and the risk of CVD. Hepatol Int 2023;17:773-91.

63. Cheng PN, Chen WJ, Hou CJ, et al. Taiwan association for the study of the liver-taiwan society of cardiology taiwan position statement for the management of metabolic dysfunction- associated fatty liver disease and cardiovascular diseases. Clin Mol Hepatol 2024;30:16-36.

64. Harrison SA, Taub R, Neff GW, et al. Resmetirom for nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled phase 3 trial. Nat Med 2023;29:2919-28.

65. Yang YY, Xie L, Zhang NP, Zhou D, Liu TT, Wu J. Updates on novel pharmacotherapeutics for the treatment of nonalcoholic steatohepatitis. Acta Pharmacol Sin 2022;43:1180-90.

66. Soto A, Spongberg C, Martinino A, Giovinazzo F. Exploring the multifaceted landscape of MASLD: a comprehensive synthesis of recent studies, from pathophysiology to organoids and beyond. Biomedicines 2024;12:397.

67. El-Diwany R, Pawlik TM, Ejaz A. Intrahepatic cholangiocarcinoma. Surg Oncol Clin N Am 2019;28:587-99.

68. Battistella S, Grasso M, Catanzaro E, et al. Evolution of liver transplantation indications: expanding horizons. Medicina (Kaunas) 2024;60:412.

69. Ioka T, Kanai M, Kobayashi S, et al. Kansai Hepatobiliary Oncology Group (KHBO). Randomized phase III study of gemcitabine, cisplatin plus S-1 versus gemcitabine, cisplatin for advanced biliary tract cancer (KHBO1401- MITSUBA). J Hepatobiliary Pancreat Sci 2023;30:102-10.

70. Kelley RK, Ueno M, Yoo C, et al. KEYNOTE-966 Investigators. Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2023;401:1853-65.

71. Tsung C, Quinn PL, Ejaz A. Management of intrahepatic cholangiocarcinoma: a narrative review. Cancers (Basel) 2024;16:739.

72. Bragazzi MC, Venere R, Ribichini E, Covotta F, Cardinale V, Alvaro D. Intrahepatic cholangiocarcinoma: evolving strategies in management and treatment. Dig Liver Dis 2024;56:383-93.

73. Cotter TG, Charlton M. Nonalcoholic steatohepatitis after liver transplantation. Liver Transpl 2020;26:141-59.

74. Hobeika C, Cauchy F, Poté N, et al. Short- and long-term outcomes of liver resection for intrahepatic cholangiocarcinoma associated with the metabolic syndrome. World J Surg 2019;43:2048-60.

75. Meijer VE, Kalish BT, Puder M, Ijzermans JN. Systematic review and meta-analysis of steatosis as a risk factor in major hepatic resection. Br J Surg 2010;97:1331-9.

76. Neal CP, Mann CD, Pointen E, et al. Influence of hepatic parenchymal histology on outcome following right hepatic trisectionectomy. J Gastrointest Surg 2012;16:2064-73.

77. Veteläinen R, van Vliet AK, van Gulik TM. Severe steatosis increases hepatocellular injury and impairs liver regeneration in a rat model of partial hepatectomy. Ann Surg 2007;245:44-50.

78. McCuskey RS, Ito Y, Robertson GR, McCuskey MK, Perry M, Farrell GC. Hepatic microvascular dysfunction during evolution of dietary steatohepatitis in mice. Hepatology 2004;40:386-93.

79. Poon RT, Fan ST. Assessment of hepatic reserve for indication of hepatic resection: how I do it. J Hepatobiliary Pancreat Surg 2005;12:31-7.

80. Cauchy F, Zalinski S, Dokmak S, et al. Surgical treatment of hepatocellular carcinoma associated with the metabolic syndrome. Br J Surg 2013;100:113-21.

81. Reddy SK, Marsh JW, Varley PR, et al. Underlying steatohepatitis, but not simple hepatic steatosis, increases morbidity after liver resection: a case-control study. Hepatology 2012;56:2221-30.

82. Cauchy F, Soubrane O, Belghiti J. Liver resection for HCC: patient's selection and controversial scenarios. Best Pract Res Clin Gastroenterol 2014;28:881-96.

83. Rinella ME, Lazarus JV, Ratziu V, et al. NAFLD Nomenclature consensus group. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Ann Hepatol 2024;29:101133.

84. Ayada I, van Kleef LA, Alferink LJM, Li P, de Knegt RJ, Pan Q. Systematically comparing epidemiological and clinical features of MAFLD and NAFLD by meta-analysis: focusing on the non-overlap groups. Liver Int 2022;42:277-87.

85. Alharthi J, Gastaldelli A, Cua IH, Ghazinian H, Eslam M. Metabolic dysfunction-associated fatty liver disease: a year in review. Curr Opin Gastroenterol 2022;38:251-60.

86. Younossi ZM, Paik JM, Stepanova M, Ong J, Alqahtani S, Henry L. Clinical profiles and mortality rates are similar for metabolic dysfunction-associated steatotic liver disease and non-alcoholic fatty liver disease. J Hepatol 2024;80:694-701.

87. Suzuki K, Tamaki N, Kurosaki M, et al. Concordance between metabolic dysfunction-associated steatotic liver disease and nonalcoholic fatty liver disease. Hepatol Res 2024;54:600-5.

88. Zou H, Ma X, Pan W, Xie Y. Comparing similarities and differences between NAFLD, MAFLD, and MASLD in the general U.S. population. Front Nutr 2024;11:1411802.

89. Guthold R, Stevens GA, Riley LM, Bull FC. Worldwide trends in insufficient physical activity from 2001 to 2016: a pooled analysis of 358 population-based surveys with 1·9 million participants. Lancet Glob Health 2018;6:e1077-86.

90. Xie X, Guo B, Xiao X, et al. Healthy dietary patterns and metabolic dysfunction-associated fatty liver disease in less-developed ethnic minority regions: a large cross-sectional study. BMC Public Health 2022;22:118.

91. Eslam M, El-Serag HB, Francque S, et al. Metabolic (dysfunction)-associated fatty liver disease in individuals of normal weight. Nat Rev Gastroenterol Hepatol 2022;19:638-51.

92. Chen F, Esmaili S, Rogers GB, et al. Lean NAFLD: a distinct entity shaped by differential metabolic adaptation. Hepatology 2020;71:1213-27.

93. Younes R, Govaere O, Petta S, et al. Caucasian lean subjects with non-alcoholic fatty liver disease share long-term prognosis of non-lean: time for reappraisal of BMI-driven approach? Gut 2022;71:382-90.

94. Wei L, Cheng X, Luo Y, et al. Lean non-alcoholic fatty liver disease and risk of incident diabetes in a euglycaemic population undergoing health check-ups: a cohort study. Diabetes Metab 2021;47:101200.

95. Ye Q, Zou B, Yeo YH, et al. Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2020;5:739-52.

96. Ito T, Ishigami M, Zou B, et al. The epidemiology of NAFLD and lean NAFLD in Japan: a meta-analysis with individual and forecasting analysis, 1995-2040. Hepatol Int 2021;15:366-79.

97. Reddy SK, Hyder O, Marsh JW, et al. Prevalence of nonalcoholic steatohepatitis among patients with resectable intrahepatic cholangiocarcinoma. J Gastrointest Surg 2013;17:748-55.

98. Eslam M, Fan JG, Mendez-Sanchez N. Non-alcoholic fatty liver disease in non-obese individuals: the impact of metabolic health. Lancet Gastroenterol Hepatol 2020;5:713-5.

99. Alharthi J, Pan Z, Gloss BS, et al. Loss of metabolic adaptation in lean MAFLD is driven by endotoxemia leading to epigenetic reprogramming. Metabolism 2023;144:155583.

100. Sinn DH, Kang D, Cho SJ, et al. Weight change and resolution of fatty liver in normal weight individuals with nonalcoholic fatty liver disease. Eur J Gastroenterol Hepatol 2021;33:e529-34.

101. Osadnik K, Osadnik T, Lonnie M, et al. Metabolically healthy obese and metabolic syndrome of the lean: the importance of diet quality. Analysis of MAGNETIC cohort. Nutr J 2020;19:19.

Cite This Article

Review
Open Access
MASLD: an emerging factor in the pathophysiology and clinical management of ICC
Zong-Long LiZong-Long Li, ... Zhao-Hui TangZhao-Hui Tang

How to Cite

Li, Z. L.; Chen J. L.; Tang Y.; Qin D. L.; Tang Z. H. MASLD: an emerging factor in the pathophysiology and clinical management of ICC. Hepatoma. Res. 2024, 10, 41. http://dx.doi.org/10.20517/2394-5079.2024.63

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
56
Downloads
26
Citations
0
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Hepatoma Research
ISSN 2454-2520 (Online) 2394-5079 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/