Sato-Espinoza K, Valdivia-Herrera M, Chotiprasidhi P, Diaz-Ferrer J. Hepatocellular carcinoma in patients without cirrhosis. World J Gastroenterol 2025; 31(23): 107100 [DOI: 10.3748/wjg.v31.i23.107100]
Corresponding Author of This Article
Javier Diaz-Ferrer, MD, Associate Professor, Hepatology Service, Department of Digestive Diseases, Hospital Nacional Edgardo Rebagliati Martins, 490 Edgardo Rebagliati Avenue, Lima 15072, Peru. jodf13@hotmail.com
Research Domain of This Article
Gastroenterology & Hepatology
Article-Type of This Article
Review
Open-Access Policy of This Article
This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Karina Sato-Espinoza, Perapa Chotiprasidhi, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55902, United States
Mayra Valdivia-Herrera, Escuela de Medicina Humana, Facultad de Ciencias de la Salud, Universidad Cientifica del Sur, Lima 15067, Peru
Javier Diaz-Ferrer, Hepatology Service, Department of Digestive Diseases, Hospital Nacional Edgardo Rebagliati Martins, Lima 15072, Peru
Javier Diaz-Ferrer, Department of Gastroenterology Service, Clinica Internacional, Lima 15036, Peru
Javier Diaz-Ferrer, Medicine Faculty, Universidad San Martin de Porres, Lima 15024, Peru
Author contributions: Sato-Espinoza K, Valdivia-Herrera M, Chotiprasidhi P, and Diaz-Ferrer J performed the methodology and wrote, reviewed, and edited the manuscript.
Conflict-of-interest statement: The authors have no conflicts of interest to declare.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Javier Diaz-Ferrer, MD, Associate Professor, Hepatology Service, Department of Digestive Diseases, Hospital Nacional Edgardo Rebagliati Martins, 490 Edgardo Rebagliati Avenue, Lima 15072, Peru. jodf13@hotmail.com
Received: March 16, 2025 Revised: April 22, 2025 Accepted: May 27, 2025 Published online: June 21, 2025 Processing time: 97 Days and 14 Hours
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, the sixth most common cancer worldwide, and the third leading cause of cancer-related death. Cirrhosis is the predominant risk factor for HCC, driven by major etiologies including hepatitis B and C, excessive alcohol consumption, and metabolic dysfunction-associated steatotic liver disease (MASLD). While approximately 80% of HCC cases occur in patients with cirrhosis, its incidence among individuals without cirrhosis has significantly increased, particularly in developed countries, driven by the rising prevalence of MASLD. The prevalence of patients with non-cirrhotic HCC varies geographically, yet data on this subgroup remain limited. Consequently, screening and clinical management guidelines for patients with non-cirrhotic HCC are underdeveloped. Current surveillance is typically not recommended for non-cirrhotic populations, except for individuals with hepatitis B, and diagnostic criteria like Liver Imaging Reporting and Data System are designed explicitly for cirrhotic or hepatitis B-associated HCC. Furthermore, treatment strategies for non-cirrhotic HCC are often extrapolated from studies focused on patients with cirrhosis, leading to gaps in knowledge regarding treatment efficacy, survival outcomes, and etiological variability in non-cirrhotic cohorts. Thus, emerging evidence must be reviewed to guide the development of enhanced diagnostic and therapeutic strategies for patients with non-cirrhotic HCC. To address these gaps, we comprehensively reviewed the epidemiology, clinical and genetic characteristics, diagnostic modalities, and therapeutic approaches for patients with non-cirrhotic HCC.
Core Tip: Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, and the incidence of HCC among individuals without cirrhosis is on the rise, correlating with increasing rates of obesity and metabolic syndrome. Targeted surveillance is advised for high-risk groups, especially those with chronic hepatitis B infection, while it remains unrecommended for individuals with metabolic dysfunction-associated steatotic liver disease. Liver biopsy is the only diagnostic method available for this population, highlighting the need for further non-invasive diagnostic tools. This review summarizes the epidemiology, clinical and genetic characteristics, diagnostic methods, and treatment options for patients with non-cirrhotic HCC.
Citation: Sato-Espinoza K, Valdivia-Herrera M, Chotiprasidhi P, Diaz-Ferrer J. Hepatocellular carcinoma in patients without cirrhosis. World J Gastroenterol 2025; 31(23): 107100
Hepatocellular carcinoma (HCC) is the most common primary liver neoplasia and the sixth leading cause of cancer-related mortality worldwide. Its prevalence varies across regions and is influenced by factors such as underlying liver disease (LD) etiology, genetic predisposition, and environmental risk factors[1]. While the majority of HCC cases arise in cirrhotic livers, a significant proportion (approximately 15%-20%) occurs in individuals without cirrhosis[2-4]. A recent epidemiological study showed a rising prevalence of non-cirrhotic HCC, particularly among individuals with metabolic dysfunction-associated steatotic LD (MASLD)[5]. This trend parallels the increasing rates of obesity and metabolic risk factors in Western countries and is further supported by studies highlighting an increasing prevalence of incidental HCC in individuals who underwent liver transplantation (LT) due to MASLD[5-7].
Current guidelines recommend biannual surveillance for individuals with liver cirrhosis[8-10]. However, individuals without cirrhosis typically do not undergo such surveillance, often resulting in diagnosis at more advanced tumor stages, which complicates clinical management. Furthermore, the lack of consensus and the limited studies on this specific population highlight the urgent need for improved risk stratification and early detection strategies.
This review provides a comprehensive overview of the current literature on the epidemiology, clinical and genetic characteristics, diagnostic modalities, and therapeutic approaches for HCC in non-cirrhotic individuals. Notably, HCC in individuals without cirrhosis represents a distinct clinical entity, with unique patterns in tumor presentation, diagnosis, treatment, prognosis, and epidemiology compared to HCC in patients with cirrhosis.
EPIDEMIOLOGY
Risk factors for HCC in individuals without cirrhosis vary across regions and populations. This section summarizes regional trends (Figure 1).
Figure 1 Geographic distribution of hepatocellular carcinoma in non-cirrhotic liver based on different etiologies.
ALD: Alcohol-associated liver disease; HBV: Hepatitis B virus; HCV: Hepatitis C virus; MASLD: Metabolic dysfunction-associated steatotic liver disease. Created in BioRender (Supplementary material).
North America
Two multicenter studies conducted in the United States reported that the most common etiology of HCC in individuals without cirrhosis was LD of unknown origin. The first study (2000-2014) found that 43% of cases had unknown etiology, followed by MASLD (26%), with hepatitis C virus (HCV) and hepatitis B virus (HBV) accounting for 8.6% and 8.3% of cases, respectively[11]. The second study (2000-2015) reported similar results: 40% unknown etiology, 25% MASLD, and 8.3% HBV, with a higher contribution from HCV (19%)[12]. A study in the United States Veterans Health Administration (2005-2015) noted a higher prevalence of metabolic syndrome in patients with non-cirrhotic HCC, especially those with MASLD or no known LD[13]. In Mexico, a single-center study (2008-2018) found that MASLD was the leading cause of non-cirrhotic HCC (63%), followed by alcohol LD (ALD) (27%) and HBV (9%)[14]. Another United States-based study analyzing MASLD cases (2015-2020) found that 15% of MASLD individuals developed HCC without cirrhosis, with a prevalence of 4.6 per 10000[15] (Figure 2A).
Figure 2 Summary of percentage by etiology of hepatocellular carcinoma in individuals without cirrhosis from studies conducted.
A: North America; B: Europe; C: Summary of percentage by etiology of individuals with hepatocellular carcinoma (HCC) from studies conducted in South America; D: Asia. 1Risk factor distribution was not mutually exclusive as many patients had more than one risk factor, and the percentage sum was more than 100%; 2Percentage of non-cirrhotic HCC by country: Peru 31%, Ecuador 28%, Chile 26%, Brazil 12%, Argentina 7%, Colombia 4%. Limitation of the study: The reported etiology was combined with that of individuals with cirrhosis; 3The data were inferred from the individuals with cirrhosis available status, but no confirmation of non-cirrhosis was reported in the study; 4Countries included in the analysis: Peru, Ecuador, Brazil, Argentina, Colombia, Uruguay. No data on the number of individuals without cirrhosis were available. The study’s limitation was that the reported etiology was combined with that of individuals with cirrhosis; 5Only data available on hepatitis B virus (HBV) infection in non-cirrhosis available. ALD: Alcohol-associated liver disease; HCV: Hepatitis C virus; MASLD: Metabolic dysfunction-associated steatotic liver disease.
Europe
In Western Europe, LD of unknown origin is also the most reported etiology for non-cirrhotic HCC. A nationwide study in Iceland (1998-2017) found that 35% of cases had unknown etiology, followed by metabolic syndrome/metabolic dysfunction-associated steatohepatitis (MASH) (31%) and ALD (22%). Viral hepatitis did not appear to be a significant etiology in this population during the study period[16]. In Sweden (2008-2018), 81% of cases were of unknown etiology, with 7.4% HCV and 3.5% MASLD[17]. Two studies from the Netherlands (2005-2012 and 2009-2020) reported similar patterns: Unknown etiology (30%-33%), MASLD (22%-28%), ALD (16%), and HBV (7%-15%)[18,19].
By contrast, in Eastern Europe, particularly Turkey, chronic HBV was identified as the predominant cause of non-cirrhotic HCC. The first Turkish study (2009-2019) reported HBV in 61% of cases, followed by MASLD or unknown LD (23%) and HCV (16%)[20]. The second Turkish study (2016-2020) found similar trends, with 62% HBV, 18% MASLD, and 12% unknown etiology[21]. The third Turkish study (2011-2021) observed nearly equal proportions of unknown etiology (47%) and HBV (43%)[22] (Figure 2B).
South America
A multinational study by the South American Liver Research Network, analyzing data from 2019 to 2021 across Argentina, Peru, Ecuador, Chile, and Colombia, identified HBV as the most common etiology of non-cirrhotic HCC (31%), followed by MASLD (28%)[23]. Another multinational study (2005-2015) involving Argentina, Brazil, Colombia, Ecuador, and Uruguay reported HCV and ALD as the leading risk factors for HCC in all countries except Peru, where HBV accounted for 34% of cases[24]. Supporting this, an earlier Peruvian study (2007-2008) found HBV in 81% of non-cirrhotic HCC cases[25]. In Argentina, a multicenter study (2009-2016) reported unknown LD as the most common etiology in individuals without cirrhosis (53%), followed by HCV (21%) and MASLD (10%)[26] (Figure 2C).
A major limitation in studies reporting high HCV-related HCC prevalence is the inconsistent definition of “non-cirrhotic” and the frequent absence of liver fibrosis staging. Without proper fibrosis assessment, individuals with advanced fibrosis (e.g., F3) may be misclassified as non-cirrhotic. As HCV-associated HCC risk increases in advanced fibrosis, these findings must be interpreted with caution. Prospective studies with standardized definitions and fibrosis staging are needed to more accurately characterize HCC risk factors in these populations.
Asia
A study from Korea (2000-2009) showed that about 59% of individuals without cirrhosis with HCC had chronic HBV infection, regardless of whether the infection was active[27]. Similarly, a study from Taiwan (2009-2011) reported that 49% of non-cirrhotic HCC cases were HBV-positive, followed by HCV (31%), and no underlying LD (18%)[28] (Figure 2D).
HBV infection, particularly genotypes B and C, is a well-established risk factor, even in the absence of cirrhosis, due to its direct oncogenic mechanisms[29,30]. By contrast, HCV increases the risk of HCC in patients with advanced fibrosis[31], and true non-cirrhotic HCC in patients with HCV infection is considered rare.
Worldwide
Aflatoxin exposure is a recognized risk factor for HCC, even in the absence of cirrhosis. The proportion of HCC cases linked to aflatoxin exposure varies widely by region, from as low as 3% in Latin America to as high as 40% in parts of Africa. This risk is particularly elevated in Sub-Saharan Africa, Southeast Asia, and China, where HBV and aflatoxin exposure are common[32].
Hereditary factors also contribute to non-cirrhotic HCC, although they account for a small proportion of cases. These include Wilson's disease, glycogen storage disease, alpha-1 antitrypsin deficiency, hereditary hemochromatosis, acute hepatic porphyria, hypercitrulinemia, and Budd-Chiari syndrome[33-37].
Healthcare professionals should consider these uncommon etiologies, especially in patients without conventional risk factors or with clinical features suggestive of metabolic or hereditary LD, or with exposure to high-risk regions.
In North America and Western Europe, the leading risk factors for non-cirrhotic HCC are LD of unknown etiology and MASLD. The adoption of the new nomenclature for MASLD and MASH raises an important question: Will the reclassification of cardiometabolic risk factors reduce the proportion of cases previously labeled as “unknown?” The updated nomenclature emphasizes the role of cardiometabolic conditions, such as obesity, type 2 diabetes (T2DM), hypertension, and dyslipidemia, which were not explicitly accounted for under NAFLD but are now recognized contributors to HCC[38,39]. This standardization may enhance diagnostic accuracy and support future research in identifying MASLD and MASH as leading causes of HCC[40].
Conversely, Asia and South America report higher prevalence of viral hepatitis, particularly HBV, as the leading risk factor for non-cirrhotic HCC. While antiviral therapies significantly reduce HCC risk, it does not eliminate it[41-44]. Additionally, HBV vaccination significantly reduces HCC risk. Current guidelines recommend universal vaccinations for newborns and unvaccinated high-risk adults. Ongoing screening and surveillance remain crucial for early detection and HCC prevention in at-risk populations.
HCV is frequently identified as the second or third most common etiology of HCC across different populations. However, the accuracy of these findings is limited by misclassification due to retrospective data and reliance on indirect cirrhosis assessments. Many studies use International Classification of Diseases codes, non-invasive scores, or clinical indicators such as esophageal varices, ascites, features of portal hypertension, Child-Pugh scores, elastography findings, or limited liver biopsy data. Patients with advanced fibrosis (e.g., F3 stage) but without overt cirrhotic-related complications may have been misclassified as non-cirrhotic, leading to incomplete or inaccurate risk assessments.
Accurate assessment of fibrosis status is essential for diagnosis, risk stratification, and characterization of HCC in individuals without cirrhosis. Mittal et al[13] proposed a classification system to identify individuals without cirrhosis using laboratory and imaging data. The model had two probability levels: Level 1 (indicating a very high probability of absence of cirrhosis) requires histological confirmation via liver biopsy. Level 2 (high probability of absence of cirrhosis) required the following criteria: Aspartate transaminase to platelet ratio index score < 1; no imaging features suggestive of cirrhosis on abdominal imaging within 3 years prior to HCC diagnosis; and at least two of the following three laboratory values within 6 months before and 4 weeks after diagnosis-albumin > 3.5 g/dL, platelet count > 200000/μL, or international normalized ratio < 1.1. Level 2 was externally validated by Gawrieh et al[11] using liver biopsy as the reference standard, demonstrating an area under the receiver operating characteristic of 0.80 (95% confidence interval: 0.74-0.86), with 62% sensitivity and 98% specificity. These findings support the utility of this model in accurately classifying patients without cirrhosis when biopsy is not feasible, and its potential to enhance early detection and prognostic evaluation in future studies.
PATHOGENESIS
Inflammatory mechanism
Inflammatory processes arising from various pathways of cellular injury play a significant role in initiating hepatic carcinogenesis. Hepatocyte injury promotes the recruitment of immune cells that secrete pro-inflammatory cytokines, including interleukin 1 (IL-1), IL-6, IL-18, and tumor necrosis factor alpha (TNF-α), with TNF-α identified as the most critical factor in HCC development[45]. These cytokines activate transcription factors such as nuclear factor kappa B (NF-kB) and signal transducer and activator of transcription 3 (STAT3), which drive hepatocyte proliferation, survival, and tumor progression.
In individuals with cirrhosis, chronic inflammation contributes to a pro-carcinogenic environment through persistent hepatocyte damage and regeneration, facilitating tumor initiation and progression. Genomic studies have demonstrated that sustained inflammatory stress can induce hepatocytes to dedifferentiate into progenitor-like cells, which can subsequently undergo malignant transformation[46] (Figure 3).
Figure 3 Inflammatory mechanism in hepatic carcinogenesis and metastasis.
IL: Interleukin; HBV: Hepatitis B virus; HCV: Hepatitis C virus; TNF-α: Tumor necrosis factor alpha. Created in BioRender (Supplementary material).
In individuals without cirrhosis with MASH, elevated levels of IL-6 and TNF-α stimulate STAT3 activation, promoting hepatic progenitor cell expansion, cellular transformation, and dysplastic changes in hepatocytes[47]. This inflammatory signaling facilitates tumor invasion and metastasis, correlating with clinical outcomes[48]. While inflammation remains a key contributor to tumorigenesis in non-cirrhotic HCC, the underlying mechanisms are more closely associated with metabolic dysregulation than with fibrosis-driven injury.
MASLD
HCC development in MASLD is multifactorial, with insulin resistance (IR) and oxidative stress playing key roles. The "two-hit" hypothesis suggests that IR constitutes the first hit, followed by oxidative stress as the second hit, which induces lipid peroxidation and cytokine-mediated hepatocellular injury. IR upregulates insulin-like growth factor 1 (IGF-1), activating phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways, which promote cell proliferation and inhibit apoptosis[49]. The PI3K pathway contributes to tumorigenesis through cyclin D1-mediated cell cycle progression, mouse double minute 2 homolog p53 inhibition, and mammalian target of rapamycin-driven cellular growth[50]. The MAPK pathway facilitates fibrosis and oncogenesis via transcriptional activation of c-fos and c-jun, which trigger the Wnt/β-catenin signal cascade[51]. The multiple parallel hit theory suggests that MASH development and its progression to HCC result from multiple factors including oxidative stress, genetic variations, abnormal lipid metabolism, mitochondrial dysfunction, immune response alterations, and gut microbiota imbalances[52] (Figure 4).
Figure 4 Hypothesis of the development of hepatocellular carcinoma in individuals with metabolic dysfunction-associated steatotic liver disease.
Akt: Protein kinase B; IGF-1: Insulin-like growth factor-1; MAPK: Mitogen-activated protein kinase; mTOR: Mammalian target of rapamycin; PI3K: Phosphoinositide 3-kinase. Created in BioRender (Supplementary material).
Obesity is a well-known risk factor for various types of cancer, including HCC[53,54]. Individuals with obesity often present an imbalance of adipokines, characterized by elevated leptin levels and reduced adiponectin levels, promoting IR and inflammation[55]. Leptin stimulates IGF-1 expression and enhances c-jun and c-fos, activation, thereby amplifying the risk of HCC development in both individuals with and without cirrhosis[56,57].
Hepatitis viral infection
HBV integrates its genome into the hepatocyte genome, resulting in chromosomal rearrangement and genomic instability. HBV encodes the hepatitis B virus X protein (HBx) protein, which is genotoxic and interacts with the tumor suppressor p53, inhibiting its function. Additionally, the HBx protein activates various pathways, including Ras, Raf, MAPK, extracellular signal-regulated kinase, and c-Jun N-terminal kinase, which regulate cell proliferation[58].
By contrast, the HCV does not integrate into the hepatocyte genome. Instead, it causes direct cellular damage through a neuroinflammatory process mediated by viral factors. Repeated cycles of hepatocyte death and regeneration allow for mutations to accumulate that can lead to tumorigenesis[58]. Key viral proteins such as nonstructural protein 5A (NS5A) and NS3 induce oxidative stress and activate NF-kB and MAPK signaling, enhancing inflammatory cytokine release and impairing apoptosis. Transcription factors such as Notch and transforming growth factor beta (TGF-β) are upregulated, with TGF-β playing an important role in promoting tumor invasion and metastasis through induction of oncogene expression[59].
Although both HBV and HCV are associated with HCC development in individuals with cirrhosis, only HBV can initiate carcinogenesis in the absence of cirrhosis through direct genomic integration and oncogenic activation. HCV-associated HCC, by contrast, typically requires advanced fibrosis or cirrhosis.
Aflatoxin B1
Aflatoxin B1, a mycotoxin produced by Aspergillus flavus, is a potent hepatocarcinogen. Upon ingestion, it is metabolized in the liver into a reactive epoxide that forms DNA adduct, leading to DNA strand breaks, base modifications, and oxidative damage. Aflatoxin B1 has been shown to induce mutations in the tumor protein p53 (TP53) tumor suppressor gene, which are strongly associated with HCC development, even in the absence of cirrhosis[60].
Alcohol
Excessive alcohol consumption plays a multifaceted role in liver carcinogenesis. Ethanol is converted to acetaldehyde, a genotoxic compound that forms DNA adducts and induces mutations[61]. Cytochrome P450 2E1-mediated metabolism of ethanol generates reactive oxygen species, leading to oxidative stress and hepatocyte injury. Additionally, alcohol impairs immune surveillance by suppressing the activity of natural killer cells and cytotoxic T cells[62].
Chronic inflammation, caused by endotoxins such as lipopolysaccharides, activates toll-like receptor 4, promoting tumorigenesis and carcinogenesis[63]. Alcohol has a synergistic effect with metabolic disorders and viral hepatitis, compounding HCC risk. It also disrupts gut microbiota, increases intestinal permeability, and promotes microbial translocation to the liver, further aggravating inflammation[64].
Recent findings implicate the activation of inflammasomes in ALD. Oxidative stress from alcohol stimulates the nucleotide-binding domain, leucine-rich-repeat family, pyrin domain-containing-3 inflammasome, leading to IL-1β and IL-18β secretion, immune cell recruitment, and hepatic inflammation. Interferon-inducible protein 2 inflammasome activation further contributes to immune dysregulation and fibrosis progression[64]. Despite these known pathways, the precise mechanisms underlying alcohol-related HCC in individuals without cirrhosis remain incompletely understood.
CLINICAL AND GENETIC CHARACTERISTICS
Clinical characteristics
The prevalence of HCC in individuals without cirrhosis ranges from 3.5% to 37.3%, depending on the study population and comparison groups[11-13,17-22,28,65-72] (Table 1).
Table 1 Non-cirrhotic hepatocellular carcinoma characteristics vs cirrhotic counterparts.
Individuals without cirrhosis with HCC are generally diagnosed at an older age compared to their cirrhotic counterparts[11,12,17,22,65,68,69,71]. However, conflicting research suggests that patients without cirrhosis may be younger than their cirrhotic counterparts[25,72,73], and other studies have found no significant age differences between the two groups[19,21,66]. This age variability appears to be influenced by differences in underlying population characteristics. Studies reporting a younger age of HCC onset often involve populations with a high prevalence of HBV infection, where routine surveillance enables earlier detection. In contrast, individuals with risk factors such as MASLD, obesity, or metabolic syndrome are typically diagnosed at an older age due to the lack of established screening protocols in these populations.
Several studies have reported that individuals with non-cirrhotic HCC have a higher prevalence of comorbidities, including hypertension, coronary artery disease, hyperlipidemia, T2DM, and metabolic syndrome, compared to their cirrhotic counterparts[12,13,65,68]. However, a few studies have described a lower prevalence of T2DM than in patients without cirrhosis compared to their cirrhotic counterpart[11,71].
Tumor characteristics
Most studies on HCC tumor characteristics have reported that individuals without cirrhosis are more likely to present with a single tumor, but with a larger size at diagnosis compared to their cirrhotic counterparts[11,12,16,17,19,20,22,28,65-67,69,71,72,74,75]. However, a study in Turkey reported no difference in tumor size and morphology between the groups[21]. At diagnosis, patients without cirrhosis often present with more advanced disease and a higher likelihood of metastasis[11,17,20,65,66,68,75,76]. They also show a higher prevalence of cases falling outside the Milan criteria[11,12,66,74,76], which dictate LT eligibility as a single tumor ≤ 5 cm, or up to three tumors each ≤ 3 cm, with no extrahepatic spread or major vessel involvement[77]. By contrast, a few studies have reported a lower prevalence of vascular infiltration and portal vein invasion in patients without cirrhosis compared to cirrhotic counterparts[78] (Table 1).
Genetic characteristics
An Italian study found a significant association between the membrane-bound O-acyltransferase 7 (MBOAT7) gene variant (rs641738) and increased risk of HCC in individuals with MASLD, identifying the T allele as the risk allele, particularly among those without advanced fibrosis (F0-F2)[78] HCV and ALD have also been shown to increase HCC risk in individuals without cirrhosis carrying the T allele, suggesting that the role of MBOAT7 in carcinogenesis extends beyond MASLD.
Mutations in telomerase reverse transcriptase (TERT) promoter are the most commonly reported mutation in non-cirrhotic liver with HCC[79], especially for individuals with MASLD[80]. One study utilizing high-throughput sequencing for genome- and transcriptome-wide profiling of non-cirrhotic HCC found mutations in TERT (55%), TP53 (28%), beta-catenin (CTNNB1) (21%), and apolipoprotein B (13%), similar to those found in cirrhotic counterparts. Despite high TERT expression, most tumors showed shortened telomeres. In individuals without cirrhosis, CTNNB1 mutations led to downregulated β-catenin expression—a mechanism distinct from that seen in cirrhosis. The study concludes that cirrhotic and non-cirrhotic HCC share similar genomic landscapes during tumor progression[81].
SURVEILLANCE AND DIAGNOSIS
Current guidelines do not recommend routine surveillance for all individuals with non-cirrhotic HCC. Instead, they specify subgroups of patients without cirrhosis who may warrant surveillance[82-86] (Table 2).
Table 2 Recommendations for hepatocellular carcinoma screening and surveillance by medical societies.
Individuals with Child-Pugh A or B with any etiology, and Child-Pugh C if awaiting LT
CT or MRI is suggested if limited liver visualization with ultrasound
Chronic HBV infection: From endemic countries (women > 50 years old and men > 40 years old); From Africa at an earlier age; Family history of HCC; PAGE-B Score ≥ 10
Individuals with cirrhosis type B and C should undergo dynamic CT/MRI every 6-12 months, which is optional
Individuals with chronic HBV and HCV
N/A
Ultrasound + tumor marker
Every 6 months for chronic hepatitis B/C and non-viral cirrhosis; Every 3-4 months for cirrhosis types B and C
HCC diagnosis in non-cirrhotic livers differs from that in individuals with cirrhosis. Currently, liver biopsy remains the only valid method for diagnosing HCC in patients without cirrhosis (Figure 5). Non-invasive imaging techniques, such as computed tomography scans or magnetic resonance imaging, are not recommended for diagnosing HCC in patients without cirrhosis, although they are commonly used in those with cirrhosis.
Figure 5 Graphical abstract summarizing the etiology, treatments, and diagnosis of hepatocellular carcinoma in individuals without cirrhosis.
MASLD: Metabolic dysfunction-associated steatotic liver disease. Created in BioRender (Supplementary material).
Scores and staging systems
Several studies evaluating HCC in both individuals with and without cirrhosis apply the same scoring and staging systems to assess liver function, guide treatment selection, and predict prognosis. However, the applicability and validity of these systems in individuals without cirrhosis remain unclear and warrant further discussion. Commonly used systems include the tumor-node-metastasis (TNM), Cancer of the Liver Italian Program, Barcelona Clinic Liver Cancer (BCLC), Japan Integrated Staging, and Hong Kong Liver Cancer (HKLC) systems. Among these, the BCLC system is the most widely used and validated, though primarily in cirrhotic populations.
The BCLC system, originally developed for patients with chronic LD, particularly cirrhosis[87], was updated in 2022 to incorporate features more applicable to non-cirrhotic individuals[88]. It utilizes tumor burden, liver function, and performance status. While individuals without cirrhosis typically have preserved liver function, they may still fall into BCLC stages 0, A, B, or C depending on tumor size, vascular invasion, and extrahepatic spread. Nonetheless, a key limitation is that the BCLC system lacks stratified criteria designed explicitly for patients without cirrhosis and has not been well validated in this subgroup. Prognosis and recurrence patterns may also differ between cirrhotic and non-cirrhotic HCC.
The Child-Pugh score was originally designed to assess cirrhosis severity and predict outcomes in patients undergoing surgery or treatment for portal hypertension[89,90]. It has not been validated in patients without cirrhosis, where preserved liver function can lead to underestimation of clinical features such as ascites and encephalopathy, resulting in potential misclassification as Class A.
The TNM staging system evaluates tumor characteristics, such as size, number, vascular invasion, lymph node involvement, and distant metastases, without accounting for liver function[91-93]. This makes it applicable to both individuals with and without cirrhosis[94,95].
The HKLC system, developed in a population largely affected by HBV, incorporates more aggressive treatment strategies[96]. While it may include individuals with and without cirrhosis, it uses the Child-Pugh score to assess liver function, making it difficult to determine whether patients without cirrhosis were explicitly included.
This section highlights the key features, limitations, and gaps in current scoring and staging systems as they apply to individuals with non-cirrhotic HCC. For a comprehensive comparison, refer to Table 3.
Table 3 Summary of the scores and stage systems in individuals with non-cirrhotic hepatocellular carcinoma.
Controversies
Limitations
Research hotspots
Scores and stage systems
BCLC
New update in 2022, introducing more flexibility and personalized treatment decisions with no strictly stage-based approach could be advantageous for individuals without cirrhosis
Primarily focus on individuals with underlying chronic liver disease, especially with cirrhosis. Not validated in non-cirrhotic individuals
Studies validated the BCLC stage systems in non-cirrhotic individuals with HCC, which evaluate treatment options, long-term outcomes, and explicitly stratified categories for those individuals are needed
Child-Pugh
The use of ascites and encephalopathy may be underestimated due to the common presentation of preserved liver functions observed in individuals without cirrhosis
Designed to evaluate the severity and prognosis in individuals with cirrhosis. Not validated in non-cirrhotic individuals
Studies that validate the Child-Pugh score in individuals with HCC without cirrhosis are needed; Creating a new score potentially without ascites and encephalopathy is needed to compare if the individuals are being underestimated
TNM
It is a valuable staging system for anatomical tumor burden, but is not sufficient alone for accurate HCC treatment guidance or outcomes
Focus on anatomical staging; Best for post-operative prognosis
Studies redefining TNM, evaluating treatment responses, and long-term outcomes are needed
HKLC
Proposed more aggressive treatments with more curative therapy but with potential risk of over-treatment
Developed based on a population predominantly with hepatitis B; Not well validated with other etiologies
Studies validating the stage system in diverse populations with different background etiologies are needed; Studies evaluating treatment options and long-term outcomes are needed
LI-RADS
Only individuals with hepatitis B or cirrhosis benefit from this, but individuals outside these criteria may be leading to a missed diagnosis. No consensus on biopsy, follow-up time, or surveillance in intermediate categories
Designed for individuals with cirrhosis or with HCC-associated hepatitis B; Not well validated with other etiologies
Studies validating the diagnosis in populations with different background etiologies are needed; Studies integrating LI-RADS with biomarkers to improve early HCC detection are needed
New diagnostic approaches
Currently, there is no consensus on reliable biomarkers or diagnostic tools, other than liver biopsy, for diagnosing HCC in individuals without cirrhosis. However, numerous ongoing studies are exploring the potential approaches in this population.
A case-control study evaluated bifucosylated tetra-antennary haptoglobin glycan (BiFc-Tetra-glycan) and alpha-fetoprotein (AFP) levels in individuals without cirrhosis with and without HCC. BiFc-tetra-glycan demonstrated a sensitivity of 71% and specificity of 96% for detecting HCC, whereas AFP exhibited a lower sensitivity (43%) but a higher specificity (99%)[97].
Another study assessed the utility of the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) to distinguish between HCC with and without cirrhosis. NLR showed no significant differences between the groups, but PLR was significantly higher in individuals without cirrhosis, with borderline predictive performance with area under the curve AUC of 0.66[98].
DNA methylation markers (DMMs), including homeobox A1, C-type lectin domain family 11 member A, AK055957, and testis-specific Y-encoded-like protein 5, have been evaluated in liver tissue from individuals with non-cirrhotic HCC. Four of these DMMs were elevated and present in 73.1% of patients with non-cirrhosis, compared to AFP > 20 ng/mL in only 40%. The combination of the four DMMs achieved a sensitivity of 78%, a specificity of 80%, and an AUC of 0.85 in distinguishing HCC from benign or hepatitis-related lesions[99].
Diagnosing HCC in individuals without cirrhosis is challenging due to the absence of universally validated biomarkers and limited sample sizes of existing studies. As artificial intelligence (AI) continues to advance, integrating machine learning tools with clinical data may enhance diagnostic accuracy, interpretability, and real-world applicability[100]. These technologies hold promise for improving early detection, prognostic assessment, and personalized treatment planning in non-cirrhotic HCC. Existing studies indicate that AFP has low sensitivity in individuals without cirrhosis, limiting its reliability as a diagnostic maker in this population. Emerging biomarkers and AI-driven tools represent key innovations in the diagnostic landscape, but prospective studies involving larger, more diverse cohorts are needed to validate these approaches and determine their clinical utility.
TREATMENT
Management of HCC in individuals without cirrhosis requires a comprehensive evaluation of etiology, tumor characteristics (macroscopic and microscopic), performance status, and surgical risk. Available treatments include hepatic resection, locoregional therapy, and systemic therapy (Figure 5). Hepatic resection is generally preferred, regardless of underlying etiology. While locoregional and systemic therapy are used in intermediate or advanced diseases stages, data on long-term outcomes remain limited. No clinical trials have stratified outcome by cirrhosis status, which limits the generalizability of findings to non-cirrhotic populations.
Hepatic resection
Hepatic resection is the most common curative treatment for patients without cirrhosis with HCC[12,16,17,19,21,22,28,66,69]. These individuals often present with preserved liver function, absence of portal hypertension, and lower risk of multifocal disease, making them more suitable surgical candidates. The majority presents with large, solitary tumors amenable to resection over LT or locoregional therapy.
Surgical procedures include anatomical resections (segmentectomy, sectionectomy, hemihepatectomy, extended hepatectomy) and non-anatomical (wedge) resections[101-103]. Postoperative outcomes are categorized using the R classification: R0 indicates no residual tumor (curative), R1 indicates microscopic residual tumor, and R2 indicates gross residual disease[104].
A study comparing surgical procedures for HCC in individuals without cirrhosis of various etiologies (MASH, viral hepatitis, and ALD) found that patients with MASH underwent more extensive liver resections and fewer segmental resections than other groups. These patients experienced higher rates of surgical complications, such as bile leaks, but had lower rates of non-surgical complications[105]. Another study, excluding individuals with viral hepatitis, reported higher rates of hemihepatectomy and extended hepatectomy, with 88% achieving R0 resection. Achieving R0 status was associated with improved cumulative survival (an increase by 4% at 1 year and 12% at 3 years)[106]. A German study found 50% of patients with non-cirrhotic HCC of various etiologies underwent extended hepatectomy, 87% achieved R0 resection, and 41% underwent lymphadenectomy at the hepatic hilus (median: Two lymph nodes removed)[107]. A Taiwanese study also reported a higher incidence of extensive hepatectomy with lower rates of postoperative hepatic decompensation[66].
A Korean study on hepatitis C-related HCC compared resection outcomes in individuals with and without cirrhosis. They found that individuals without cirrhosis had a higher rate of anatomic resections (96%) and major hepatectomies, but lower rates of laparoscopic surgery. No significant differences were found in transfusion rates, blood loss, operative time, postoperative mortality, or hospital stay[108]. Another study similarly reported no differences in hepatic venous pressure gradients, postoperative complications, or length of admission between the two groups[75].
A multicenter European study compared anatomical liver resection (ALR)-removal of the tumor with the corresponding liver segment-to non-ALR (NALR), which preserves more parenchyma. ALR was performed more frequently (71%) than NALR (29%). Postoperative outcomes showed no significant difference in intensive care unit stay (average 5 days), but moderate to life-threatening complications were more frequent in the NALR group, while ALR had a higher incidence of liver insufficiency[102].
Hepatic resection is the preferred treatment for HCC in non-cirrhotic livers, regardless of the underlying etiology. However, the choice of surgical technique and patient-specific risk factors significantly influence complication risk, highlighting the importance of individualized treatment planning.
Locoregional therapy
While hepatic resection remains the primary treatment for HCC in individuals without cirrhosis, several studies have reported the use of locoregional and systemic therapies in patients with non-cirrhotic HCC.
Locoregional treatments, including transarterial chemoembolization (TACE), transarterial radioembolization (TARE), and radiofrequency ablation (RFA), are commonly used in patients with cirrhosis with early to intermediate-stage HCC[109,110]. Systemic therapy is typically reserved for more advanced diseases[111,112]. However, the efficacy of these therapies in patients without cirrhosis remains unclear and warrants further investigation.
A nationwide study in Iceland found that 41% of individuals with non-cirrhotic HCC received non-surgical treatments, including TACE, RFA, and sorafenib, although, none underwent LT[16]. In a United States study for individuals with non-cirrhotic HCC, 33% received TACE, 20% TARE, and 19% systemic therapy[65]. A Turkish study reported 63% of individuals with non-cirrhotic HCC received palliative treatment, of whom 70% received TACE, 23% systemic therapy, and 7% TARE[20]. Similarly, in Mexico, 35% of patients without cirrhosis received non-surgical therapy: 26% systemic (sorafenib), 23% RFA, and 12% TACE. This study identified that older patients (> 70 years) with multiple comorbidities (e.g., hypertension, diabetes) were more likely to receive RFA, although no significant difference in survival was found between RFA and TACE[14]. By contrast, a Scottish study found improved survival with TACE in patients without cirrhosis compared to those with cirrhosis, with no differences observed for other treatments[70].
Despite these findings, direct comparative data on locoregional therapies in non-cirrhotic HCC are limited. Without standardized guidelines, treatment decisions-whether curative, palliative, or as a bridge to surgery-vary by institutional protocols and resource availability.
Systemic therapy
Systemic therapy is recommended for patients with advanced-stage HCC (BCLC stage C). particularly those who are ineligible for locoregional therapies or have failed initial treatment. According to European Association for the Study of the Liver guidelines[8], individuals with advanced HCC, preserved lived function (Child-Pugh class A), and Eastern Cooperative Oncology Group (ECOG) performance status of 0-1 should receive systemic therapy, including at least one programmed cell death protein 1/program cell death ligand 1 inhibitor, unless contraindicated. This recommendation applies regardless of tumor size, location, or number of lesions. The presence or absence of cirrhosis significantly influences tolerance to systemic therapies (Table 4).
Table 4 Clinical comparison of individuals with and without cirrhosis with hepatocellular carcinoma in systemic therapy.
Cirrhotic
Non-cirrhotic
Liver function
Impaired (Child-Pugh A, B, C)
Preserved
Therapeutic flexibility
Limited
Higher freedom for the TKI and IO combination
Toxicity risk
High (decompensation, bleeding, and encephalopathy)
Low (better tolerance)
Clinical trial inclusion
Most Child-Pugh A (B or C are typically excluded)
No well-classified
TKI use
Reduced dose in Child-Pugh B, and avoided in Child-Pugh C
Full dose (according to guidelines)
IO
Can be used in Child-Pugh A, but has uncertain use in Child-Pugh B or C
Well tolerated (first-line treatment)
Treatment goals
Controlling the tumor while maintaining liver function
Aggressive tumor control; Potential cure
Liver transplant eligibility
Yes, it is within the criteria (e.g., Milan criteria)
Usually not applicable
Prognosis
Hepatic functional reserve is more critical than tumor stage
Determined by tumor size and stage
The Sorafenib HCC Assessment Randomized Protocol (SHARP), a Phase 3 study, demonstrated the efficacy of sorafenib in patients with advanced HCC[113]. A Phase 2 randomized, double-blind, placebo-controlled trial conducted in the Asia-Pacific region[114] confirmed sorafenib’s effectiveness. The REFLECT trial, a Phase 3 multinational randomized non-inferiority study, established that lenvatinib is non-inferior to sorafenib for unresectable HCC[115]. Recently, IMBRAVE150 showed that atezolizumab plus bevacizumab significantly improved overall and progression-free survival compared to sorafenib in untreated patients with locally advanced or metastatic HCC[116]. Similarly, the HIMALAYA trial demonstrated that a combination of durvalumab and tremelimumab as first-line therapy for advanced HCC resulted in superior overall survival (OS) vs sorafenib[117].
A recent Phase 3 trial, camrelizumab plus rivoceranib vs sorafenib as first-line therapy for unresectable HCC (CARES-310), found that camrelizumab plus rivoceranib improved OS compared to sorafenib, particularly among individuals with HBV infection[118]. Interestingly, the median OS in the sorafenib arm was higher than that in the SHARP study, potentially due to differences in population characteristics, including the proportion of patients without cirrhosis.
It is important to note that all systemic therapy trials primarily included patients with compensated cirrhosis (Child-Pugh class A) and an ECOG performance status of 0-1 (Table 5). As a result, the efficacy and safety of these treatments in individuals without cirrhosis remain poorly understood. Additionally, previous studies did not adequately address the inappropriate application of the Child-Pugh score, which was specifically designed for patients with cirrhosis, thereby limiting the applicability of these findings to non-cirrhotic individuals in real-world settings. This raises concerns about extrapolating such results to non-cirrhotic populations, whose treatments and outcomes remain unclear.
Table 5 Summary of available clinical trials on systemic therapy in individuals with hepatocellular carcinoma.
Excluded patients at high risk for GI bleeding, including varices
Patients with recurrent HCC after LT are often excluded from clinical trials. However, several descriptive studies suggest that systemic therapies such as sorafenib, regorafenib, and lenvatinib may provide some clinical benefit in patients without cirrhosis[8,119-121]. By contrast, immunotherapy is generally not recommended in this population due to the risk of allograft rejection and immune-related complications.
OUTCOMES
The literature presents mixed findings on survival outcomes individuals with HCC, with and without cirrhosis, across metrics such as regarding OS, recurrence-free survival (RFS), disease-free survival (DFS), and progression-free survival (PFS).
Multiple studies have reported no significant difference in OS between individuals with and without cirrhotic HCC[16,18,20,65,66,68,70,75]. Among these, outcome variability was observed for other survival endpoints. For instance, a study on unresectable HCC found similar PFS between groups[65], while another showed no significant differences in RFS or variceal bleeding-free survival following resection[66]. A Dutch study examining cryptogenic HCC found it associated with worse outcomes compared to viral or non-viral etiologies; however, OS remained comparable between cirrhotic and non-cirrhotic individuals. Independent predictors of survival in non-cirrhotics included age, tumor stage, and treatment strategy[18].
Conversely, several studies have reported superior OS in non-cirrhotic individuals[11,12,21,22,28,69,71,76]. A Turkish study found that patients without cirrhosis showed better DFS and PFS, with curative treatment predicting OS and vascular invasion predicting DFS[22]. Another study in the same population identified portal vein thrombosis, elevated AFP, and cryptogenic etiology as independent risk factors for poor survival[21]. A multicenter French study reported that age, liver function, tumor burden, and BCLC classification were significant predictors of OS[71]. Conversely, A Taiwanese study found that age > 70 years, American Joint Committee on Cancer TNM stage III-IV, serum AFP > 200 ng/mL, and non-curative treatment were associated with increased mortality in patients with and without cirrhosis[28].
By contrast, two studies reported worse OS in patients without cirrhosis. A Swedish study identified age, ECOG performance status, tumor size, number of lesions, and treatment modality (e.g., resection or ablation) as key survival predictors[17]. The European Liver Transplant Registry found that individuals without cirrhosis had a higher mortality risk (hazard ratio = 1.37) compared to cirrhotic counterparts[72].
The variability in outcomes across studies likely reflects differences in study populations, geographic regions, and etiologies. Research from the United States, Europe, and Asia has highlighted the role of ethnic background and underlying LD in shaping outcomes. Additionally, the predominance of retrospective study designs limits control for confounders. Despite this heterogeneity, most studies indicate either improved or similar outcomes in non-cirrhotic patients. Commonly reported independent risk factors include age, tumor burden, AFP levels, and cryptogenic etiology.
CONCLUSION
HCC in individuals without cirrhosis is primarily associated with underlying LD of unknown etiology, MASLD, chronic HBV and HCV infection, and aflatoxin exposure. The prevalence of these risk factors varies by demographic region, raising concerns about the increasing prevalence of HCC associated with MASLD, metabolic syndrome, and obesity – conditions that are rising globally. Current guidelines do not recommend routine HCC surveillance for all individuals without cirrhosis but only advise screening for high-risk populations, including those with chronic hepatitis B and a family history of HCC, particularly among Asian and African populations. Accurate assessment of liver fibrosis status is essential for diagnosing and correctly classifying patients at high risk. Liver biopsy is the only validated method for diagnosing HCC in patients without cirrhosis, highlighting the need to further explore new non-invasive diagnostic tools. Additionally, the absence of valid staging and prognostic models complicates clinical management. The most common treatment performed is hepatic resection, while locoregional therapies, and systemic therapies are used in intermediate or advanced disease stages, data on long-term outcomes remained limited. The interpretation of these results is challenging since evaluating individuals using the BCLC and Child-Pugh scoring systems may underestimate prognosis in this population. Additionally, no clinical trials have differentiated outcomes based on cirrhosis status, making it challenging to apply the results to the non-cirrhotic population. Further research, particularly retrospective studies incorporating diverse cohorts and standardized diagnostic criteria, is essential to improve our understanding and management of HCC in non-cirrhotic individuals.
Footnotes
Provenance and peer review: Invited article; Externally peer reviewed.
Peer-review model: Single blind
Specialty type: Gastroenterology and hepatology
Country of origin: Peru
Peer-review report’s classification
Scientific Quality: Grade A, Grade B, Grade B, Grade B
Novelty: Grade A, Grade A, Grade B, Grade B
Creativity or Innovation: Grade A, Grade A, Grade B, Grade C
Scientific Significance: Grade A, Grade A, Grade B, Grade C
P-Reviewer: Li MY; Watanabe T; Wu HM S-Editor: Li L L-Editor: Filipodia P-Editor: Zhao S
Younossi Z, Stepanova M, Ong JP, Jacobson IM, Bugianesi E, Duseja A, Eguchi Y, Wong VW, Negro F, Yilmaz Y, Romero-Gomez M, George J, Ahmed A, Wong R, Younossi I, Ziayee M, Afendy A; Global Nonalcoholic Steatohepatitis Council. Nonalcoholic Steatohepatitis Is the Fastest Growing Cause of Hepatocellular Carcinoma in Liver Transplant Candidates.Clin Gastroenterol Hepatol. 2019;17:748-755.e3.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 599][Cited by in RCA: 569][Article Influence: 94.8][Reference Citation Analysis (0)]
Debes JD, Chan AJ, Balderramo D, Kikuchi L, Gonzalez Ballerga E, Prieto JE, Tapias M, Idrovo V, Davalos MB, Cairo F, Barreyro FJ, Paredes S, Hernandez N, Avendaño K, Diaz Ferrer J, Yang JD, Carrera E, Garcia JA, Mattos AZ, Hirsch BS, Gonçalves PT, Carrilho FJ, Roberts LR. Hepatocellular carcinoma in South America: Evaluation of risk factors, demographics and therapy.Liver Int. 2018;38:136-143.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 63][Cited by in RCA: 62][Article Influence: 8.9][Reference Citation Analysis (0)]
Bustíos Sánchez C, Díaz Ferrer J, Román Vargas R, Dávalos Moscol M, Zumaeta Villena E. Características clínico - epidemiológicas del Carcinoma Hepatocelular y su tratamiento en el departamento del aparato digestivo del HNERM ES-SALUD [Clinical - Epidemiological characteristics of the Hepatocellular Carcinoma and treatment in the departament of digestive system diseases of the National Hospital "Eduardo Rebagliatti Martins" (HNERM) - ESSALUD].Rev Gastroenterol Peru. 2009;29:17-23.
[PubMed] [DOI]
Xiao S, Liu Y, Fu X, Chen T, Xie W. Modifiable Risk Factors for Hepatocellular Carcinoma in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease: A Meta-Analysis.Am J Med. 2024;137:1072-1081.e32.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 4][Reference Citation Analysis (0)]
Liu CH, Cheng PN, Fang YJ, Chen CY, Kao WY, Lin CL, Yang SS, Shih YL, Peng CY, Chang YP, Huang SC, Su TH, Tseng TC, Liu CJ, Chen PJ, Kao JH. Risk of de novo HCC in patients with MASLD following direct-acting antiviral-induced cure of HCV infection.J Hepatol. 2025;82:582-593.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 9][Cited by in RCA: 13][Article Influence: 13.0][Reference Citation Analysis (0)]
Tanaka Y, Ogawa E, Huang CF, Toyoda H, Jun DW, Tseng CH, Hsu YC, Enomoto M, Takahashi H, Furusyo N, Yeh ML, Iio E, Yasuda S, Lam CP, Lee DH, Haga H, Yoon EL, Ahn SB, Wong G, Nakamuta M, Nomura H, Tsai PC, Jung JH, Song DS, Dang H, Maeda M, Henry L, Cheung R, Yuen MF, Ueno Y, Eguchi Y, Tamori A, Yu ML, Hayashi J, Nguyen MH; REAL-C Investigators. HCC risk post-SVR with DAAs in East Asians: findings from the REAL-C cohort.Hepatol Int. 2020;14:1023-1033.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 52][Cited by in RCA: 48][Article Influence: 9.6][Reference Citation Analysis (0)]
De Minicis S, Agostinelli L, Rychlicki C, Sorice GP, Saccomanno S, Candelaresi C, Giaccari A, Trozzi L, Pierantonelli I, Mingarelli E, Marzioni M, Muscogiuri G, Gaggini M, Benedetti A, Gastaldelli A, Guido M, Svegliati-Baroni G. HCC development is associated to peripheral insulin resistance in a mouse model of NASH.PLoS One. 2014;9:e97136.
[RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)][Cited by in Crossref: 60][Cited by in RCA: 78][Article Influence: 7.1][Reference Citation Analysis (0)]
Lamb C, Tham J, Goh TL, Barclay S, Priest M, Forrest EH, Fraser A, Kay D, Kasthuri R, Evans J, Stanley AJ. Comparison between patient characteristics, aetiology and outcomes in patients with and without cirrhosis with hepatocellular carcinoma diagnosed in a regional centre.Frontline Gastroenterol. 2024;15:380-386.
[RCA] [PubMed] [DOI] [Full Text][Cited by in RCA: 1][Reference Citation Analysis (0)]
Vitellius C, Desjonqueres E, Lequoy M, Amaddeo G, Fouchard I, N'Kontchou G, Canivet CM, Ziol M, Regnault H, Lannes A, Oberti F, Boursier J, Ganne-Carrie N. MASLD-related HCC: Multicenter study comparing patients with and without cirrhosis.JHEP Rep. 2024;6:101160.
[RCA] [PubMed] [DOI] [Full Text][Cited by in RCA: 5][Reference Citation Analysis (0)]
Pommergaard HC, Rostved AA, Adam R, Rasmussen A, Salizzoni M, Bravo MAG, Cherqui D, De Simone P, Houssel-Debry P, Mazzaferro V, Soubrane O, García-Valdecasas JC, Prous JF, Pinna AD, O'Grady J, Karam V, Duvoux C, Thygesen LC. Mortality after Transplantation for Hepatocellular Carcinoma: A Study from the European Liver Transplant Registry.Liver Cancer. 2020;9:455-467.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 6][Cited by in RCA: 9][Article Influence: 1.8][Reference Citation Analysis (0)]
Busch F, De Paepe KN, Gibbs P, Allison M, Hoare M, See TC. The clinical value of the hepatic venous pressure gradient in patients undergoing hepatic resection for hepatocellular carcinoma with or without liver cirrhosis.Open Med (Wars). 2024;19:20230851.
[RCA] [PubMed] [DOI] [Full Text][Cited by in RCA: 1][Reference Citation Analysis (0)]
Omata M, Cheng AL, Kokudo N, Kudo M, Lee JM, Jia J, Tateishi R, Han KH, Chawla YK, Shiina S, Jafri W, Payawal DA, Ohki T, Ogasawara S, Chen PJ, Lesmana CRA, Lesmana LA, Gani RA, Obi S, Dokmeci AK, Sarin SK. Asia-Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update.Hepatol Int. 2017;11:317-370.
[RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)][Cited by in Crossref: 1628][Cited by in RCA: 1613][Article Influence: 201.6][Reference Citation Analysis (0)]
Kokudo N, Takemura N, Hasegawa K, Takayama T, Kubo S, Shimada M, Nagano H, Hatano E, Izumi N, Kaneko S, Kudo M, Iijima H, Genda T, Tateishi R, Torimura T, Igaki H, Kobayashi S, Sakurai H, Murakami T, Watadani T, Matsuyama Y. Clinical practice guidelines for hepatocellular carcinoma: The Japan Society of Hepatology 2017 (4th JSH-HCC guidelines) 2019 update.Hepatol Res. 2019;49:1109-1113.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 414][Cited by in RCA: 225][Article Influence: 37.5][Reference Citation Analysis (0)]
Suddle A, Reeves H, Hubner R, Marshall A, Rowe I, Tiniakos D, Hubscher S, Callaway M, Sharma D, See TC, Hawkins M, Ford-Dunn S, Selemani S, Meyer T. British Society of Gastroenterology guidelines for the management of hepatocellular carcinoma in adults.Gut. 2024;73:1235-1268.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 2][Reference Citation Analysis (0)]
Benson AB, D'Angelica MI, Abbott DE, Anaya DA, Anders R, Are C, Bachini M, Borad M, Brown D, Burgoyne A, Chahal P, Chang DT, Cloyd J, Covey AM, Glazer ES, Goyal L, Hawkins WG, Iyer R, Jacob R, Kelley RK, Kim R, Levine M, Palta M, Park JO, Raman S, Reddy S, Sahai V, Schefter T, Singh G, Stein S, Vauthey JN, Venook AP, Yopp A, McMillian NR, Hochstetler C, Darlow SD. Hepatobiliary Cancers, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology.J Natl Compr Canc Netw. 2021;19:541-565.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 354][Cited by in RCA: 146][Article Influence: 36.5][Reference Citation Analysis (0)]
Reig M, Forner A, Rimola J, Ferrer-Fàbrega J, Burrel M, Garcia-Criado Á, Kelley RK, Galle PR, Mazzaferro V, Salem R, Sangro B, Singal AG, Vogel A, Fuster J, Ayuso C, Bruix J. BCLC strategy for prognosis prediction and treatment recommendation: The 2022 update.J Hepatol. 2022;76:681-693.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 1904][Cited by in RCA: 279][Article Influence: 93.0][Reference Citation Analysis (58)]
Zhu X, Zhou H. Neutrophil-to-Lymphocyte Ratio Can Distinguish Patients with Liver Cirrhosis from Healthy People but Cannot Distinguish Patients with Cirrhotic Hepatocellular Carcinoma from Patients with Liver Cirrhosis.J Hepatocell Carcinoma. 2022;9:1127-1136.
[RCA] [PubMed] [DOI] [Full Text][Reference Citation Analysis (0)]
Yuan H. Clinical decision making: Evolving from the hypothetico‐deductive model to knowledge‐enhanced machine learning.Med Adv. 2024;2:375-379.
[PubMed] [DOI] [Full Text]
Zeindler J, Hess GF, von Heesen M, Aegerter N, Reber C, Schmitt AM, Muenst S, Bolli M, Soysal SD, Kollmar O. Anatomic versus non-anatomic liver resection for hepatocellular carcinoma-A European multicentre cohort study in cirrhotic and non-cirrhotic patients.Cancer Med. 2024;13:e6981.
[RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)][Cited by in Crossref: 6][Reference Citation Analysis (0)]
Penzkofer L, Mittler J, Heinrich S, Wachter N, Straub BK, Kloeckner R, Stoehr F, Gairing SJ, Bartsch F, Lang H. Outcome after Resection for Hepatocellular Carcinoma in Noncirrhotic Liver-A Single Centre Study.J Clin Med. 2022;11:5802.
[RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)][Reference Citation Analysis (0)]
Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, Luo R, Feng J, Ye S, Yang TS, Xu J, Sun Y, Liang H, Liu J, Wang J, Tak WY, Pan H, Burock K, Zou J, Voliotis D, Guan Z. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial.Lancet Oncol. 2009;10:25-34.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 3854][Cited by in RCA: 4277][Article Influence: 251.6][Reference Citation Analysis (0)]
Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, Baron A, Park JW, Han G, Jassem J, Blanc JF, Vogel A, Komov D, Evans TRJ, Lopez C, Dutcus C, Guo M, Saito K, Kraljevic S, Tamai T, Ren M, Cheng AL. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial.Lancet. 2018;391:1163-1173.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 3128][Cited by in RCA: 2397][Article Influence: 342.4][Reference Citation Analysis (1)]
Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO, Li D, Verret W, Xu DZ, Hernandez S, Liu J, Huang C, Mulla S, Wang Y, Lim HY, Zhu AX, Cheng AL; IMbrave150 Investigators. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma.N Engl J Med. 2020;382:1894-1905.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 2542][Cited by in RCA: 4556][Article Influence: 911.2][Reference Citation Analysis (2)]
Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, Sukeepaisarnjaroen W, Kang YK, Van Dao T, De Toni EN, Rimassa L, Breder V, Vasilyev A, Heurgué A, Tam VC, Mody K, Thungappa SC, Ostapenko Y, Yau T, Azevedo S, Varela M, Cheng AL, Qin S, Galle PR, Ali S, Marcovitz M, Makowsky M, He P, Kurland JF, Negro A, Sangro B. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma.NEJM Evid. 2022;1:EVIDoa2100070.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 416][Reference Citation Analysis (0)]
Qin S, Chan SL, Gu S, Bai Y, Ren Z, Lin X, Chen Z, Jia W, Jin Y, Guo Y, Hu X, Meng Z, Liang J, Cheng Y, Xiong J, Ren H, Yang F, Li W, Chen Y, Zeng Y, Sultanbaev A, Pazgan-Simon M, Pisetska M, Melisi D, Ponomarenko D, Osypchuk Y, Sinielnikov I, Yang TS, Liang X, Chen C, Wang L, Cheng AL, Kaseb A, Vogel A; CARES-310 Study Group. Camrelizumab plus rivoceranib versus sorafenib as first-line therapy for unresectable hepatocellular carcinoma (CARES-310): a randomised, open-label, international phase 3 study.Lancet. 2023;402:1133-1146.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 343][Reference Citation Analysis (2)]
Invernizzi F, Iavarone M, Zavaglia C, Mazza S, Maggi U, Cesarini L, Antonelli B, Airoldi A, Manini MA, Sangiovanni A, Rossi G, Donato MF, Saverio Belli L, Lampertico P. Experience With Early Sorafenib Treatment With mTOR Inhibitors in Hepatocellular Carcinoma Recurring After Liver Transplantation.Transplantation. 2020;104:568-574.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 21][Cited by in RCA: 41][Article Influence: 8.2][Reference Citation Analysis (0)]
Gomez-Martin C, Bustamante J, Castroagudin JF, Salcedo M, Garralda E, Testillano M, Herrero I, Matilla A, Sangro B. Efficacy and safety of sorafenib in combination with mammalian target of rapamycin inhibitors for recurrent hepatocellular carcinoma after liver transplantation.Liver Transpl. 2012;18:45-52.
[RCA] [PubMed] [DOI] [Full Text][Cited by in Crossref: 122][Cited by in RCA: 125][Article Influence: 9.6][Reference Citation Analysis (0)]