Skip Navigation
Skip to contents

JLC : Journal of Liver Cancer

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Liver Cancer > Volume 25(2); 2025 > Article
Review Article
Navigating liver cancer with mouse models: a comprehensive overview of HCC experimental systems
Ga-Young Kim1,2*orcid, Dokyung Kim1*orcid, Jaehyun Jeon1orcid, Wonhyo Seo1,2†orcid, Seol Hee Park3†orcid
Journal of Liver Cancer 2025;25(2):239-250.
DOI: https://doi.org/10.17998/jlc.2025.08.21
Published online: August 22, 2025

1Lab of Hepatotoxicity, Ewha Womans University College of Pharmacy, Seoul, Korea

2Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Korea

3Department of Companion Animal Health, Hanyang Women’s University, Seoul, Korea

Corresponding author: Wonhyo Seo, Graduate School of Pharmaceutical Sciences, Ewha Womans University College of Pharmacy, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea E-mail: wonhyoseo@ewha.ac.kr
Corresponding author: Seol Hee Park, Department of Companion Animal Health, Hanyang Women’s University, 200 Salgoji-gil, Seongdong-gu, Seoul 04763, Korea E-mail: seolhee@hywoman.ac.kr
*These two authors contributed equally to this work as first authors.
†These two authors contributed equally to this work as corresponding authors.
• Received: June 6, 2025   • Revised: August 12, 2025   • Accepted: August 21, 2025

© 2025 The Korean Liver Cancer Association.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 1,495 Views
  • 76 Downloads
prev next
  • Hepatocellular carcinoma (HCC) is the most prevalent primary hepatic malignancy and is globally the third leading cause of cancerrelated deaths. Despite significant advancements in diagnostic techniques and therapeutic interventions, HCC prognosis remains poor due to asymptomatic progression, frequent recurrence, and inadequate treatment responsiveness. The development of HCC is closely linked to chronic liver diseases, such as hepatitis B and C infections, alcoholic liver disease, and metabolic dysfunctionassociated steatotic liver disease (MASLD). To better understand hepatocarcinogenesis and support therapeutic development, a range of animal models have been established. Among these animal models, mice are extensively utilized because of their genetic manipulability, physiological resemblance to humans, and relatively short experimental timelines. The most well-established protocol for analyzing the onset and progression of HCC is the diethylnitrosamine (DEN)-induced HCC model. Additionally, carbon tetrachloride (CCl4)-induced HCC models, DEN+CCl4 combination HCC models, MASLD HCC mouse models (STAMTM), alcoholassociated HCC models, hydrodynamics-based transfection systems, and orthotopic HCC transplantation approaches also provide distinct advantages for exploring specific elements of HCC pathophysiology. Unfortunately, due to the complexity and heterogeneity of human HCC, no single animal model can accurately recapitulate the disease. Therefore, careful selection or combination of appropriate mouse models for specific research objectives is crucial to enhance the translational value of preclinical studies. This review provides a comprehensive overview of the mouse models currently employed in HCC research, highlighting their respective strengths and limitations. Such understanding and application of these HCC models are essential for advancing mechanistic insights and fostering the development of novel therapeutic strategies.
Hepatocellular carcinoma (HCC), the most common primary liver malignancy, accounts for 75-85% of liver cancer cases and is the third leading cause of cancer-related death.1,2 Despite advances in diagnostic and therapeutic applications, HCC prognosis remains poor due to its typically late-stage diagnosis, high recurrence rates, and limited therapeutic options. The incidence of HCC is strongly linked to underlying chronic liver diseases, including hepatitis B virus and hepatitis C virus (HCV) infections, alcohol-associated liver disease, and metabolic dysfunction- associated steatotic liver disease (MASLD).3,4 Additional risk factors such as aflatoxin exposure, obesity, and diabetes can also exacerbate HCC progression.
Animal models of HCC have been proposed in order to better understand its complicated pathogenesis and to explore novel therapeutic approaches. These models aimed to recapitulate the primary characteristics of human HCC, including genetic mutations, the tumor microenvironment, and disease progression. Among these models, diethylnitrosamine (DEN)-induced mouse model is one of the most extensively characterized and widely used systems to study HCC pathogenesis.5 Other commonly employed models include chronic carbon tetrachloride (CCl4)-induced HCC models,6 combination of DEN+CCl4-induced HCC models,7 MASLD-associated HCC (STAMTM-HCC) models,8 alcohol-associated HCC models,9 hydrodynamics-based transfection (HBT) models,10 and orthotopic transplantation models.11 Each of these models provide unique insights into specific facets of HCC pathogenesis, albeit with inherent strengths and limitations. However, no single animal model can fully represent the complexity of human HCC, which results from intricate genetic, environmental, and metabolic interactions.12 Furthermore, translating animal study findings into human clinical applications remains a significant challenge due to interspecies variations in liver physiology and tumor biology. Therefore, to increase the translational relevance of preclinical studies, it is crucial to carefully select and combine appropriate animal models based on specific research questions.
Given the diversity and utility of existing models, this review aims to provide a comprehensive overview of the principal animal models employed in HCC research, with a particular focus on their advantages and limitations. By summarizing the current landscape of HCC modeling, this review aims to guide researchers in selecting the most appropriate models for their studies, and to facilitate the translation of experimental findings into meaningful clinical applications.
The carcinogenic effects of DEN are primarily driven by its metabolic activation, reactive oxygen species (ROS) production, and eventual DNA damage, which promotes genetic mutations. DEN is a potent alkylating agent that induces DNA damage by ethylating the N7 position of guanine, thereby forming N7-ethylguanine adducts. Such lesions compromise the integrity of DNA by causing depurination, generating apurinic sites, and promoting base mispairing. Modified guanine erroneously couples with thymine instead of cytosine, and the accumulation of such errors fosters hepatocellular carcinogenesis. CYP2E1, a key cytochrome P450 enzyme, metabolizes DEN to reactive intermediates and promotes ROS generation, including hydrogen peroxide (H2O2) and hydroxyl radicals (OH·).13,14 Exposure to DEN frequently induces mutations in the Ha-ras and B-raf oncogenes, and the Ctnnb1 gene, which plays a central role in the Wnt/β-catenin signaling pathway.13,15,16 These mutations contribute to tumor initiation and progression by driving aberrant cell proliferation and survival, disrupting the normal regulatory mechanisms of hepatocytes. The extent of DEN-induced hepatocarcinogenesis is strongly influenced by the genetic background of the mouse strain, highlighting the importance of careful strain selection to ensure experimental relevance and reproducibility (Table 1).14 Furthermore, the age and sex of the mice significantly influence the progression of DEN-induced HCC. Due to the elevated hepatic DNA synthesis during this developmental stage, weanling mice exhibit heightened susceptibility to hepatocarcinogenesis.13,14,17 Moreover, HCC develops at an early stage in all male mice (100%), whereas the incidence in female mice is limited to approximately 30%.18 These disparities are partly explained by the hepatoprotective effects of estrogens, which attenuate hepatocarcinogenesis via estrogen-dependent suppression of interleukin (IL)-6 production in Kupffer cells.19
DEN can be administered to rodents via multiple routes, including drinking water, diet, oral gavage, or inhalation.13,14,20,21 However, intraperitoneal administration is the most commonly employed protocol, as it enables precise dosage control and ensures consistency across experimental animals. Multiple intraperitoneal injections of DEN accelerate hepatic tumor proliferation (Table 1).22,23 In a study exploring HCC induction via drinking water rather than the conventional intraperitoneal injection, mice received DEN-containing water (0.95 g/mL) for 6 consecutive days followed by a day of regular water, and this cycle was repeated for 24 weeks.24
The DEN-induced HCC model provides the advantage of consistent liver tumor induction, particularly in male mice. While a single DEN administration in mice does not capture intermediate pathogenic processes, multiple DEN administrations have been demonstrated to induce fibrosis and chronic liver inflammation, more accurately reflecting human HCC progression.14,25 Although the DEN-induced HCC model is widely used, tumor formation typically requires a prolonged period (32-48 weeks), and its translational relevance may be limited due to interspecies differences in immune responses between rodents and humans.22
Chronic CCl4-induced liver injury is recognized as a well-established experimental model for studying liver disease owing to its potent hepatotoxic effects. Administered CCl4 is metabolized in the liver by CYP2E1, generating highly reactive trichloromethyl free radical (CCl3·).26,27 Produced reactive CCl3· free radical damages the endoplasmic reticulum, leading to lipid accumulation, inhibition of protein synthesis, and disruption of mixed-function oxidase activity. The reactive CCl3· free radical further undergoes further oxygenation to form trichloromethylperoxy radicals (CCl3OO·), which exacerbate hepatocyte damage by promoting lipid peroxidation.27,28 This leads to the degradation of polyunsaturated fatty acids in cellular membranes, resulting in increased membrane permeability. Additionally, the administration of CCl4 activates Kupffer cell, leading to the secretion of cytokines such as tumor necrosis factor (TNF)-α, IL-1β, IL-6, and transforming growth factor (TGF)-β.29 These cytokines, in turn, stimulate oxidant production in hepatocytes, resulting in increased ROS generation and thereby exacerbating oxidative stress and hepatic injury.26,30 CCl4-induced liver injury begins with an initial phase of predominant necrosis, characterized by elevated liver-specific enzymes and reduced pseudocholinesterase activity. This is followed by an intermediate phase of significant hepatic steatosis, marked by increased serum triglyceride and aspartate aminotransferase levels and progressive decline in liver function (intermediate phase). In the third phase, continued elevation in aspartate aminotransferase, hydroxyproline, and triglyceride levels indicates ongoing hepatic damage and fibrogenesis. In the final stage, further decline in pseudocholinesterase levels and the onset of liver atrophy reflect severe, progressive hepatic dysfunction linked to sustained fibrotic progression. Susceptibility to CCl4-induced liver injury is significantly impacted on the genetic background of the mouse strain, which should be carefully considered when designing experimental models of hepatic fibrosis (Table 2).26,31,32 Although a definitive link between age and susceptibility to CCl4 exposure has not been firmly established, growing evidence suggests that the risk of hepatic injury increases with aging.33 This upregulated inflammatory reaction accelerates the development of fibrosis in older animals, even while there is no alteration in the proliferative capacity of hepatocytes following CCl4 injury.33 A recent study revealed that ovariectomy exacerbated CCl4-induced liver injury in mice, whereas estrogen supplementation mitigated the ovariectomy-associated hepatic damage.34 The estrogen-mediated attenuation of CCl4-induced liver injury suggests that the extent of hepatic damage may differ between sexes. Thus, to minimize the variability from hormonal fluctuations, liver injury studies commonly employ male rodents.
The degree of hepatic fibrosis and the development of HCC are significantly influenced by the frequency and duration of CCl4 administration. When administered intraperitoneally to C57BL/6 mice twice a week for 6 weeks or three times a week for 4 weeks, CCl4 caused substantial intrahepatic collagen deposition that, according to the Desmet/Scheuer method, resembled stage 3 human fibrosis. Highly susceptible strains such as BALB/c mice can develop fibrosis after just 4 weeks of treatment. However, establishment of the CCl4-induced HCC model is still challenging because of the complex pathogenic mechanisms of hepatotoxins, which typically require extended dosing (e.g., 0.2 mL/kg twice weekly for 28 weeks) to promote tumor formation (Table 2).35 The subsequent administration of CCl4 enables the study of intermediate stages such as inflammation, fibrosis, and necrosis without requiring transgenic mice, making the CCl4-induced HCC mouse model highly cost-effective. Although CCl4-mediated HCC models are simple and reproducible, the lack of established protocols (e.g., treatment period/ dose) and inconsistent outcomes pose limitations. Furthermore, CCl4-induced HCC development requires multiple injections over extended periods (to 28 weeks), and the chemical’s high toxicity necessitates stringent safety precautions for researchers.
Combined administration of DEN and CCl4 accelerates HCC progression more effectively than administration of either agent alone. As previously mentioned, single dose DEN injection generates DNA adducts,13,14,17 while subsequent administration of CCl4 promotes ROS and endotoxin production, cumulatively aggravating hepatic pathology and more closely mirroring the clinical progression of human HCC.13,14 In this setting, DEN is usually administered once at a dose ranging from 2.5 to 100.0 mg/kg, depending on the age and genetic makeup of the mice used in the experiment.36 Research has revealed that even a small dose of DEN (1 mg/kg) combined with 0.2 mL/kg of CCl4 could induce HCC by 5 months of age.7 Another study demonstrated that a single DEN injection (100 mg/kg) in 6- to 14-week-old mice, followed by 6-12 biweekly injections of CCl4 (0.5 mL/kg) induced HCC development (Table 3).14 The combined administration of DEN and CCl4 induces the full spectrum of hepatic pathophysiological stages including inflammation, fibrosis, and cirrhosis, which are characteristic features of human liver cancer. Given the inherently slow progression of hepatic tumor, however, the combined administration of DEN and CCl4 provides the advantage of reducing the experimental time required to induce HCC. Moreover, this mouse model is also cost-effective than genetically engineered mouse models and achieves 100% liver tumor formation, ensuring high reproducibility and reliability. The relative simplicity of the experimental techniques boosts its practicality for researchers to understand HCC pathogenesis.
MASLD-associated HCC mouse model was developed to more precisely recapitulate the clinicopathological spectrum from hepatic steatosis to metabolic dysfunction-associated hepatitis (MASH), fibrosis, and HCC in the presence of type 2 diabetes. This model was commercialized and registered by SMC Laboratories (Tokyo, Japan) and is frequently referred to as the STAMTM model in scientific literature. This model addresses the limitations of conventional approaches, including genetically modified models, leptin-deficient models, methionine- and choline-deficient diet models, and long-term high-fat diet (HFD) models.37 The STAMTM mouse model was established by administering a single subcutaneous injection of 200 μg streptozotocin (STZ) to neonatal male C57BL/6J mice 2 days after birth, followed by an HFD starting at 4 weeks of age (Table 4).38 Low-dose STZ induces mild islet inflammation and destruction, contributing to diabetic conditions within 4 weeks.37 STZ is selectively transported into pancreatic β-cells via GLUT2, where it exerts cytotoxic effects through DNA alkylation, culminating in β-cell death.39 STZ induced cytotoxicity causes metabolic disturbances, including hyperglycemia, impaired glucose tolerance, and reduced glucose responsiveness due to decreased insulin secretion.40 Once these diabetes characteristics were established, the animals were subjected to an HFD to further accelerate disease progression. The composition of the HFD used in the STAMTM model can differ based on the study’s purpose. In the STAMTM model, HFDs typically account for 45% to 75% of the total calorie intake and may include additional components such as fructose or cholesterol.41 These HFDs usually consist of 60 kcal% fat, 20 kcal% protein, and 20 kcal% carbohydrates42,43 or a diet that contains 45 kcal% fat, 20 kcal% protein and 35 kcal% carbohydrates.41,44 Consequently, MASH develops by 8 weeks, followed by fibrosis and cirrhosis around 12 weeks, and HCC between 16 and 20 weeks of age.45,46 In particular, hyperglycemia and impaired insulin secretion facilitate fatty acid esterification and subsequent triglyceride accumulation after 6 weeks of treatment.47 Elevated triglyceride levels that overwhelm β-oxidation capability activate alternative metabolic pathways, resulting in the production of lipotoxic compounds such as ceramides and cholesterol, which subsequently promote ROS generation in hepatocytes.40 Over the course of 8 weeks, persistent hepatic inflammation activates Kupffer cells via damage-associated molecular patterns and facilitates immune cell infiltration, which results in the release of pro-inflammatory cytokines such as IL-6, interferon-γ, TNF-α, and CCL2.48 By weeks 10 to 12, this chronic inflammatory milieu triggers the expression of fibrogenic mediators, including TGF-β1, matrix metalloproteinase-9, tissue inhibitor of metalloproteinase-1, IL-4, and IL-10, which activate hepatic stellate cells (HSC) and advance hepatic fibrosis.37,40,49 After 20 weeks of exposure, all mice developed HCC and aberrant activation of the PI3K/Akt/mTOR signaling pathway. This disruption activates downstream effectors such as nuclear factor-κB and the Wnt/β-catenin pathway, thereby promoting tumor cell proliferation, migration, and invasion.40,50,51 Notably, there has been evidence of a sex-specific discrepancy in the STAMTM model in mouse models. All male mice developed HCC, whereas female mice did not.37 Female mice only exhibited fatty liver, mild liver inflammation, and decreased phagocytosis of macrophages. In addition, the female mice did not exhibit MASH-based persistent accumulation of macrophages and fibroblasts in the pericentral zones, indicating that the recruitment of fibroblasts around macrophages is the crucial characteristic for the advancement of diabetes-induced HCC.37 Although the STAMTM model effectively demonstrated HCC progression in patients with diabetes, it still has several limitations. The STAMTM model is induced using STZ, which results in a near complete loss of pancreatic insulin production.45 The model is frequently associated with weight loss and insulin deficiency due to the effects of STZ treatment features not typically observed in human MASH-HCC patients. In human MASH-HCC, obesity and insulin resistance boost de novo lipogenesis. However, reduced insulin secretion in the STAMTM model may result in insufficient activation of de novo lipogenesis, restricting its ability to accurately replicate the metabolic factors implicated in disease progression.47 Despite the limitations, the STAMTM model demonstrates approximately 100% reproducibility with a well-defined disease progression in a comparatively brief experimental period.37 Given its high reproducibility and ability to replicate the clinicopathological process in male MASH-HCC patients, this model offers valuable insights into the link between metabolic disorders and carcinoma, serving as a platform for developing novel therapeutic strategies.
Alcohol is a well-established etiological factor implicated in a broad spectrum of liver disorders, ranging from simple steatosis to liver cancer. Alcohol-associated HCC is caused by a variety of mechanisms, including the dysregulation of alcohol metabolism, induction of CYP2E1, impaired hepatocyte regeneration, and associated nutritional inadequacies.52,53 Imbalanced hepatic homeostasis promotes lipid peroxidation and structural/functional damage to cellular membranes, nucleic acids, and proteins, which ultimately results in hepatocyte death. Chronic alcohol exposure further amplifies the CYP2E1 expression and enzymatic activity, increasing the accumulation of toxic metabolites and promoting progressive hepatic injury.54,55 Additionally, ethanol-induced hypoxia and activation of the hypoxia-inducible transcription factors further induce hepatocyte death, escalating profibrotic responses such as HSC activation.56 The ongoing hepatocyte death and production of reactive metabolites accelerate genomic instability, accelerating the progression from dysplastic lesions to malignant tumors.57 Beyond the direct effects of alcohol, alcoholic liver disease (ALD) development is further impacted by gut barrier dysfunction, which promotes the translocation of endotoxins into the circulation.58 The elevated endotoxin level further promotes the activation of HSCs, causing excessive extracellular matrix deposition, fibrosis, cirrhosis, and eventually HCC.57
Previous studies have shown that aging is a critical determinant in the experimental models of alcohol exposure. Adolescent mice demonstrate elevated ethanol intake and tolerance, whereas older mice are more vulnerable to ALD, which is attributed to the diminished CYP2E1 enzymatic activity and the downregulation of SIRT1 in both hepatocytes and HSCs.59 Therefore, to optimize metabolic responsiveness while minimizing age-related susceptibility, the use of 8- to 11-week-old mice is recommended in rodent models to reduce alcohol-induced mortality.57 Additionally, due to differences in alcohol metabolism, hormone regulation, and antioxidant defense, mouse strain and sex differences also exert a substantial impact on the development of alcohol-associated HCC (Table 5).60-62 Females are typically more susceptible to alcohol-induced hepatotoxicity due to elevated endotoxin levels, potentially driven by estradiol-mediated reductions in lipoprotein levels. However, estrogen also raises hepatic glutathione levels, providing defense against oxidative stress. In contrast, males may experience more rapid alcohol-associated HCC progression because of lower antioxidant capacity, highlighting a complex interplay of hormonal effects. Given the complexity of the many contributing factors, females may experience a more pronounced liver injury due to heightened susceptibility to advanced HCC progression. This complexity makes it especially challenging to make direct comparisons of sex-based vulnerability in ALD-associated HCC.
Based on previous studies, the combined exposure of alcohol and carcinogens synergistically drives liver injury, progressing from inflammation to liver fibrosis and HCC. To investigate alcohol-associated liver carcinogenesis, several mouse models have been developed by integrating ethanol administration with agents such as DEN, CCl4, or HCV, thereby closely simulating the pathological characteristics of alcohol-associated HCC.52,63-69 In the DEN+alcohol model, C57BL/6 male mice receive intraperitoneal injections of DEN (75 mg/kg for 3 weeks, followed by 100 mg/kg for another 3 weeks), with a 4% Lieber-DeCarli ethanol diet introduced at week 8. This regimen induces early HCC development as early as week 12, significantly earlier than in DEN-only models.68 An alternative protocol employs a single low-dose DEN injection (10 mg/kg) at 2 weeks of age, followed by 4.8% ethanol feeding for 3-7 weeks, leading to visible tumor formation by weeks 19-20.66,69 In the DEN+CCl4+alcohol model, mice are sequentially administered DEN (100 mg/kg via intraperitoneal injection, followed by 50 mg/kg via oral administration), CCl4 (up to 8 mL/kg by oral gavage), and 9% ethanol in drinking water, resulting in progressive induction of hepatitis (by day 60), fibrosis (by day 90), cirrhosis (by day 120), and HCC (by day 150).66 In the HCV+alcohol model, transgenic mice expressing the HCV NS5A gene under the apoE promoter are chronically administered a 3.5% ethanol Lieber-DeCarli diet (Table 5).63 This mild but sustained alcohol exposure amplifies NS5A-mediated Toll-like receptor 4 signaling and promotes liver tumorigenesis, with 23% of mice developing HCC within a year. These mouse models effectively provide a valuable platform for elucidating alcohol-associated molecular and cellular mechanisms, especially the interplay of environmental carcinogens, genetic alterations, and alcohol-induced oxidative injury.63-70
Despite these advantages, several limitations hinder the complete replication of human ALD-associated HCC pathology in these mouse models. Significantly, mice metabolize alcohol seven times faster than humans and tend to lower their intake once acetaldehyde accumulates, making it difficult to establish a mode of chronic alcohol consumption.71 Additionally, the forced administration of alcohol -often through liquid diets- can induce metabolic alterations unrelated to alcohol exposure.57 Disease progression modeling is further constrained by the inability to maintain long-term alcohol consumption.70 Importantly, in humans, alcohol consumption is frequently sporadic and modulated by individual dietary patterns and lifestyle, which contrasts with the sustained and standardized alcohol exposure protocols commonly employed in animal models. Furthermore, although these models replicate alcohol-induced liver damage leading to HCC, the underlying genetic mutations and molecular pathways often diverge from those observed in humans, raising concerns about their translational relevance. Variability in alcohol dosing protocols, administration methods, and interstrain genetic differences further complicates reproducibility and undermines experimental reliability (Table 5).
HBT is a gene delivery technique involving the rapid intravenous administration of a large volume of naked DNA plasmids, typically via the tail vein. The resulting hydrodynamic pressure transiently disrupts the capillary endothelium and induces pore formation in the plasma membranes of adjacent parenchymal cells, thereby facilitating the entry of plasmid DNA into the intracellular compartment. Subsequently, the membrane pores reseal, effectively trapping the plasmid DNA within the cells. The internalized plasmids are subsequently transcribed, resulting in the expression of oncogenes or other functional genes that promote tumorigenesis.10 Notably, the liver preferentially uptakes the majority of the injected plasmid DNA, leading to effective transfection in approximately 10-40% of hepatocytes. This strategy has been successfully utilized for efficient hepatic gene delivery in mice and has enabled the in depth investigation of liver cancer through the HBT-induced HCC mouse model. Although plasmid DNA is degraded within a few days following hydrodynamic transfection, sustained transgene expression is crucial for effective tumor induction. Using the Sleeping Beauty (SB) transposon method, which promotes stable transgene incorporation into the host genome, long-term gene expression was achieved (Table 6).10,72 This integration enables persistent oncogene expression, thereby promoting tumor development. The resulting model replicates the key genetic alterations observed in human HCC, offering a robust and biologically relevant platform for investigating HCC pathogenesis and evaluating potential therapeutic interventions. Specifically, DNA plasmids are prepared by recombinant DNA technology by constructing and amplifying plasmids containing the target gene.73 The DNA plasmid solution, corresponding to 8-10% of the mouse’s body weight, is rapidly injected via the tail vein.10,73 When combined with the plasmid, the SB transposon technology allows for stable integration and sustained expression of the target gene in hepatocytes for extended transgene expression.10 The onset and progression of liver tumor development vary based on the specific gene introduced. For instance, Fah-deficient mice administered with the corresponding plasmid begin to develop liver nodules approximately 20 weeks following injection, while those receiving a plasmid expressing Myc generally develop liver tumors within 5 to 8 weeks.72
Compared to transgenic mice, the HBT-induced HCC model is highly efficient and relatively simple to establish. The DNA plasmid can be easily reconstructed through the recombinant DNA technique, which takes a shorter period of time. Because of its great reproducibility, this model enables rapid assessment of the experimental findings. One of the primary limitations of the HBT-induced HCC model is the transient expression of non-integrating DNA plasmids. However, this limitation can be addressed by employing the SB transposon system, which facilitates stable genomic integration and prolonged transgene expression. Furthermore, the successful establishment of HCC models using this system requires careful selection of oncogenes or tumor suppressor genes that accurately recapitulate the molecular pathogenesis of HCC.
The orthotopic HCC implantation mouse model is a widely employed preclinical platform that closely recapitulates the tumor microenvironment characteristic of human liver cancer.74 The orthotopic HCC mouse model is established by implanting HCC cells or tumor tissue directly into the liver of mice, enabling tumor growth within its native microenvironment.74,75 In this model, either HCC cell suspensions or tumor tissue fragments are directly implanted into the liver of mice, enabling tumor growth within the native hepatic architecture. There are two main implantation techniques: cell suspension injection, where cultured HCC cells (HepG2, Huh7, Hepa1-6, or HCA-1) are directly transplanted into the liver parenchyma, and tumor fragment transplantation, which involves surgically embedding pieces of pre-existing tumors into the hepatic tissue. While the former provides a simpler and more rapid approach to induce tumor formation (typically within 1-2 weeks), the latter better preserves tumor heterogeneity and stromal architecture.76
The orthotopic mouse model for HCC induction offers critical advantages, including rapid and reproducible tumor formation due to direct hepatic injection of cancer cells, comparatively low technical difficulty, and accurate recapitulation of the human tumor microenvironment (Table 7). Furthermore, a key advantage of the orthotopic model is its ability to reflect the vascular, stromal, and immune components of the hepatic microenvironment, rendering it more physiologically relevant than subcutaneous xenograft models. This model is particularly useful for investigating tumor progression, angiogenesis, immune cell interactions, and metastasis.75,77,78 However, the model necessitates substantial surgical expertise to ensure precise injection and to prevent tumor cell leakage into the peritoneal cavity, which might result in ectopic tumor growth. Furthermore, tracking tumor progression in vivo can be challenging unless the tumor cells are genetically labeled with bioluminescent or fluorescent reporters. Despite these limitations, the orthotopic HCC model remains a translationally relevant tool for investigating liver cancer biology and evaluating therapeutic strategies in a setting that closely mirrors human disease.
Given the ethical and practical limitations on human experimentation, the development of optimal animal models is critical for elucidating the pathophysiology of liver cancer. Mice are widely employed due to the substantial similarity between mouse and human pathogenic pathways, enabling the study of disease progression and the evaluation of therapeutic strategies with high translational relevance. Mouse models produce highly reproducible results, ensuring consistent and reliable data for preclinical research. Moreover, their relatively short lifespan allows for rapid observation of disease progression, thereby reducing the overall duration of experiments. In this review, we compared various mouse models, including those induced by DEN, CCl4, DEN+CCl4, STZ+HFD, chronic alcohol feeding, hydrodynamic transfection, as well as orthotopic implantation (Table 8; Fig. 1). Each model presents distinct advantages and offers insights into diverse aspects of HCC pathophysiology. Given the heterogeneity of disease progression and underlying mechanisms, the selection of an appropriate model customized to the specific research objective is essential for yielding relevant and interpretable results.
As no single animal model perfectly recapitulates all human HCC characteristics, meticulous assessment of each model’s strengths and limitations is necessary in both basic and translational research. Ultimately, the strategic selection of HCC mouse models enables the comprehensive exploration of disease mechanisms and therapeutic interventions, thereby advancing the overall understanding of HCC.

Acknowledgements

The figures were generated using BioRender.com (http://www.biorender.com).

Conflicts of Interest

The authors have no conflicts of interest to disclose.

Ethics Statement

This review article is fully based on articles that have already been published and does not involve additional participants. Therefore, IRB approval is not necessary.

Funding Statement

This work was supported by grants from the National Research Foundation of Korea (NRF) (No. 2021R1A6C101A442 and 2022R1C1C1008912), the Supporting Program of the Korean Association for the Study of the Liver, and the Korean Liver Foundation.

Data Availability

Not available.

Author Contributions

Conceptualization: SHP

Supervision: WS

Writing - first draft: DK, JJ

Writing - review & editing: GYK

Figure 1.
A comprehensive overview of HCC mouse models. Schematic illustration of representative experimental systems used to study HCC pathogenesis and therapeutic strategies. Models include DEN-induced HCC mouse model, CCl4-induced HCC mouse model, combined DEN+CCl4-induced HCC mouse model, MASLD-associated HCC mouse model, alcohol-associated HCC mouse model, HBTinduced HCC mouse model, and orthotopic HCC implantation mouse model (using either HCC cell line suspension or tumor tissue). These systems differ in their etiological relevance, pathological progression, and applicability for mechanistic and preclinical studies. DEN, diethylnitrosamine; HCC, hepatocellular carcinoma; HBT, hydrodynamics-based transfection; CCl4, carbon tetrachloride; MASLD, metabolic dysfunction-associated steatotic liver disease.
jlc-2025-08-21f1.jpg
Table 1.
Genetic background and administration protocol for DEN-induced hepatocarcinogenesis
Single DEN injection Multiple DEN injection
Frequency Once 2-3 times per week
Duration 32-48 weeks 24-40 weeks
Intermediate Processes Only HCC observed Inflammation, fibrosis, and HCC observed
Dosage 25 mg/kg (2-week-old mouse)25 or 90 mg/kg (6-week-old mouse)23 35 mg/kg (6-week-old mouse)79 or 20 mg/kg (1 week) → 30 mg/kg (1 week) → 50 mg/kg of DEN (6 weeks)17

Highly susceptible strain, C3H, CBA; intermediate strain, FVB, SM/J, P/J, CE/J, LP, and APR/J; highly resistant strain, BALB/c, C57BL/6, 129, and SWR.14

Table 2.
Genetic background and comparison of experimental protocols for inducing liver pathology via multiple intraperitoneal injections or oral gavage of CCl4
Multiple intraperitoneal injections Oral gavage77,78
Frequency 2-3 times per week26,35 3 times per week
Duration To 28 weeks (when used individually)35 To 12 weeks
Intermediate processes Every intermediate process observed (inflammation, fibrosis, and HCC)13 Intermediate processes observed (fibrosis and cirrhosis)
Dosage 0.2-1.0 mL/kg with olive oil 150 μL*

Highly susceptible strain, BALB/c; intermediate strain, C57BL/6, DBA/232; highly resistant strain, FVB/N, C3H/He, AKR.26,31

CCl4, carbon tetrachloride; HCC, hepatocellular carcinoma.

* Oral gavage may not lead to HCC development.

Table 3.
Comprehensive overview of the experimental parameters and pathological characteristics associated with the combined administration of DEN and CCl4 in mouse models of hepatocarcinogenesis
Combined administration of DEN+CCl4 (intraperitoneal injection)
Frequency Single injection of DEN+multiple (1-2 times per week) CCl4 injections
Duration 5-10 months
HCC growth 100%13,19
Intermediate processes Every intermediate step is observed (fibrosis, inflammation, necrosis, and HCC)
Dosage 1-100 mg/kg of DEN and 0.2-0.5 mg/kg of CCl4 (depending on the age, sex, and genetic background of the mice)

DEN, diethylnitrosamine; CCl4, carbon tetrachloride; HCC, hepatocellular carcinoma.

Table 4.
Overview of the experimental parameters and disease progression in a mouse model of hepatocarcinogenesis induced by STZ administration and HFD
Subcutaneous Injection of STZ+HFD37,38,41-43,45,46
Frequency Single injection of STZ (2 days after birth)+HFD (postnatal day 28)
Duration 16-20 weeks
Intermediate processes Every intermediate process observed (inflammation, fibrosis, and HCC)
Dosage 200 μg of STZ

STZ, streptozotocin; HFD, high-fat diet; HCC, hepatocellular carcinoma.

Table 5.
Genetic background and the administration protocols of key experimental conditions in mouse models of alcohol-associated HCC, including treatment regimens, durations, dosages, and administration methods
DEN+alcohol68,69 DEN+CCl4+alcohol66 HCV+alcohol63
Frequency Multiple injections of DEN+4% EtOH-containing Lieber DeCarli liquid diet Single injection of DEN+4.8% EtOH-containing Lieber DeCarli liquid diet DEN injected twice+multiple injections of CCl4+9% alcohol in drinking water HCV (NS5A) transgenic+3.5% EtOH-containing Lieber DeCarli liquid diet
Duration To 12 weeks 19-20 weeks To 5 months To 12 months
Intermediate processes Intermediate processes observed (inflammation, fibrosis, and HCC) Intermediate processes observed (inflammation, fibrosis, and HCC) Intermediate processes observed (inflammation, fibrosis, and HCC) Intermediate processes observed (inflammation, fibrosis, and HCC)
Dosage 100 mg/kg DEN or 75 mg/kg DEN68 Low-dose DEN injection (10 mg/kg) 100 mg/kg DEN (IP) → 5 mL/kg CCl4 (twice a week, PO) → 50 mg/kg DEN (PO) → 8 mL/kg CCl4 (twice a week, PO) -
Method IP injection IP injection IP injection (1st DEN), gavage (2nd DEN, CCl4) -

Highly alcohol-preferring strain, C57BL/6NCrl; alcohol-preferring strain, C57BL/6J; alcohol-resistant strain, BALB/c, DBA/2.

HCC, hepatocellular carcinoma; DEN, diethylnitrosamine; CCl4, carbon tetrachloride; HCV, hepatitis C virus; EtOH, ethanol; IP, intraperitoneal; PO, oral administration.

Table 6.
Comparative analysis of HBT with and without the SB transposon
HBT without SB transposon HBT with SB transposon
Frequency Once, 5-9 seconds of rapid injection10,73 Once, 5-9 Seconds of rapid injection10,73
Duration DNA degrades within several days72 Degrades much later than the classic HBT (without SB transposon)10
Dosage 8-10% of the body weight10,73 8-10% of the body weight10,73
Method Hydrodynamic tail vein injection Hydrodynamic tail vein injection
Special notes The liver should not undergo fibrosis (lowers the absorption of DNA)80 The liver should not undergo fibrosis (lowers the absorption of DNA)80

HBT, hydrodynamics-based transfection; SB, Sleeping Beauty.

Table 7.
Comparison of HCC cell line injections and tissue transplantation in an orthotopic HCC mouse model
Human HCC cell line injection77 Tissue transplantation74
Onset period 1-2 weeks At least 3 months after implantation
Pre-existing conditions Fibrosis and cirrhosis Not required
Dosage 1×106 HCA-1 cells 2×2 mm liver tissue
Method Intrahepatic injection Median laparotomy

HCC, hepatocellular carcinoma.

Table 8.
Comprehensive summary of the advantages, limitations, and experimental applications of mouse models for investigating HCC
Models Advantage Limitation Application
DEN High tumor incidence with simple protocol Long duration of HCC development (>40 weeks) Ideal for studying “genetic mutation-driven HCC pathogenesis, and tumor initiation”
Precise dosage control Lack fibrosis/inflammation (single dose)
Useful for studying genetic mutations Effective mainly in males
Mechanisms partly differ from human HCC
CCl4 Shows clear pathological stages in experiments High toxicity of CCl4 Best suited for investigating “pathogenesis of liver fibrosis and chronic inflammation-mediated HCC progression”
Reproduces fibrosis and HCC progression Complex pathogenic mechanisms
Variable reproducibility (by age and sex)
DEN+CCl4 Accelerated HCC progression Toxicity and safety issue Recommended when modeling “inflammation- and fibrosis-driven HCC” closely resembling human chronic liver disease
Improved clinical relevance Complexity in mechanistic interpretation
Suitable for investigating the entire pathological spectrum of liver disease (from inflammation to HCC)
STZ+HFD Rapid experimental timeline Weight loss and lack of obesity Optimal for studying “MASLD/MASH-associated HCC in diabetic, non-obese context”, appropriate for metabolic and anti-MASH therapeutic research
Applicable to diabetes-associated liver cancer Impaired modeling of lipogenesis
Enables immune-metabolic interaction studies Limited metabolic diversity
Alcohol Clinically relevant disease progression (from inflammation to HCC) Alcohol administration alone is hard to induce HCC Applied in studies focusing on “alcohol-associated liver disease and its carcinogenic mechanisms”, especially sex- and strain-specific vulnerability
Efficient tumor induction timeline Difficult to maintain long-term alcohol consumption
Lack of human-like metabolic comorbidities strain, sex and protocol variability
HBT High efficiency and rapid tumor induction Potential for off-target effects Useful for “testing gene function, oncogene overexpression, or tumor microenvironment manipulation” in vivo
Stable gene expression via SB transposon Limited reflection of tumor microenvironment
Customizable genetic modeling
Orthotopic Physiological relevance Tumor leaking risk Employed for “studies of tumor growth in physiological liver environment, metastasis, microenvironment interactions, and therapeutic interventions”
Rapid and reproducible tumor formation In vivo tracking difficulties (requires fluorescent tagging system)
Low technical complexity (for cell suspension)

HCC, hepatocellular carcinoma; DEN, diethylnitrosamine; CCl4, carbon tetrachloride; STZ, streptozotocin; HFD, high-fat diet; MASLD, metabolic dysfunction-associated steatotic liver disease; MASH, metabolic dysfunction-associated hepatitis; HBT, hydrodynamics-based transfection; SB, Sleeping Beauty.

  • 1. Chidambaranathan-Reghupaty S, Fisher PB, Sarkar D. Hepatocellular carcinoma (HCC): epidemiology, etiology and molecular classification. Adv Cancer Res 2021;149:1−61.PubMed
  • 2. Kim DY. Changing etiology and epidemiology of hepatocellular carcinoma: Asia and worldwide. J Liver Cancer 2024;24:62−70.ArticlePubMedPMCPDF
  • 3. Gallo P, Silletta M, Prinzi FL, Farolfi T, Coppola A. Hepatocellular carcinoma and non-alcoholic fatty liver disease: a modern context for an ancient disease. J Clin Med 2023;12:4605. ArticlePubMedPMC
  • 4. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:6. ArticlePubMedPDF
  • 5. Schulien I, Hasselblatt P. Diethylnitrosamine-induced liver tumorigenesis in mice. Methods Cell Biol 2021;163:137−152.PubMed
  • 6. Fujii T, Fuchs BC, Yamada S, Lauwers GY, Kulu Y, Goodwin JM, et al. Mouse model of carbon tetrachloride induced liver fibrosis: histopathological changes and expression of CD133 and epidermal growth factor. BMC Gastroenterol 2010;10:79. ArticlePubMedPMCPDF
  • 7. Uehara T, Pogribny IP, Rusyn I. The DEN and CCl4-induced mouse model of fibrosis and inflammation-associated hepatocellular carcinoma. Curr Protoc Pharmacol 2014;66:14.30.1−14.30.10.ArticlePubMedPMCPDF
  • 8. Jeong BK, Choi WI, Choi W, Moon J, Lee WH, Choi C, et al. A male mouse model for metabolic dysfunction-associated steatotic liver disease and hepatocellular carcinoma. Nat Commun 2024;15:6506. ArticlePubMedPMCPDF
  • 9. Ambade A, Satishchandran A, Szabo G. Alcoholic hepatitis accelerates early hepatobiliary cancer by increasing stemness and miR-122-mediated HIF-1α activation. Sci Rep 2016;6:21340. ArticlePubMedPMCPDF
  • 10. Chen X, Calvisi DF. Hydrodynamic transfection for generation of novel mouse models for liver cancer research. Am J Pathol 2014;184:912−923.ArticlePubMedPMC
  • 11. Lee TK, Na KS, Kim J, Jeong HJ. Establishment of animal models with orthotopic hepatocellular carcinoma. Nucl Med Mol Imaging 2014;48:173−179.ArticlePubMedPMCPDF
  • 12. Blidisel A, Marcovici I, Coricovac D, Hut F, Dehelean CA, Cretu OM. Experimental models of hepatocellular carcinoma-a preclinical perspective. Cancers (Basel) 2021;13:3651. ArticlePubMedPMC
  • 13. Caviglia JM, Schwabe RF. Mouse models of liver cancer. Methods Mol Biol 2015;1267:165−183.ArticlePubMed
  • 14. Tolba R, Kraus T, Liedtke C, Schwarz M, Weiskirchen R. Diethylnitrosamine (DEN)-induced carcinogenic liver injury in mice. Lab Anim 2015;49 Suppl 1:59−69.ArticlePDF
  • 15. Wang X, Liu Y, Zhang S, Zhang J, Lin X, Liang Y, et al. Genomic and transcriptomic analyses of chemical hepatocarcinogenesis aggravated by oncoprotein loss. Hepatology 2025;81:1181−1196.ArticlePubMed
  • 16. Jaworski M, Buchmann A, Bauer P, Riess O, Schwarz M. B-raf and Haras mutations in chemically induced mouse liver tumors. Oncogene 2005;24:1290−1295.ArticlePubMedPDF
  • 17. Memon A, Pyao Y, Jung Y, Lee JI, Lee WK. A modified protocol of diethylnitrosamine administration in mice to model hepatocellular carcinoma. Int J Mol Sci 2020;21:5461. ArticlePubMedPMC
  • 18. Smiriglia A, Lorito N, Serra M, Perra A, Morandi A, Kowalik MA. Sex difference in liver diseases: how preclinical models help to dissect the sexrelated mechanisms sustaining NAFLD and hepatocellular carcinoma. iScience 2023;26:108363. ArticlePubMedPMC
  • 19. Prieto J. Inflammation, HCC and sex: IL-6 in the centre of the triangle. J Hepatol 2008;48:380−381.ArticlePubMed
  • 20. Yang B, Zhang J, Wang J, Fan W, Barbier-Torres L, Yang X, et al. CSNK2A1-mediated MAX phosphorylation upregulates HMGB1 and IL-6 expression in cholangiocarcinoma progression. Hepatol Commun 2023;7:e00144.ArticlePubMedPMC
  • 21. Iatropoulos MJ, Jeffrey AM, Schlüter G, Enzmann HG, Williams GM. Bioassay of mannitol and caprolactam and assessment of response to diethylnitrosamine in heterozygous p53-deficient (+/-) and wild type (+/+) mice. Arch Toxicol 2001;75:52−58.ArticlePubMedPDF
  • 22. Kurma K, Manches O, Chuffart F, Sturm N, Gharzeddine K, Zhang J, et al. DEN-induced rat model reproduces key features of human hepatocellular carcinoma. Cancers (Basel) 2021;13:4981. ArticlePubMedPMC
  • 23. Rignall B, Braeuning A, Buchmann A, Schwarz M. Tumor formation in liver of conditional β-catenin-deficient mice exposed to a diethylnitrosamine/phenobarbital tumor promotion regimen. Carcinogenesis 2011;32:52−57.ArticlePubMed
  • 24. Liu ZH, Bai MJ, Li JQ, Yan SH, Ye XW, Bu JG, et al. Correlation between inflammatory cytokines and liver cancer stem cell markers in DEN-induced liver cancer rats. Eur Rev Med Pharmacol Sci 2021;25:710−721.PubMed
  • 25. Schneider C, Teufel A, Yevsa T, Staib F, Hohmeyer A, Walenda G, et al. Adaptive immunity suppresses formation and progression of diethylnitrosamine-induced liver cancer. Gut 2012;61:1733−1743.ArticlePubMedPMC
  • 26. Scholten D, Trebicka J, Liedtke C, Weiskirchen R. The carbon tetrachloride model in mice. Lab Anim 2015;49 Suppl 1:4−11.ArticlePDF
  • 27. Ritesh KR, Suganya A, Dileepkumar HV, Rajashekar Y, Shivanandappa T. A single acute hepatotoxic dose of CCl4 causes oxidative stress in the rat brain. Toxicol Rep 2015;2:891−895.ArticlePubMedPMC
  • 28. Ezhilarasan D, Karthikeyan S, Najimi M, Vijayalakshmi P, Bhavani G, Rani MJ. Preclinical liver toxicity models: advantages, limitations and recommendations. Toxicology 2025;511:154020. ArticlePubMed
  • 29. Taniguchi M, Takeuchi T, Nakatsuka R, Watanabe T, Sato K. Molecular process in acute liver injury and regeneration induced by carbon tetrachloride. Life Sci 2004;75:1539−1549.ArticlePubMed
  • 30. Dong S, Chen QL, Song YN, Sun Y, Wei B, Li XY, et al. Mechanisms of CCl4-induced liver fibrosis with combined transcriptomic and proteomic analysis. J Toxicol Sci 2016;41:561−572.ArticlePubMed
  • 31. Santos NP, Colaço AA, Oliveira PA. Animal models as a tool in hepatocellular carcinoma research: a review. Tumour Biol 2017;39:1010428317695923. ArticlePubMedPDF
  • 32. Zheng HC, Xue H, Yun WJ. An overview of mouse models of hepatocellular carcinoma. Infect Agent Cancer 2023;18:49. ArticlePubMedPMCPDF
  • 33. Mahrouf-Yorgov M, de l'Hortet AC, Cosson C, Slama A, Abdoun E, Guidotti JE, et al. Increased susceptibility to liver fibrosis with age is correlated with an altered inflammatory response. Rejuvenation Res 2011;14:353−363.ArticlePubMed
  • 34. Chen ZY, Panga MJ, Zhang X, Qiao S, Chen S, Appiah C, et al. Estrogen alleviates liver fibrosis and restores metabolic homeostasis in ovariectomy-induced liver injury and carbon tetrachloride (CCl4) exposure. Eur J Pharmacol 2024;978:176774. ArticlePubMed
  • 35. Seo W, Gao Y, He Y, Sun J, Xu H, Feng D, et al. ALDH2 deficiency promotes alcohol-associated liver cancer by activating oncogenic pathways via oxidized DNA-enriched extracellular vesicles. J Hepatol 2019;71:1000−1011.ArticlePubMedPMC
  • 36. Fuentes-Hernández S, Alarcón-Sánchez BR, Guerrero-Escalera D, Montes-Aparicio AV, Castro-Gil MP, Idelfonso-García OG, et al. Chronic administration of diethylnitrosamine to induce hepatocarcinogenesis and to evaluate its synergistic effect with other hepatotoxins in mice. Toxicol Appl Pharmacol 2019;378:114611. ArticlePubMed
  • 37. Fujii M, Shibazaki Y, Wakamatsu K, Honda Y, Kawauchi Y, Suzuki K, et al. A murine model for non-alcoholic steatohepatitis showing evidence of association between diabetes and hepatocellular carcinoma. Med Mol Morphol 2013;46:141−152.ArticlePubMedPDF
  • 38. Fang T, Wang H, Pan X, Little PJ, Xu S, Weng J. Mouse models of nonalcoholic fatty liver disease (NAFLD): pathomechanisms and pharmacotherapies. Int J Biol Sci 2022;18:5681−5697.ArticlePubMedPMC
  • 39. Szkudelski T. The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res 2001;50:537−546.ArticlePubMed
  • 40. Márquez-Quiroga LV, Arellanes-Robledo J, Vásquez-Garzón VR, Villa-Treviño S, Muriel P. Models of nonalcoholic steatohepatitis potentiated by chemical inducers leading to hepatocellular carcinoma. Biochem Pharmacol 2022;195:114845. ArticlePubMed
  • 41. Lau JKC, Zhang X, Yu J. Animal models of non-alcoholic fatty liver diseases and its associated liver cancer. Adv Exp Med Biol 2018;1061:139−147.ArticlePubMed
  • 42. Ito M, Suzuki J, Tsujioka S, Sasaki M, Gomori A, Shirakura T, et al. Longitudinal analysis of murine steatohepatitis model induced by chronic exposure to high-fat diet. Hepatol Res 2007;37:50−57.ArticlePubMed
  • 43. Oligschlaeger Y, Shiri-Sverdlov R. NAFLD Preclinical models: more than a handful, less of a concern? Biomedicines 2020;8:28. ArticlePubMedPMC
  • 44. Lau JK, Zhang X, Yu J. Animal models of non-alcoholic fatty liver disease: current perspectives and recent advances. J Pathol 2017;241:36−44.ArticlePubMedPMCPDF
  • 45. Oniciu DC, Hashiguchi T, Shibazaki Y, Bisgaier CL. Gemcabene downregulates inflammatory, lipid-altering and cell-signaling genes in the STAMTM model of NASH. PLoS One 2018;13:e0194568.ArticlePubMedPMC
  • 46. Saito K, Uebanso T, Maekawa K, Ishikawa M, Taguchi R, Nammo T, et al. Characterization of hepatic lipid profiles in a mouse model with nonalcoholic steatohepatitis and subsequent fibrosis. Sci Rep 2015;5:12466. ArticlePubMedPMCPDF
  • 47. Sasaki Y, Asahiyama M, Tanaka T, Yamamoto S, Murakami K, Kamiya W, et al. Pemafibrate, a selective PPARα modulator, prevents non-alcoholic steatohepatitis development without reducing the hepatic triglyceride content. Sci Rep 2020;10:7818. ArticlePubMedPMCPDF
  • 48. van der Windt DJ, Sud V, Zhang H, Varley PR, Goswami J, Yazdani HO, et al. Neutrophil extracellular traps promote inflammation and development of hepatocellular carcinoma in nonalcoholic steatohepatitis. Hepatology 2018;68:1347−1360.ArticlePubMedPMCPDF
  • 49. Saito T, Muramatsu M, Ishii Y, Saigo Y, Konuma T, Toriniwa Y, et al. Pathophysiological analysis of the progression of hepatic lesions in STAM mice. Physiol Res 2017;66:791−799.ArticlePubMed
  • 50. Chen J, Chen J, Huang J, Li Z, Gong Y, Zou B, et al. HIF-2α upregulation mediated by hypoxia promotes NAFLD-HCC progression by activating lipid synthesis via the PI3K-AKT-mTOR pathway. Aging (Albany NY) 2019;11:10839−10860.ArticlePubMedPMC
  • 51. Fishman P, Cohen S, Itzhak I, Amer J, Salhab A, Barer F, et al. The A3 adenosine receptor agonist, namodenoson, ameliorates non‑alcoholic steatohepatitis in mice. Int J Mol Med 2019;44:2256−2264.ArticlePubMedPMC
  • 52. Mercer KE, Hennings L, Ronis MJ. Alcohol consumption, Wnt/β-catenin signaling, and hepatocarcinogenesis. Adv Exp Med Biol 2015;815:185−195.ArticlePubMedPMC
  • 53. Seitz HK, Pöschl G, Simanowski UA. Alcohol and cancer. Recent Dev Alcohol 1998;14:67−95.ArticlePubMed
  • 54. Mercer KE, Hennings L, Sharma N, Lai K, Cleves MA, Wynne RA, et al. Alcohol consumption promotes diethylnitrosamine-induced hepatocarcinogenesis in male mice through activation of the Wnt/β-catenin signaling pathway. Cancer Prev Res (Phila) 2014;7:675−685.ArticlePubMedPMCPDF
  • 55. Leung TM, Lu Y. Alcoholic liver disease: from CYP2E1 to CYP2A5. Curr Mol Pharmacol 2017;10:172−178.ArticlePubMedPMC
  • 56. Roychowdhury S, Chiang DJ, McMullen MR, Nagy LE. Moderate, chronic ethanol feeding exacerbates carbon-tetrachloride-induced hepatic fibrosis via hepatocyte-specific hypoxia inducible factor 1α. Pharmacol Res Perspect 2014;2:e00061.ArticlePubMedPMCPDF
  • 57. Guo F, Zheng K, Benedé-Ubieto R, Cubero FJ, Nevzorova YA. The lieberdecarli diet-a flagship model for experimental alcoholic liver disease. Alcohol Clin Exp Res 2018;42:1828−1840.ArticlePubMedPDF
  • 58. Szabo G, Bala S. Alcoholic liver disease and the gut-liver axis. World J Gastroenterol 2010;16:1321−1329.ArticlePubMedPMC
  • 59. Ramirez T, Li YM, Yin S, Xu MJ, Feng D, Zhou Z, et al. Aging aggravates alcoholic liver injury and fibrosis in mice by downregulating sirtuin 1 expression. J Hepatol 2017;66:601−609.ArticlePubMedPMC
  • 60. Bizzaro D, Becchetti C, Trapani S, Lavezzo B, Zanetto A, D'Arcangelo F, et al. Influence of sex in alcohol-related liver disease: pre-clinical and clinical settings. United European Gastroenterol J 2023;11:218−227.ArticlePubMedPMCPDF
  • 61. Bertola A, Mathews S, Ki SH, Wang H, Gao B. Mouse model of chronic and binge ethanol feeding (the NIAAA model). Nat Protoc 2013;8:627−637.ArticlePubMedPMCPDF
  • 62. Poley W. Alcohol-preferring and alcohol-avoiding C57BL mice. Behav Genet 1972;2:245−248.ArticlePubMedPDF
  • 63. Machida K, Tsukamoto H, Mkrtchyan H, Duan L, Dynnyk A, Liu HM, et al. Toll-like receptor 4 mediates synergism between alcohol and HCV in hepatic oncogenesis involving stem cell marker Nanog. Proc Natl Acad Sci U S A 2009;106:1548−1553.ArticlePubMedPMC
  • 64. Yao P, Zhang Z, Liu H, Jiang P, Li W, Du W. p53 protects against alcoholic fatty liver disease via ALDH2 inhibition. EMBO J 2023;42:e112304.PubMedPMC
  • 65. Ha NB, Yao F. Alcohol and hepatocellular carcinoma. Clin Liver Dis 2024;28:633−646.ArticlePubMed
  • 66. Xin B, Cui Y, Wang Y, Wang L, Yin J, Zhang L, et al. Combined use of alcohol in conventional chemical-induced mouse liver cancer model improves the simulation of clinical characteristics of human hepatocellular carcinoma. Oncol Lett 2017;14:4722−4728.ArticlePubMedPMC
  • 67. Brandon-Warner E, Walling TL, Schrum LW, McKillop IH. Chronic ethanol feeding accelerates hepatocellular carcinoma progression in a sex-dependent manner in a mouse model of hepatocarcinogenesis. Alcohol Clin Exp Res 2012;36:641−653.ArticlePubMedPMC
  • 68. Ambade A, Satishchandran A, Gyongyosi B, Lowe P, Szabo G. Adult mouse model of early hepatocellular carcinoma promoted by alcoholic liver disease. World J Gastroenterol 2016;22:4091−4108.ArticlePubMedPMC
  • 69. Zhao J, O'Neil M, Vittal A, Weinman SA, Tikhanovich I. PRMT1-dependent macrophage IL-6 production is required for alcohol-induced HCC progression. Gene Expr 2019;19:137−150.ArticlePubMedPMC
  • 70. Zhu L, Li HD, Xu JJ, Li JJ, Cheng M, Meng XM, et al. Advancements in the alcohol-associated liver disease model. Biomolecules 2022;12:1035. ArticlePubMedPMC
  • 71. Brandon-Warner E, Schrum LW, Schmidt CM, McKillop IH. Rodent models of alcoholic liver disease: of mice and men. Alcohol 2012;46:715−725.ArticlePubMedPMC
  • 72. Ju HL, Han KH, Lee JD, Ro SW. Transgenic mouse models generated by hydrodynamic transfection for genetic studies of liver cancer and preclinical testing of anti-cancer therapy. Int J Cancer 2016;138:1601−1608.ArticlePubMed
  • 73. Wu LL, Wang HY, Chen PJ. Hydrodynamic HBV transfection mouse model. Methods Mol Biol 2017;1540:227−235.ArticlePubMed
  • 74. Armengol C, Tarafa G, Boix L, Solé M, Queralt R, Costa D, et al. Orthotopic implantation of human hepatocellular carcinoma in mice: analysis of tumor progression and establishment of the BCLC-9 cell line. Clin Cancer Res 2004;10:2150−2157.PubMed
  • 75. Lambertucci F, Li S, Motiño O, Montégut L, Nogueira-Recalde U, Chen H, et al. Orthotopic model of hepatocellular carcinoma in mice. Methods Mol Biol 2024;2769:1−13.ArticlePubMed
  • 76. Céspedes MV, Espina C, García-Cabezas MA, Trias M, Boluda A, del Pulgar MTG, et al. Orthotopic microinjection of human colon cancer cells in nude mice induces tumor foci in all clinically relevant metastatic sites. Am J Pathol 2007;170:1077−1085.ArticlePubMedPMC
  • 77. Reiberger T, Chen Y, Ramjiawan RR, Hato T, Fan C, Samuel R, et al. An orthotopic mouse model of hepatocellular carcinoma with underlying liver cirrhosis. Nat Protoc 2015;10:1264−1274.ArticlePubMedPMCPDF
  • 78. Muñoz NM, Dupuis C, Williams M, Dixon K, McWatters A, Zhang J, et al. Immune modulation by molecularly targeted photothermal ablation in a mouse model of advanced hepatocellular carcinoma and cirrhosis. Sci Rep 2022;12:14449. PubMedPMC
  • 79. Santos NP, Oliveira PA, Arantes-Rodrigues R, Faustino-Rocha AI, Colaço A, Lopes C, et al. Cytokeratin 7/19 expression in N-diethylnitrosamine-induced mouse hepatocellular lesions: implications for histogenesis. Int J Exp Pathol 2014;95:191−198.ArticlePubMedPMCPDF
  • 80. Yeikilis R, Gal S, Kopeiko N, Paizi M, Pines M, Braet F, et al. Hydrodynamics based transfection in normal and fibrotic rats. World J Gastroenterol 2006;12:6149−6155.ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      • ePub LinkePub Link
      • XML DownloadDownload Citation
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        Navigating liver cancer with mouse models: a comprehensive overview of HCC experimental systems
        J Liver Cancer. 2025;25(2):239-250.   Published online August 22, 2025
        Close
      • XML DownloadXML Download
      Navigating liver cancer with mouse models: a comprehensive overview of HCC experimental systems
      Image
      Figure 1. A comprehensive overview of HCC mouse models. Schematic illustration of representative experimental systems used to study HCC pathogenesis and therapeutic strategies. Models include DEN-induced HCC mouse model, CCl4-induced HCC mouse model, combined DEN+CCl4-induced HCC mouse model, MASLD-associated HCC mouse model, alcohol-associated HCC mouse model, HBTinduced HCC mouse model, and orthotopic HCC implantation mouse model (using either HCC cell line suspension or tumor tissue). These systems differ in their etiological relevance, pathological progression, and applicability for mechanistic and preclinical studies. DEN, diethylnitrosamine; HCC, hepatocellular carcinoma; HBT, hydrodynamics-based transfection; CCl4, carbon tetrachloride; MASLD, metabolic dysfunction-associated steatotic liver disease.
      Navigating liver cancer with mouse models: a comprehensive overview of HCC experimental systems
      Single DEN injection Multiple DEN injection
      Frequency Once 2-3 times per week
      Duration 32-48 weeks 24-40 weeks
      Intermediate Processes Only HCC observed Inflammation, fibrosis, and HCC observed
      Dosage 25 mg/kg (2-week-old mouse)25 or 90 mg/kg (6-week-old mouse)23 35 mg/kg (6-week-old mouse)79 or 20 mg/kg (1 week) → 30 mg/kg (1 week) → 50 mg/kg of DEN (6 weeks)17
      Multiple intraperitoneal injections Oral gavage77,78
      Frequency 2-3 times per week26,35 3 times per week
      Duration To 28 weeks (when used individually)35 To 12 weeks
      Intermediate processes Every intermediate process observed (inflammation, fibrosis, and HCC)13 Intermediate processes observed (fibrosis and cirrhosis)
      Dosage 0.2-1.0 mL/kg with olive oil 150 μL*
      Combined administration of DEN+CCl4 (intraperitoneal injection)
      Frequency Single injection of DEN+multiple (1-2 times per week) CCl4 injections
      Duration 5-10 months
      HCC growth 100%13,19
      Intermediate processes Every intermediate step is observed (fibrosis, inflammation, necrosis, and HCC)
      Dosage 1-100 mg/kg of DEN and 0.2-0.5 mg/kg of CCl4 (depending on the age, sex, and genetic background of the mice)
      Subcutaneous Injection of STZ+HFD37,38,41-43,45,46
      Frequency Single injection of STZ (2 days after birth)+HFD (postnatal day 28)
      Duration 16-20 weeks
      Intermediate processes Every intermediate process observed (inflammation, fibrosis, and HCC)
      Dosage 200 μg of STZ
      DEN+alcohol68,69 DEN+CCl4+alcohol66 HCV+alcohol63
      Frequency Multiple injections of DEN+4% EtOH-containing Lieber DeCarli liquid diet Single injection of DEN+4.8% EtOH-containing Lieber DeCarli liquid diet DEN injected twice+multiple injections of CCl4+9% alcohol in drinking water HCV (NS5A) transgenic+3.5% EtOH-containing Lieber DeCarli liquid diet
      Duration To 12 weeks 19-20 weeks To 5 months To 12 months
      Intermediate processes Intermediate processes observed (inflammation, fibrosis, and HCC) Intermediate processes observed (inflammation, fibrosis, and HCC) Intermediate processes observed (inflammation, fibrosis, and HCC) Intermediate processes observed (inflammation, fibrosis, and HCC)
      Dosage 100 mg/kg DEN or 75 mg/kg DEN68 Low-dose DEN injection (10 mg/kg) 100 mg/kg DEN (IP) → 5 mL/kg CCl4 (twice a week, PO) → 50 mg/kg DEN (PO) → 8 mL/kg CCl4 (twice a week, PO) -
      Method IP injection IP injection IP injection (1st DEN), gavage (2nd DEN, CCl4) -
      HBT without SB transposon HBT with SB transposon
      Frequency Once, 5-9 seconds of rapid injection10,73 Once, 5-9 Seconds of rapid injection10,73
      Duration DNA degrades within several days72 Degrades much later than the classic HBT (without SB transposon)10
      Dosage 8-10% of the body weight10,73 8-10% of the body weight10,73
      Method Hydrodynamic tail vein injection Hydrodynamic tail vein injection
      Special notes The liver should not undergo fibrosis (lowers the absorption of DNA)80 The liver should not undergo fibrosis (lowers the absorption of DNA)80
      Human HCC cell line injection77 Tissue transplantation74
      Onset period 1-2 weeks At least 3 months after implantation
      Pre-existing conditions Fibrosis and cirrhosis Not required
      Dosage 1×106 HCA-1 cells 2×2 mm liver tissue
      Method Intrahepatic injection Median laparotomy
      Models Advantage Limitation Application
      DEN High tumor incidence with simple protocol Long duration of HCC development (>40 weeks) Ideal for studying “genetic mutation-driven HCC pathogenesis, and tumor initiation”
      Precise dosage control Lack fibrosis/inflammation (single dose)
      Useful for studying genetic mutations Effective mainly in males
      Mechanisms partly differ from human HCC
      CCl4 Shows clear pathological stages in experiments High toxicity of CCl4 Best suited for investigating “pathogenesis of liver fibrosis and chronic inflammation-mediated HCC progression”
      Reproduces fibrosis and HCC progression Complex pathogenic mechanisms
      Variable reproducibility (by age and sex)
      DEN+CCl4 Accelerated HCC progression Toxicity and safety issue Recommended when modeling “inflammation- and fibrosis-driven HCC” closely resembling human chronic liver disease
      Improved clinical relevance Complexity in mechanistic interpretation
      Suitable for investigating the entire pathological spectrum of liver disease (from inflammation to HCC)
      STZ+HFD Rapid experimental timeline Weight loss and lack of obesity Optimal for studying “MASLD/MASH-associated HCC in diabetic, non-obese context”, appropriate for metabolic and anti-MASH therapeutic research
      Applicable to diabetes-associated liver cancer Impaired modeling of lipogenesis
      Enables immune-metabolic interaction studies Limited metabolic diversity
      Alcohol Clinically relevant disease progression (from inflammation to HCC) Alcohol administration alone is hard to induce HCC Applied in studies focusing on “alcohol-associated liver disease and its carcinogenic mechanisms”, especially sex- and strain-specific vulnerability
      Efficient tumor induction timeline Difficult to maintain long-term alcohol consumption
      Lack of human-like metabolic comorbidities strain, sex and protocol variability
      HBT High efficiency and rapid tumor induction Potential for off-target effects Useful for “testing gene function, oncogene overexpression, or tumor microenvironment manipulation” in vivo
      Stable gene expression via SB transposon Limited reflection of tumor microenvironment
      Customizable genetic modeling
      Orthotopic Physiological relevance Tumor leaking risk Employed for “studies of tumor growth in physiological liver environment, metastasis, microenvironment interactions, and therapeutic interventions”
      Rapid and reproducible tumor formation In vivo tracking difficulties (requires fluorescent tagging system)
      Low technical complexity (for cell suspension)
      Table 1. Genetic background and administration protocol for DEN-induced hepatocarcinogenesis

      Highly susceptible strain, C3H, CBA; intermediate strain, FVB, SM/J, P/J, CE/J, LP, and APR/J; highly resistant strain, BALB/c, C57BL/6, 129, and SWR.14

      Table 2. Genetic background and comparison of experimental protocols for inducing liver pathology via multiple intraperitoneal injections or oral gavage of CCl4

      Highly susceptible strain, BALB/c; intermediate strain, C57BL/6, DBA/232; highly resistant strain, FVB/N, C3H/He, AKR.26,31

      CCl4, carbon tetrachloride; HCC, hepatocellular carcinoma.

      Oral gavage may not lead to HCC development.

      Table 3. Comprehensive overview of the experimental parameters and pathological characteristics associated with the combined administration of DEN and CCl4 in mouse models of hepatocarcinogenesis

      DEN, diethylnitrosamine; CCl4, carbon tetrachloride; HCC, hepatocellular carcinoma.

      Table 4. Overview of the experimental parameters and disease progression in a mouse model of hepatocarcinogenesis induced by STZ administration and HFD

      STZ, streptozotocin; HFD, high-fat diet; HCC, hepatocellular carcinoma.

      Table 5. Genetic background and the administration protocols of key experimental conditions in mouse models of alcohol-associated HCC, including treatment regimens, durations, dosages, and administration methods

      Highly alcohol-preferring strain, C57BL/6NCrl; alcohol-preferring strain, C57BL/6J; alcohol-resistant strain, BALB/c, DBA/2.

      HCC, hepatocellular carcinoma; DEN, diethylnitrosamine; CCl4, carbon tetrachloride; HCV, hepatitis C virus; EtOH, ethanol; IP, intraperitoneal; PO, oral administration.

      Table 6. Comparative analysis of HBT with and without the SB transposon

      HBT, hydrodynamics-based transfection; SB, Sleeping Beauty.

      Table 7. Comparison of HCC cell line injections and tissue transplantation in an orthotopic HCC mouse model

      HCC, hepatocellular carcinoma.

      Table 8. Comprehensive summary of the advantages, limitations, and experimental applications of mouse models for investigating HCC

      HCC, hepatocellular carcinoma; DEN, diethylnitrosamine; CCl4, carbon tetrachloride; STZ, streptozotocin; HFD, high-fat diet; MASLD, metabolic dysfunction-associated steatotic liver disease; MASH, metabolic dysfunction-associated hepatitis; HBT, hydrodynamics-based transfection; SB, Sleeping Beauty.


      JLC : Journal of Liver Cancer
      TOP