The Ewha Medical Journal
Ewha Womans University College of Medicine and Ewha Medical Research Institute
Review

The histopathological and molecular heterogeneity of hepatocellular carcinoma: a narrative review

Wonju Chung1https://orcid.org/0009-0006-8239-9968, Haeryoung Kim1,*https://orcid.org/0000-0002-4205-9081
1Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
*Corresponding author: Haeryoung Kim, Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, 103 Daehak-no, Jongno-gu, Seoul 03080, Korea, E-mail: haeryoung.kim@snu.ac.kr

© Copyright 2024 Ewha Womans University College of Medicine and Ewha Medical Research Institute. 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.

Received: Aug 19, 2024; Accepted: Sep 26, 2024

Published Online: Oct 31, 2024

Abstract

Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related deaths worldwide, with poor clinical outcomes due to challenges in early detection and limited efficacy of current treatments such as receptor tyrosine kinase inhibitors and immunotherapy. HCC exhibits significant heterogeneity at both histopathological and molecular levels, complicating its management but offering potential for personalized therapeutic approaches. This review outlines the morpho-molecular heterogeneity of HCC and summarizes various histological subtypes, including steatohepatitic, clear cell, macrotrabecular-massive, scirrhous, lymphocyte-rich, and fibrolamellar HCCs. Each subtype possesses distinct clinical, histological, and molecular features; for instance, steatohepatitic HCC is associated with metabolic dysfunction and shows IL-6/JAK/STAT activation, while clear cell HCCs often have IDH1 mutations and favorable prognosis. The macrotrabecular-massive subtype is linked to poor outcomes and TP53 mutations, whereas scirrhous HCCs express stemness markers and have TSC1/TSC2 mutations. Lymphocyte-rich HCCs are characterized by immune cell infiltration and better prognosis. CTNNB1-mutated HCCs show specific morphological features and may benefit from targeted therapies. Understanding these subtypes and associated molecular alterations is crucial for developing effective diagnostic and therapeutic strategies, including potential predictive biomarkers and personalized treatments. Additionally, the identification of patterns like vessels-encapsulating-tumor-clusters offers prognostic implications and may guide therapeutic decisions. Recent molecular studies have enhanced our comprehension of HCC heterogeneity, laying the groundwork for more personalized approaches. Pathologists play a vital role in recognizing these subtypes, aiding in prognosis prediction and treatment planning. Advances in digital pathology and artificial intelligence may further facilitate biomarker research, ultimately improving patient outcomes in HCC management.

Keywords: Carcinoma; hepatocellular; Liver; Pathology

Introduction

Hepatocellular carcinoma (HCC) is the most common primary liver cancer, accounting for 75%–80% of primary liver malignancies [1]. HCC mostly develops in the background of chronic liver disease, the most common etiologies being HBV or HCV infection, and chronic alcohol abuse. During the recent year, metabolic syndrome has become another major risk factor for HCC, even in Asian countries [2,3].

The clinical outcome of patients with HCC is generally poor, mainly due to difficulties in early detection and limited treatment options for advanced disease [4,5]. Currently, systemic therapy, such as receptor tyrosine kinase inhibitors and immunotherapy, is the treatment of choice for the patients with unresectable HCC [5]. However, the efficacy of systemic therapy in HCC is still unsatisfactory with survival benefit of 1–3 months, objective response rate less than 30%, and a high incidence of adverse events [6,7]. In this context, understanding the heterogeneity of HCC will play a key role in developing effective diagnostic and therapeutic strategies, by offering potential predictive biomarkers and personalized approaches for HCC management.

Ethics statement

It is a literature database-based review; therefore, neither approval by the institutional review board nor obtainment of informed consent was required.

Morpho-molecular heterogeneity of hepatocellular carcinoma

HCC is typically composed of tumor cells showing hepatocytic differentiation with variable degrees of atypia [8]. While HCC recapitulates the cytoarchitectural morphology of the liver to varying extents, there are alterations in the hepatic microarchitecture such as loss of portal tracts, reduction or loss of the reticulin framework, and thickening of the hepatic plates. Neoangiogenesis occurs in HCCs, in the form of sinusoidal capillarization and unpaired arteries [8]. These changes also result in the characteristic imaging features, such as the early enhancement on contrast enhanced CT/MRI.

However, there is considerable heterogeneity of HCC, in the microscopic growth patterns, cytological features, and histological grade. About 50% of HCCs demonstrate mixed patterns of growth: trabecular, pseudoglandular, solid, and macrotrabecular (Fig. 1) [9]. In addition, while most HCCs demonstrate cytological features that recapitulate those of normal hepatocytes (i.e. polygonal cells with abundant eosinophilic cytoplasm), some HCCs show extensive areas with clear cell change, fatty change, and cholestasis. Cytoplasmic inclusions (e.g., hyaline bodies, Mallory-Denk bodies, and pale bodies) may also be seen in some tumor cells [8]. Histological grading is currently performed according to either the four-tiered modified Edmondson and Steiner system or the three-tiered World Health Organization (WHO) grading system [911].

emj-47-4-54-g1
Fig. 1. Microscopic features of a typical hepatocellular carcinoma (HCC). (A) Non-neoplastic hepatocytes (lower half) and HCC tumor cells (upper half) are separated by a fibrous capsule. (B) Trabecular pattern HCC with steatosis. (C) Pseudoglandular pattern HCC with cholestasis (Hematoxylin-Eosin stain, original magnification ×100 [A,B], ×200 [C]).
Download Original Figure

Recent advances in genomic techniques have unraveled the heterogeneity in the mutational landscape of HCC [12,13]. The most frequently mutated genes include TERT promoter, TP53, CTNNB1, ARID1A, ARID2, JAK1, ALB, AXIN1, NFE2L2, and RPS6KA3 [12]. In addition, gene expression profiling studies have suggested several molecular subclasses of HCC that correlate with the clinicopathological features, providing the foundation for an integrated morphological-molecular classification of HCC [14]. In the past two decades, there have been many efforts to establish a subclassification system that better categorizes HCCs with distinct clinical, histological, and molecular features (Table 1) [13,1519]. HCC can be subclassified into two major groups, the proliferative class and the non-proliferative class. The proliferative class is characterized by high chromosomal instability and TP53 mutations, and is associated with poor histological differentiation, frequent vascular invasion, increased alpha-fetoprotein (AFP) level, and overall poor clinical outcome [16,18]. On the other hand, the non-proliferative class displays chromosomal stability and a well-differentiated phenotype with less frequent vascular invasion [16,18]. CTNNB1-mutated HCCs belong to the latter group: these demonstrate frequent cholestasis and less immune cell infiltration on histology [14,20].

Table 1. Integrated morphological-molecular classification of hepatocellular carcinoma
Classification system Proliferative class Non-proliferative class
Lee et al. [15] Cluster A Cluster B
Boyault et al. [16] G1, G2, G3 G4, G5, G6
Chiang et al. [17] Proliferation Interferon, Poly7, CTNNB1
Hoshida et al. [18] S1, S2 S3
TCGA Research Network [13] Immune high and intermediate Immune excluded
Sia et al. [19] iClust1, iClust3 iClust2
Clinical features Poor clinical outcome, high AFP, HBV, frequent vascular invasion Improved clinical outcome, low AFP, HCV, low vascular invasion
Histological features Poorly differentiated Well differentiated
Molecular alterations Chromosomal instability, TP53 mutations, FGF19 amplification Chromosomal stability, CTNNB1 mutations, TERT promoter mutations

AFP, alpha-fetoprotein.

Download Excel Table

Currently, approximately 35% of HCC can be further subclassified into histological subtypes with distinct morphological, clinicopathological and molecular characteristics [9]. The following section will summarize the clinicopathological and molecular features of these different subtypes.

Steatohepatitic hepatocellular carcinoma

The steatohepatitic subtype of HCC demonstrates the key histological features of non-neoplastic steatohepatitis, including steatosis, pericellular fibrosis, cell ballooning, inflammation, and Mallory-Denk bodies, and these features occupy a major portion (>50%) of the tumor (Fig. 2) [21]. This subtype has been more frequently identified in patients with underlying metabolic dysfunction-associated steatotic liver disease and alcohol abuse, and its relative frequency has been reported to be between 5% and 20% [3,21]. Steatohepatitic HCC has been associated with less frequent vascular invasion and satellite nodules; however, its prognosis appears to be similar to that of conventional HCC [14]. Key molecular alterations associated with steatohepatitic HCC include IL-6/JAK/STAT activation, while CTNNB1, TERT promoter and TP53 mutations have been found to be less frequent in these tumors [14].

emj-47-4-54-g2
Fig. 2. Steatohepatitic hepatocellular carcinoma (HCC). (A) The tumor demonstrates a yellow hue on macroscopy reflecting the lipid component. (B) High power magnification showing the diffuse steatosis of tumor cells, tumor cell ballooning (arrow), some inflammatory cells, and pericellular fibrosis (Hematoxylin-Eosin stain, original magnification ×400).
Download Original Figure

Clear cell hepatocellular carcinoma

By definition, in clear cell HCCs, more than 80% of tumor cells demonstrate abundant clear cytoplasm (Fig. 3). The clear cytoplasm is a result of glycogen accumulation; however, some tumor cells may appear clear due to lipid droplets, and some degree of steatosis is acceptable for this diagnosis [22]. The relative frequency of clear cell HCC has been estimated to be around 3%–7%. Clear cell HCCs are generally well-differentiated tumors with a favorable prognosis [23]. One study has reported that clear cell HCCs demonstrate higher frequency of IDH1 mutation, although this mutation is not sufficient to define the subtype [24].

emj-47-4-54-g3
Fig. 3. Clear cell hepatocellular carcinoma (HCC). Most of the tumor cells demonstrate clear cytoplasm due to glycogen accumulation (Hematoxylin-Eosin stain, original magnification ×200).
Download Original Figure

Macrotrabecular-massive hepatocellular carcinoma

The macrotrabecular-massive subtype of HCC is an HCC in which more than 50% of the tumor cells assume a macrotrabecular growth pattern, defined as large trabeculae that are more than 6–10 cells thick (Fig. 4) [25]. This subtype accounts for approximately 5% of all HCCs and has been strongly associated with elevated serum AFP levels, high-grade cytological atypia, extensive lymphovascular invasion, more frequent distant metastasis, and a poor prognosis [25,26]. In addition, the vessels-encapsulating-tumor-clusters (VETC) pattern of neoangiogenesis, which has been associated with metastatic dissemination of HCC, is often enriched in this subtype (Fig. 4) [27,28]. TP53 mutations and FGF19 amplifications have been more frequently identified in the macrotrabecular-massive subtype of HCC [14].

emj-47-4-54-g4
Fig. 4. Macrotrabecular-massive hepatocellular carcinoma (HCC; A) and vessels-encapsulating-tumor clusters (VETC) pattern (B). (A) Macrotrabecular-massive HCC demonstrating thick tumor cell trabeculae, of more than 10-cell thickness (Hematoxylin-Eosin stain, original magnification ×100). (B) CD34 immunostain highlighting the VETC pattern, where the CD34-positive endothelial cells completely surround tumor cell clusters.
Download Original Figure

Scirrhous hepatocellular carcinoma

This subtype is characterized by dense intratumoral fibrous stroma (Fig. 5). The scirrhous subtype has a relative frequency of 4% and often mimics intrahepatic cholangiocarcinoma on imaging [29]. Expression of immunohistochemical markers associated with stemness (e.g., cytokeratin ([CK]) 7, CK19, and epithelial cell adhesion molecule) is often seen in scirrhous HCCs, and increased expression of cholangiocarcinoma-like and stem-cell-like genes have been identified by gene expression profiling, consistent with the intermediate characteristic of this subtype [30,31]. Furthermore, scirrhous HCC is associated with frequent TSC1/TSC2 mutations and transforming growth factor-β signaling activation [14,30].

emj-47-4-54-g5
Fig. 5. Scirrhous hepatocellular carcinoma (HCC). (A) The tumor appears as a firm, yellowish-white and lobulated mass on gross examination, mimicking an intrahepatic cholangiocarcinoma. (B) Dense intratumoral fibrosis is evident at low power magnification (Hematoxylin-Eosin stain, original magnification ×40). (C) Immunohistochemical expression of cytokeratin 19 is seen in a few tumor cells.
Download Original Figure

Lymphocyte-rich hepatocellular carcinoma

The lymphocyte-rich subtype demonstrates massive intratumoral infiltration of lymphocytes, which outnumber the tumor cells in most microscopic fields. This subtype is rare, accounting for less than 1% of all HCCs, but has received much attention as it has been associated with a favorable clinical outcome [32]. The lymphocyte-rich subtype is associated with increased programmed death-ligand 1 expression and focal amplification of chromosome 11q13.3, which is related to the immune checkpoint signature (CD274, PDCD1, BTLA, CTLA4, HAVCR2, IDO1, and LAG3) [3235]. Interestingly, although this subtype is also known as “lymphoepithelioma-like HCC”, it is not associated with Epstein-Barr virus infection, unlike the lymphoepithelioma-like tumors arising in other organs, such as the nasopharynx and stomach [32].

Fibrolamellar hepatocellular carcinoma

Fibrolamellar carcinoma, or fibrolamellar HCC, consists of strands of large eosinophilic tumor cells with abundant cytoplasm and prominent nucleoli, and separated by dense intratumoral bands of fibrosis [36]. Fibrolamellar carcinoma accounts for approximately 1% of all HCC, occurs in younger patients (median age of 25 years), and the background liver is non-cirrhotic [37]. The prognosis of fibrolamellar carcinoma appears to be better than that of conventional HCC arising in cirrhotic livers, but similar to that of HCC in non-cirrhotic livers [37]. DNAJB1-PRKACA gene fusion has been identified in >95% of cases, and fluorescence in situ hybridization for PRKACA gene rearrangement is a useful ancillary test in confirming the diagnosis [38]. Expression of CK7 and CD68 in the tumor cells is another characteristic of fibrolamellar carcinoma [38].

CTNNB1-mutated hepatocellular carcinoma

CTNNB1 mutations have been reported in approximately 20%–40% of HCCs [39]. CTNNB1 encodes β-catenin, which plays a key role in the WNT signaling pathway that regulates liver function and zonation [40]. In addition, bile salt transporter expression is dysregulated in these tumors, histologically manifested by frequent intratumoral cholestasis. Some CTNNB1-mutated HCCs may be diagnosed by gadoxetic acid-enhanced MRI, due to the upregulation of the organic anion transporting polypeptide 1B3 (OATP1B3) [41]. Histologically, CTNNB1-mutated HCCs are typically well-differentiated tumors with microtrabecular and/or pseudoglandular growth patterns, intratumoral cholestasis, and less immune cell infiltration compared to non-CTNNB1-mutated HCCs [14,20]. However, CTNNB1-mutated HCCs are not morphologically homogeneous, with approximately 40% not demonstrating the “classic CTNNB1 morphology” [42]. Immunohistochemical studies for β-catenin (nuclear expression) and glutamine synthetase (diffuse, strong and homogeneous expression) may serve as useful surrogate markers for CTNNB1 mutation.

Hepatocellular carcinoma with stemness-related marker expression

HCC with stemness-related marker expression, or progenitor HCC, is defined as HCC expressing stemness-related markers, e.g., CK19, in >5% of the tumor cells [15]. This subset of HCCs differ from combined hepatocellular-cholangiocarcinoma, as they are morphologically compatible with HCC, and there is no evidence of glandular differentiation or mucin production in these tumors. They are associated with increased serum AFP levels, frequent vascular invasion, poor histological differentiation, high recurrence rate, resistance to systemic chemotherapy and locoregional treatment, and overall poor prognosis [43]. HCCs with stemness-related marker expression more frequently demonstrate TP53 mutations and chromosomal instability, and increased PD-L1 expression [33,34].

Hepatocellular carcinomas with vessels-encapsulating-tumor-clusters pattern

The VETC phenotype is defined by the presence of VETC pattern in more than 55% of the tumor area, characterized by CD34-positive vessels that encapsulate and isolate individual tumor clusters, forming a cobweb-like pattern (Fig. 4) [27,28,44]. The VETC pattern is often found in the macrotrabecular-massive subtype of HCC (7.8%) and is associated with aggressive behavior and metastatic dissemination [27,28]. It has been reported that VETC pattern is related to a novel mechanism of metastasis, independent of epithelial-to-mesenchymal transition [44]. Furthermore, patients with VETC-positive HCC have shown greater survival benefits from sorafenib therapy compared to those with VETC-negative HCC, suggesting that the VETC pattern may serve as a potential predictive marker for sorafenib response [28]. Correlation between the VETC pattern on histology and a rim arterial phase hyperenhancement in arterial phase imaging suggests a role for imaging in the prognostication of HCC [45].

Other rare histological subtypes of HCC have been described. The chromophobe subtype of HCC has tumor cells with clear to pale cytoplasm and mainly bland nuclei with focal areas of striking nuclear atypia [46]. Chromophobe subtype is strongly associated with alternative lengthening of telomeres, a telomerase-independent mechanism of telomere maintenance, which can be detected by fluorescence in situ hybridization [46]. Its prognosis is currently known to be similar to that of conventional HCC [47]. Neutrophil-rich HCC is characterized by marked intratumoral neutrophilic infiltration, granulocyte colony-stimulating factor production by tumor cells, and a poor prognosis [47]. The tumor cells are often poorly differentiated, and focal sarcomatoid differentiation can be observed [48].

Conclusion

Recent molecular studies have significantly enhanced our understanding of the morphological and molecular heterogeneity of HCC, providing the foundation for more effective and personalized treatment strategies. Pathologists are becoming increasingly aware of the histomorphological heterogeneity of HCC, and the specification of the various subtypes of HCC has helped pathologists understand the histology of HCC in more detail and the various differential diagnoses and diagnostic pitfalls for each variant. The correlation between the histomorphology and the molecular and biological features suggests the role of histology in the prediction of therapeutic response and prognosis. This may be further facilitated by the recent advances in digital pathology and artificial intelligence-based biomarker research.

Authors' contributions

Project administration: not applicable

Conceptualization: Chung W, Kim H

Methodology & data curation: not applicable

Funding acquisition: Kim H

Writing – original draft: Chung W

Writing – review & editing: Chung W, Kim H

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Funding

This was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NRF-2022R1A2C2010348).

Data availability

Not applicable.

Acknowledgments

Not applicable.

Supplementary materials

Not applicable.

References

1.

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6):394-424

2.

Kim DY. Changing etiology and epidemiology of hepatocellular carcinoma: Asia and worldwide. J Liver Cancer 2024; 24(1):62-70

3.

Daher D, Dahan KSE, Singal AG. Non-alcoholic fatty liver disease-related hepatocellular carcinoma. J Liver Cancer 2023; 23(1):127-142

4.

Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet 2012; 379(9822):1245-1255

5.

Gordan JD, Kennedy EB, Abou-Alfa GK, Beg MS, Brower ST, Gade TP, et al. Systemic therapy for advanced hepatocellular carcinoma: ASCO guideline. J Clin Oncol 2020; 38(36):4317-4345

6.

Doycheva I, Thuluvath PJ. Systemic therapy for advanced hepatocellular carcinoma: an update of a rapidly evolving field. J Clin Exp Hepatol 2019; 9(5):588-596

7.

Song YG, Yoo JJ, Kim SG, Kim YS. Complications of immunotherapy in advanced hepatocellular carcinoma. J Liver Cancer 2024; 24(1):9-16

8.

Burt AD, Ferrell LD, Hübscher SG. MacSween's pathology of the liver. Amsterdam: Elsevier Health Sciences. 2022.

9.

Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, et al. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020; 76(2):182-188

10.

Edmondson HA, Steiner PE. Primary carcinoma of the liver: a study of 100 cases among 48,900 necropsies. Cancer 1954; 7(3):462-503

11.

Martins-Filho SN, Paiva C, Azevedo RS, Alves VAF. Histological grading of hepatocellular carcinoma: a systematic review of literature. Front Med 2017; 4:193

12.

Lee JS. The mutational landscape of hepatocellular carcinoma. Clin Mol Hepatol 2015; 21(3):220-229

13.

The Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 2017; 169(7):1327-1341.E23.

14.

Calderaro J, Couchy G, Imbeaud S, Amaddeo G, Letouzé E, Blanc JF, et al. Histological subtypes of hepatocellular carcinoma are related to gene mutations and molecular tumour classification. J Hepatol 2017; 67(4):727-738

15.

Lee JS, Heo J, Libbrecht L, Chu IS, Kaposi-Novak P, Calvisi DF, et al. A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progenitor cells. Nat Med 2006; 12(4):410-416

16.

Boyault S, Rickman DS, de Reyniès A, Balabaud C, Rebouissou S, Jeannot E, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 2007; 45(1):42-52

17.

Chiang DY, Villanueva A, Hoshida Y, Peix J, Newell P, Minguez B, et al. Focal gains of VEGFA and molecular classification of hepatocellular carcinoma. Cancer Res 2008; 68(16):6779-6788

18.

Hoshida Y, Nijman SMB, Kobayashi M, Chan JA, Brunet JP, Chiang DY, et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res 2009; 69(18):7385-7392

19.

Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C, Castro de Moura M, et al. Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology 2017; 153(3):812-826

20.

Audard V, Grimber G, Elie C, Radenen B, Audebourg A, Letourneur F, et al. Cholestasis is a marker for hepatocellular carcinomas displaying β-catenin mutations. J Pathol 2007; 212(3):345-352

21.

Salomao M, Yu WM, Brown RS, Emond JC, Lefkowitch JH. Steatohepatitic hepatocellular carcinoma (SH-HCC): a distinctive histological variant of HCC in hepatitis C virus-related cirrhosis with associated NAFLD/NASH. Am J Surg Pathol 2010; 34(11):1630-1636

22.

Bannasch P, Ribback S, Su Q, Mayer D. Clear cell hepatocellular carcinoma: origin, metabolic traits and fate of glycogenotic clear and ground glass cells. Hepatobiliary Pancreatic Dis Int 2017; 16(6):570-594

23.

Li T, Fan J, Qin LX, Zhou J, Sun HC, Qiu SJ, et al. Risk factors, prognosis, and management of early and late intrahepatic recurrence after resection of primary clear cell carcinoma of the liver. Ann Surg Oncol 2011; 18(7):1955-1963

24.

Lee JH, Shin DH, Park WY, Shin N, Kim A, Lee HJ, et al. IDH1 R132C mutation is detected in clear cell hepatocellular carcinoma by pyrosequencing. World J Surg Oncol 2017; 15(1):82

25.

Jeon Y, Benedict M, Taddei T, Jain D, Zhang X. Macrotrabecular hepatocellular carcinoma: an aggressive subtype of hepatocellular carcinoma. Am J Surg Pathol 2019; 43(7):943-948

26.

Kumar D, Hafez O, Jain D, Zhang X. Can primary hepatocellular carcinoma histomorphology predict extrahepatic metastasis?. Hum Pathol 2021; 113:39-46

27.

Renne SL, Woo HY, Allegra S, Rudini N, Yano H, Donadon M, et al. Vessels encapsulating tumor clusters (VETC) is a powerful predictor of aggressive hepatocellular carcinoma. Hepatology 2020; 71(1):183-195

28.

Fang JH, Xu L, Shang LR, Pan CZ, Ding J, Tang YQ, et al. Vessels that encapsulate tumor clusters (VETC) pattern is a predictor of sorafenib benefit in patients with hepatocellular carcinoma. Hepatology 2019; 70(3):824-839

29.

Kurogi M, Nakashima O, Miyaaki H, Fujimoto M, Kojiro M. Clinicopathological study of scirrhous hepatocellular carcinoma. J Gastroenterol Hepatol 2006; 21(9):1470-1477

30.

Seok JY, Na DC, Woo HG, Roncalli M, Kwon SM, Yoo JE, et al. A fibrous stromal component in hepatocellular carcinoma reveals a cholangiocarcinoma-like gene expression trait and epithelial-mesenchymal transition. Hepatology 2012; 55(6):1776-1786

31.

Matsuura S, Aishima S, Taguchi K, Asayama Y, Terashi T, Honda H, et al. 'Scirrhous' type hepatocellular carcinomas: a special reference to expression of cytokeratin 7 and hepatocyte paraffin 1. Histopathology 2005; 47(4):382-390

32.

Chan AWH, Tong JHM, Pan Y, Chan SL, Wong GLH, Wong VWS, et al. Lymphoepithelioma-like hepatocellular carcinoma: an uncommon variant of hepatocellular carcinoma with favorable outcome. Am J Surg Pathol 2015; 39(3):304-312

33.

Calderaro J, Rousseau B, Amaddeo G, Mercey M, Charpy C, Costentin C, et al. Programmed death ligand 1 expression in hepatocellular carcinoma: relationship with clinical and pathological features. Hepatology 2016; 64(6):2038-2046

34.

Nishida N, Sakai K, Morita M, Aoki T, Takita M, Hagiwara S, et al. Association between genetic and immunological background of hepatocellular carcinoma and expression of programmed cell death-1. Liver Cancer 2020; 9(4):426-439

35.

Chan AWH, Zhang Z, Chong CCN, Tin EKY, Chow C, Wong N. Genomic landscape of lymphoepithelioma-like hepatocellular carcinoma. J Pathol 2019; 249(2):166-172

36.

Torbenson M. Fibrolamellar carcinoma: 2012 update. Scientifica 2012; 2012:743790

37.

El-Serag HB, Davila JA. Is fibrolamellar carcinoma different from hepatocellular carcinoma? A US population-based study. Hepatology 2004; 39(3):798-803

38.

Graham RP, Yeh MM, Lam-Himlin D, Roberts LR, Terracciano L, Cruise MW, et al. Molecular testing for the clinical diagnosis of fibrolamellar carcinoma. Mod Pathol 2018; 31(1):141-149

39.

Gougelet A, Torre C, Veber P, Sartor C, Bachelot L, Denechaud PD, et al. T-cell factor 4 and β-catenin chromatin occupancies pattern zonal liver metabolism in mice. Hepatology 2014; 59(6):2344-2357

40.

Monga SP. β-Catenin signaling and roles in liver homeostasis, injury, and tumorigenesis. Gastroenterology 2015; 148(7):1294-1310

41.

Ueno A, Masugi Y, Yamazaki K, Komuta M, Effendi K, Tanami Y, et al. OATP1B3 expression is strongly associated with Wnt/β-catenin signalling and represents the transporter of gadoxetic acid in hepatocellular carcinoma. J Hepatol 2014; 61(5):1080-1087

42.

Torbenson M, McCabe CE, O'Brien DR, Yin J, Bainter T, Tran NH, et al. Morphological heterogeneity in beta-catenin–mutated hepatocellular carcinomas: implications for tumor molecular classification. Hum Pathol 2022; 119:15-27

43.

Rhee H, Kim H, Park YN. Clinico-radio-pathological and molecular features of hepatocellular carcinomas with Keratin 19 expression. Liver Cancer 2020; 9(6):663-681

44.

Fang JH, Zhou HC, Zhang C, Shang LR, Zhang L, Xu J, et al. A novel vascular pattern promotes metastasis of hepatocellular carcinoma in an epithelial–mesenchymal transition–independent manner. Hepatology 2015; 62(2):452-465

45.

Hwang SH, Rhee H. Radiologic features of hepatocellular carcinoma related to prognosis. J Liver Cancer 2023; 23(1):143-156

46.

Wood LD, Heaphy CM, Daniel HDJ, Naini BV, Lassman CR, Arroyo MR, et al. Chromophobe hepatocellular carcinoma with abrupt anaplasia: a proposal for a new subtype of hepatocellular carcinoma with unique morphological and molecular features. Mod Pathol 2013; 26(12):1586-1593

47.

Torbenson MS. Hepatocellular carcinoma: making sense of morphological heterogeneity, growth patterns, and subtypes. Hum Pathol 2021; 112:86-101

48.

Amano H, Itamoto T, Emoto K, Hino H, Asahara T, Shimamoto F. Granulocyte colony-stimulating factor-producing combined hepatocellular/cholangiocellular carcinoma with sarcomatous change. J Gastroenterol 2005; 40(12):1158-1159