1
|
Zhang T, Niu N, Taddei T, Jain D, Zhang X. Clinicopathologic features and prognosis of steatohepatitic hepatocellular carcinoma based on varying cutoffs of tumoral steatohepatitic changes. Am J Clin Pathol 2025; 163:411-418. [PMID: 39418121 DOI: 10.1093/ajcp/aqae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVES Steatohepatitic hepatocellular carcinoma (SH-HCC) is currently recognized as a distinct histologic subtype of HCC. The prognosis and specific criteria for determining the amount of steatohepatitis required to define SH-HCC are still unclear. METHODS After excluding all recognized HCC subtypes from 505 HCC cases (2010-2019), the remaining cases were categorized as conventional HCC (CV-HCC) (n = 223). The cases classified as SH-HCC (n = 171) were further divided into groups based on the percentage of steatohepatitis: 5% or more, 30% or more, and 50% or more. RESULTS Hepatitis C virus infection was the predominant underlying liver disease in both the CV-HCC and SH-HCC groups. Metabolic dysfunction-associated steatotic liver disease (formerly nonalcoholic fatty liver disease) was more prevalent in all cases of SH-HCC with different steatohepatitic cutoffs than in cases of CV-HCC. There were no differences in the stage of fibrosis of the background liver between the CV-HCC and SH-HCC groups. SH-HCC with different cutoffs exhibited a notable increase in the presence of glycogenated nuclei, Mallory-Denk bodies, and hyaline globules in tumor cells. Survival analysis did not reveal substantial differences in overall survival between the CV-HCC and SH-HCC groups and among patients with SH-HCC with different steatohepatitis cutoffs. CONCLUSIONS The degree of intratumoral steatohepatitis in patients with SH-HCC does not appear to be a notable prognostic factor. The presence of steatohepatitis in the tumor is better recognized as 1 of the histopathologic patterns of HCC.
Collapse
Affiliation(s)
| | | | - Tamar Taddei
- Section of Digestive Diseases. Yale University School of Medicine. New Haven, CT, US
| | | | | |
Collapse
|
2
|
Guest RV, Goeppert B, Nault JC, Sia D. Morphomolecular Pathology and Genomic Insights into the Cells of Origin of Cholangiocarcinoma and Combined Hepatocellular-Cholangiocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:345-361. [PMID: 39341365 PMCID: PMC11841493 DOI: 10.1016/j.ajpath.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024]
Abstract
Cholangiocarcinomas are a highly heterogeneous group of malignancies that, despite recent progress in the understanding of their molecular pathogenesis and clinical management, continue to pose a major challenge to public health. The traditional view posits that cholangiocarcinomas derive from the neoplastic transformation of cholangiocytes lining the biliary tree. However, increasing genetic and experimental evidence has recently pointed to a more complex, and nuanced, scenario for the potential cell of origin of cholangiocarcinomas. Hepatocytes as well as hepatic stem/progenitor cells are being considered as additional potential sources, depending on microenvironmental contexts, including liver injury. The hypothesis of potentially diverse cells of origin for cholangiocarcinoma, albeit controversial, is certainly not surprising given the plasticity of the cells populating the liver as well as the existence of liver cancer subtypes with mixed histologic and molecular features. This review carefully examines the current pathologic, genomic, and experimental evidence supporting the existence of multiple cells of origin of liver and biliary tract cancers, with particular focus on cholangiocarcinoma and combined hepatocellular-cholangiocarcinoma.
Collapse
Affiliation(s)
- Rachel V Guest
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Benjamin Goeppert
- Institute of Pathology, RKH Klinikum Ludwigsburg, Ludwigsburg, Germany; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jean-Charles Nault
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Team "Functional Genomics of Solid Tumors", Equipe labellisée Ligue Nationale Contre le Cancer, Labex OncoImmunology, Paris, France; Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, Bobigny, France
| | - Daniela Sia
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
3
|
Andrade R, Perez-Rojas J, da Silva SG, Miskinyte M, Quaresma MC, Frazão LP, Peixoto C, Cubells A, Montalvá EM, Figueiredo A, Cipriano A, Gonçalves-Reis M, Proença D, Folgado A, Pereira-Leal JB, Oliveira RC, Pinto-Marques H, Tralhão JG, Berenguer M, Cardoso J. HepatoPredict Accurately Selects Hepatocellular Carcinoma Patients for Liver Transplantation Regardless of Tumor Heterogeneity. Cancers (Basel) 2025; 17:500. [PMID: 39941867 PMCID: PMC11816190 DOI: 10.3390/cancers17030500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths rising worldwide. This is leading to an increased demand for liver transplantation (LT), the most effective treatment for HCC in its initial stages. However, current patient selection criteria are limited in predicting recurrence and raise ethical concerns about equitable access to care. This study aims to enhance patient selection by refining the HepatoPredict (HP) tool, a machine learning-based model that combines molecular and clinical data to forecast LT outcomes. METHODS The updated HP algorithm was trained on a two-center dataset and assessed against standard clinical criteria. Its prognostic performance was evaluated through accuracy metrics, with additional analyses considering tumor heterogeneity and potential sampling bias. RESULTS HP outperformed all clinical criteria, particularly regarding negative predictive value, addressing critical limitations in existing selection strategies. It also demonstrated improved differentiation of recurrence-free and overall survival outcomes. Importantly, the prognostic accuracy of HP remained largely unaffected by intra-nodule and intra-patient heterogeneity, indicating its robustness even when biopsies were taken from smaller or non-dominant nodules. CONCLUSIONS These findings support the usage of HP as a valuable tool for optimizing LT candidate selection, promoting fair organ allocation and enhancing patient outcomes through integrated analysis of molecular and clinical data.
Collapse
Affiliation(s)
- Rita Andrade
- Surgery Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal; (R.A.); (J.G.T.)
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal;
| | - Judith Perez-Rojas
- Pathology Service, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.C.); (E.M.M.); (M.B.)
- Instituto de Investigación Sanitaria La Fe (ISS La Fe), 46026 Valencia, Spain
| | - Sílvia Gomes da Silva
- Hepato-Biliary-Pancreatic and Transplantation Centre, Hospital Curry Cabral, Unidade Local de Saúde de São José, 1069-166 Lisbon, Portugal; (S.G.d.S.)
- NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Migla Miskinyte
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - Margarida C. Quaresma
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - Laura P. Frazão
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - Carolina Peixoto
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - Almudena Cubells
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.C.); (E.M.M.); (M.B.)
- Instituto de Investigación Sanitaria La Fe (ISS La Fe), 46026 Valencia, Spain
- Hepatology Unit, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Eva M. Montalvá
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.C.); (E.M.M.); (M.B.)
- Instituto de Investigación Sanitaria La Fe (ISS La Fe), 46026 Valencia, Spain
- Liver Transplantation and Surgery Unit, Hospital Universitari I Politècnic La Fe, 46026 Valencia, Spain
- Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain
| | - António Figueiredo
- Pathology Service, Hospital Curry Cabral, Unidade Local de Saúde de São José, 1069-166 Lisbon, Portugal;
| | - Augusta Cipriano
- Pathology Department, Unidade Local de Saúde de Coimbra, 3004-561 Coimbra, Portugal;
| | - Maria Gonçalves-Reis
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - Daniela Proença
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - André Folgado
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - José B. Pereira-Leal
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| | - Rui Caetano Oliveira
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal;
- Pathology Department, Unidade Local de Saúde de Coimbra, 3004-561 Coimbra, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), 3000-548 Coimbra, Portugal
- Centro de Investigação em Meio Ambiente, Genética e Oncobiologia (CIMAGO), 3001-301 Coimbra, Portugal
- Centro Académico e Clínico (CAC), 3004-531 Coimbra, Portugal
| | - Hugo Pinto-Marques
- Hepato-Biliary-Pancreatic and Transplantation Centre, Hospital Curry Cabral, Unidade Local de Saúde de São José, 1069-166 Lisbon, Portugal; (S.G.d.S.)
- NOVA Medical School, 1169-056 Lisbon, Portugal
| | - José Guilherme Tralhão
- Surgery Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal; (R.A.); (J.G.T.)
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), 3000-548 Coimbra, Portugal
- Centro de Investigação em Meio Ambiente, Genética e Oncobiologia (CIMAGO), 3001-301 Coimbra, Portugal
- Centro Académico e Clínico (CAC), 3004-531 Coimbra, Portugal
| | - Marina Berenguer
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain; (A.C.); (E.M.M.); (M.B.)
- Instituto de Investigación Sanitaria La Fe (ISS La Fe), 46026 Valencia, Spain
- Hepatology Unit, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain
| | - Joana Cardoso
- Ophiomics Precision Medicine, 1600-514 Lisbon, Portugal; (M.M.); (M.C.Q.); (L.P.F.); (C.P.); (M.G.-R.); (D.P.); (A.F.); (J.B.P.-L.)
| |
Collapse
|
4
|
Yu J, Wu S, Gong J, Deng W, Xiao Z, Wu L, Long H. Prediction of Hepatocellular Carcinoma Prognosis and Immunotherapy Response Using Mitochondrial Dysregulation Features. J Cell Mol Med 2025; 29:e70389. [PMID: 39910622 PMCID: PMC11798730 DOI: 10.1111/jcmm.70389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major contributor to cancer-related deaths globally. Although there have been improvements in identifying treating the disease, patient outcomes are still unfavourable because of the significant variation in HCC. Mitochondrial-related genes (MRGs) are crucial in tumour metabolism, cell death and immune response, emerging as potential therapeutic targets. We analysed 2030 MRGs using TCGA, GEO and HCCDB18 databases. Differentially expressed genes were identified using edgeR and limma, and enrichment analysis was performed via the clusterProfiler package. A prognostic model was built using machine learning algorithms and evaluated using LOOCV. Immune infiltration was assessed with CIBERSORT, EPIC, MCPCounter and TIMER algorithms, and drug sensitivity was analysed using the CTRP and PRISM datasets. MRG expression levels are significantly associated with worse outcomes in HCC patients outperformed conventional clinical indicators in immune response revealed that individuals at high risk exhibited weaker immune responses, characterised by reduced immune scores, and elevated levels of CD8+ T cells and macrophages. Notably, high-risk patients also displayed heightened susceptibility to chemotherapy agents such as paclitaxel and irinotecan. Abnormal MRG expression serves as a significant biomarker for HCC prognosis. The developed model accurately predicts disease progression and can guide personalised treatment, especially for immune and chemotherapeutic therapies. Further validation with broader clinical samples is needed.
Collapse
Affiliation(s)
- Jia Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
| | - Shengli Wu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Jinglong Gong
- Department of Hepatobiliary Surgery, Hunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal UniversityChangshaHunanChina
| | - Wenbing Deng
- Department of Gastroenterology, The First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
| | - Zhongsheng Xiao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
| | - LiangLiang Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
| | - Hong Long
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
- Faculty of Data ScienceCity University of MacauMacau SARChina
| |
Collapse
|
5
|
Saler CHA, Shuai S, Beckervordersandforth JC, Rennspiess D, Roemen G, Gevers T, Stoehr‐Kleinegris MCF, Bouwense SAW, Dewulf MJL, Coolsen MME, Bemelmans MHA, Damink SWO, Winnepenninckx V, zur Hausen A, Kramer M, Samarska IV. Clinicopathological Study on Morphological Subtypes of Hepatocellular Carcinoma: A Single Tertiary Referral Center Experience. Cancer Rep (Hoboken) 2025; 8:e70127. [PMID: 39953652 PMCID: PMC11828739 DOI: 10.1002/cnr2.70127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/09/2024] [Accepted: 01/16/2025] [Indexed: 02/17/2025] Open
Abstract
AIM We aimed to analyze hepatocellular carcinoma (HCC) morphological subtypes characterized according to the WHO classification and the International Collaboration on Cancer Reporting (ICCR) recommendations, and their prognostic features in a Dutch population. METHODS AND RESULTS This retrospective study in a tertiary referral center included the histopathological revision of 62 HCC resection specimens, obtained from 22 female and 40 male patients (median age: 67 years), in a period between 2011 and 2021 at the Maastricht University Medical Center +. Clinical data, morphological subtypes, growth pattern (GP), tumor grade, tumor extension, margins, and vascular and perineural invasion were collected. Eighteen cases were assigned a specific morphologic subtype and steatohepatic HCC was the most common in our cohort. Twenty-one tumors classified as conventional type HCC (HCC-NOS), commonly exhibiting two concurrent GPs. Twenty-three cases revealed a heterogeneous morphologic differentiation, compromising the combination of HCC-NOS with another morphologic subtype, most frequently a steatohepatitic component. Comparison of HCC-NOS and HCC with heterogeneous morphology did not show significant differences in the main clinicopathological characteristics and survival. CONCLUSION Although the most common morphologic subtype was steatohepatitic HCC, the majority of cases demonstrated multiple morphologic patterns. In case of HCC-NOS, heterogeneous GPs were often observed. Therefore, a histomorphological diagnosis based on a single tumor biopsy specimen may lead to incorrect classification of HCC. Sufficient tumor sampling of HCC resection specimens is required for the complete evaluation of all histomorphological features followed by correct subclassification in order to meet the clinical needs regarding prognostic relevance and patient follow-up.
Collapse
Affiliation(s)
- C. H. A. Saler
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
- Department of Internal MedicineGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - S. Shuai
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - J. C. Beckervordersandforth
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - D. Rennspiess
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - G. Roemen
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - T. Gevers
- Department of Internal MedicineGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - M. C. F. Stoehr‐Kleinegris
- Department of Internal MedicineGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - S. A. W. Bouwense
- Department of SurgerySchool of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht UniversityMaastrichtthe Netherlands
| | - M. J. L. Dewulf
- Department of SurgerySchool of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht UniversityMaastrichtthe Netherlands
| | - M. M. E. Coolsen
- Department of SurgerySchool of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht UniversityMaastrichtthe Netherlands
| | - M. H. A. Bemelmans
- Department of SurgerySchool of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht UniversityMaastrichtthe Netherlands
| | - S. W. Olde Damink
- Department of SurgerySchool of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht UniversityMaastrichtthe Netherlands
| | - V. Winnepenninckx
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - A. zur Hausen
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - M. Kramer
- Department of Internal MedicineGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| | - I. V. Samarska
- Department of PathologyGROW‐School for Oncology and Reproduction, Maastricht University Medical Center +Maastrichtthe Netherlands
| |
Collapse
|
6
|
Benderski K, Schneider P, Kordeves P, Fichter M, Schunke J, De Lorenzi F, Durak F, Schrörs B, Akilli Ö, Kiessling F, Bros M, Diken M, Grabbe S, Schattenberg JM, Lammers T, Sofias AM, Kaps L. A hepatocellular carcinoma model with and without parenchymal liver damage that integrates technical and pathophysiological advantages for therapy testing. Pharmacol Res 2025; 211:107560. [PMID: 39730106 DOI: 10.1016/j.phrs.2024.107560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Hepatocellular Carcinoma (HCC) is the most common form of primary liver cancer, with cirrhosis being its strongest risk factor. Interestingly, an increasing number of HCC cases is also observed without cirrhosis. We developed an HCC model via intrasplenic injection of highly tumorigenic HCC cells, which, due to cellular tropism, invade the liver and allow for a controllable disease progression. Specifically, C57BL/6JRj mice were intrasplenically inoculated with Dt81Hepa1-6 HCC cells, with a subgroup pre-treated with CCl4 to induce cirrhosis (C-HCC). At four weeks post-inoculation, mice were sacrificed, and diseased livers were analyzed via histology, flow cytometry, and RT-qPCR to profile the extracellular matrix (ECM), angiogenesis, and immune cells. In addition, tumor-bearing mice were treated with the first-line therapy, AtezoBev, to assess therapeutic responsiveness of the model. Dt81Hepa1-6 cells displayed similar gene expression as human HCC. After intrasplenic injection, all mice developed multifocal disease. C-HCC mice had a significantly higher tumor load than non-cirrhotic HCC mice. Both HCC and C-HCC models displayed extensive ECM formation, increased levels of vascularization, and immune cell infiltration compared to healthy and non-cancerous cirrhotic livers. AtezoBev treatment produced robust antitumor efficacy, validating the model's suitability for therapy testing. In conclusion, we established a rapidly developing and high-yield HCC model through a simple intrasplenic injection, with or without cirrhotic damage. The model overexpressed key human HCC genes and showed high responsiveness to first-line treatment. Our model uniquely combines all the above-mentioned features, promoting its use towards HCC therapy testing.
Collapse
Affiliation(s)
- Karina Benderski
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Paul Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Panayiotis Kordeves
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Michael Fichter
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Jenny Schunke
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany
| | - Feyza Durak
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Barbara Schrörs
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Özlem Akilli
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Jörn M Schattenberg
- Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrberger Strasse 100, Saarbrücken 66123, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Leonard Kaps
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrberger Strasse 100, Saarbrücken 66123, Germany.
| |
Collapse
|
7
|
Yuan Y, Sun W, Xie J, Zhang Z, Luo J, Han X, Xiong Y, Yang Y, Zhang Y. RNA nanotherapeutics for hepatocellular carcinoma treatment. Theranostics 2025; 15:965-992. [PMID: 39776807 PMCID: PMC11700867 DOI: 10.7150/thno.102964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality worldwide, particularly due to the limited effectiveness of current therapeutic options for advanced-stage disease. The efficacy of traditional treatments is often compromised by the intricate liver microenvironment and the inherent heterogeneity. RNA-based therapeutics offer a promising alternative, utilizing the innovative approach of targeting aberrant molecular pathways and modulating the tumor microenvironment. The integration of nanotechnology in this field, through the development of advanced nanocarrier delivery systems, especially lipid nanoparticles (LNPs), polymer nanoparticles (PNPs), and bioinspired vectors, enhances the precision and efficacy of RNA therapies. This review highlights the significant progress in RNA nanotherapeutics for HCC treatment, covering micro RNA (miRNA), small interfering RNA (siRNA), message RNA (mRNA), and small activating RNA (saRNA) mediated gene silencing, therapeutic protein restoration, gene activation, cancer vaccines, and concurrent therapy. It further comprehensively discusses the prevailing challenges within this therapeutic landscape and provides a forward-looking perspective on the potential of RNA nanotherapeutics to transform HCC treatment.
Collapse
Affiliation(s)
- Yihang Yuan
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
- Department of General Surgery Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School Nanjing University, Nanjing 210008, China
| | - Weijie Sun
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Jiaqi Xie
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
| | - Ziheng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Jing Luo
- Department of Urology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiangfei Han
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yongfu Xiong
- Department of Hepatobiliary Surgery, Academician (Expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong 637600, China
| | - Yang Yang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
| | - Yang Zhang
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
8
|
Li X, Li L, He N, Kou D, Chen S, Song H, Yan X. Pathomics signatures and cuproptosis-related genes signatures for prediction of prognosis in patients with hepatocellular carcinoma. Transl Cancer Res 2024; 13:5473-5483. [PMID: 39525011 PMCID: PMC11543060 DOI: 10.21037/tcr-24-350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/14/2024] [Indexed: 11/16/2024]
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignant tumor with high heterogeneity and poor prognosis, so early prediction and treatment are still difficult. Cuproptosis is a newly discovered type of programmed cell death that has been shown to be closely related to the occurrence and progression of HCC. Cancer morphology is influenced by genetic drivers, and computational pathology methods typically use tissue images such as entire slide images as input to predict clinical or genetic features. Therefore, the comprehensive analysis of pathological features and genomic data provides a feasible way to explore the potential mechanism of the tumor. The objective of this study was to develop a prediction model for HCC prognosis based on the pathomics signatures (PS) and the genomics signatures (GS). Methods A dataset comprising 315 HCC patients was randomly divided into a training set (n=200) and a validation set (n=115). Prognostic models related to PS and GS were constructed by univariate and multivariate Cox regression analyses and least absolute shrinkage and selection operator (LASSO) regression analysis. Kaplan-Meier survival analysis, receiver operating characteristic (ROC) curve, univariate and multivariate Cox analyses, and nomogram were used to evaluate the predictive performance of the prognostic model. The prognostic value of the model was internally validated. Results A prognostic model incorporating clinical features, PS, and GS was developed using Cox regression analysis and LASSO regression analyses. Kaplan-Meier survival analysis revealed statistically significant differences in survival time between high-risk and low-risk subgroups in both the training and validation datasets (PS: P=0.003 and <0.001, respectively; GS: P=0.008 and 0.004, respectively). The time-dependent ROC curve showed favorable predictive value for survival in both the training and validation sets. The area under the ROC curves at 1, 3, and 5 years was 0.750, 0.830, and 0.870 in the training set, and 0.780, 0.810, and 0.760 in the validation set, respectively. A nomogram model based on the risk model score could effectively predict the survival probability of HCC patients. The calibration curves further demonstrated the good predictive capability of the nomogram model. Conclusions The prognostic model incorporating PS and GS could effectively predict the prognosis of HCC patients.
Collapse
Affiliation(s)
- Xiaoliang Li
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Lina Li
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Nan He
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Dan Kou
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Shizhao Chen
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Hui Song
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Xiang Yan
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
9
|
Safri F, Nguyen R, Zerehpooshnesfchi S, George J, Qiao L. Heterogeneity of hepatocellular carcinoma: from mechanisms to clinical implications. Cancer Gene Ther 2024; 31:1105-1112. [PMID: 38499648 PMCID: PMC11327108 DOI: 10.1038/s41417-024-00764-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common types of primary liver cancer. Current treatment options have limited efficacy against this malignancy, primarily owing to difficulties in early detection and the inherent resistance to existing drugs. Tumor heterogeneity is a pivotal factor contributing significantly to treatment resistance and recurrent manifestations of HCC. Intratumoral heterogeneity is an important aspect of the spectrum of complex tumor heterogeneity and contributes to late diagnosis and treatment failure. Therefore, it is crucial to thoroughly understand the molecular mechanisms of how tumor heterogeneity develops. This review aims to summarize the possible molecular dimensions of tumor heterogeneity with an emphasis on intratumoral heterogeneity, evaluate its profound impact on the diagnosis and therapeutic strategies for HCC, and explore the suitability of appropriate pre-clinical models that can be used to best study tumor heterogeneity; thus, opening new avenues for cancer treatment.
Collapse
Affiliation(s)
- Fatema Safri
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Shadi Zerehpooshnesfchi
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
10
|
Bao M, Wu A. Understanding the heterogeneity in liver hepatocellular carcinoma with a special focus on malignant cell through single-cell analysis. Discov Oncol 2024; 15:241. [PMID: 38913193 PMCID: PMC11196498 DOI: 10.1007/s12672-024-01115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is the most common form of liver cancer globally and remains a major cause of cancer-related deaths. HCC exhibits significant intra-tumoral and interpatient heterogeneity, impacting treatment efficacy and patient prognosis. METHODS We acquired transcriptome data from the TCGA and ICGC databases, as well as liver cancer chip data from the GEO database, and processed the data for subsequent analysis. We also obtained single cell data from the GEO database and performed data analysis using the Seurat package. To further investigate epithelial cell subgroups and their copy number variations, we used the Seurat workflow for subgroup classification and the InferCNV software for CNV analysis, utilizing endothelial cells as a reference. Pseudo-time analysis and transcription factor analysis of epithelial cells were performed using the monocle2 and SCENIC software, respectively. To assess intercellular communication, we employed the CellChat package to identify potential ligand-receptor interactions. We also analyzed gene expression differences and conducted enrichment analysis using the limma and clusterProfiler packages. Additionally, we established tumor-related risk characteristics using Cox analysis and Lasso regression, and predicted immunotherapy response using various datasets. RESULTS The samples were classified into 23 clusters, with malignant epithelial cells being the majority. Trajectory analysis revealed the differentiation states of the malignant epithelial cells, with cluster 1 being in the terminal state. Functional analysis revealed higher aggressiveness and epithelial-mesenchymal transition (EMT) scores in cluster 1, indicating a higher propensity for metastasis. RBP4+ tumor cells were highly enriched with hypoxia process and intensive cell-to-cell communication. A prognostic model was established, and immune infiltration analysis showed increased infiltration in the high-risk group. TP53 demonstrated significant differences in mutation rate between the two risk groups. Validation analysis confirmed the up-regulation of model genes, including AKR1B10, ARL6IP4, ATP6V0B, and BSG in tumor tissues. CONCLUSION A prognostic model was established based on HCC malignant cell associated gene signature, displaying decent prognosis guiding effectiveness in the multiple cohorts. The study provided comprehensive insights into the heterogeneity and potential therapeutic targets of LIHC.
Collapse
Affiliation(s)
- Mengmeng Bao
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
11
|
Ding F, Huang M, Ren P, Zhang J, Lin Z, Sun Y, Liang C, Zhao X. Quantitative information from gadobenate dimeglumine-enhanced MRI can predict proliferative subtype of solitary hepatocellular carcinoma: a multicenter retrospective study. Eur Radiol 2024; 34:2445-2456. [PMID: 37691080 DOI: 10.1007/s00330-023-10227-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/18/2023] [Accepted: 07/15/2023] [Indexed: 09/12/2023]
Abstract
OBJECTIVES To investigate the value of quantitative parameters derived from gadobenate dimeglumine-enhanced magnetic resonance imaging (MRI) for predicting molecular subtype of hepatocellular carcinoma (HCC) and overall survival. METHODS This multicenter retrospective study included 218 solitary HCC patients who underwent gadobenate dimeglumine-enhanced MRI. All HCC lesions were resected and pathologically confirmed. The lesion-to-liver contrast enhancement ratio (LLCER) and lesion-to-liver contrast (LLC) were measured in the hepatobiliary phase. Potential risk factors for proliferative HCC were assessed by logistic regression. The ability of LLCER and LLC to predict proliferative HCC was assessed by the receiver operating characteristic (ROC) curve. Prognostic factors were evaluated using the Cox proportional hazards regression model for survival outcomes. RESULTS LLCER was an independent predictor of proliferative HCC (odds ratio, 0.015; 95% confidence interval [CI], 0.008-0.022; p < 0.001). The area under the ROC curve was 0.812 (95% CI, 0.748-0.877), higher than that of LLC, alpha-fetoprotein > 100 ng/ml, satellite nodules, and rim arterial phase hyperenhancement (all p ≤ 0.001). HCC patients with LLCER < -4.59% had a significantly higher incidence of proliferative HCC than those with the LLCER ≥ -4.59%. During the follow-up period, LLCER was an independent predictor of overall survival (hazard ratio, 0.070; 95% CI, 0.015-0.324; p = 0.001) in HCC patients. CONCLUSIONS Gadobenate dimeglumine-enhanced quantitative parameter in the hepatobiliary phase can predict the proliferative subtype of solitary HCC with a moderately high accuracy. CLINICAL RELEVANCE STATEMENT Quantitative information from gadobenate dimeglumine-enhanced MRI can provide crucial information on hepatocellular carcinoma subtypes. It might be valuable to design novel therapeutic strategies, such as targeted therapies or immunotherapy. KEY POINTS • The lesion-to-liver contrast enhancement ratio (LLCER) is an independent predictor of proliferative hepatocellular carcinoma (HCC). • The ability of LLCER to predict proliferative HCC outperformed lesion-to-liver contrast, alpha-fetoprotein > 100 ng/ml, satellite nodules, and rim arterial phase hyperenhancement. • HCC patients with LLCER < -4.59% had a significantly higher incidence of proliferative HCC than those with the LLCER ≥ -4.59%.
Collapse
Affiliation(s)
- Feier Ding
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong Province, China
| | - Min Huang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong Province, China
| | - Ping Ren
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong Province, China
| | - Junlei Zhang
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong Province, China
| | - Zhengyu Lin
- Department of Interventional Radiology, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, Fujian Province, China
| | - Yan Sun
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250021, Shandong Province, China
| | - Changhu Liang
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong Province, China.
| | - Xinya Zhao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong Province, China.
| |
Collapse
|
12
|
Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies: Role of tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol 2024; 80:515-530. [PMID: 38104635 PMCID: PMC10923008 DOI: 10.1016/j.jhep.2023.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/27/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
The diagnosis and management of hepatocellular carcinoma (HCC) have improved significantly in recent years. With the introduction of immunotherapy-based combination therapy, there has been a notable expansion in treatment options for patients with unresectable HCC. Simultaneously, innovative molecular tests for early detection and management of HCC are emerging. This progress prompts a key question: as liquid biopsy techniques rise in prominence, will they replace traditional tissue biopsies, or will both techniques remain relevant? Given the ongoing challenges of early HCC detection, including issues with ultrasound sensitivity, accessibility, and patient adherence to surveillance, the evolution of diagnostic techniques is more relevant than ever. Furthermore, the accurate stratification of HCC is limited by the absence of reliable biomarkers which can predict response to therapies. While the advantages of molecular diagnostics are evident, their potential has not yet been fully harnessed, largely because tissue biopsies are not routinely performed for HCC. Liquid biopsies, analysing components such as circulating tumour cells, DNA, and extracellular vesicles, provide a promising alternative, though they are still associated with challenges related to sensitivity, cost, and accessibility. The early results from multi-analyte liquid biopsy panels are promising and suggest they could play a transformative role in HCC detection and management; however, comprehensive clinical validation is still ongoing. In this review, we explore the challenges and potential of both tissue and liquid biopsy, highlighting that these diagnostic methods, while distinct in their approaches, are set to jointly reshape the future of HCC management.
Collapse
Affiliation(s)
- Brandon M Lehrich
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Josephine Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA
| | - Satdarshan P Monga
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| | - Renumathy Dhanasekaran
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA.
| |
Collapse
|
13
|
Chen D, Aierken A, Li H, Chen R, Ren L, Wang K. Identification of subclusters and prognostic genes based on glycolysis/gluconeogenesis in hepatocellular carcinoma. Front Immunol 2023; 14:1232390. [PMID: 37881434 PMCID: PMC10597634 DOI: 10.3389/fimmu.2023.1232390] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Background This study aimed to examine glycolysis/gluconeogenesis-related genes in hepatocellular carcinoma (HCC) and evaluate their potential roles in HCC progression and immunotherapy response. Methods Data analyzed in this study were collected from GSE14520, GSE76427, GSE174570, The Cancer Genome Atlas (TCGA), PXD006512, and GSE149614 datasets, metabolic pathways were collected from MSigDB database. Differentially expressed genes (DEGs) were identified between HCC and controls. Differentially expressed glycolysis/gluconeogenesis-related genes (candidate genes) were obtained and consensus clustering was performed based on the expression of candidate genes. Bioinformatics analysis was used to evaluate candidate genes and screen prognostic genes. Finally, the key results were tested in HCC patients. Results Thirteen differentially expressed glycolysis/gluconeogenesis-related genes were validated in additional datasets. Consensus clustering analysis identified two distinct patient clusters (C1 and C2) with different prognoses and immune microenvironments. Immune score and tumor purity were significantly higher in C1 than in C2, and CD4+ memory activated T cell, Tfh, Tregs, and macrophage M0 were higher infiltrated in HCC and C1 group. The study also identified five intersecting DEGs from candidate genes in TCGA, GSE14520, and GSE141198 as prognostic genes, which had a protective role in HCC patient prognosis. Compared with the control group, the prognostic genes all showed decreased expression in HCC patients in RT-qPCR and Western blot analyses. Flow cytometry verified the abnormal infiltration level of immune cells in HCC patients. Conclusion Results showed that glycolysis/gluconeogenesis-related genes were associated with patient prognosis, immune microenvironment, and response to immunotherapy in HCC. It suggests that the model based on five prognostic genes may valuable for predicting the prognosis and immunotherapy response of HCC patients.
Collapse
Affiliation(s)
- Dan Chen
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Ayinuer Aierken
- Department of Hepatobiliary Hydatid Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hui Li
- Central Laboratory, Xinjiang Medical University, Urumqi, China
| | - Ruihua Chen
- Center of Animal Experiments, Xinjiang Medical University, Urumqi, China
| | - Lei Ren
- Department of Burns, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Kai Wang
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
14
|
Choi JH, Thung SN. Advances in Histological and Molecular Classification of Hepatocellular Carcinoma. Biomedicines 2023; 11:2582. [PMID: 37761023 PMCID: PMC10526317 DOI: 10.3390/biomedicines11092582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer characterized by hepatocellular differentiation. HCC is molecularly heterogeneous with a wide spectrum of histopathology. The prognosis of patients with HCC is generally poor, especially in those with advanced stages. HCC remains a diagnostic challenge for pathologists because of its morphological and phenotypic diversity. However, recent advances have enhanced our understanding of the molecular genetics and histological subtypes of HCC. Accurate diagnosis of HCC is important for patient management and prognosis. This review provides an update on HCC pathology, focusing on molecular genetics, histological subtypes, and diagnostic approaches.
Collapse
Affiliation(s)
- Joon Hyuk Choi
- Department of Pathology, Yeungnam University College of Medicine, Daegu 42415, Republic of Korea
| | - Swan N. Thung
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA;
| |
Collapse
|
15
|
Kim Y, Yang H, Lee D. Cell line-specific features of 3D chromatin organization in hepatocellular carcinoma. Genomics Inform 2023; 21:e19. [PMID: 37704209 PMCID: PMC10326539 DOI: 10.5808/gi.23015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/14/2023] [Indexed: 07/08/2023] Open
Abstract
Liver cancer, particularly hepatocellular carcinoma (HCC), poses a significant global threat to human lives. To advance the development of innovative diagnostic and treatment approaches, it is essential to examine the hidden features of HCC, particularly its 3D genome architecture, which is not well understood. In this study, we investigated the 3D genome organization of four HCC cell lines-Hep3B, Huh1, Huh7, and SNU449-using in situ Hi-C and assay for transposase-accessible chromatin sequencing. Our findings revealed that HCC cell lines had more long-range interactions, both intra-and interchromosomal, compared to human mammary epithelial cells (HMECs). Unexpectedly, HCC cell lines displayed cell line-specific compartmental modifications at the megabase (Mb) scale, which could potentially be leveraged in determining HCC subtypes. At the sub-Mb scale, we observed decreases in intra-TAD (topologically associated domain) interactions and chromatin loops in HCC cell lines compared to HMECs. Lastly, we discovered a correlation between gene expression and the 3D chromatin architecture of SLC8A1, which encodes a sodium-calcium antiporter whose modulation is known to induce apoptosis by comparison between HCC cell lines and HMECs. Our findings suggest that HCC cell lines have a distinct 3D genome organization that is different from those of normal and other cancer cells based on the analysis of compartments, TADs, and chromatin loops. Overall, we take this as evidence that genome organization plays a crucial role in cancer phenotype determination. Further exploration of epigenetics in HCC will help us to better understand specific gene regulation mechanisms and uncover novel targets for cancer treatment.
Collapse
Affiliation(s)
- Yeonwoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Hyeokjun Yang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Daeyoup Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| |
Collapse
|
16
|
Chang X, Tian C, Jia Y, Cai Y, Yan P. MLXIPL promotes the migration, invasion, and glycolysis of hepatocellular carcinoma cells by phosphorylation of mTOR. BMC Cancer 2023; 23:176. [PMID: 36809979 PMCID: PMC9945719 DOI: 10.1186/s12885-023-10652-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is associated with a high occurrence, mortality, and poor prognosis. MLX interacting protein like (MLXIPL) is an important regulator of glucolipid metabolism and is involved in tumor progression. We aimed to clarify the role of MLXIPL in HCC and its underlying mechanisms. METHODS The level of MLXIPL was predicted using bioinformatic analysis and verified using quantitative real-time PCR (qPCR), immunohistochemical analysis, and western blot. We assessed the effects of MLXIPL on biological behaviors using the cell counting kit-8, colony formation, and Transwell assay. Glycolysis was evaluated using the Seahorse method. The interaction between MLXIPL and mechanistic target of rapamycin kinase (mTOR) was confirmed using RNA immunoprecipitation and co-immunoprecipitation. mTOR expression was detected in HCC cells using qPCR, immunofluorescence analysis, and western blot. RESULTS The results showed that MLXIPL levels were elevated in both HCC tissues and HCC cell lines. Knockdown of MLXIPL impeded HCC cell growth, invasion, migration, and glycolysis. Moreover, MLXIPL combined with mTOR to induce phosphorylation of mTOR. Activated mTOR abrogated the effects on cellular processes induced by MLXIPL. CONCLUSION MLXIPL promoted the malignant progression of HCC by activating phosphorylation of mTOR, suggesting an important role of the combination of MLXIPL and mTOR in HCC.
Collapse
Affiliation(s)
- Xiaowei Chang
- grid.508540.c0000 0004 4914 235XDepartment of General Surgery, The First Affiliated Hospital of Xi’an Medical University, No. 48, Fenghao West Road, Lianhu District, 710077 Xi’an, Shaanxi China
| | - Chang Tian
- grid.508540.c0000 0004 4914 235XDepartment of Clinical Laboratory, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi China
| | - Yuanyuan Jia
- grid.508540.c0000 0004 4914 235XDepartment of Faculty Development and Teaching Evaluation Office, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi China
| | - Yu Cai
- grid.508540.c0000 0004 4914 235XDepartment of General Surgery, The First Affiliated Hospital of Xi’an Medical University, No. 48, Fenghao West Road, Lianhu District, 710077 Xi’an, Shaanxi China
| | - Pu Yan
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, No. 48, Fenghao West Road, Lianhu District, 710077, Xi'an, Shaanxi, China.
| |
Collapse
|
17
|
Yan ZQ, Ding SY, Chen P, Liu HP, Chang ML, Shi SY. A water-soluble polysaccharide from Eucommia folium: the structural characterization and anti-tumor activity in vivo. Glycoconj J 2022; 39:759-772. [PMID: 36342595 DOI: 10.1007/s10719-022-10086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
In this study, a water-soluble polysaccharide from Eucommia folium was extracted by hot water and purified using Sephadex G-200 gel columns. The results showed that the purified fraction (EFP) has a molecular weight of 9.98 × 105 Da and consisted of rhamnose, arabinose, galactose, glucose, mannose, xylose, galacturonic acid, and glucuronic acid (molar ratio: 0.226: 1.739: 2.183: 1: 0.155: 0.321: 0.358: 0.047). The combination of infrared spectroscopy and NMR analysis proved that EFP is an acidic polysaccharide whose main chain consists of α-L-Araf-(1 → , → 3,5)-α-Araf-(1 → , → 3)-β-Galp-(1 → , → 3,6)-β-Glcp-(1 → , → 2)-α-D-Manp-(1 → , → 4)-α-GalpA-(1 → , → 2,4)-α-Rhap-(1 → . In addition, the in vivo antitumoral activity of EFP was studied using a H22 tumor-bearing mice model. EFP effectively inhibited tumor growth in mice following intragastric administration. By Combining with the results of the apoptosis assay and JC-1 staining analysis, we confirmed that EFP induces apoptosis through the mitochondrial pathway. Furthermore, cell cycle analysis demonstrated that EFP blocks the cell cycle at S phase.
Collapse
Affiliation(s)
- Zhi-Qian Yan
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Science and Engineering, Tianjin University of Science &Technology, Tianjin, 300457, People's Republic of China
| | - Su-Yun Ding
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Science and Engineering, Tianjin University of Science &Technology, Tianjin, 300457, People's Republic of China
| | - Pei Chen
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Science and Engineering, Tianjin University of Science &Technology, Tianjin, 300457, People's Republic of China
| | - Hui-Ping Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Science and Engineering, Tianjin University of Science &Technology, Tianjin, 300457, People's Republic of China.
| | - Meng-Li Chang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Science and Engineering, Tianjin University of Science &Technology, Tianjin, 300457, People's Republic of China
| | - Shu-Yuan Shi
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Science and Engineering, Tianjin University of Science &Technology, Tianjin, 300457, People's Republic of China
| |
Collapse
|
18
|
Li Y, Jia M, Cao L, Yu J, Gao H, Sun PL. Silva cumulative score and its relationship with prognosis in Endocervical adenocarcinoma. BMC Cancer 2022; 22:1172. [PMID: 36376880 PMCID: PMC9661810 DOI: 10.1186/s12885-022-10270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
Background The Silva system has been demonstrated to have a good predictive value of lymph node metastasis (LNM) in endocervical adenocarcinoma (EAC). Tumours were classified based on the highest identified pattern of invasion in this system, this may not exactly reflect the true situation when it presents with a “mixed pattern” in some cases. Recent study has shown that patients with lymphovascular invasion (LVI) have worse prognosis in EAC. Here we design a Silva cumulative score (SCS) system which also combined the LVI status to explore its prognostic role in EAC patients. Methods A total of 120 patients with EAC were included in this study. Clinicopathological characteristics were retrospectively retrieved from the medical records and follow-up data were obtained. The clinicopathological information included age at diagnosis, depth of invasion (DOI), LNM, LVI, Silva classification, and SCS. SCS is a classification system based on the sum score of different Silva pattern which is founded on morphological phenomena. The relationships between the pathological characteristics and prognoses were analyzed. Results According to the Silva system, 11 (9.2%), 22 (18.3%) and 87 (72.5%) patients had patterns A, B, and C, respectively. Patients with pattern C had the highest incidence of LVI and LNM (p < 0.05). Although the Kaplan-Meier curves demonstrated that survival decreased with increasing Silva classification for A-C cancers, there was no statistically significant difference [disease-free survival (DFS): p = 0.181; overall survival (OS): p = 0.205]. There were 45 cases presented as mixed-type of Silva patterns. According to the SCS, 23 cases (19.2%) were rated as grade I, 31 cases (25.8%) as grade II and 66 (55.0%) cases as grade III. Patients with SCS grade III had the highest incidence of LVI and LNM (p < 0.05). Kaplan–Meier analysis revealed that patients with higher SCS had significantly shorter DFS and OS than those with lower SCS (p < 0.05). High SCS was an independent predictor of poorer OS and DFS (p < 0.05) in patients with EAC. Conclusions The application of the Silva system could effectively predict the LNM of patients and may be helpful in selecting an appropriate surgical procedure. The SCS system we designed showed a good predictive value for DFS and OS in EAC.
Collapse
|
19
|
de Souza AJS, Malheiros AP, da Silva VL, da Silva TC, Cogliati B, de Sá LRM. Hepatocellular Carcinoma in a Free-Ranging Three-Toed Sloth (Bradypus variegatus). Animals (Basel) 2022; 12:ani12151921. [PMID: 35953911 PMCID: PMC9367539 DOI: 10.3390/ani12151921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
The increasing interest of tumors in wildlife is important for biodiversity conservation and for monitoring environmental agents and/or contaminants with potential impact on human health. Here we described the occurrence of hepatocellular carcinoma (HCC) in noncirrhotic liver of a free-ranging three-toed sloth (Bradypus variegatus) from the Atlantic Forest biome in Brazil. The HCC showed a moderate mononuclear inflammatory infiltrate within the tumor tissue but with no inflammation and fibrosis in the adjacent liver tissue. Upon immunohistochemistry, neoplastic cells were diffusely positive for HepPar-1 and glutamine-synthetase presenting an irregular and random immunostaining pattern; β-catenin was positive in the cytoplasmic membrane of malignant hepatocytes; and cytokeratin 19 immunostaining was restricted to bile duct epithelial cells. The liver tissue was negative for HBV-like and HCV-like viruses assessed by molecular tests. The potential similarity of pathogenesis may reinforce the need for research on environmental and/or infectious agents associated with HCC that may contribute to the understanding of cancer in wildlife.
Collapse
Affiliation(s)
- Alex Junior Souza de Souza
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, SP, Brazil; (A.J.S.d.S.); (T.C.d.S.); (B.C.)
| | | | | | - Tereza Cristina da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, SP, Brazil; (A.J.S.d.S.); (T.C.d.S.); (B.C.)
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, SP, Brazil; (A.J.S.d.S.); (T.C.d.S.); (B.C.)
| | - Lilian Rose Marques de Sá
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, SP, Brazil; (A.J.S.d.S.); (T.C.d.S.); (B.C.)
- Correspondence:
| |
Collapse
|
20
|
Etiology, Pathogenesis, Diagnosis, and Practical Implications of Hepatocellular Neoplasms. Cancers (Basel) 2022; 14:cancers14153670. [PMID: 35954333 PMCID: PMC9367411 DOI: 10.3390/cancers14153670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC), a major global contributor of cancer death, usually arises in a background of chronic liver disease, as a result of molecular changes that deregulate important signal transduction pathways. Recent studies have shown that certain molecular changes of hepatocarcinogenesis are associated with clinicopathologic features and prognosis, suggesting that subclassification of HCC is practically useful. On the other hand, subclassification of hepatocellular adenomas (HCAs), a heterogenous group of neoplasms, has been well established on the basis of genotype–phenotype correlations. Histologic examination, aided by immunohistochemistry, is the gold standard for the diagnosis and subclassification of HCA and HCC, while clinicopathologic correlation is essential for best patient management. Advances in clinico-radio-pathologic correlation have introduced a new approach for the diagnostic assessment of lesions arising in advanced chronic liver disease by imaging (LI-RADS). The rapid expansion of knowledge concerning the molecular pathogenesis of HCC is now starting to produce new therapeutic approaches through precision oncology. This review summarizes the etiology and pathogenesis of HCA and HCC, provides practical information for their histologic diagnosis (including an algorithmic approach), and addresses a variety of frequently asked questions regarding the diagnosis and practical implications of these neoplasms.
Collapse
|
21
|
LncRNA ZNF674-AS1 Hinders Proliferation and Invasion of Hepatic Carcinoma Cells through the Glycolysis Pathway. JOURNAL OF ONCOLOGY 2022; 2022:8063382. [PMID: 35874626 PMCID: PMC9300364 DOI: 10.1155/2022/8063382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/18/2022]
Abstract
Purpose. Long noncoding RNAs (lncRNAs) play important roles in regulating various functions of cells at the levels of transcription and posttranscription. Extensive investigations have illustrated that lncRNAs are critical in the glucose metabolism of tumor cells, but their mechanisms of action need to be further explored. This study evaluates the role of lncRNA ZNF674-AS1 on the apoptosis and proliferation of human hepatic carcinoma cells in vitro through the glucose metabolism and its related mechanisms. Methods. Real-time quantitative PCR was employed for detecting the level of expressions for lncRNA ZNF674-AS1 in liver cancer tissues (25 cases), paracancerous tissues, and liver cancer cell lines. The lncRNA ZNF674-AS1 high expression cell strain was constructed by the lentiviral overexpression vector. CCK-8, plate colony formation, transwell assay, lactate production, glucose consumption, and ATP levels were used to detect the change of cell proliferation, colony formation, migration, and invasion, as well as glycolytic capability. Western blot was carried out to detect the expression of HK2, PFKL, PKM2, GLUT1, and PKM1, which are the key proteins of glycolysis in cells. Result. The lncRNA ZNF674-AS1 was undesirably expressed in liver cancer cell lines and tissues. Cell function assessments showed that compared with the blank control group (vector), overexpression of lncRNA ZNF674-AS1 could substantially hinder the proliferation, colony formation, migration, and invasion capability of liver cancer cells. Furthermore, overexpression of lncRNA ZNF674-AS1 could inhibit cell glycolysis (inhibit glucose consumption and reduce intracellular lactate and ATP levels) by inhibiting the expression of key proteins (such as PFKL, HK2, PKM2, and GLUT1) in the process of glycolysis. Conclusion. As a tumor repressor gene, lncRNA ZNF674-AS1 inhibits the expression of key proteins in glycolysis to inhibit glycolysis level, thereby inhibiting cell migration and proliferation. Therefore, lncRNA ZNF674-AS1 could be a potent therapeutic target or a novel diagnostic molecule for patients suffering from liver cancer.
Collapse
|
22
|
Katabathina VS, Khanna L, Surabhi VR, Minervini M, Shanbhogue K, Dasyam AK, Prasad SR. Morphomolecular Classification Update on Hepatocellular Adenoma, Hepatocellular Carcinoma, and Intrahepatic Cholangiocarcinoma. Radiographics 2022; 42:1338-1357. [PMID: 35776676 DOI: 10.1148/rg.210206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hepatocellular adenomas (HCAs), hepatocellular carcinomas (HCCs), and intrahepatic cholangiocarcinomas (iCCAs) are a highly heterogeneous group of liver tumors with diverse pathomolecular features and prognoses. High-throughput gene sequencing techniques have allowed discovery of distinct genetic and molecular underpinnings of these tumors and identified distinct subtypes that demonstrate varied clinicobiologic behaviors, imaging findings, and complications. The combination of histopathologic findings and molecular profiling form the basis for the morphomolecular classification of liver tumors. Distinct HCA subtypes with characteristic imaging findings and complications include HNF1A-inactivated, inflammatory, β-catenin-activated, β-catenin-activated inflammatory, and sonic hedgehog HCAs. HCCs can be grouped into proliferative and nonproliferative subtypes. Proliferative HCCs include macrotrabecular-massive, TP53-mutated, scirrhous, clear cell, fibrolamellar, and sarcomatoid HCCs and combined HCC-cholangiocarcinoma. Steatohepatitic and β-catenin-mutated HCCs constitute the nonproliferative subtypes. iCCAs are classified as small-duct and large-duct types on the basis of the level of bile duct involvement, with significant differences in pathogenesis, molecular signatures, imaging findings, and biologic behaviors. Cross-sectional imaging modalities, including multiphase CT and multiparametric MRI, play an essential role in diagnosis, staging, treatment response assessment, and surveillance. Select imaging phenotypes can be correlated with genetic abnormalities, and identification of surrogate imaging markers may help avoid genetic testing. Improved understanding of morphomolecular features of liver tumors has opened new areas of research in the targeted therapeutics and management guidelines. The purpose of this article is to review imaging findings of select morphomolecular subtypes of HCAs, HCCs, and iCCAs and discuss therapeutic and prognostic implications. Online supplemental material is available for this article. ©RSNA, 2022.
Collapse
Affiliation(s)
- Venkata S Katabathina
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., L.K.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (V.R.S., S.R.P.); Departments of Pathology (M.M.) and Radiology (A.K.D.), University of Pittsburgh Medical Center, Pittsburgh, Pa; and Department of Radiology, NYU Medical Center, New York, NY (K.S.)
| | - Lokesh Khanna
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., L.K.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (V.R.S., S.R.P.); Departments of Pathology (M.M.) and Radiology (A.K.D.), University of Pittsburgh Medical Center, Pittsburgh, Pa; and Department of Radiology, NYU Medical Center, New York, NY (K.S.)
| | - Venkateswar R Surabhi
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., L.K.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (V.R.S., S.R.P.); Departments of Pathology (M.M.) and Radiology (A.K.D.), University of Pittsburgh Medical Center, Pittsburgh, Pa; and Department of Radiology, NYU Medical Center, New York, NY (K.S.)
| | - Marta Minervini
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., L.K.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (V.R.S., S.R.P.); Departments of Pathology (M.M.) and Radiology (A.K.D.), University of Pittsburgh Medical Center, Pittsburgh, Pa; and Department of Radiology, NYU Medical Center, New York, NY (K.S.)
| | - Krishna Shanbhogue
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., L.K.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (V.R.S., S.R.P.); Departments of Pathology (M.M.) and Radiology (A.K.D.), University of Pittsburgh Medical Center, Pittsburgh, Pa; and Department of Radiology, NYU Medical Center, New York, NY (K.S.)
| | - Anil K Dasyam
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., L.K.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (V.R.S., S.R.P.); Departments of Pathology (M.M.) and Radiology (A.K.D.), University of Pittsburgh Medical Center, Pittsburgh, Pa; and Department of Radiology, NYU Medical Center, New York, NY (K.S.)
| | - Srinivasa R Prasad
- From the Department of Radiology, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 (V.S.K., L.K.); Department of Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (V.R.S., S.R.P.); Departments of Pathology (M.M.) and Radiology (A.K.D.), University of Pittsburgh Medical Center, Pittsburgh, Pa; and Department of Radiology, NYU Medical Center, New York, NY (K.S.)
| |
Collapse
|
23
|
Torbenson M, McCabe CE, O’Brien DR, Yin J, Bainter T, Tran NH, Yasir S, Chen ZE, Dhanasekaran R, Ahn KS, Roberts LR, Wang C. Morphological heterogeneity in beta-catenin-mutated hepatocellular carcinomas: implications for tumor molecular classification. Hum Pathol 2022; 119:15-27. [PMID: 34592239 PMCID: PMC9258524 DOI: 10.1016/j.humpath.2021.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 01/03/2023]
Abstract
Beta-catenin (CTNNB1) is commonly mutated in hepatocellular carcinoma (HCC). CTNNB1-mutated HCC has important clinical correlates, such as being immune cold and less likely to respond to immune checkpoint inhibitor therapies. It remains unclear, however, if they are a morphologically homogenous group of tumors. To better understand the association between the morphology, CTNNB1 mutations, and other molecular features, a detailed study of 338 The Cancer Genome Atlas cases was performed. A characteristic histological morphology was strongly associated with CTNNB1 mutations but was present in only 58% of CTNNB1-mutated HCCs. Tumors with APC mutations tended to have the classic morphology; those with AXIN mutations did not. Pseudoglands are a key feature of the classic morphology, and they were associated with CTNNB1 mutations, male gender, specific CTNNB1 mutation site, and lack of TP53 mutations. Differential gene expression analysis stratified by the presence/absence of pseudoglands identified 60 differentially expressed genes (FDR <5%); clustering according to these differentially expressed genes revealed three groups of tumors, one with pseudoglands and a strong association with genes regulated by Wnt signaling; within this group, TP53 mutations were associated with a loss of the typical morphology of CTNNB1-mutated HCCs. When stratified by gender, further differential gene expression showed Wnt-regulated genes were associated with pseudoglands in men but not women. These findings indicate HCC with CTNNB1 mutations are morphologically heterogeneous, with gene penetrance for morphology dependent in part on gender, specific CTNNB1 mutations, and co-occurring TP53 mutations. This heterogeneity has important implications for the classification of HCC.
Collapse
Affiliation(s)
- Michael Torbenson
- Mayo Clinic Department of Pathology and Laboratory Medicine, United States,Corresponding author. Department of Pathology and Laboratory Medicine, Mayo Clinic, 200 First Street, SW, Rochester, MN 55905, USA. (M. Torbenson)
| | - Chantal E. McCabe
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Daniel R. O’Brien
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Jun Yin
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, United States
| | - Tiffany Bainter
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, United States
| | - Nguyen H. Tran
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Saba Yasir
- Mayo Clinic Department of Pathology and Laboratory Medicine, United States
| | - Zongming Eric Chen
- Mayo Clinic Department of Pathology and Laboratory Medicine, United States
| | - Renu Dhanasekaran
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, United States
| | - Keun Soo Ahn
- Department of Surgery, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
24
|
Deldar Abad Paskeh M, Mirzaei S, Ashrafizadeh M, Zarrabi A, Sethi G. Wnt/β-Catenin Signaling as a Driver of Hepatocellular Carcinoma Progression: An Emphasis on Molecular Pathways. J Hepatocell Carcinoma 2021; 8:1415-1444. [PMID: 34858888 PMCID: PMC8630469 DOI: 10.2147/jhc.s336858] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Liver cancers cause a high rate of death worldwide and hepatocellular carcinoma (HCC) is considered as the most common primary liver cancer. HCC remains a challenging disease to treat. Wnt/β-catenin signaling pathway is considered a tumor-promoting factor in various cancers; hence, the present review focused on the role of Wnt signaling in HCC, and its association with progression and therapy response based on pre-clinical and clinical evidence. The nuclear translocation of β-catenin enhances expression level of genes such as c-Myc and MMPs in increasing cancer progression. The mutation of CTNNB1 gene encoding β-catenin and its overexpression can lead to HCC progression. β-catenin signaling enhances cancer stem cell features of HCC and promotes their growth rate. Furthermore, β-catenin prevents apoptosis in HCC cells and increases their migration via triggering EMT and upregulating MMP levels. It is suggested that β-catenin signaling participates in mediating drug resistance and immuno-resistance in HCC. Upstream mediators including ncRNAs can regulate β-catenin signaling in HCC. Anti-cancer agents inhibit β-catenin signaling and mediate its proteasomal degradation in HCC therapy. Furthermore, clinical studies have revealed the role of β-catenin and its gene mutation (CTNBB1) in HCC progression. Based on these subjects, future experiments can focus on developing novel therapeutics targeting Wnt/β-catenin signaling in HCC therapy.
Collapse
Affiliation(s)
- Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, Turkey
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul, 34396, Turkey
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
25
|
Wang JY, Wang XK, Zhu GZ, Zhou X, Yao J, Ma XP, Wang B, Peng T. Distinct diagnostic and prognostic values of Glypicans gene expression in patients with hepatocellular carcinoma. BMC Cancer 2021; 21:462. [PMID: 33902495 PMCID: PMC8073913 DOI: 10.1186/s12885-021-08104-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
Backgroud In our current work, we aimed to investigate the expressions of glypican (GPC) family genes at the mRNA level and assess their prognostic significances in patients with hepatocellular carcinoma (HCC). Methods The pathological roles of GPC family genes were examined using bioinformatics analysis. The diagnostic values of GPC genes were explored with the Gene Expression Profiling Interactive Analysis. Moreover, the mRNA expression and prognostic values of GPC genes were assessed via the KM plotter database. Results Our data showed that the expression of GPC-3 was dramatically increased in the liver tumor tissue. Moreover, the expressions of the other five GPC family members were not significantly different between the tumor and normal liver tissues (P > 0.05). Furthermore, the up-regulation of GPC-1 at the mRNA level was dramatically correlated to the reduced overall survival (OS) for all HCC patients (hazard ratio = 2.03, 95% confidence intervals =1.44–2.87, P = 4.1e-05) compared with its low-expression group. Besides, the prognosis of the Caucasians was related to most GPC family genes, while the prognosis of the Asian race was only related to the expression of GPC-2. Besides, for pathological factors, including stage, grade, AJCC, and vascular invasion, the higher the pathological grade and vascular invasiveness, the lower the expression levels of GPC family genes (P < 0.05). Finally, the expression levels of GPC-1, 2, and 3 in the hepatitis group were related to the poor prognosis of HCC in the risk factor (alcohol consumption and hepatitis) subgroup (P < 0.05). Conclusions Our findings indicated that GPC-3 was dysregulated in HCC compared with paracancerous tissues. The expression of GPC-1 could be used as a potent predictive index for the general prognosis of HCC. The pathology, patients, and risk factors might affect the prognostic value of GPC family genes in HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08104-z.
Collapse
Affiliation(s)
- Jian-Yao Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6#, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.,Department of General Surgery, Shenzhen Children's Hospital, Yi Tian Road 7019#, Shenzhen, 518026, Guangdong Province, People's Republic of China
| | - Xiang-Kun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6#, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Guang-Zhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6#, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6#, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jun Yao
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, Dong Men Bei Road 1017#, Shenzhen, 518020, Guangdong Province, People's Republic of China.
| | - Xiao-Peng Ma
- Department of General Surgery, Shenzhen Children's Hospital, Yi Tian Road 7019#, Shenzhen, 518026, Guangdong Province, People's Republic of China.
| | - Bin Wang
- Department of General Surgery, Shenzhen Children's Hospital, Yi Tian Road 7019#, Shenzhen, 518026, Guangdong Province, People's Republic of China.
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Road 6#, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|