Published online Jan 27, 2025. doi: 10.4254/wjh.v17.i1.102273
Revised: November 12, 2024
Accepted: November 27, 2024
Published online: January 27, 2025
Processing time: 84 Days and 19.2 Hours
In this review, we explore the application of next-generation sequencing in liver cancer research, highlighting its potential in modern oncology. Liver cancer, particularly hepatocellular carcinoma, is driven by a complex interplay of genetic, epigenetic, and environmental factors. Key genetic alterations, such as mutations in TERT, TP53, and CTNNB1, alongside epigenetic modifications such as DNA methylation and histone remodeling, disrupt regulatory pathways and promote tumorigenesis. Environmental factors, including viral infections, alcohol consum
Core Tip: This review delves into the complexities of hepatocellular carcinoma (HCC); a highly aggressive form of liver cancer with a low 5-year survival rate and limited treatment options. It examines various factors contributing to HCC, highlights the need for improved early detection using biomarkers and advanced techniques like next-generation sequencing, and emphasizes the importance of understanding the molecular mechanisms involved, particularly the Wnt/β-catenin signaling pathway, to provide evidence for the development of more effective and personalized therapies.
- Citation: Shi J, Zhu X, Yang JB. Advances and challenges in molecular understanding, early detection, and targeted treatment of liver cancer. World J Hepatol 2025; 17(1): 102273
- URL: https://www.wjgnet.com/1948-5182/full/v17/i1/102273.htm
- DOI: https://dx.doi.org/10.4254/wjh.v17.i1.102273
Liver cancer, primarily hepatocellular carcinoma (HCC), is a severe global health issue. According to the World Health Organization, it ranks sixth in prevalence and fourth in cancer-related mortality worldwide. Characterized by its rapid progression and poor prognosis, the 5-year survival rate for liver cancer patients is a dismal 3%, severely impacting their quality of life. Current treatment options, including surgical interventions, interventional therapies, local ablation, and targeted immunotherapies, have limited effectiveness, with survival rates remaining low for many patients even after treatment[1-3].
The origins of liver cancer are frequently tied to underlying conditions, which vary significantly across different regions. Major contributing factors include hepatitis B virus (HBV) infection, poor dietary habits, sedentary lifestyles, and exposure to fungal toxins[4]. Despite advancements in understanding these risk factors, the molecular mechanisms driving liver cancer remain poorly defined[5-8]. This lack of clarity hampers the development of effective treatments and underscores the need for more comprehensive research into the pathogenesis of HCC[9]. Efforts have focused on elu
Early-stage liver cancer is often asymptomatic, making it challenging to diagnose until the disease has progressed to an advanced stage[19]. This emphasizes the need for effective screening programs and predictive models to identify at-risk individuals and detect cancer at a more treatable stage. Advances in molecular biology have opened new avenues for early detection and prediction of liver cancer. Biomarkers such as alpha-fetoprotein (AFP)[20-22], des-gamma-carboxy prothrombin (DCP)[23,24], and the glypican-3 (GPC3) protein[25,26] have shown promise in identifying early-stage HCC[27]. AFP, which was identified over 60 years ago, remains the most common serological marker of HCC[28]. The AFP gene, part of the albumin gene family, includes two distinct enhancer and silencer regions. In liver cancer, inhibition of the enhancer or deletion of the silencer can impair these regulatory elements, causing activation of the AFP promoter and leading to the overexpression of AFP, then the expression level and the protein level of AFP serve as a valuable biomarker for liver cancer[29]. DCP has been extensively studied over the past two decades, primarily in Asian countries, where it is recognized as a useful marker for liver tumors. Elevated DCP levels are commonly observed in HCC patients[30]. Normalization of DCP levels following effective cancer treatment strongly suggests that the tumor is the primary source of its production. GPC3 is a membrane proteoglycan that is typically absent in healthy adult liver but is overexpressed in HCC[31]. This overexpression is linked to a poor prognosis and contributes to HCC progression by interacting with Wnt signaling proteins and growth factors[32]. However, AFP, DCP, and GPC3 have notable limitations. AFP and DCP may lack specificity and sensitivity, often showing elevated levels in various liver conditions, while GPC3 may not be consistently expressed in all HCC cases and can be affected by the tumor microenvironment.
In addition to these classic biomarkers, non-classic markers like gamma-glutamyl transferase (GGT) and interleukin (IL)-8 have shown potential in assisting with tumor diagnosis and prognostication. Elevated GGT levels are associated with liver inflammation and oxidative stress, and recent studies have linked high GGT levels with poorer outcomes in HCC patients, suggesting its role as a marker of liver tumor progression and therapeutic resistance[33-35]. Similarly, IL-8, a pro-inflammatory cytokine, is elevated in the serum of HCC patients and has been implicated in promoting angiogenesis and immune evasion in the tumor microenvironment[36]. Elevated IL-8 levels are correlated with more aggressive HCC phenotypes and are associated with poor prognosis, making IL-8 a valuable indicator in assessing tumor progression and patient prognosis[36-38]. The addition of these non-classic markers to conventional biomarker panels may enhance diagnostic accuracy and provide more nuanced prognostic information, ultimately improving personalized treatment approaches for HCC.
Next-generation sequencing (NGS) has emerged as a transformative tool in this context, offering unprecedented insights into the molecular underpinnings of liver cancer[39-41]. NGS enables high-throughput and high-precision analysis of the genome, transcriptome, and epigenome, revealing critical mutations, gene expression patterns, and epigenetic modifications that drive tumorigenesis. circulating tumor DNA[42,43] and miRNAs[44,45] are emerging as potential noninvasive biomarkers for liver cancer screening. These biomarkers can be detected through blood tests, offering a feasible and less-invasive alternative to traditional diagnostic methods. By elucidating these molecular mechanisms, NGS provides valuable information for early detection, prognostic assessment, and the identification of novel therapeutic targets. The application of NGS in liver cancer research is rapidly advancing our understanding of the disease and holds promise for developing more effective and personalized treatment strategies.
HCC is frequently associated with paraneoplastic syndromes, which manifest through metabolic abnormalities, including altered serum lipid and blood glucose levels[46-48]. These metabolic changes are byproducts of liver dysfunction and reflect the systemic impact of HCC. For instance, dyslipidemia is often seen in HCC due to the central role of the liver in lipid metabolism, while hyperglycemia may arise as a paraneoplastic syndrome or from insulin resistance exacerbated by chronic liver disease[49,50]. Elevated blood glucose levels, especially in patients with underlying diabetes, have been linked to poorer HCC prognosis and heightened risk of recurrence after treatment[51,52]. Paraneoplastic syndromes, such as erythrocytosis[53,54], hypercalcemia[55,56], and hypoglycemia[57-59], are significant clinical considerations as they often emerge alongside advanced HCC and complicate disease management.
Liver cancer is a complex disease with multiple etiological factors and molecular mechanisms contributing to its development and progression. The underlying mechanisms can be categorized into genetic, epigenetic, and environmental factors, all of which interact to drive hepatocarcinogenesis. Genetic changes, including point mutations, gene amplifications, alterations in promoter regions, and copy number variations often triggered by viral infections or exposure to hepatotoxic stress transform hepatocytes and contribute to tumor development. Key oncogenes and tumor suppressor genes are frequently altered in HCC[60]. Alcohol-related HCC exhibits significant enrichment in genetic alterations involving CTNNB1, TERT, CDKN2A, SMARCA2 and HGF. Mutations in the TERT promoter, activate telomerase expression, occur in 60% of cases, are among the most common genetic alterations in liver cancer[61,62]. Additionally, mutations in TP53, a crucial tumor suppressor, and CTNNB1, which encodes β-catenin, are frequently observed. These mutations disrupt cell cycle regulation, apoptosis, and Wnt/β-catenin signaling, promoting uncontrolled cell proliferation and survival. CTNNB1 and TP53 mutations are mutually exclusive and define two HCC subtypes. CTNNB1-mutant tumors are large, well-differentiated, and cholestatic, with microtrabecular and pseudoglandular patterns and few inflammatory infiltrates. TP53-mutant tumors are poorly differentiated, compact, multinucleated, pleomorphic, and often show vascular invasion[62]. Epigenetic alterations, including histone, DNA, and RNA modifications, also contribute to liver cancer development[63]. Aberrant methylation of tumor suppressor gene promoters leads to their silencing, while global hypomethylation can activate oncogenes. Histone modification, including methylation, phosphorylation, acetylation, ubiquitylation, and sumoylation, controls gene expression by altering chromatin structure. Acetylation and methylation play a role in chromatin remodeling and gene expression regulation[64]. Alterations in histone acetylation and methylation enzymes affect gene expression in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), contributing to changes in hepatic metabolism, apoptosis, and progression to HCC. DNA epigenetics indicate modifications that affect gene expression without altering the DNA sequence. Aberrant hypermethylation of tumor suppressor genes in HCC, including CDH1, RASSF1A, GSTP, SOCS1, SFRP1, and PTEN, as well as CDK inhibitors such as p16INK4A, p21 and p27kip1, results in the loss of cell cycle checkpoints and promotes increased cell proliferation[65]. m6A methylation has been identified in HBV RNA during viral infections. Recent research shows that m6A factors can influence the viral life cycle and significantly impact virus–host interactions. m6A modification can regulate viral replication both directly, by adding m6A to viral genomic RNA or mRNA, and indirectly, by affecting the expression of genes or the stability of RNA involved in viral replication[66]. Chronic liver inflammation, often due to HBV or hepatitis C virus infection, is a significant risk factor for HCC[67,68]. Persistent inflammation induces a regenerative response in hepatocytes, increasing the risk of genetic and epigenetic alterations. Additionally, liver fibrosis and cirrhosis, resulting from chronic liver injury, create a pro-tumorigenic environment[69,70]. Activated hepatic stellate cells and the extracellular matrix components promote tumor cell proliferation, invasion, and angiogenesis[71]. Metabolic disorders, such as NAFLD and NASH, are increasingly recognized as significant contributors to liver cancer[72,73]. Insulin resistance, obesity, and lipid accumulation in hepatocytes lead to oxidative stress and chronic inflammation, promoting hepatocarcinogenesis[4]. Dietary factors, including aflatoxin exposure from contaminated food, also play a role in liver cancer development[74]. Aflatoxin B1, a potent carcinogen, induces mutations in the TP53 gene, further enhancing cancer risk[75]. The tumor microenvironment in the liver plays a crucial role in cancer progression[13]. Immune cells, fibroblasts, endothelial cells, and the extracellular matrix interact with tumor cells, influencing their behavior. Tumor-associated macrophages and myeloid-derived suppressor cells create an immunosuppressive environment, aiding tumor evasion from immune surveillance[76]. Liver cancer arises from a complex interplay of genetic, epigenetic, and environmental factors. A comprehensive understanding of these mechanisms is crucial for developing effective therapeutic strategies. Integrating these factors is essential for advancing early detection methods and optimizing targeted therapeutic approaches.
Despite significant advances in liver cancer research, a deeper understanding of the molecular and cellular components of the tumor microenvironment is essential, as it plays a central role in the progression of HCC. The tumor microenvironment comprises a complex network of cancer cells, innate and adaptive immune cells, stromal cells, endothelial cells, and cancer-associated fibroblasts[77-79]. Within this environment, macrophages infiltrate[9,80], fibroblasts proliferate[81,82], and angiogenesis is promoted[83]; all of which contribute to tumor formation, survival, and metastasis. Efforts are underway to develop targeted treatments that inhibit angiogenesis, cellular adhesion, and immune cell infiltration in the tumor microenvironment. Although earlier research predominantly focused on adaptive immune cells, particularly T lymphocytes due to their cytotoxic potential against cancer cells, recent therapies have shifted toward targeting innate immune responses[78,84]. This shift has led to innovative approaches, such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T cell therapies[85-87]. In treating advanced HCC, immunotherapeutics, such as monoclonal antibodies targeting cytotoxic T-lymphocyte-associated protein (CTLA)-4 and programmed cell death protein (PD)-1, have shown promise by inhibiting immune checkpoints, thereby enhancing immune responses against tumor cells[88-90]. Notablr’s unique immunosuppressive environment, which is tolerogenic to gut-derived microbial metabolites, presents challenges in achieving effective immune responses. Since 2017, ICIs such as pembrolizumab[91,92] and nivolumab[93,94], which target PD-1, have been approved as second-line therapies for advanced HCC, despite demonstrating only limited improvements in survival compared to sorafenib. Additionally, the Food and Drug Administration has approved a combination of nivolumab and ipilimumab (an anti-CTLA-4 antibody) for advanced HCC. However, these treatments yield responses in only 20%-30% of patients, highlighting an urgent need to identify biomarkers to predict therapy response and to develop combination treatments that could overcome resistance[95].
The Wnt/β-catenin signaling pathway, essential for embryonic development and cellular processes, is often dysregulated in liver cancers[18]. The pathway includes canonical and non-canonical routes, with the canonical pathway involving β-catenin, a key factor in tumorigenesis encoded by CTNNB1. Mutations in this pathway are common in liver tumors, and
The canonical Wnt/β-catenin signaling pathway is crucial in HCC pathogenesis. β-catenin activation occurs in 20%-35% of HCC cases[101], often due to mutations in the CTNNB1[102], which encode β-catenin and block its degradation, leading to its accumulation and nuclear translocation[103]. These mutations result in tumors with distinct features, including large size and well-differentiated structures[104]. While β-catenin alone is insufficient to induce HCC in mouse models, its activation can cooperate with other oncogenic pathways. In the mouse model, combined activation of insulin receptor substrate-1/mitogen-activated protein kinase and Wnt/β-catenin pathways are crucial for liver cancer development. Mice with a single transgene did not develop tumors, but double transgenic mice exhibited increased hepatocellular dysplasia and progressed to HCC. All transgenic lines showed significantly elevated mRNA levels of IGF-1, Wnt1, and Wnt3[105]. In another mouse model with a mutant β-catenin allele, isolated Wnt-activating β-catenin mutations are insufficient for liver cancer development. However, when combined with HRAS mutations, HCC develops with 100% incidence. While HRAS mutations alone cause large cell dysplasia, β-catenin mutations promote clonal expansion and progression to HCC[106]. In a mouse model with combined expression of c-MET-V5 and mutant β-catenin-Myc, HCC developed with significant similarity to clinical cases[107]. Additionally, mutations in AXIN1, present in 3%-16% of HCC cases, are often associated with HBV infection and chromosomal instability. Beyond genetic mutations, epigenetic factors and non-coding RNAs also contribute to aberrant Wnt/β-catenin activation in HCC. For example, SFRP3 promoter hypermethylation in HCC cell lines and tissues correlates with reduced SFRP3 expression. Demethylation restores SFRP3 levels, suggesting that hypermethylation contributes to HCC by downregulating SFRP3 and enhancing Wnt signaling[108]. miR-300 and various long noncoding RNAs modulate Wnt/β-catenin signaling, influencing tumor progression[109]. The tumor microenvironment, including autophagy and exosomes, further impacts Wnt/β-catenin signaling, affecting cancer cell metabolism, proliferation, and metastasis.
NGS has become crucial for identifying genetic variations and biomarkers in liver cancer. This technology enables comprehensive profiling of cancer genomes and transcriptomes, rapidly pinpointing potential driver mutations and therapeutic targets. NGS applications, including targeted NGS[110], whole-exome sequencing (WES)[111], and whole-genome sequencing (WGS)[112], aid in diagnosing, treatment planning, and risk assessment for liver cancer. Biomarkers for liver disease identified with protein and NGS were as summarized in Table 1.
Applied method | Biomarkers (Target proteins and genes) | Ref. |
Detection of protein content | AFP, DCP, GPC3, GGT, IL-8 | Zhao et al[20]; Hu et al[21]; Hanif et al[22]; Lai et al[23]; Bertino et al[24]; Zhou et al[25]; Shimizu et al[26] |
Epigenetic modification | CDH1, RASSF1A, GSTP, SOCS1, SFRP1, PTEN, p16, p21, p27 and m6A methylation of HBV RNA | Raggi and Invernizzi[65]; Kostyusheva et al[66] |
Targeted NGS | TP53, TERT and TSC2 | Lin et al[113] |
WGS | CDKN2A, CCND1, APC, TERT, ASH1L, NCOR1, and MACROD2, ARID1A, TTC28, LRP1B and BCL9 | Fujimoto et al[118]; Zhou et al[119] |
WES | PCK2, HUWE1, ARID1A, RPS6KA3, NFE2L2, IRF2, APOB, P53, Keap1, CDKN1A, XPO1 and HIST1H1E | Gnirke et al[114]; Liu et al[115]; Guichard et al[116]; Zhou et al[117] |
RNA-seq | PTPRC and FOXP3 | Zhang et al[121] |
Targeted NGS: Targeted NGS is designed to focus on specific genes associated with liver cancer. These panels provide a detailed analysis of known cancer-related mutations, making them a cost-effective and rapid option for clinical use. However, their limitation lies in their narrow scope, as they miss mutations outside the targeted genes, potentially leading to incomplete risk assessments and missed novel biomarkers.
An ultra-deep targeted NGS was applied to analyze the 31 spatially distinct regions from 11 resected multifocal HCC samples. Truncal and branchy drivers were both economically and effectively identified. TP53 and TERT were the most commonly altered truncal drivers in multifocal HCC, and TSC2 was the most frequently mutated branchy driver[113].
WES covers: Whole protein-coding exons of the human genome (30-40 Mb, approximately 1%-2% of the human genome)[114] capture a broad range of genetic mutations relevant to liver cancer. WES was the most common platform for the Cancer Genome Atlas projects. The coverage of the target regions is usually more than 100 ×, which enables more accurate detection of detection of single nucleotide variants and short indels.
By whole-exome sequencing of a pleomorphic cell-type HCC tissue and its matched normal tissue, it is suggested that PCK2 and HUWE1 are associated with carcinoma cell proliferation in HCC[115].
WES identified 24 HCC tumors, revealing 135 homozygous deletions and 994 somatic mutations in genes with predicted functional consequences. Additionally, new recurrent alterations were discovered in four genes: ARID1A, RPS6KA3, NFE2L2, and IRF2[116]. Cell lines from HCC patients were analyzed using WES, showing an average of 16.55 (range, 15.38%-18.17%) heterogeneous mutations in each line. The main observed substitutions were C:G > T:A and T:A > C:G transitions, Seven reported driver gene mutations were observed in the three HCC cells, including APOB, P53, Keap1, CDKN1A, XPO1, ARID1A and HIST1H1E[117].
WGS: WGS provides a comprehensive view of the entire genome, including both coding and non-coding regions, structural variations, and epigenetic changes. This method identifies all types of genetic alterations, including novel and unexpected variants. The genetic changes were analyzed in 300 liver cancers, including point mutations, structural variations, and virus integrations. There was a correlation between structural variations and DNA replication timing, impacting known cancer-related genes such as CDKN2A, CCND1, APC, and TERT, as well as new genes such as ASH1L, NCOR1, and MACROD2, altering gene expression. The analysis identified recurrent structural variations targeting key driver genes such as TERT, CDKN2A, APC, and ARID1A, as well as newly identified genes such as TTC28, MACROD2, and LRP1B, leading to changes in gene expression in liver cancer genomes[118]. WGS of 40 pairs of primary and early-recurrent HBV-related HCC tumors from patients showed that the mutations and copy-number gains in BCL9 activated Wnt/β-catenin signaling and created an immune-excluded tumor microenvironment. BCL9 could be a new therapeutic target for recurrent HCC[119].
RNA sequencing: RNA sequencing (RNA-seq) has become a powerful tool in defining the transcriptomic changes related to HCC. A transcriptome-wide study of HCC in a group of United States Caucasian patients using RNA-seq technology revealed significant up-regulation of oxidative phosphorylation as the primary signal in HCC development, and signals related to DNA damage were also identified in HCC samples[120]. Single-cell RNA-seq (scRNA-seq) is a powerful tool for studying cellular components, cell-cell interactions, transition in cellular status, and clonal evolution of the tumor. A recent study conducted scRNA-seq on samples taken from eight patients with HCC. Researchers were able to identify three distinct HCC subtypes characterized by varying immune statuses. This analysis also shed light on the expression levels of chemokines and cytokines, as well as the metabolic features unique to each subtype, they also use PTPRC and FOXP3 to differentiate HCCs into the three subtypes[121]. According to a study on the spatial heterogeneity of the immune microenvironment in HCC, the researchers observed a gradual increase in the number of CD4 + effector memory T cells from the non-tumor region to the tumor core. Conversely, CD8 + effector memory T cells showed a decreasing trend in the same regions[122].
The integration of comprehensive molecular studies with emerging sequencing technologies is key to tackling the complexities of liver cancer. Continued research in this domain is essential for developing effective screening programs, early detection methodologies, and novel therapeutic approaches, ultimately aiming to improve survival rates and quality of life for liver cancer patients.
1. | Darvesh AS, Aggarwal BB, Bishayee A. Curcumin and liver cancer: a review. Curr Pharm Biotechnol. 2012;13:218-228. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 165] [Cited by in F6Publishing: 165] [Article Influence: 12.7] [Reference Citation Analysis (0)] |
2. | Gravitz L. Liver cancer. Nature. 2014;516:S1. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 48] [Cited by in F6Publishing: 77] [Article Influence: 7.7] [Reference Citation Analysis (0)] |
3. | Bruix J, Han KH, Gores G, Llovet JM, Mazzaferro V. Liver cancer: Approaching a personalized care. J Hepatol. 2015;62:S144-S156. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 217] [Cited by in F6Publishing: 206] [Article Influence: 20.6] [Reference Citation Analysis (0)] |
4. | Marengo A, Rosso C, Bugianesi E. Liver Cancer: Connections with Obesity, Fatty Liver, and Cirrhosis. Annu Rev Med. 2016;67:103-117. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 308] [Cited by in F6Publishing: 452] [Article Influence: 45.2] [Reference Citation Analysis (0)] |
5. | Liang N, Yang T, Huang Q, Yu P, Liu C, Chen L, Wang Q, Wang G, He X. Mechanism of cancer stemness maintenance in human liver cancer. Cell Death Dis. 2022;13:394. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 13] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
6. | Liu F, Liao Z, Zhang Z. MYC in liver cancer: mechanisms and targeted therapy opportunities. Oncogene. 2023;42:3303-3318. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 1] [Reference Citation Analysis (0)] |
7. | Pan H, Fu X, Huang W. Molecular mechanism of liver cancer. Anticancer Agents Med Chem. 2011;11:493-499. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 33] [Cited by in F6Publishing: 34] [Article Influence: 2.4] [Reference Citation Analysis (0)] |
8. | Wang M, Tao H, Huang P. Clinical significance of LARGE1 in progression of liver cancer and the underlying mechanism. Gene. 2021;779:145493. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 1] [Article Influence: 0.3] [Reference Citation Analysis (0)] |
9. | Cheng K, Cai N, Zhu J, Yang X, Liang H, Zhang W. Tumor-associated macrophages in liver cancer: From mechanisms to therapy. Cancer Commun (Lond). 2022;42:1112-1140. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 145] [Article Influence: 48.3] [Reference Citation Analysis (0)] |
10. | Craig AJ, von Felden J, Villanueva A. Molecular profiling of liver cancer heterogeneity. Discov Med. 2017;24:117-125. [PubMed] [Cited in This Article: ] |
11. | Zhu S, Jin Y, Zhou M, Li L, Song X, Su X, Liu B, Shen J. KK-LC-1, a biomarker for prognosis of immunotherapy for primary liver cancer. BMC Cancer. 2024;24:811. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
12. | Esquea EM, Young RG, Reginato MJ. Fructose promotes liver cancer via microbial acetate-induced O-GlcNAcylation. Trends Endocrinol Metab. 2024;35:88-90. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
13. | Yu LX, Schwabe RF. The gut microbiome and liver cancer: mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol. 2017;14:527-539. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 334] [Cited by in F6Publishing: 378] [Article Influence: 47.3] [Reference Citation Analysis (0)] |
14. | Begolli R, Chatziangelou M, Samiotaki M, Goutas A, Barda S, Goutzourelas N, Kevrekidis DP, Malea P, Trachana V, Liu M, Lin X, Kollatos N, Stagos D, Giakountis A. Transcriptome and proteome analysis reveals the anti-cancer properties of Hypnea musciformis marine macroalga extract in liver and intestinal cancer cells. Hum Genomics. 2023;17:71. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 6] [Reference Citation Analysis (0)] |
15. | Wang YC, Tian ZB, Tang XQ. Bioinformatics screening of biomarkers related to liver cancer. BMC Bioinformatics. 2021;22:521. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 7] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
16. | Banaudha KK, Verma M. Epigenetic biomarkers in liver cancer. Methods Mol Biol. 2015;1238:65-76. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 9] [Cited by in F6Publishing: 8] [Article Influence: 0.8] [Reference Citation Analysis (0)] |
17. | Tabnak P, Ghasemi Y, Natami M, Khorram R, Ebrahimnezhad M. Role of m(6)A modification in dysregulation of Wnt/β-catenin pathway in cancer. Biomed Pharmacother. 2023;157:114023. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 8] [Reference Citation Analysis (0)] |
18. | He S, Tang S. WNT/β-catenin signaling in the development of liver cancers. Biomed Pharmacother. 2020;132:110851. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 65] [Cited by in F6Publishing: 176] [Article Influence: 35.2] [Reference Citation Analysis (0)] |
19. | Chinnappan R, Makhzoum T, Arai M, Hajja A, Abul Rub F, Alodhaibi I, Alfuwais M, Elahi MA, Alshehri EA, Ramachandran L, Mani NK, Abrahim S, Mir MS, Al-Kattan K, Mir TA, Yaqinuddin A. Recent Advances in Biosensor Technology for Early-Stage Detection of Hepatocellular Carcinoma-Specific Biomarkers: An Overview. Diagnostics (Basel). 2024;14. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
20. | Zhao K, Zhou X, Xiao Y, Wang Y, Wen L. Research Progress in Alpha-fetoprotein-induced Immunosuppression of Liver Cancer. Mini Rev Med Chem. 2022;22:2237-2243. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 6] [Reference Citation Analysis (0)] |
21. | Hu X, Chen R, Wei Q, Xu X. The Landscape Of Alpha Fetoprotein In Hepatocellular Carcinoma: Where Are We? Int J Biol Sci. 2022;18:536-551. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 7] [Cited by in F6Publishing: 83] [Article Influence: 27.7] [Reference Citation Analysis (0)] |
22. | Hanif H, Ali MJ, Susheela AT, Khan IW, Luna-Cuadros MA, Khan MM, Lau DT. Update on the applications and limitations of alpha-fetoprotein for hepatocellular carcinoma. World J Gastroenterol. 2022;28:216-229. [PubMed] [DOI] [Cited in This Article: ] [Cited by in CrossRef: 36] [Cited by in F6Publishing: 98] [Article Influence: 32.7] [Reference Citation Analysis (9)] |
23. | Lai Q, Iesari S, Levi Sandri GB, Lerut J. Des-gamma-carboxy prothrombin in hepatocellular cancer patients waiting for liver transplant: a systematic review and meta-analysis. Int J Biol Markers. 2017;32:e370-e374. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 15] [Cited by in F6Publishing: 34] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
24. | Bertino G, Ardiri AM, Calvagno GS, Bertino N, Boemi PM. Prognostic and diagnostic value of des-γ-carboxy prothrombin in liver cancer. Drug News Perspect. 2010;23:498-508. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 32] [Cited by in F6Publishing: 36] [Article Influence: 2.6] [Reference Citation Analysis (0)] |
25. | Zhou F, Shang W, Yu X, Tian J. Glypican-3: A promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med Res Rev. 2018;38:741-767. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 144] [Cited by in F6Publishing: 221] [Article Influence: 27.6] [Reference Citation Analysis (0)] |
26. | Shimizu Y, Suzuki T, Yoshikawa T, Endo I, Nakatsura T. Next-Generation Cancer Immunotherapy Targeting Glypican-3. Front Oncol. 2019;9:248. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 54] [Cited by in F6Publishing: 80] [Article Influence: 13.3] [Reference Citation Analysis (0)] |
27. | Chen J, Niu C, Yang N, Liu C, Zou SS, Zhu S. Biomarker discovery and application-An opportunity to resolve the challenge of liver cancer diagnosis and treatment. Pharmacol Res. 2023;189:106674. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 8] [Reference Citation Analysis (0)] |
28. | European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol. 2018;69:182-236. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 5593] [Cited by in F6Publishing: 5585] [Article Influence: 797.9] [Reference Citation Analysis (0)] |
29. | Galle PR, Foerster F, Kudo M, Chan SL, Llovet JM, Qin S, Schelman WR, Chintharlapalli S, Abada PB, Sherman M, Zhu AX. Biology and significance of alpha-fetoprotein in hepatocellular carcinoma. Liver Int. 2019;39:2214-2229. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 192] [Cited by in F6Publishing: 340] [Article Influence: 56.7] [Reference Citation Analysis (0)] |
30. | Liebman HA, Furie BC, Tong MJ, Blanchard RA, Lo KJ, Lee SD, Coleman MS, Furie B. Des-gamma-carboxy (abnormal) prothrombin as a serum marker of primary hepatocellular carcinoma. N Engl J Med. 1984;310:1427-1431. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 395] [Cited by in F6Publishing: 402] [Article Influence: 9.8] [Reference Citation Analysis (16)] |
31. | Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 2019;47:W556-W560. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1991] [Cited by in F6Publishing: 2604] [Article Influence: 434.0] [Reference Citation Analysis (0)] |
32. | Kolluri A, Ho M. The Role of Glypican-3 in Regulating Wnt, YAP, and Hedgehog in Liver Cancer. Front Oncol. 2019;9:708. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 47] [Cited by in F6Publishing: 69] [Article Influence: 11.5] [Reference Citation Analysis (0)] |
33. | Sung KC, Ryu S, Kim BS, Cheong ES, Park DI, Kim BI, Kwon MJ, Wild SH, Byrne CD. γ-Glutamyl Transferase Is Associated with Mortality Outcomes Independently of Fatty Liver. Clin Chem. 2015;61:1173-1181. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 15] [Cited by in F6Publishing: 16] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
34. | Huang M, Chen Z, Qin S, Zhou J, Huang Y, Peng S, Huang P, Lin Y, Alenzi M, Huang J, Lin J, Chen Z, Huang M, Liu G. Gamma-glutamyl transferase to aspartate aminotransferase ratio (GSR) predicts prognoses in patients with colorectal cancer with liver metastasis after microwave ablation. BMC Gastroenterol. 2024;24:327. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
35. | Su TH, Huang SC, Chen CL, Hsu SJ, Liao SH, Hong CM, Tseng TC, Liu CH, Yang HC, Wu YM, Liu CJ, Chen PJ, Kao JH. Pre-operative gamma-glutamyl transferase levels predict outcomes in hepatitis B-related hepatocellular carcinoma after curative resection. J Formos Med Assoc. 2023;122:1008-1017. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Reference Citation Analysis (0)] |
36. | Sun F, Wang J, Sun Q, Li F, Gao H, Xu L, Zhang J, Sun X, Tian Y, Zhao Q, Shen H, Zhang K, Liu J. Interleukin-8 promotes integrin β3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 2019;38:449. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 35] [Cited by in F6Publishing: 92] [Article Influence: 15.3] [Reference Citation Analysis (0)] |
37. | Rey I, Effendi-Ys R, Sukatendel K. The Comparison of Serum Interleukin-8 Levels Based on Severity of Liver Cirrhosis. Med Arch. 2024;78:92-94. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
38. | Bi H, Zhang Y, Wang S, Fang W, He W, Yin L, Xue Y, Cheng Z, Yang M, Shen J. Interleukin-8 promotes cell migration via CXCR1 and CXCR2 in liver cancer. Oncol Lett. 2019;18:4176-4184. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 7] [Cited by in F6Publishing: 11] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
39. | Zhang BL, Ji X, Yu LX, Gao Y, Xiao CH, Liu J, Zhao DX, Le Y, Diao GH, Sun JY, Li GH, Lei GL, Yu P, Wang RL, Wu JZ, Yang PH, Yan J, Li JY, Xu JJ, Zhang SG, Tian H. Somatic mutation profiling of liver and biliary cancer by targeted next generation sequencing. Oncol Lett. 2018;16:6003-6012. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 4] [Reference Citation Analysis (0)] |
40. | Budhu A, Wang XW. At the cancer steering wheel: defining key genomic drivers of liver cancer with next generation sequencing. J Hepatol. 2012;56:1412-1414. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
41. | Marquardt JU, Andersen JB. Next-generation sequencing: application in liver cancer-past, present and future? Biology (Basel). 2012;1:383-394. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 12] [Article Influence: 1.1] [Reference Citation Analysis (0)] |
42. | Ye Q, Ling S, Zheng S, Xu X. Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. Mol Cancer. 2019;18:114. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 116] [Cited by in F6Publishing: 225] [Article Influence: 37.5] [Reference Citation Analysis (0)] |
43. | Kopystecka A, Patryn R, Leśniewska M, Budzyńska J, Kozioł I. The Use of ctDNA in the Diagnosis and Monitoring of Hepatocellular Carcinoma-Literature Review. Int J Mol Sci. 2023;24. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 11] [Reference Citation Analysis (0)] |
44. | Hochreuter MY, Dall M, Treebak JT, Barrès R. MicroRNAs in non-alcoholic fatty liver disease: Progress and perspectives. Mol Metab. 2022;65:101581. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 45] [Reference Citation Analysis (0)] |
45. | Wang X, He Y, Mackowiak B, Gao B. MicroRNAs as regulators, biomarkers and therapeutic targets in liver diseases. Gut. 2021;70:784-795. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 139] [Cited by in F6Publishing: 253] [Article Influence: 63.3] [Reference Citation Analysis (0)] |
46. | Ong Y, Huey CWT, Shelat VG. Paraneoplastic syndromes in hepatocellular carcinoma: a review. Expert Rev Gastroenterol Hepatol. 2022;16:449-471. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 4] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
47. | Burciu C, Sirli R, Bende R, Vuletici D, Miutescu B, Moga T, Bende F, Popescu A, Sporea I, Koppandi O, Miutescu E, Iovanescu D, Danila M. Paraneoplastic Syndromes in Hepatocellular Carcinoma, Epidemiology, and Survival: A Retrospective Seven Years Study. Medicina (Kaunas). 2024;60. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
48. | Qu Q, Wang S, Chen S, Zhou L, Rui J. Prognostic Role and Significance of Paraneoplastic Syndromes in Hepatocellular Carcinoma. The American Surgeon™. 2014;80:191-196. [DOI] [Cited in This Article: ] [Cited by in Crossref: 14] [Cited by in F6Publishing: 14] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
49. | Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: An update. Metabolism. 2016;65:1109-1123. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 275] [Cited by in F6Publishing: 406] [Article Influence: 45.1] [Reference Citation Analysis (0)] |
50. | Shin HS, Jun BG, Yi SW. Impact of diabetes, obesity, and dyslipidemia on the risk of hepatocellular carcinoma in patients with chronic liver diseases. Clin Mol Hepatol. 2022;28:773-789. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 30] [Reference Citation Analysis (0)] |
51. | Liu MX, Jin L, Sun SJ, Liu P, Feng X, Cheng ZL, Liu WR, Guan KL, Shi YH, Yuan HX, Xiong Y. Metabolic reprogramming by PCK1 promotes TCA cataplerosis, oxidative stress and apoptosis in liver cancer cells and suppresses hepatocellular carcinoma. Oncogene. 2018;37:1637-1653. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 73] [Cited by in F6Publishing: 127] [Article Influence: 18.1] [Reference Citation Analysis (0)] |
52. | Yoo JJ, Cho EJ, Han K, Heo SS, Kim BY, Shin DW, Yu SJ. Glucose Variability and Risk of Hepatocellular Carcinoma in Patients with Diabetes: A Nationwide Population-Based Study. Cancer Epidemiol Biomarkers Prev. 2021;30:974-981. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 6] [Cited by in F6Publishing: 12] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
53. | Legros L, Pascale A, Guettier C, Eftekhari P, Merabet YB, Stang M, Bossevot R, Goldschmidt E, Ulusakarya A, Morisset S, Lewin M, Samuel D, Rosmorduc O. Progressive erythrocytosis under lenvatinib treatment in patients with advanced hepatocellular carcinoma. Cancer Chemother Pharmacol. 2023;91:337-344. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
54. | Ke S, Chen S, Dong Z, Hong CS, Zhang Q, Tang L, Yang P, Zhai J, Yan H, Shen F, Zhuang Z, Wen W, Wang H. Erythrocytosis in hepatocellular carcinoma portends poor prognosis by respiratory dysfunction secondary to mitochondrial DNA mutations. Hepatology. 2017;65:134-151. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 14] [Cited by in F6Publishing: 20] [Article Influence: 2.5] [Reference Citation Analysis (0)] |
55. | Newman NB, Jabbour SK, Hon JD, Berman JJ, Malik D, Carpizo D, Moss RA. Hepatocellular Carcinoma Without Cirrhosis Presenting With Hypercalcemia: Case Report and Literature Review. J Clin Exp Hepatol. 2015;5:163-166. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 11] [Cited by in F6Publishing: 10] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
56. | Bashir AM, Mohamed AH, Mohamed HN, Ibrahim IG. Severe Hypercalcemia as an Initial Presentation of Advanced Hepatocellular Carcinoma: A Case Report. Cancer Manag Res. 2022;14:1577-1580. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
57. | Iqbal R, Solipuram D, Mohammed YN, Bajwa AT, Irfan A, Jafar A, Rehman Z, Islam ZU. Impact of Hypoglycemia on Hospitalized Patients With Hepatocellular Carcinoma. Cureus. 2024;16:e64673. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
58. | Yu J, Liang X, Chen Y, Huang F, Fan W, Xue J, Li C. Hepatocellular carcinoma with worsened hypoglycemia after transarterial chemoembolization: a case report and systematic review. Int J Clin Exp Pathol. 2020;13:3167-3173. [PubMed] [Cited in This Article: ] |
59. | Gómez Torres KM, Molina Villalba C, Estévez Escobar M. Lenvatinib in severe hypoglycemia associated with hepatocellular carcinoma. Rev Esp Enferm Dig. 2021;113:799. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 1] [Article Influence: 0.3] [Reference Citation Analysis (0)] |
60. | Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. Genetic Landscape and Biomarkers of Hepatocellular Carcinoma. Gastroenterology. 2015;149:1226-1239.e4. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 744] [Cited by in F6Publishing: 891] [Article Influence: 89.1] [Reference Citation Analysis (0)] |
61. | Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, Couchy G, Meiller C, Shinde J, Soysouvanh F, Calatayud AL, Pinyol R, Pelletier L, Balabaud C, Laurent A, Blanc JF, Mazzaferro V, Calvo F, Villanueva A, Nault JC, Bioulac-Sage P, Stratton MR, Llovet JM, Zucman-Rossi J. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 2015;47:505-511. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1006] [Cited by in F6Publishing: 1260] [Article Influence: 126.0] [Reference Citation Analysis (0)] |
62. | Calderaro J, Couchy G, Imbeaud S, Amaddeo G, Letouzé E, Blanc JF, Laurent C, Hajji Y, Azoulay D, Bioulac-Sage P, Nault JC, Zucman-Rossi J. Histological subtypes of hepatocellular carcinoma are related to gene mutations and molecular tumour classification. J Hepatol. 2017;67:727-738. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 373] [Cited by in F6Publishing: 489] [Article Influence: 61.1] [Reference Citation Analysis (0)] |
63. | Nagaraju GP, Dariya B, Kasa P, Peela S, El-Rayes BF. Epigenetics in hepatocellular carcinoma. Semin Cancer Biol. 2022;86:622-632. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 7] [Cited by in F6Publishing: 115] [Article Influence: 38.3] [Reference Citation Analysis (0)] |
64. | Rajan PK, Udoh UA, Sanabria JD, Banerjee M, Smith G, Schade MS, Sanabria J, Sodhi K, Pierre S, Xie Z, Shapiro JI, Sanabria J. The Role of Histone Acetylation-/Methylation-Mediated Apoptotic Gene Regulation in Hepatocellular Carcinoma. Int J Mol Sci. 2020;21. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 18] [Cited by in F6Publishing: 32] [Article Influence: 6.4] [Reference Citation Analysis (0)] |
65. | Raggi C, Invernizzi P. Methylation and liver cancer. Clin Res Hepatol Gastroenterol. 2013;37:564-571. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 13] [Cited by in F6Publishing: 14] [Article Influence: 1.2] [Reference Citation Analysis (0)] |
66. | Kostyusheva A, Brezgin S, Glebe D, Kostyushev D, Chulanov V. Host-cell interactions in HBV infection and pathogenesis: the emerging role of m6A modification. Emerg Microbes Infect. 2021;10:2264-2275. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 30] [Cited by in F6Publishing: 25] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
67. | Yang YM, Kim SY, Seki E. Inflammation and Liver Cancer: Molecular Mechanisms and Therapeutic Targets. Semin Liver Dis. 2019;39:26-42. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 218] [Cited by in F6Publishing: 263] [Article Influence: 43.8] [Reference Citation Analysis (0)] |
68. | Liu S, Yang X. Intestinal flora plays a role in the progression of hepatitis-cirrhosis-liver cancer. Front Cell Infect Microbiol. 2023;13:1140126. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 4] [Reference Citation Analysis (0)] |
69. | Moon AM, Singal AG, Tapper EB. Contemporary Epidemiology of Chronic Liver Disease and Cirrhosis. Clin Gastroenterol Hepatol. 2020;18:2650-2666. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 470] [Cited by in F6Publishing: 612] [Article Influence: 122.4] [Reference Citation Analysis (0)] |
70. | Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev. 2017;121:27-42. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 530] [Cited by in F6Publishing: 931] [Article Influence: 116.4] [Reference Citation Analysis (0)] |
71. | Xu Y, Zhao W, Xu J, Li J, Hong Z, Yin Z, Wang X. Activated hepatic stellate cells promote liver cancer by induction of myeloid-derived suppressor cells through cyclooxygenase-2. Oncotarget. 2016;7:8866-8878. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 44] [Cited by in F6Publishing: 65] [Article Influence: 8.1] [Reference Citation Analysis (0)] |
72. | Sun L, Cai J, Gonzalez FJ. The role of farnesoid X receptor in metabolic diseases, and gastrointestinal and liver cancer. Nat Rev Gastroenterol Hepatol. 2021;18:335-347. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 80] [Cited by in F6Publishing: 200] [Article Influence: 50.0] [Reference Citation Analysis (0)] |
73. | Yahoo N, Dudek M, Knolle P, Heikenwälder M. Role of immune responses in the development of NAFLD-associated liver cancer and prospects for therapeutic modulation. J Hepatol. 2023;79:538-551. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 41] [Reference Citation Analysis (0)] |
74. | Zhou Y, Li Y, Zhou T, Zheng J, Li S, Li HB. Dietary Natural Products for Prevention and Treatment of Liver Cancer. Nutrients. 2016;8:156. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 172] [Cited by in F6Publishing: 177] [Article Influence: 19.7] [Reference Citation Analysis (0)] |
75. | Cao W, Yu P, Yang K, Cao D. Aflatoxin B1: metabolism, toxicology, and its involvement in oxidative stress and cancer development. Toxicol Mech Methods. 2022;32:395-419. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 4] [Cited by in F6Publishing: 75] [Article Influence: 18.8] [Reference Citation Analysis (0)] |
76. | Lu C, Rong D, Zhang B, Zheng W, Wang X, Chen Z, Tang W. Current perspectives on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol Cancer. 2019;18:130. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 142] [Cited by in F6Publishing: 266] [Article Influence: 44.3] [Reference Citation Analysis (0)] |
77. | Donne R, Lujambio A. The liver cancer immune microenvironment: Therapeutic implications for hepatocellular carcinoma. Hepatology. 2023;77:1773-1796. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 114] [Cited by in F6Publishing: 204] [Article Influence: 102.0] [Reference Citation Analysis (0)] |
78. | Lawal G, Xiao Y, Rahnemai-Azar AA, Tsilimigras DI, Kuang M, Bakopoulos A, Pawlik TM. The Immunology of Hepatocellular Carcinoma. Vaccines (Basel). 2021;9. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 9] [Cited by in F6Publishing: 34] [Article Influence: 8.5] [Reference Citation Analysis (0)] |
79. | Muppala S. Significance of the Tumor Microenvironment in Liver Cancer Progression. Crit Rev Oncog. 2020;25:1-9. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 5] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
80. | Wei C, Yang C, Wang S, Shi D, Zhang C, Lin X, Liu Q, Dou R, Xiong B. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol Cancer. 2019;18:64. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 238] [Cited by in F6Publishing: 513] [Article Influence: 85.5] [Reference Citation Analysis (1)] |
81. | Giguelay A, Turtoi E, Khelaf L, Tosato G, Dadi I, Chastel T, Poul MA, Pratlong M, Nicolescu S, Severac D, Adenis A, Sgarbura O, Carrère S, Rouanet P, Quenet F, Ychou M, Pourquier D, Colombo PE, Turtoi A, Colinge J. The landscape of cancer-associated fibroblasts in colorectal cancer liver metastases. Theranostics. 2022;12:7624-7639. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
82. | Deng S, Cheng D, Wang J, Gu J, Xue Y, Jiang Z, Qin L, Mao F, Cao Y, Cai K. MYL9 expressed in cancer-associated fibroblasts regulate the immune microenvironment of colorectal cancer and promotes tumor progression in an autocrine manner. J Exp Clin Cancer Res. 2023;42:294. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 2] [Reference Citation Analysis (0)] |
83. | Qiu S, Xie L, Lu C, Gu C, Xia Y, Lv J, Xuan Z, Fang L, Yang J, Zhang L, Li Z, Wang W, Xu H, Li B, Xu Z. Gastric cancer-derived exosomal miR-519a-3p promotes liver metastasis by inducing intrahepatic M2-like macrophage-mediated angiogenesis. J Exp Clin Cancer Res. 2022;41:296. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 93] [Reference Citation Analysis (0)] |
84. | Vogt A, Sadeghlar F, Ayub TH, Schneider C, Möhring C, Zhou T, Mahn R, Bartels A, Praktiknjo M, Kornek MT, Toma M, Schmidt-Wolf IGH, Branchi V, Matthaei H, Kalff JC, Strassburg CP, Gonzalez-Carmona MA. Alpha-Fetoprotein- and CD40Ligand-Expressing Dendritic Cells for Immunotherapy of Hepatocellular Carcinoma. Cancers (Basel). 2021;13. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 15] [Article Influence: 3.8] [Reference Citation Analysis (0)] |
85. | Dai H, Tong C, Shi D, Chen M, Guo Y, Chen D, Han X, Wang H, Wang Y, Shen P. Efficacy and biomarker analysis of CD133-directed CAR T cells in advanced hepatocellular carcinoma: a single-arm, open-label, phase II trial. Oncoimmunology. 2020;9:1846926. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 24] [Cited by in F6Publishing: 91] [Article Influence: 18.2] [Reference Citation Analysis (0)] |
86. | First-Ever CAR T-cell Therapy Approved in U.S. Cancer Discov. 2017;7:OF1. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 33] [Cited by in F6Publishing: 44] [Article Influence: 5.5] [Reference Citation Analysis (0)] |
87. | Mullard A. FDA approves second BCMA-targeted CAR-T cell therapy. Nat Rev Drug Discov. 2022;21:249. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 5] [Article Influence: 1.7] [Reference Citation Analysis (0)] |
88. | Xiao X, Lao XM, Chen MM, Liu RX, Wei Y, Ouyang FZ, Chen DP, Zhao XY, Zhao Q, Li XF, Liu CL, Zheng L, Kuang DM. PD-1hi Identifies a Novel Regulatory B-cell Population in Human Hepatoma That Promotes Disease Progression. Cancer Discov. 2016;6:546-559. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 185] [Cited by in F6Publishing: 240] [Article Influence: 26.7] [Reference Citation Analysis (0)] |
89. | Guo X, Jiang H, Shi B, Zhou M, Zhang H, Shi Z, Du G, Luo H, Wu X, Wang Y, Sun R, Li Z. Disruption of PD-1 Enhanced the Anti-tumor Activity of Chimeric Antigen Receptor T Cells Against Hepatocellular Carcinoma. Front Pharmacol. 2018;9:1118. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 113] [Cited by in F6Publishing: 138] [Article Influence: 19.7] [Reference Citation Analysis (0)] |
90. | Shi F, Shi M, Zeng Z, Qi RZ, Liu ZW, Zhang JY, Yang YP, Tien P, Wang FS. PD-1 and PD-L1 upregulation promotes CD8(+) T-cell apoptosis and postoperative recurrence in hepatocellular carcinoma patients. Int J Cancer. 2011;128:887-896. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 291] [Cited by in F6Publishing: 359] [Article Influence: 25.6] [Reference Citation Analysis (0)] |
91. | Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, Verslype C, Zagonel V, Fartoux L, Vogel A, Sarker D, Verset G, Chan SL, Knox J, Daniele B, Webber AL, Ebbinghaus SW, Ma J, Siegel AB, Cheng AL, Kudo M; KEYNOTE-224 investigators. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol. 2018;19:940-952. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1184] [Cited by in F6Publishing: 1776] [Article Influence: 253.7] [Reference Citation Analysis (0)] |
92. | Kudo M, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer DH, Verslype C, Zagonel V, Fartoux L, Vogel A, Sarker D, Verset G, Chan SL, Knox J, Daniele B, Yau T, Gurary EB, Siegel AB, Wang A, Cheng AL, Zhu AX; KEYNOTE-224 Investigators. Updated efficacy and safety of KEYNOTE-224: a phase II study of pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib. Eur J Cancer. 2022;167:1-12. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 62] [Article Influence: 20.7] [Reference Citation Analysis (0)] |
93. | Yau T, Kang YK, Kim TY, El-Khoueiry AB, Santoro A, Sangro B, Melero I, Kudo M, Hou MM, Matilla A, Tovoli F, Knox JJ, Ruth He A, El-Rayes BF, Acosta-Rivera M, Lim HY, Neely J, Shen Y, Wisniewski T, Anderson J, Hsu C. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients With Advanced Hepatocellular Carcinoma Previously Treated With Sorafenib: The CheckMate 040 Randomized Clinical Trial. JAMA Oncol. 2020;6:e204564. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 505] [Cited by in F6Publishing: 861] [Article Influence: 172.2] [Reference Citation Analysis (0)] |
94. | El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, Kim TY, Choo SP, Trojan J, Welling TH Rd, Meyer T, Kang YK, Yeo W, Chopra A, Anderson J, Dela Cruz C, Lang L, Neely J, Tang H, Dastani HB, Melero I. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet. 2017;389:2492-2502. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2536] [Cited by in F6Publishing: 3141] [Article Influence: 392.6] [Reference Citation Analysis (1)] |
95. | Yau T, Kang Y, Kim T, El-khoueiry AB, Santoro A, Sangro B, Melero I, Kudo M, Hou M, Matilla A, Tovoli F, Knox JJ, He AR, El-rayes BF, Acosta-rivera M, Neely J, Shen Y, Baccan C, Dela Cruz CM, Hsu C. Nivolumab (NIVO) + ipilimumab (IPI) combination therapy in patients (pts) with advanced hepatocellular carcinoma (aHCC): Results from CheckMate 040. J Clin Oncol. 2019;37:4012-4012. [DOI] [Cited in This Article: ] [Cited by in Crossref: 153] [Cited by in F6Publishing: 155] [Article Influence: 25.8] [Reference Citation Analysis (0)] |
96. | Shree Harini K, Ezhilarasan D, Mani U. Molecular insights on intracellular Wnt/β-catenin signaling in alcoholic liver disease. Cell Biochem Funct. 2024;42:e3916. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
97. | Song J, Xie C, Jiang L, Wu G, Zhu J, Zhang S, Tang M, Song L, Li J. Transcription factor AP-4 promotes tumorigenic capability and activates the Wnt/β-catenin pathway in hepatocellular carcinoma. Theranostics. 2018;8:3571-3583. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 30] [Cited by in F6Publishing: 56] [Article Influence: 8.0] [Reference Citation Analysis (0)] |
98. | Clevers H, Nusse R. Wnt/β-catenin signaling and disease. Cell. 2012;149:1192-1205. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3740] [Cited by in F6Publishing: 4253] [Article Influence: 327.2] [Reference Citation Analysis (0)] |
99. | Tacchelly-Benites O, Wang Z, Yang E, Lee E, Ahmed Y. Toggling a conformational switch in Wnt/β-catenin signaling: regulation of Axin phosphorylation. The phosphorylation state of Axin controls its scaffold function in two Wnt pathway protein complexes. Bioessays. 2013;35:1063-1070. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 22] [Cited by in F6Publishing: 24] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
100. | Schaefer KN, Peifer M. Wnt/Beta-Catenin Signaling Regulation and a Role for Biomolecular Condensates. Dev Cell. 2019;48:429-444. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 148] [Cited by in F6Publishing: 123] [Article Influence: 20.5] [Reference Citation Analysis (0)] |
101. | Russell JO, Monga SP. Wnt/β-Catenin Signaling in Liver Development, Homeostasis, and Pathobiology. Annu Rev Pathol. 2018;13:351-378. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 178] [Cited by in F6Publishing: 296] [Article Influence: 37.0] [Reference Citation Analysis (0)] |
102. | Lu LC, Shao YY, Lee YH, Hsieh MS, Hsiao CH, Lin HH, Kao HF, Ma YY, Yen FC, Cheng AL, Hsu CH. β-catenin (CTNNB1) mutations are not associated with prognosis in advanced hepatocellular carcinoma. Oncology. 2014;87:159-166. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 20] [Cited by in F6Publishing: 28] [Article Influence: 2.5] [Reference Citation Analysis (0)] |
103. | Monga SP. β-Catenin Signaling and Roles in Liver Homeostasis, Injury, and Tumorigenesis. Gastroenterology. 2015;148:1294-1310. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 363] [Cited by in F6Publishing: 498] [Article Influence: 49.8] [Reference Citation Analysis (0)] |
104. | Calderaro J, Ziol M, Paradis V, Zucman-Rossi J. Molecular and histological correlations in liver cancer. J Hepatol. 2019;71:616-630. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 198] [Cited by in F6Publishing: 321] [Article Influence: 53.5] [Reference Citation Analysis (0)] |
105. | Longato L, de la Monte S, Kuzushita N, Horimoto M, Rogers AB, Slagle BL, Wands JR. Overexpression of insulin receptor substrate-1 and hepatitis Bx genes causes premalignant alterations in the liver. Hepatology. 2009;49:1935-1943. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 50] [Cited by in F6Publishing: 63] [Article Influence: 3.9] [Reference Citation Analysis (0)] |
106. | Harada N, Oshima H, Katoh M, Tamai Y, Oshima M, Taketo MM. Hepatocarcinogenesis in mice with beta-catenin and Ha-ras gene mutations. Cancer Res. 2004;64:48-54. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 145] [Cited by in F6Publishing: 151] [Article Influence: 7.2] [Reference Citation Analysis (0)] |
107. | Zhan N, Michael AA, Wu K, Zeng G, Bell A, Tao J, Monga SP. The Effect of Selective c-MET Inhibitor on Hepatocellular Carcinoma in the MET-Active, β-Catenin-Mutated Mouse Model. Gene Expr. 2018;18:135-147. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 12] [Cited by in F6Publishing: 11] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
108. | Lin YW, Shih YL, Lien GS, Suk FM, Hsieh CB, Yan MD. Promoter methylation of SFRP3 is frequent in hepatocellular carcinoma. Dis Markers. 2014;2014:351863. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 12] [Cited by in F6Publishing: 15] [Article Influence: 1.4] [Reference Citation Analysis (0)] |
109. | Wang R, Yu Z, Chen F, Xu H, Shen S, Chen W, Chen L, Su Q, Zhang L, Bi J, Zeng W, Li W, Huang X, Wang Q. miR-300 regulates the epithelial-mesenchymal transition and invasion of hepatocellular carcinoma by targeting the FAK/PI3K/AKT signaling pathway. Biomed Pharmacother. 2018;103:1632-1642. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 25] [Cited by in F6Publishing: 34] [Article Influence: 4.9] [Reference Citation Analysis (0)] |
110. | Xia H, Zhang Z, Luo C, Wei K, Li X, Mu X, Duan M, Zhu C, Jin L, He X, Tang L, Hu L, Guan Y, Lam DCC, Yang J. MultiPrime: A reliable and efficient tool for targeted next-generation sequencing. Imeta. 2023;2:e143. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 7] [Cited by in F6Publishing: 4] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
111. | Jelin AC, Vora N. Whole Exome Sequencing: Applications in Prenatal Genetics. Obstet Gynecol Clin North Am. 2018;45:69-81. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 44] [Cited by in F6Publishing: 75] [Article Influence: 10.7] [Reference Citation Analysis (0)] |
112. | Zhao EY, Jones M, Jones SJM. Whole-Genome Sequencing in Cancer. Cold Spring Harb Perspect Med. 2019;9. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 30] [Cited by in F6Publishing: 29] [Article Influence: 4.8] [Reference Citation Analysis (0)] |
113. | Lin J, Zhao S, Wang D, Song Y, Che Y, Yang X, Mao J, Xie F, Long J, Bai Y, Yang X, Zhang L, Bian J, Lu X, Sang X, Pan J, Wang K, Zhao H. Targeted Next-Generation Sequencing Combined With Circulating-Free DNA Deciphers Spatial Heterogeneity of Resected Multifocal Hepatocellular Carcinoma. Front Immunol. 2021;12:673248. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 2] [Article Influence: 0.5] [Reference Citation Analysis (0)] |
114. | Gnirke A, Melnikov A, Maguire J, Rogov P, LeProust EM, Brockman W, Fennell T, Giannoukos G, Fisher S, Russ C, Gabriel S, Jaffe DB, Lander ES, Nusbaum C. Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing. Nat Biotechnol. 2009;27:182-189. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1052] [Cited by in F6Publishing: 1036] [Article Influence: 64.8] [Reference Citation Analysis (0)] |
115. | Liu YX, Zhang SF, Ji YH, Guo SJ, Wang GF, Zhang GW. Whole-exome sequencing identifies mutated PCK2 and HUWE1 associated with carcinoma cell proliferation in a hepatocellular carcinoma patient. Oncol Lett. 2012;4:847-851. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 10] [Cited by in F6Publishing: 13] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
116. | Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, Calderaro J, Bioulac-Sage P, Letexier M, Degos F, Clément B, Balabaud C, Chevet E, Laurent A, Couchy G, Letouzé E, Calvo F, Zucman-Rossi J. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012;44:694-698. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 962] [Cited by in F6Publishing: 1094] [Article Influence: 84.2] [Reference Citation Analysis (0)] |
117. | Zhou Q, Li Z, Song L, Mu D, Wang J, Tian L, Liao Y. Whole-exome mutational landscape of metastasis in patient-derived hepatocellular carcinoma cells. Genes Dis. 2020;7:380-391. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 5] [Cited by in F6Publishing: 5] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
118. | Fujimoto A, Furuta M, Totoki Y, Tsunoda T, Kato M, Shiraishi Y, Tanaka H, Taniguchi H, Kawakami Y, Ueno M, Gotoh K, Ariizumi S, Wardell CP, Hayami S, Nakamura T, Aikata H, Arihiro K, Boroevich KA, Abe T, Nakano K, Maejima K, Sasaki-Oku A, Ohsawa A, Shibuya T, Nakamura H, Hama N, Hosoda F, Arai Y, Ohashi S, Urushidate T, Nagae G, Yamamoto S, Ueda H, Tatsuno K, Ojima H, Hiraoka N, Okusaka T, Kubo M, Marubashi S, Yamada T, Hirano S, Yamamoto M, Ohdan H, Shimada K, Ishikawa O, Yamaue H, Chayama K, Miyano S, Aburatani H, Shibata T, Nakagawa H. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat Genet. 2016;48:500-509. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 465] [Cited by in F6Publishing: 518] [Article Influence: 57.6] [Reference Citation Analysis (0)] |
119. | Zhou SL, Zhou ZJ, Song CL, Xin HY, Hu ZQ, Luo CB, Luo YJ, Li J, Dai Z, Yang XR, Shi YH, Wang Z, Huang XW, Fan J, Zhou J. Whole-genome sequencing reveals the evolutionary trajectory of HBV-related hepatocellular carcinoma early recurrence. Signal Transduct Target Ther. 2022;7:24. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1] [Cited by in F6Publishing: 10] [Article Influence: 3.3] [Reference Citation Analysis (0)] |
120. | Liu Y, Al-Adra DP, Lan R, Jung G, Li H, Yeh MM, Liu YZ. RNA sequencing analysis of hepatocellular carcinoma identified oxidative phosphorylation as a major pathologic feature. Hepatol Commun. 2022;6:2170-2181. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 9] [Cited by in F6Publishing: 7] [Article Influence: 2.3] [Reference Citation Analysis (0)] |
121. | Zhang Q, Lou Y, Yang J, Wang J, Feng J, Zhao Y, Wang L, Huang X, Fu Q, Ye M, Zhang X, Chen Y, Ma C, Ge H, Wang J, Wu J, Wei T, Chen Q, Wu J, Yu C, Xiao Y, Feng X, Guo G, Liang T, Bai X. Integrated multiomic analysis reveals comprehensive tumour heterogeneity and novel immunophenotypic classification in hepatocellular carcinomas. Gut. 2019;68:2019-2031. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 154] [Cited by in F6Publishing: 215] [Article Influence: 35.8] [Reference Citation Analysis (0)] |
122. | Zheng B, Wang D, Qiu X, Luo G, Wu T, Yang S, Li Z, Zhu Y, Wang S, Wu R, Sui C, Gu Z, Shen S, Jeong S, Wu X, Gu J, Wang H, Chen L. Trajectory and Functional Analysis of PD-1(high) CD4(+)CD8(+) T Cells in Hepatocellular Carcinoma by Single-Cell Cytometry and Transcriptome Sequencing. Adv Sci (Weinh). 2020;7:2000224. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 59] [Cited by in F6Publishing: 67] [Article Influence: 13.4] [Reference Citation Analysis (0)] |