Skip Navigation
Skip to contents

JLC : Journal of Liver Cancer

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Liver Cancer > Volume 25(1); 2025 > Article
Review Article
Molecular and immune landscape of hepatocellular carcinoma for therapeutic development
Hiroyuki Suzukiorcid, Sumit Mishraorcid, Subhojit Paulorcid, Yujin Hoshidaorcid
Journal of Liver Cancer 2025;25(1):9-18.
DOI: https://doi.org/10.17998/jlc.2024.12.02
Published online: December 6, 2024

Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA

Corresponding author: Yujin Hoshida, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA E-mail: Yujin.Hoshida@UTSouthwestern.edu
• Received: October 30, 2024   • Revised: November 26, 2024   • Accepted: December 2, 2024

© 2025 The Korean Liver Cancer Association.

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

  • 2,141 Views
  • 302 Downloads
  • 2 Citations
prev next
  • Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with an estimated 750,000 deaths in 2022. Recent emergence of molecular targeted agents and immune checkpoint inhibitors and their combination therapies have been transforming HCC care, but their prognostic impact in advanced-stage disease remains unsatisfactory. In addition, their application to early-stage disease is still an unmet need. Omics profiling studies have elucidated recurrent and heterogeneously present molecular aberrations involved in pro-cancer tumor (immune) microenvironment that may guide therapeutic strategies. Recurrent aberrations such somatic mutations in TERT promoter and TP53 have been regarded undruggable, but recent studies have suggested that these may serve as new classes of therapeutic targets. HCC markers such as alpha-fetoprotein, glypican-3, and epithelial cell adhesion molecule have also been explored as therapeutic targets. These molecular features may be utilized as biomarkers to guide the application of new approaches as companion biomarkers to maximize therapeutic benefits in patients who are likely to benefit from the therapies, while minimizing unnecessary harm in patients who will not respond. The explosive number of new agents in the pipelines have posed challenges in their clinical testing. Novel clinical trial designs guided by predictive biomarkers have been proposed to enable their efficient and cost-effective evaluation. These new developments collectively facilitate clinical translation of personalized molecular-targeted therapies in HCC and substantially improve prognosis of HCC patients.
Globally, liver cancer, predominantly hepatocellular carcinoma (HCC), ranks as the third most common cause of cancer-related mortality. In 2022, it was responsible for approximately 750,000 fatalities worldwide.1,2 The emergence of technologies based on next-generation sequencing has significantly enhanced our comprehension of the molecular and immunological characteristics of HCC.3 In addition, the treatment landscape for advanced-stage HCC has undergone drastic changes due to the approval of molecular targeted agents (MTAs) and immune checkpoint inhibitors (ICIs) in the past 5 years.4 Although the introduction of these new therapeutic options is promising, their impact on survival rates in advanced-stage HCC remains modest, with median survival still falling short of 2 years. Experimental studies and genomic data are anticipated to provide molecular insights that will soon influence the clinical management of patients with HCC. However, the optimal sequencing of these novel therapies remains uncertain. Recent pre-clinical and clinical investigations have indicated that the response to these molecularly targeted treatments may be affected by clinical factors, such as the etiology of liver disease. Therefore, an in-depth understanding the molecular pathogenesis of HCC in specific clinical scenarios is essential to optimize the application of novel therapies for maximum effectiveness. This review serves as a roadmap for clinical trials and the implementation of new therapeutic approaches by offering a comprehensive overview of the molecular and immunological landscape of HCC.
Extensive multicenter studies have performed in-depth genomic phenotyping of human HCC tumors and the tumor microenvironment (TME)/tumor immune microenvironment (TIME), identifying key molecular characteristics driving/supporting cancer initiation/progression (Fig. 1A).5 A thorough grasp of the intricacies and diversity within the molecular and immune landscape of HCC and its TME/TIME will offer insights for personalized treatment approaches. The variations within and between tumors arise not only from inherent epi/genetic abnormalities in cancer cells but also from shifts in the makeup and stromal components within the TME/TIME. Comprehensive multi-omic examination of whole tissue, individual cells/nuclei, and spatially mapped tissue has uncovered a wide range of molecular aberrations in HCC tumors and their microenvironment.6-16 Some molecular pathways/hallmarks can be targeted using approved or tested clinical agents, while others, previously considered untreatable, are now being explored as novel therapeutic targets (Fig. 1B). These include frequently mutated regions (e.g., TERT promoter [TERTp], TP53, CTNNB1). In this context, we discuss current and reported molecular and immune mechanisms that could serve as potential therapeutic strategies.6-11,17
Telomere maintenance mechanisms (TMM)
The disruption of TMM, including the reactivation of telomerase, is a crucial aspect in achieving replicative immortality, which is considered one of the fundamental characteristics in multiple cancer types including HCC.18,19 Somatic alterations in the TERTp are the most common structural changes found in human HCC, occurring in 40-60% of cases, thus, these modifications can take the form of nucleotide substitutions, insertions or deletions, and the integration of viral DN.6-11,17,20,21 The occurrence of somatic TERTp mutations in early-stage HCC, dysplastic nodules of varying grades, and cell-free DNA from cirrhosis patients without clinically detectable HCC indicates their role in the initial stages of HCC formation. This finding also suggests that detecting TERTp mutations could be valuable for assessing risk and/or identifying HCC in its early phases.22-27 The reactivation of telomerase was linked to worse recurrence-free survival, indicating its role in disease advancement.28-30 Conversely, a recent case-control investigation discovered a novel single nucleotide polymorphism in the TERT gene (rs2242652) that appeared to protect against HCC in patients with alcohol-induced cirrhosis.31 This suggests that telomere length may be a factor in the molecular heterogeneity of HCC tumors.32 The precise mechanistic involvement of these mutations in HCC initiation and progression remains unclear. Nevertheless, due to the high frequency of TERTp mutations in HCC and other cancers, TMM has emerged as a potential therapeutic target.33 For example, pre-clinical HCC models have shown promising anti-HCC effects using antisense oligonucleotides that target TERT transcripts.32 Additionally, a nucleoside analog called 6-thio-dG (THIO) has been found to cause telomeric DNA damage in telomerase-activated HCC cells, stimulate innate and adaptive anti-tumor immunity, and improve the response to ICIs in mice.34 These TMM-directed therapeutic approaches, along with others, may prove valuable in the clinical management of individuals at risk for HCC.
Wnt/beta-catenin
The Wnt signaling is an essential pathway in liver development and homeostasis, regulated by a key transcriptional mediator, beta-catenin encoded by the CTNNB1 gene. CTNNB1 is among the most frequently mutated genes in approximately 20-40% of HCC tumors.35 In contrast to other cancer types where CTNNB1 mutations are associated with induction of oncogenic canonical Wnt signaling, these mutations are accompanied with induction of glutamate-ammonia ligase (also known as glutamine synthase) encoded by GLUL gene, more differentiated histology, immune exclusion, and better prognosis in HCC.35-37 Of note, canonical Wnt signaling can be induced in HCC tumors without CTNNB1 mutations.35,38 Wnt/beta-catenin pathway is now actively explored as a therapeutic target in multiple cancer types and non-cancer conditions by utilizing various strategies.39-41 An ongoing phase I/II trial is evaluating a dickkopf-1 (DKK1) inhibitor in patients whose tumors display positive glutamine synthase immunostaining as a surrogate marker of CTNNB1 mutations (NCT03645980).
TP53
TP53 gene, which encodes the p53 tumor protein, is frequently mutated in advanced-stage HCC and is linked to aflatoxin B1 food contamination.11,42-48 Loss-of-function mutations in this tumor suppressor gene and its related genes (TP73L, TP63, CDKN2A, MDM2/MDM4) are associated with aggressive cancer characteristics, including stemness features and resistance to therapy.49-53 A study using a rodent model suggested that p53 function may be compromised in early HCC development through CD44/STAT3 signaling, independent of somatic mutations.54 Another rodent model demonstrated that TP53 loss works in conjunction with c-MET to promote liver cancer development.55 Researchers are actively investigating therapeutic approaches to restore p53 function in pre-clinical studies and early-phase clinical trials. These methods include refolding mutant p53 and inhibiting MDM2, which may soon be applicable to HCC cases with TP53 mutations.56,57 Additionally, adenoviral delivery of the p53 tumor suppressor protein has been evaluated in solid tumors without dominant-negative TP53 mutations (NCT03544723).
Alpha-fetoprotein (AFP)
While the limited positivity of AFP (around 30%) restricts its use as a tumor detection biomarker, it may still prove valuable as an indicator of specific biological/clinical characteristics and treatment responses. Research has demonstrated a connection between high AFP levels and tumor progression, resistance to cell death, and blood vessel formation. These effects might be countered by medications targeting relevant molecular pathways, although the precise mechanisms of therapeutic response, such as in the case of ramucirumab, are not always well understood. Recent investigations have also indicated that AFP might aid in tumor immune evasion by suppressing dendritic cells, natural killer (NK) cells, and T-cells.58 While AFP itself has weak immunogenicity, AFP-targeted immune engineering strategies, including chimeric antigen receptor (CAR) T cell therapy and specific peptide enhanced affinity receptor (SPEAR) T cell therapy, have undergone phase I/II clinical trials.59 Peptide vaccines targeting have been developed AFP and other cancer-related proteins and neoantigens, including WT-I, ROBO1, FOXM1, HER3, and mutant TP53.60
Glypican-3 (GPC3)
GPC3, a heparan sulfate proteoglycan on cell surfaces, serves as an immunohistochemical indicator for HCC.61 Epithelial cell adhesion molecule (EpCAM), a marker for hepatic stem/progenitor cells on cell surfaces, is highly expressed in various gastrointestinal cancers, including HCC.62,63 GPC3 (and EpCAM), like AFP, is expressed in a subset of HCC tumors exhibiting stemness characteristics, poor outcomes, and a strong association with the Hoshida S2 subtype (Fig. 1A).38,63,64 GPC3 is also an ideal target for immune-engineered approaches, such as CAR T-cell therapy, currently evaluated in multiple ongoing phase I/II trials (Table 1).64 Codrituzumab (GC33), a humanized monoclonal antibody targeting human GPC3, did not demonstrate anti-tumor effects in a phase II trial. However, it may be useful as a molecular imaging probe when combined with the positron emission tomography radiotracer, I-124.65,66 ERY974, a bispecific antibody designed to redirect T cells to GPC3-positive tumor cells by targeting GPC3/CD3, showed general tolerability in patients with advanced solid tumors during a phase I trial.67
Suppressive immune checkpoints
ICIs, including those targeting programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) pathways, have become crucial elements in the systemic treatment of cancer, including HCC. These agents work by modulating the interactions between tumor and immune cells and are often used in combination with MTAs or other ICIs. There is still no approved companion biomarker for ICI-based therapy specifically for HCC, but several immune-related features such as PD-L1 expression, tumor mutation burden (TMB), and DNA repair defects (e.g., deficient mismatch repair and high microsatellite instability) have been approved for patient selection in several solid cancers such as non-small cell lung cancer.68 In ongoing clinical trials for HCC patients, several molecular features, pembrolizumab (anti-PD-1) plus futibatinib (fibroblast growth factor receptor [FGFR]1-4 inhibitor) have been tested in fibroblast growth factor [FGF]19-positive HCC tumors in phase II trials (NCT04828486). However, despite their importance, reliable biomarkers for predicting patient response to these therapies have not yet been identified.69-71 Currently, a clinical trial (NCT04802876) is evaluating spartalizumab, an anti-PD-1 antibody, in patients exhibiting elevated PDCD1 mRNA expression (encoding PD-1). Recent clinical studies have also focused on targeting established molecular characteristics of HCC, recruiting patients whose tumors display specific molecular alterations. PD-0332991, an inhibitor of CDK4/6 cell cycle regulators, has been studied in HCC patients with intact retinoblastoma (RB) protein (NCT01356628). In HCC patients treated with atezolizumab (anti-PD-L1) and bevacizumab (anti-vascular endothelial growth factor [VEFG]), an increase in Ki-67/PD-1/CD8-positive T cells and TIGIT-positive T cells at 3 weeks after initiation of the therapy was associated with superior progression-free and overall survival, suggesting their role as non-invasive predictive biomarkers of response.72
Vessels that encapsulate tumor clusters (VETC)
VETC pattern is a pathological finding observed in a certain percentage of HCCs and has been demonstrated to correlate with poor patient prognosis.73 VETC-positive HCCs have also been reported to develop under the influence of angiopoietin 2, and the potential efficacy of tyrosine kinase inhibitors has been investigated.74 A study showed that unresectable VETC-positive HCC may benefit from treatment with sorafenib.75 A clinical trial is currently in progress to evaluate the treatment of VETC-high expressing HCC with a combination of an ICI and a tyrosine kinase inhibitor (NCT06461936, NCT06311929).
The positive outcomes of these trials could potentially pave the way for the clinical application of personalized, biomarker-guided treatments for HCC across various therapeutic approaches. This advancement may enable a more scientifically rational and effective tumor management strategy while reducing the risk of unnecessary adverse effects in patients who unlikely benefit from these therapies.
Although ongoing and planned clinical trials still tend to enroll all-comer patients, biomarker-driven trials have increased in recent years, coinciding with the active development of MTAs.76 Predictive biomarkers, which indicate therapeutic response, play a crucial role in innovative clinical trial methodologies like adaptive designs, facilitating the translation of novel therapeutic agents and strategies into clinical practice.35,77
Biomarker enrichment trials seek to enhance the detection of therapeutic benefits by focusing on a subset of clinically eligible patients who test positive for a biologically rational target biomarker (Fig. 2). In these trials, only biomarker-positive patients are randomly assigned to either the study drug or placebo arm. These trials aim to demonstrate the superiority of the biomarker enrichment approach over the traditional allcomer strategy. The efficacy of experimental therapies and strategies is evaluated using endpoints relevant to specific clinical contexts, such as anti-tumor response, disease control, survival advantage, and cost-effectiveness of interventions. To efficiently assess the growing number of new MTAs across multiple cancer types and histologies, master protocol designs have been developed.78 One such design is the basket/bucket trial, which recruits patients with the same molecular target, regardless of tissue type or histology, potentially including HCC alongside other cancer types. Umbrella trials assess various molecularly targeted therapies based on the presence of corresponding biomarkers. Platform trials employ multi-arm/stage designs with an open-ended timeline to evaluate multiple experimental treatments against a shared control group (Fig. 2).
HCC remains the third leading cause of cancer-related mortality, and there are large racial, ethnic, and interindividual disparities in HCC risk and survival.1,2 Our knowledge of whether molecular and genetic factors contribute to these observed differences is limited by a lack of biological samples. Biospecimen collection is the process of collecting, processing, and storing tissue, blood (e.g., ctDNA), or urine samples for research purposes; biospecimens collected through this process are utilized to elucidate the genetic and molecular factors contributing to HCC and to develop novel biomarker assays of significant importance.79,80 These studies facilitate the development of companion biomarkers based on test results that examine the expression of target molecules, the presence of genetic mutations, and genetic polymorphisms of drug-metabolizing enzymes to predict the efficacy and safety of specific drugs. Furthermore, this research will lead to the development of innovative biomarker-focused trial designs, which will result in improved patient selection, optimizing the application of novel therapies and maximizing their efficacy.

Acknowledgement

The figures were created using BioRender.com.

Conflicts of Interest

Yujin Hoshida is shareholder for Alentis Therapeutics and Espervita Therapeutics, advises Helio Genomics, Espertiva Therapeutics, Roche Diagnostics, and Elevar Therapeutics.

Ethics Statement

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

Funding Statement

This study was supported by US National Institutes of Health (CA233794, CA255621, CA282178, CA288375, CA283935), European Commission (ERC-AdG-2020-101021417), Cancer Prevention and Research Institute of Texas (RR180016, RP200554), Uehara Memorial Foundation. The funders had no role in this article.

Data Availability

Not applicable.

Author Contributions

Conceptualization: HS, SM, SP, and YH

Data curation: HS, SM, SP, and YH

Funding acquisition: HS, SM, SP, and YH

Methodology: HS, SM, SP, and YH

Project administration: HS, SM, SP, and YH

Resources: HS, SM, SP, and YH

Supervision: HS, SM, SP, and YH

Visualization: HS, SM, SP, and YH

Writing - original draft: HS, SM, SP, and YH

Writing - review & editing: HS, SM, SP, and YH

Figure 1.
Therapeutic strategies according to molecular and immune landscape of HCC. (A) Molecular subtypes of HCC tumors and associated histological and molecular features. (B) Molecular and cellular targets for approved drugs and therapeutic strategies in development in HCC. CAR-T cells, chimeric antigen receptor T cells; GPC3, glypican-3; AFP, alpha-fetoprotein; EpCAM, epithelial cell adhesion molecule; neoAg, neo antigen; VEGF, vascular endothelial growth factor; FGF, fibroblast growth factor; PDGFβ, platelet-derived growth factor beta; EGF, epidermal growth factor; HGF, hepatocyte growth factor; APC, antigen presenting cell; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; VEGFR, vascular endothelial growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; EGFR, epidermal growth factor receptor; MET, MET proto-oncogene (hepatocyte growth factor receptor); PD-L1, programmed cell death ligand 1; mTOR, mechanistic target of rapamycin; JAK/STAT, Janus kinase/signal transducer and activator of transcription; MAPK, mitogen-activated protein kinase; PI3K/Akt, phosphatidylinositol 3-kinase/protein kinase B; PD-1, programmed cell death protein 1; CDK, cyclin-dependent kinase; DKK1, dickkopf-1; Rb, retinoblastoma; HCC, hepatocellular carcinoma; TGFβ, transforming growth factor beta; TGFβR, transforming growth factor beta receptor; TERT, telomerase reverse transcriptase.
jlc-2024-12-02f1.jpg
Figure 2.
Study designs for biomarker-guided therapeutic clinical trial in HCC. HCC, hepatocellular carcinoma.
jlc-2024-12-02f2.jpg
Table 1.
Ongoing biomarker-guided therapeutic trials in HCC
Clinical context Phase Trial type Agent Class of agent Biomarker-based eligibility NCT number Last updated
Neo-adjuvant theapy II Biomarker enrichment trial Lenvatinib Multi-kinase inhibitor AFP>100 ng/mL, infiltrative/ macrotrabecular pattern NCT05113186 2024
Adjuvant therapy I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06560827 2024
II Biomarker enrichment trial Sintilimab +/- lenvatinib Anti-PD-1 +/- multi-kinase inhibitor VETC positive NCT06461936 2024
IV Biomarker enrichment trial Anti-PD-1 +/- lenvatinib Anti-PD-1 +/- multi-kinase inhibitor VETC positive NCT06311929 2024
First-line systemic therapy I Basket/bucket trial TSR-022 Anti-TIM-3 TIM-3-positive NCT02817633 2024
I/II Biomarker enrichment trial DKN-01 DKK1 inhibitor Glutamine synthetase IHC-positive NCT03645980 2020
First or second-line systemic therapy I Biomarker enrichment trial AFPc332 T cells Engineered T cells AFP≥100 ng/mL NCT03132792 2024
I Biomarker enrichment trial CT0180 cells Engineered T cells GPC3 IHC-positive NCT04756648 2023
I Basket/bucket trial BOXR1030 T cells Engineered T cells GPC3 IHC-positive NCT05120271 2024
I/II Basket/bucket trial CAR-T/TCR-T cells Engineered T cells DR5 or EGFRvIII IHC-positive NCT03941626 2021
II Basket/bucket trial 32 molecular-targeted agents Molecular-targeted agents Positive targets by tumor DNA-seq, IHC NCT02465060 2024
II Basket/bucket trial Spartalizumab Anti-PD-1 PD-1 mRNA, high expression NCT04802876 2023
II Basket/bucket trial Ad-p53 + approved ICI Viral gene therapy + ICI p53 wild type or IHC-negative NCT03544723 2020
Second-line systemic therapy I Biomarker enrichment trial C-TCR055 Engineered T cells AFP IHC-positive NCT04368182 2020
I Biomarker enrichment trial TC-CAR031 Engineered T cells GPC3 IHC-positive NCT05155189 2024
I Basket/bucket trial EpCAM CAR-T cells Engineered T cells EpCAM-positive NCT05028933 2024
I Basket/bucket trial AFP CAR-T cells Engineered T cells AFP IHC-positive or AFP≥400 ng/mL NCT06515314 2024
I Basket/bucket trial p53 MVA vaccine + pembrolizumab MVA vaccine + anti-PD-1 p53 IHC-positive or gene mutation NCT02432963 2024
I Basket/bucket trial HER2 CAR-macrophages Engineered macrophages HER2-positive NCT04660929 2024
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05003895 2024
I Biomarker enrichment trial GPC3 CAR-T cells + fludarabine + cytoxan Engineered T cells + STAT1 inhibitor + alkylating agent GPC3 IHC-positive NCT05103631 2023
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06461624 2024
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06144385 2024
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05620706 2022
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05926726 2023
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05344664 2022
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05783570 2024
I Biomarker enrichment trial GPC3 CAR-NK cells Engineered NK cells GPC3-positive NCT05845502 2024
I/II Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06084884 2024
I/II Biomarker enrichment trial B7H3 CAR-T cells Engineered T cells B7H3-positive NCT05323201 2024
I/II Biomarker enrichment trial ECT204 T cells Engineered T cells GPC3 IHC-positive NCT04864054 2024
I/II Biomarker enrichment trial HBV-TCR T cells (LioCyx-M) +/- lenvatinib Engineered T cells +/- multi-kinase inhibitor HLA class I profile matching NCT05195294 2022
II Biomarker enrichment trial PD-0332991 CDK4/6 inhibitor RB-positive NCT01356628 2023
II Biomarker enrichment trial Futibatinib + pembrolizumab FGFR inhibitor + anti-PD-1 FGF19 mRNA or IHC-positive NCT04828486 2024
Third-line systemic therapy I/II Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06590246 2024
Not defined I Basket/bucket trial IL-15 and IL-21 armored GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06198296 2024
I Biomarker enrichment trial GPC3/TGF-β CAR-T cells Engineered T cells GPC3 WB or IHC-positive NCT03198546 2024
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05070156 2023
I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06478693 2024
I/II Basket/bucket trial HMBD-001 Anti-HER3 HER3-positive NCT05057013 2023
I/II Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05652920 2024
I/II Biomarker enrichment trial RZ-001 + valganciclovir Trans-splicing ribozyme + antiviral hTERT-positive NCT05595473 2022

ClinicalTrials.gov accessed in October 2024.

HCC, hepatocellular carcinoma; NCT, National Clinical Trial; AFP, alpha-fetoprotein; GPC3, glypican-3; CAR, chimeric antigen receptor; IHC, immunohistochemistry; PD-1, programmed death-1; VETC, vessels encapsulating tumor clusters; TIM-3, T cell immunoglobulin and mucin containing protein-3; DKK1, dickkopf-1; TCR, T cell receptor; DR5, death receptor 5; EGFR, epidermal growth factor receptor; DNA-seq, DNA sequencing; Ad, adenoviral; C-TCR, AFP specific T cell receptor transduced T cells; ICI, immune checkpoint inhibitor; EpCAM, epithelial cell adhesion molecule; MVA, modified vaccinia virus ankara; HER, human epidermal growth factor receptor; STAT1, signal transducer and activator of transcription 1; NK, natural killer; HBV, hepatitis B virus; HLA, human leukocyte antigen; CDK, cyclin-dependent kinase; RB, retinoblastoma; FGFR, fibroblast growth factor receptor; FGF, fibroblast growth factor; IL, interleukin; TGF-β, transforming growth factor beta; WB, western blotting; hTERT, human telomerase reverse transcriptase.

  • 1. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229−263.ArticlePubMed
  • 2. Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol 2022;77:1598−1606.ArticlePubMedPMC
  • 3. Suzuki H, Fujiwara N, Singal AG, Baumert TF, Chung RT, Kawaguchi T, et al. Prevention of liver cancer in the era of next-generation antivirals and obesity epidemic. Hepatology 2025;Jan 14 doi: 10.1097/HEP.0000000000001227. [Epub ahead of print].Article
  • 4. Reig M, Forner A, Rimola J, Ferrer-Fàbrega J, Burrel M, Garcia-Criado Á, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol 2022;76:681−693.ArticlePubMedPMC
  • 5. Donne R, Lujambio A. The liver cancer immune microenvironment: therapeutic implications for hepatocellular carcinoma. Hepatology 2023;77:1773−1796.ArticlePubMedPMCPDF
  • 6. Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 2017;169:1327−1341.e23.PubMedPMC
  • 7. Fujimoto A, Furuta M, Totoki Y, Tsunoda T, Kato M, Shiraishi Y, et al. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat Genet 2016;48:500−509.ArticlePubMed
  • 8. Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet 2015;47:505−511.ArticlePubMedPMCPDF
  • 9. Totoki Y, Tatsuno K, Covington KR, Ueda H, Creighton CJ, Kato M, et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat Genet 2014;46:1267−1273.ArticlePubMedPDF
  • 10. Huang J, Deng Q, Wang Q, Li KY, Dai JH, Li N, et al. Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma. Nat Genet 2012;44:1117−1121.ArticlePubMedPDF
  • 11. Nault JC, Martin Y, Caruso S, Hirsch TZ, Bayard Q, Calderaro J, et al. Clinical impact of genomic diversity from early to advanced hepatocellular carcinoma. Hepatology 2020;71:164−182.ArticlePubMedPDF
  • 12. Zhang Q, He Y, Luo N, Patel SJ, Han Y, Gao R, et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 2019;179:829−845.e20.ArticlePubMed
  • 13. Lu Y, Yang A, Quan C, Pan Y, Zhang H, Li Y, et al. A single-cell atlas of the multicellular ecosystem of primary and metastatic hepatocellular carcinoma. Nat Commun 2022;13:4594. ArticlePubMedPMCPDF
  • 14. Sharma A. A single-cell atlas of hepatocellular carcinoma. J Glob Oncol 2019;Suppl 5:46. Article
  • 15. Sun Y, Wu L, Zhong Y, Zhou K, Hou Y, Wang Z, et al. Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell 2021;184:404−421.e16.ArticlePubMed
  • 16. Yao F, Zhan Y, Li C, Lu Y, Chen J, Deng J, et al. Single-cell RNA sequencing reveals the role of phosphorylation-related genes in hepatocellular carcinoma stem cells. Front Cell Dev Biol 2022;9:734287. ArticlePubMedPMC
  • 17. Jhunjhunwala S, Jiang Z, Stawiski EW, Gnad F, Liu J, Mayba O, et al. Diverse modes of genomic alteration in hepatocellular carcinoma. Genome Biol 2014;15:436. ArticlePubMedPMCPDF
  • 18. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov 2022;12:31−46.ArticlePubMedPDF
  • 19. Sieverling L, Hong C, Koser SD, Ginsbach P, Kleinheinz K, Hutter B, et al. Genomic footprints of activated telomere maintenance mechanisms in cancer. Nat Commun 2020;11:733. ArticlePubMedPMC
  • 20. Sze KM, Ho DW, Chiu YT, Tsui YM, Chan LK, Lee JM, et al. Hepatitis B virus-telomerase reverse transcriptase promoter integration harnesses host ELF4, resulting in telomerase reverse transcriptase gene transcription in hepatocellular carcinoma. Hepatology 2021;73:23−40.ArticlePubMedPMCPDF
  • 21. Li CL, Hsu CL, Lin YY, Ho MC, Hu RH, Chen CL, et al. HBV DNA integration into telomerase or MLL4 genes and TERT promoter point mutation as three independent signatures in subgrouping HBV-related HCC with distinct features. Liver Cancer 2023;13:41−55.ArticlePubMedPMCPDF
  • 22. Torrecilla S, Sia D, Harrington AN, Zhang Z, Cabellos L, Cornella H, et al. Trunk mutational events present minimal intra- and inter-tumoral heterogeneity in hepatocellular carcinoma. J Hepatol 2017;67:1222−1231.ArticlePubMed
  • 23. Nault JC, Mallet M, Pilati C, Calderaro J, Bioulac-Sage P, Laurent C, et al. High frequency of telomerase reverse-transcriptase promoter somatic mutations in hepatocellular carcinoma and preneoplastic lesions. Nat Commun 2013;4:2218. ArticlePubMedPMCPDF
  • 24. Trung NT, Hoan NX, Trung PQ, Binh MT, Van Tong H, Toan NL, et al. Clinical significance of combined circulating TERT promoter mutations and miR-122 expression for screening HBV-related hepatocellular carcinoma. Sci Rep 2020;10:8181. ArticlePubMedPMCPDF
  • 25. Jiao J, Watt GP, Stevenson HL, Calderone TL, Fisher-Hoch SP, Ye Y, et al. Telomerase reverse transcriptase mutations in plasma DNA in patients with hepatocellular carcinoma or cirrhosis: Prevalence and risk factors. Hepatol Commun 2018;2:718−731.ArticlePubMedPMCPDF
  • 26. Lee YT, Fujiwara N, Yang JD, Hoshida Y. Risk stratification and early detection biomarkers for precision HCC screening. Hepatology 2023;78:319−362.ArticlePubMedPMCPDF
  • 27. Mishima M, Takai A, Takeda H, Iguchi E, Nakano S, Fujii Y, et al. TERT upregulation promotes cell proliferation via degradation of p21 and increases carcinogenic potential. J Pathol 2024;264:318−331.PubMed
  • 28. Yu JI, Choi C, Ha SY, Park CK, Kang SY, Joh JW, et al. Clinical importance of TERT overexpression in hepatocellular carcinoma treated with curative surgical resection in HBV endemic area. Sci Rep 2017;7:12258. ArticlePubMedPMCPDF
  • 29. Cheng Y, Huang M, Xie W, Gao C, Cai S, Ji J, et al. Chromosome 8q24 amplification predicts prognosis for patients with advanced hepatocellular carcinoma (HCC). J Clin Oncol 2019;37 Suppl 15:e15654. Article
  • 30. Li X, Xu W, Kang W, Wong SH, Wang M, Zhou Y, et al. Genomic analysis of liver cancer unveils novel driver genes and distinct prognostic features. Theranostics 2018;8:1740−1751.ArticlePubMedPMC
  • 31. Buch S, Innes H, Lutz PL, Nischalke HD, Marquardt JU, Fischer J, et al. Genetic variation in TERT modifies the risk of hepatocellular carcinoma in alcohol-related cirrhosis: results from a genome-wide case-control study. Gut 2023;72:381−391.ArticlePubMedPMC
  • 32. Ningarhari M, Caruso S, Hirsch TZ, Bayard Q, Franconi A, Védie AL, et al. Telomere length is key to hepatocellular carcinoma diversity and telomerase addiction is an actionable therapeutic target. J Hepatol 2021;74:1155−1166.ArticlePubMed
  • 33. Gao J, Pickett HA. Targeting telomeres: advances in telomere maintenance mechanism-specific cancer therapies. Nat Rev Cancer 2022;22:515−532.ArticlePubMedPDF
  • 34. Mender I, Siteni S, Barron S, Flusche AM, Kubota N, Yu C, et al. Activating an adaptive immune response with a telomerase-mediated telomere targeting therapeutic in hepatocellular carcinoma. Mol Cancer Ther 2023;22:737−750.ArticlePubMedPMCPDF
  • 35. Dhanasekaran R, Suzuki H, Lemaitre L, Kubota N, Hoshida Y. Molecular and immune landscape of hepatocellular carcinoma to guide therapeutic decision-making. Hepatology 2025;81:1038−1057.ArticlePubMed
  • 36. Xu C, Xu Z, Zhang Y, Evert M, Calvisi DF, Chen X. β-catenin signaling in hepatocellular carcinoma. J Clin Invest 2022;132:e154515.ArticlePubMedPMC
  • 37. Tan PS, Nakagawa S, Goossens N, Venkatesh A, Huang T, Ward SC, et al. Clinicopathological indices to predict hepatocellular carcinoma molecular classification. Liver Int 2016;36:108−118.ArticlePubMedPMCPDF
  • 38. Hoshida Y, Nijman SM, Kobayashi M, Chan JA, Brunet JP, Chiang DY, et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res 2009;69:7385−7392.ArticlePubMedPMCPDF
  • 39. Song P, Gao Z, Bao Y, Chen L, Huang Y, Liu Y, et al. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J Hematol Oncol 2024;17:46. ArticlePubMedPMCPDF
  • 40. Selvaggi F, Catalano T, Cotellese R, Aceto GM. Targeting Wnt/β-catenin pathways in primary liver tumours: from microenvironment signaling to therapeutic agents. Cancers (Basel) 2022;14:1912. ArticlePubMedPMC
  • 41. Duspara K, Bojanic K, Pejic JI, Kuna L, Kolaric TO, Nincevic V, et al. Targeting the Wnt signaling pathway in liver fibrosis for drug options: an update. J Clin Transl Hepatol 2021;9:960−971.ArticlePubMedPMC
  • 42. Bouaoun L, Sonkin D, Ardin M, Hollstein M, Byrnes G, Zavadil J, et al. TP53 variations in human cancers: new lessons from the IARC TP53 database and genomics data. Hum Mutat 2016;37:865−876.ArticlePubMed
  • 43. Teramoto T, Satonaka K, Kitazawa S, Fujimori T, Hayashi K, Maeda S. p53 gene abnormalities are closely related to hepatoviral infections and occur at a late stage of hepatocarcinogenesis. Cancer Res 1994;54:231−235.PubMed
  • 44. Zhang W, He H, Zang M, Wu Q, Zhao H, Lu LL, et al. Genetic features of aflatoxin-associated hepatocellular carcinoma. Gastroenterology 2017;153:249−262.e2.ArticlePubMed
  • 45. Chaisaingmongkol J, Budhu A, Dang H, Rabibhadana S, Pupacdi B, Kwon SM, et al. Common molecular subtypes among Asian hepatocellular carcinoma and cholangiocarcinoma. Cancer Cell 2017;32:57−70.e3.ArticlePubMedPMC
  • 46. Candia J, Bayarsaikhan E, Tandon M, Budhu A, Forgues M, Tovuu LO, et al. The genomic landscape of Mongolian hepatocellular carcinoma. Nat Commun 2020;11:4383. ArticlePubMedPMCPDF
  • 47. Boyault S, Rickman DS, de Reyniès A, Balabaud C, Rebouissou S, Jeannot E, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 2007;45:42−52.ArticlePubMed
  • 48. Désert R, Rohart F, Canal F, Sicard M, Desille M, Renaud S, et al. Human hepatocellular carcinomas with a periportal phenotype have the lowest potential for early recurrence after curative resection. Hepatology 2017;66:1502−1518.ArticlePubMedPDF
  • 49. Kancherla V, Abdullazade S, Matter MS, Lanzafame M, Quagliata L, Roma G, et al. Genomic analysis revealed new oncogenic signatures in TP53-mutant hepatocellular carcinoma. Front Genet 2018;9:2. ArticlePubMedPMC
  • 50. Yang T, Choi Y, Joh JW, Cho SK, Kim DS, Park SG. Phosphorylation of p53 serine 15 is a predictor of survival for patients with hepatocellular carcinoma. Can J Gastroenterol Hepatol 2019;2019:9015453. ArticlePubMedPMCPDF
  • 51. Ma Z, Guo D, Wang Q, Liu P, Xiao Y, Wu P, et al. Lgr5-mediated p53 Repression through PDCD5 leads to doxorubicin resistance in hepatocellular carcinoma. Theranostics 2019;9:2967−2983.ArticlePubMedPMC
  • 52. Kopanja D, Huang S, Al Raheed MRH, Guzman G, Raychaudhuri P. p19Arf inhibits aggressive progression of H-ras-driven hepatocellular carcinoma. Carcinogenesis 2018;39:318−326.ArticlePubMedPMC
  • 53. Zhu ZZ, Bao LL, Zhao K, Xu Q, Zhu JY, Zhu KX, et al. Copy number aberrations of multiple genes identified as prognostic markers for extrahepatic metastasis-free survival of patients with hepatocellular carcinoma. Curr Med Sci 2019;39:759−765.ArticlePubMedPDF
  • 54. Dhar D, Antonucci L, Nakagawa H, Kim JY, Glitzner E, Caruso S, et al. Liver cancer initiation requires p53 inhibition by CD44-enhanced growth factor signaling. Cancer Cell 2018;33:1061−1077.e6.ArticlePubMedPMC
  • 55. Zhou Y, Cui G, Xu H, Chun J, Yang D, Zhang Z, et al. Loss of TP53 cooperates with c-MET overexpression to drive hepatocarcinogenesis. Cell Death Dis 2023;14:476. ArticlePubMedPMCPDF
  • 56. Duffy MJ, Tang M, Rajaram S, O’Grady S, Crown J. Targeting mutant p53 for cancer treatment: moving closer to clinical use? Cancers (Basel) 2022;14:4499. ArticlePubMedPMC
  • 57. Zhou X, Singh M, Santos GS, Guerlavais V, Carvajal LA, Aivado M, et al. Pharmacologic activation of p53 triggers viral mimicry response thereby abolishing tumor immune evasion and promoting antitumor immunity. Cancer Discov 2021;11:3090−3105.ArticlePubMedPMCPDF
  • 58. Lu Y, Lin B, Li M. The role of alpha-fetoprotein in the tumor microenvironment of hepatocellular carcinoma. Front Oncol 2024;14:1363695. ArticlePubMedPMC
  • 59. Hussein MS, Li Q, Mao R, Peng Y, He Y. TCR T cells overexpressing c-Jun have better functionality with improved tumor infiltration and persistence in hepatocellular carcinoma. Front Immunol 2023;14:1114770. ArticlePubMedPMC
  • 60. Charneau J, Suzuki T, Shimomura M, Fujinami N, Nakatsura T. Peptide-based vaccines for hepatocellular carcinoma: a review of recent advances. J Hepatocell Carcinoma 2021;8:1035−1054.ArticlePubMedPMCPDF
  • 61. Zheng X, Liu X, Lei Y, Wang G, Liu M. Glypican-3: a novel and promising target for the treatment of hepatocellular carcinoma. Front Oncol 2022;12:824208. ArticlePubMedPMC
  • 62. Terris B, Cavard C, Perret C. EpCAM, a new marker for cancer stem cells in hepatocellular carcinoma. J Hepatol 2010;52:280−281.ArticlePubMed
  • 63. Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology 2009;136:1012−1024.ArticlePubMedPMC
  • 64. Tremosini S, Forner A, Boix L, Vilana R, Bianchi L, Reig M, et al. Prospective validation of an immunohistochemical panel (glypican 3, heat shock protein 70 and glutamine synthetase) in liver biopsies for diagnosis of very early hepatocellular carcinoma. Gut 2012;61:1481−1487.ArticlePubMed
  • 65. Abou-Alfa GK, Puig O, Daniele B, Kudo M, Merle P, Park JW, et al. Randomized phase II placebo controlled study of codrituzumab in previously treated patients with advanced hepatocellular carcinoma. J Hepatol 2016;65:289−295.ArticlePubMed
  • 66. Carrasquillo JA, O’Donoghue JA, Beylergil V, Ruan S, Pandit-Taskar N, Larson SM, et al. I-124 codrituzumab imaging and biodistribution in patients with hepatocellular carcinoma. EJNMMI Res 2018;8:20. ArticlePubMedPMCPDF
  • 67. Komatsu SI, Kayukawa Y, Miyazaki Y, Kaneko A, Ikegami H, Ishiguro T, et al. Determination of starting dose of the T cell-redirecting bispecific antibody ERY974 targeting glypican-3 in first-in-human clinical trial. Sci Rep 2022;12:12312. ArticlePubMedPMCPDF
  • 68. Yamaguchi H, Hsu JM, Sun L, Wang SC, Hung MC. Advances and prospects of biomarkers for immune checkpoint inhibitors. Cell Rep Med 2024;5:101621. ArticlePubMedPMC
  • 69. Sangro B, Melero I, Wadhawan S, Finn RS, Abou-Alfa GK, Cheng AL, et al. Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J Hepatol 2020;73:1460−1469.ArticlePubMedPMC
  • 70. Yau T, Park JW, Finn RS, Cheng AL, Mathurin P, Edeline J, et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol 2022;23:77−90.ArticlePubMed
  • 71. Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, et al. Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase III trial. J Clin Oncol 2020;38:193−202.ArticlePubMed
  • 72. Han JW, Kang MW, Lee SK, Yang H, Kim JH, Yoo JS, et al. Dynamic peripheral T-cell analysis identifies on-treatment prognostic biomarkers of atezolizumab plus bevacizumab in hepatocellular carcinoma. Liver Cancer 2024;Sep 2 doi: 10.1159/000541181. [Epub ahead of print].ArticlePDF
  • 73. Renne SL, Woo HY, Allegra S, Rudini N, Yano H, Donadon M, et al. Vessels encapsulating tumor clusters (VETC) is a powerful predictor of aggressive hepatocellular carcinoma. Hepatology 2020;71:183−195.ArticlePubMedPDF
  • 74. Liu K, Dennis C, Prince DS, Marsh-Wakefield F, Santhakumar C, Gamble JR, et al. Vessels that encapsulate tumour clusters vascular pattern in hepatocellular carcinoma. JHEP Rep 2023;5:100792. ArticlePubMedPMC
  • 75. Fang JH, Xu L, Shang LR, Pan CZ, Ding J, Tang YQ, et al. Vessels that encapsulate tumor clusters (VETC) pattern is a predictor of sorafenib benefit in patients with hepatocellular carcinoma. Hepatology 2019;70:824−839.ArticlePubMedPDF
  • 76. Vogel A, Meyer T, Sapisochin G, Salem R, Saborowski A. Hepatocellular carcinoma. Lancet 2022;400:1345−1362.ArticlePubMed
  • 77. Pallmann P, Bedding AW, Choodari-Oskooei B, Dimairo M, Flight L, Hampson LV, et al. Adaptive designs in clinical trials: why use them, and how to run and report them. BMC Med 2018;16:29. ArticlePubMedPMCPDF
  • 78. Subbiah V. The next generation of evidence-based medicine. Nat Med 2023;29:49−58.ArticlePubMedPDF
  • 79. Campani C, Imbeaud S, Couchy G, Ziol M, Hirsch TZ, Rebouissou S, et al. Circulating tumour DNA in patients with hepatocellular carcinoma across tumour stages and treatments. Gut 2024;73:1870−1882.ArticlePubMed
  • 80. Chen VL, Xu D, Wicha MS, Lok AS, Parikh ND. Utility of liquid biopsy analysis in detection of hepatocellular carcinoma, determination of prognosis, and disease monitoring: a systematic review. Clin Gastroenterol Hepatol 2020;18:2879−2902.e9.ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Paracancerous binuclear hepatocytes assessed by computer program is a novel biomarker for short term recurrence of hepatocellular carcinoma after surgery
      Yifan Zhang, Yiquan Lu, Nan Wang, Fengjie Hao, Yongjun Chen, Xiaochun Fei, Junqing Wang
      Scientific Reports.2025;[Epub]     CrossRef
    • Differential Infiltration of T-Cell Populations in Tumor and Liver Tissues Predicts Recurrence-Free Survival in Surgically Resected Hepatocellular Carcinoma
      Eun Ji Jang, Ho Joong Choi, Young Kyoung You, Deok Hwa Seo, Mi Hyun Kwon, Keungmo Yang, Jaejun Lee, Jeong Won Jang, Seung Kew Yoon, Ji Won Han, Pil Soo Sung
      Cancers.2025; 17(9): 1548.     CrossRef

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

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Molecular and immune landscape of hepatocellular carcinoma for therapeutic development
      J Liver Cancer. 2025;25(1):9-18.   Published online December 6, 2024
      Close
    • XML DownloadXML Download
    Related articles
    Molecular and immune landscape of hepatocellular carcinoma for therapeutic development
    Image Image
    Figure 1. Therapeutic strategies according to molecular and immune landscape of HCC. (A) Molecular subtypes of HCC tumors and associated histological and molecular features. (B) Molecular and cellular targets for approved drugs and therapeutic strategies in development in HCC. CAR-T cells, chimeric antigen receptor T cells; GPC3, glypican-3; AFP, alpha-fetoprotein; EpCAM, epithelial cell adhesion molecule; neoAg, neo antigen; VEGF, vascular endothelial growth factor; FGF, fibroblast growth factor; PDGFβ, platelet-derived growth factor beta; EGF, epidermal growth factor; HGF, hepatocyte growth factor; APC, antigen presenting cell; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; VEGFR, vascular endothelial growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; EGFR, epidermal growth factor receptor; MET, MET proto-oncogene (hepatocyte growth factor receptor); PD-L1, programmed cell death ligand 1; mTOR, mechanistic target of rapamycin; JAK/STAT, Janus kinase/signal transducer and activator of transcription; MAPK, mitogen-activated protein kinase; PI3K/Akt, phosphatidylinositol 3-kinase/protein kinase B; PD-1, programmed cell death protein 1; CDK, cyclin-dependent kinase; DKK1, dickkopf-1; Rb, retinoblastoma; HCC, hepatocellular carcinoma; TGFβ, transforming growth factor beta; TGFβR, transforming growth factor beta receptor; TERT, telomerase reverse transcriptase.
    Figure 2. Study designs for biomarker-guided therapeutic clinical trial in HCC. HCC, hepatocellular carcinoma.
    Molecular and immune landscape of hepatocellular carcinoma for therapeutic development
    Clinical context Phase Trial type Agent Class of agent Biomarker-based eligibility NCT number Last updated
    Neo-adjuvant theapy II Biomarker enrichment trial Lenvatinib Multi-kinase inhibitor AFP>100 ng/mL, infiltrative/ macrotrabecular pattern NCT05113186 2024
    Adjuvant therapy I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06560827 2024
    II Biomarker enrichment trial Sintilimab +/- lenvatinib Anti-PD-1 +/- multi-kinase inhibitor VETC positive NCT06461936 2024
    IV Biomarker enrichment trial Anti-PD-1 +/- lenvatinib Anti-PD-1 +/- multi-kinase inhibitor VETC positive NCT06311929 2024
    First-line systemic therapy I Basket/bucket trial TSR-022 Anti-TIM-3 TIM-3-positive NCT02817633 2024
    I/II Biomarker enrichment trial DKN-01 DKK1 inhibitor Glutamine synthetase IHC-positive NCT03645980 2020
    First or second-line systemic therapy I Biomarker enrichment trial AFPc332 T cells Engineered T cells AFP≥100 ng/mL NCT03132792 2024
    I Biomarker enrichment trial CT0180 cells Engineered T cells GPC3 IHC-positive NCT04756648 2023
    I Basket/bucket trial BOXR1030 T cells Engineered T cells GPC3 IHC-positive NCT05120271 2024
    I/II Basket/bucket trial CAR-T/TCR-T cells Engineered T cells DR5 or EGFRvIII IHC-positive NCT03941626 2021
    II Basket/bucket trial 32 molecular-targeted agents Molecular-targeted agents Positive targets by tumor DNA-seq, IHC NCT02465060 2024
    II Basket/bucket trial Spartalizumab Anti-PD-1 PD-1 mRNA, high expression NCT04802876 2023
    II Basket/bucket trial Ad-p53 + approved ICI Viral gene therapy + ICI p53 wild type or IHC-negative NCT03544723 2020
    Second-line systemic therapy I Biomarker enrichment trial C-TCR055 Engineered T cells AFP IHC-positive NCT04368182 2020
    I Biomarker enrichment trial TC-CAR031 Engineered T cells GPC3 IHC-positive NCT05155189 2024
    I Basket/bucket trial EpCAM CAR-T cells Engineered T cells EpCAM-positive NCT05028933 2024
    I Basket/bucket trial AFP CAR-T cells Engineered T cells AFP IHC-positive or AFP≥400 ng/mL NCT06515314 2024
    I Basket/bucket trial p53 MVA vaccine + pembrolizumab MVA vaccine + anti-PD-1 p53 IHC-positive or gene mutation NCT02432963 2024
    I Basket/bucket trial HER2 CAR-macrophages Engineered macrophages HER2-positive NCT04660929 2024
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05003895 2024
    I Biomarker enrichment trial GPC3 CAR-T cells + fludarabine + cytoxan Engineered T cells + STAT1 inhibitor + alkylating agent GPC3 IHC-positive NCT05103631 2023
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06461624 2024
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06144385 2024
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05620706 2022
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05926726 2023
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05344664 2022
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05783570 2024
    I Biomarker enrichment trial GPC3 CAR-NK cells Engineered NK cells GPC3-positive NCT05845502 2024
    I/II Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06084884 2024
    I/II Biomarker enrichment trial B7H3 CAR-T cells Engineered T cells B7H3-positive NCT05323201 2024
    I/II Biomarker enrichment trial ECT204 T cells Engineered T cells GPC3 IHC-positive NCT04864054 2024
    I/II Biomarker enrichment trial HBV-TCR T cells (LioCyx-M) +/- lenvatinib Engineered T cells +/- multi-kinase inhibitor HLA class I profile matching NCT05195294 2022
    II Biomarker enrichment trial PD-0332991 CDK4/6 inhibitor RB-positive NCT01356628 2023
    II Biomarker enrichment trial Futibatinib + pembrolizumab FGFR inhibitor + anti-PD-1 FGF19 mRNA or IHC-positive NCT04828486 2024
    Third-line systemic therapy I/II Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06590246 2024
    Not defined I Basket/bucket trial IL-15 and IL-21 armored GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06198296 2024
    I Biomarker enrichment trial GPC3/TGF-β CAR-T cells Engineered T cells GPC3 WB or IHC-positive NCT03198546 2024
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05070156 2023
    I Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT06478693 2024
    I/II Basket/bucket trial HMBD-001 Anti-HER3 HER3-positive NCT05057013 2023
    I/II Biomarker enrichment trial GPC3 CAR-T cells Engineered T cells GPC3 IHC-positive NCT05652920 2024
    I/II Biomarker enrichment trial RZ-001 + valganciclovir Trans-splicing ribozyme + antiviral hTERT-positive NCT05595473 2022
    Table 1. Ongoing biomarker-guided therapeutic trials in HCC

    ClinicalTrials.gov accessed in October 2024.

    HCC, hepatocellular carcinoma; NCT, National Clinical Trial; AFP, alpha-fetoprotein; GPC3, glypican-3; CAR, chimeric antigen receptor; IHC, immunohistochemistry; PD-1, programmed death-1; VETC, vessels encapsulating tumor clusters; TIM-3, T cell immunoglobulin and mucin containing protein-3; DKK1, dickkopf-1; TCR, T cell receptor; DR5, death receptor 5; EGFR, epidermal growth factor receptor; DNA-seq, DNA sequencing; Ad, adenoviral; C-TCR, AFP specific T cell receptor transduced T cells; ICI, immune checkpoint inhibitor; EpCAM, epithelial cell adhesion molecule; MVA, modified vaccinia virus ankara; HER, human epidermal growth factor receptor; STAT1, signal transducer and activator of transcription 1; NK, natural killer; HBV, hepatitis B virus; HLA, human leukocyte antigen; CDK, cyclin-dependent kinase; RB, retinoblastoma; FGFR, fibroblast growth factor receptor; FGF, fibroblast growth factor; IL, interleukin; TGF-β, transforming growth factor beta; WB, western blotting; hTERT, human telomerase reverse transcriptase.


    JLC : Journal of Liver Cancer
    TOP