Clin Mol Hepatol > Volume 31(1); 2025 > Article
Yim, Lee, Kim, Lee, Kaseb, and Lee: Correspondence to editorial 2 on “Genomic biomarkers to predict response to atezolizumab plus bevacizumab immunotherapy in hepatocellular carcinoma: insights from the IMbrave150 trial”
Dear Editor,
We appreciate Dr. Nishida for his interest on our recent study [1] and insightful editorial on personalized approaches to treating hepatocellular carcinoma (HCC) using immune checkpoint inhibitors (ICIs) [2]. Dr. Nishida highlights the significant advances made in treatment for HCC patients based on the tumor immune microenvironment (TiME) and the critical need for reliable molecular biomarkers to enhance overall therapeutic outcomes. We would like to extend this discussion by highlighting the importance of integrating single-cell level data, pathology, and medical imaging data to further refine biomarkers for immunotherapy responses.
Expanding on Dr. Nishida’s insights, we agree that detailed characterization of the tumor microenvironment (TME) is vital for predicting responses to ICIs. However, current transcriptome-based approaches, including the immune signature score (ISS10), while promising, may overlook critical nuances of tumor heterogeneity. Single cell RNA-sequencing (scRNA-seq) technologies offer an unprecedented level of detail by allowing us to analyze individual cells within the context of the TME [3]. This enables the identification of specific immune cell populations that may drive immunosuppression or resistance to ICIs. Furthermore, the dynamic profiling facilitated by scRNA-seq allows us to monitor these immune populations over time, assessing how they evolve in response to treatment and providing real-time insights into therapeutic efficacy. Therefore, integrating scRNA-seq data into predictive biomarkers could refine patient stratification and help overcome the challenge of immune resistance in HCC.
In addition to molecular data, the integration of advanced pathology and medical imaging techniques could significantly enhance our understanding of the spatial architecture of the TME. As HCC exhibit profound heterogeneity in immune cell infiltration, spatial transcriptomics and proteomics using imaging mass cytometry could help map immune cell distributions across different regions of the tumor [4]. This would allow for a more nuanced understanding of how immune effector cells interact with tumor cells and how immunosuppressive cells are positioned within the tumor landscape. Moreover, medical imaging technologies such as radiomics could provide non-invasive means of assessing tumor features, such as immune infiltration, angiogenesis, and tumor vasculature, that correlate with responses to ICIs [5]. Combining these imaging data with molecular biomarkers would give us a more comprehensive tool set for predicting immunotherapy outcomes in realworld clinical settings.
Another critical aspect of HCC biomarker research that requires further attention is the role of cancer etiology. HCC is a highly heterogeneous disease with distinct etiological factors, such as hepatitis B virus (HBV), hepatitis C virus (HCV), alcohol consumption, and non-alcoholic steatohepatitis (NASH), contributing to its development [6,7]. These etiologies have profound effects on the TME, immune cell composition, and the mutational landscape of the tumor. For example, HBV-driven HCC often exhibits higher levels of T-cell exhaustion, whereas HCV-associated HCC is characterized by chronic inflammation and increased infiltration of immune cells [8,9]. Incorporating etiology data into biomarker studies could help refine the predictive power of existing molecular signatures. Moreover, it could lead to the discovery of etiology-specific biomarkers that predict responses to ICIs more accurately.
Recent advancements in personalized cancer vaccines have shown significant promise in improving outcomes for patients with HCC, as demonstrated in a recent phase 1/2 trial [10]. In this study, a personalized neoantigen vaccine was combined with pembrolizumab (anti-PD-1 antibody) in patients with advanced HCC. The trial demonstrated that the vaccine, tailored to each patient’s specific tumor mutations, elicited strong immune responses and showed encouraging clinical activity. By targeting tumor-specific neoantigens, these vaccines have the potential to overcome the immunosuppressive tumor microenvironment seen in the “immune-excluded” or “immune-desert” subtypes of HCC [11]. The study also highlighted that combining the neoantigen vaccine with pembrolizumab enhanced T-cell infiltration into tumors, improving the efficacy of immune checkpoint inhibition. This approach represents a promising strategy for turning “cold” tumors into “hot” ones, thereby increasing their sensitivity to immunotherapy.
We remain grateful for Dr. Nishida’s insightful comments and expert perspective. We see significant potential in combining scRNA-seq technologies, pathology, and medical imaging data to advance biomarker discovery. Additionally, personalized cancer vaccines represent a promising new direction in HCC treatment. We believe that combining these approaches with deep learning and artificial intelligence could address the complex heterogeneity of the tumor microenvironment and lead to more targeted and effective immunotherapies for HCC.

ACKNOWLEDGMENTS

This work is supported by the NIH/NCI under award numbers R01CA237327, P50CA217674, and P30CA016672; The University of Texas MD Anderson Cancer Center Institutional Research Grant (IRG) Program; The University of Texas MD Anderson Cancer Center Institutional Bridge Funds; the Duncan Family Institute for Cancer Prevention and Risk Assessment Seed Funding Research Program at MD Anderson Cancer Center.

FOOTNOTES

Authors’ contribution
Conception and design of the work: Sun Young Yim, Ju-Seog Lee. Drafting the article: Sun Young Yim, Sung Hwan Lee, Ju-Seog Lee. Critical revision of the article: All authors.
Conflicts of Interest
The authors have no conflicts to disclose.

Abbreviations

HBV
hepatitis B virus
HCC
hepatocellular carcinoma
HCV
hepatitis C virus
ICIs
immune checkpoint inhibitors
NASH
non-alcoholic steatohepatitis
scRNA-seq
single cell RNA-sequencing
TME
tumor microenvironment

REFERENCES

1. Yim SY, Lee SH, Baek SW, Sohn B, Jeong YS, Kang SH, et al. Genomic biomarkers to predict response to atezolizumab plus bevacizumab immunotherapy in hepatocellular carcinoma: insights from the IMbrave150 trial. Clin Mol Hepatol 2024;30:807-823.
pmid pmc
2. Nishida N. Personalized approaches to the treatment of hepatocellular carcinoma using immune checkpoint inhibitors: Editorial on “Genomic biomarkers to predict response to atezolizumab plus bevacizumab immunotherapy in hepatocellular carcinoma: Insights from the IMbrave150 trial”. Clin Mol Hepatol 2025;31:311-315.
crossref pmid pdf
3. Ramachandran P, Matchett KP, Dobie R, Wilson-Kanamori JR, Henderson NC. Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis. Nat Rev Gastroenterol Hepatol 2020;17:457-472.
crossref pmid pdf
4. Vandereyken K, Sifrim A, Thienpont B, Voet T. Methods and applications for single-cell and spatial multi-omics. Nat Rev Genet 2023;24:494-515.
crossref pmid pmc pdf
5. Xue C, Zhou Q, Xi H, Zhou J. Radiomics: a review of current applications and possibilities in the assessment of tumor microenvironment. Diagn Interv Imaging 2023;104:113-122.
crossref pmid
6. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:6.
crossref pmid pdf
7. Sankar K, Gong J, Osipov A, Miles SA, Kosari K, Nissen NN, et al. Recent advances in the management of hepatocellular carcinoma. Clin Mol Hepatol 2024;30:1-15.
crossref pmid pmc pdf
8. Cho HJ, Cheong JY. Role of immune cells in patients with hepatitis B virus-related hepatocellular carcinoma. Int J Mol Sci 2021;22:8011.
crossref pmid pmc
9. Axley P, Ahmed Z, Ravi S, Singal AK. Hepatitis C virus and hepatocellular carcinoma: a narrative review. J Clin Transl Hepatol 2018;6:79-84.
crossref pmid pmc
10. Yarchoan M, Gane EJ, Marron TU, Perales-Linares R, Yan J, Cooch N, et al. Personalized neoantigen vaccine and pembrolizumab in advanced hepatocellular carcinoma: a phase 1/2 trial. Nat Med 2024;30:1044-1053.
crossref pmid pmc pdf
11. Montironi C, Castet F, Haber PK, Pinyol R, Torres-Martin M, Torrens L, et al. Inflamed and non-inflamed classes of HCC: a revised immunogenomic classification. Gut 2023;72:129-140.
crossref

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: cmh_journal@ijpnc.com
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 5411
TOTAL : 2380083
Close layer