Clin Mol Hepatol > Volume 31(1); 2025 > Article
Li, Wen, Xu, Yan, Han, and Yu: Comprehensive analysis of transcriptomic biomarkers for predicting response to atezolizumab plus bevacizumab immunotherapy in hepatocellular carcinoma
Dear Editor,
We read with great interest the article by Yim et al. [1], which explores the use of an immune signature score (ISS) and its refined 10-gene version (ISS10) to predict responses to Atezolizumab plus Bevacizumab (Atezo/Bev or A+T) therapy. The use of immune checkpoint inhibitors (ICIs) has revolutionized clinical care across cancer types through the potential for durable responses [2]. Atezo/Bev has dramatically altered the treatment landscape and is now the first-line treatment of advanced hepatocellular carcinoma (HCC) [3]. However, despite these advancements, the majority of HCC patients do not benefit, as the objective response rate (ORR) for single-agent ICI in HCC is around 15%, while the ORR for Atezo/Bev combination therapy remains at approximately 30% [4]. Therefore, there is an urgent need to develop predictive biomarkers for response to both ICI single-agent and combination therapies.
The response to ICI is influenced by the spatial and temporal variability of host, tumor, and tumor microenvironment (TME) [5]. While multiple predictive biomarkers for single-agent ICI response have been established, robust predictive biomarkers for Atezo/Bev are still lacking. Moreover, whether the immunotherapy response predictive signatures (IRPS) derived from other ICI agents and cancer types can be applied to Atezo/Bev in HCC remains largely unknown. To address this gap, we conducted a comprehensive analysis to identify Atezolizumab plus Bevacizumab response predictive signatures (ABRPS) based on transcriptomics.
First, we conducted an extensive literature review to gather ICI response indicators [6,7], resulting in the collection of 23 reported transcriptomic IPRS (Supplementary Methods and Supplementary Table 1). The size of these gene sets ranged from 2 to 186, with a median of 14 genes. Most of these signatures were derived from TME and exhibited some overlap (Supplementary Fig. 1). In our recent study, we developed a transcriptomic signature based on the fatty acid degradation (FAD) pathway, which has been shown to predict response to targeted therapy, transarterial chemoembolization (TACE), ICI monotherapy and combination therapies [8]. Notably, the FAD signature, which captures the metabolic features of tumor genomics, shares no common genes with other TME-based IRPS.
We then categorized HCC tumors within the IMbrave150 cohort into high and low signature subtypes based on the ssGSEA score of the corresponding signatures. Significant correlations were observed among the 23 IRPS, as shown by the correlation matrix calculated using ssGSEA scores (Supplementary Fig. 2A), revealing four distinct clusters (Supplementary Fig. 2B). Both the Cox proportional hazards model and Kaplan–Meier plots were employed to identify ABRPS (Supplementary Methods, Supplementary Fig. 3). Surprisingly, despite these 23 IRPS being derived from various cancer types and different ICI agents, 22 of them were identified as ABRPS when overall survival (OS) was used as the clinical outcome, and 15 were identified as ABRPS when progression-free survival (PFS) was used. Furthermore, 14 out of the 23 IRPS were identified as ABRPS even when applying a more stringent criterion that requires significance in both the Cox model and the log-rank test for both OS and PFS (Fig. 1A, 1B, Supplementary Fig. 3).
We further defined the hazard ratio (HR) score to evaluate and compare the predictive performance of the 23 IRPS in predicting response to Atezo/Bev, with a higher HR score indicating superior predictive accuracy. The ABRS (Atezolizumab plus Bevacizumab Response Signature), a 10-gene signature specifically developed from responders and non-responders in IMbrave150 trials [9], achieved the highest HR score, as anticipated. Notably, our FAD signature exhibited comparable efficacy (Fig. 1C). Interestingly, significantly different HR scores for OS and PFS were observed for signatures such as FAD, IMPRES, and C_ECM_ up, despite using the same dataset. This discrepancy may partly be due to these signatures identifying particularly aggressive phenotype of HCC (Supplementary Fig. 2). Additionally, we directly correlated these 23 signatures with the ORR to Atezo/Bev. Sixteen IRPS, including ABRS and FAD, showed a significant correlation with Atezo/Bev response (Supplementary Table 2). In contrast, only four IRPS, including the FAD signature, were associated with response to Sorafenib (Supplementary Table 3).
Identifying potential ICI predictive biomarkers is essential for minimizing immune-related adverse events and reducing treatment costs. We collected 23 transcriptomic biomarkers and showed that many of these IRPS, developed for other ICIs, are also predictive of response to the Atezo/Bev combination. We previously demonstrated that the HCC TME-based Inflamed signature, which captures the functional activity of immune cells [10], could predict response to anti-PD-1 therapy across various cancers [11]. Here, we have reaffirmed this concept using the Atezo/Bev dataset. Additionally, this study underscores the superior predictive performance of our HCC-cell-based FAD signature in predicting responses to Atezo/Bev.
There are several limitations of our study. Firstly, our analysis is based on a retrospective cohort. Secondly, despite our extensive literature review, it is possible that some relevant IRPS were overlooked. Thirdly, the precise underlying molecular mechanisms of ABRPS remain to be fully elucidated.
In summary, we conducted a comprehensive study to identify predictive biomarkers for Atezo/Bev, which not only complements but also extends the work of Yim. To the best of our knowledge, this is the first intergrated study to analyze and compare transcriptomic biomarkers for Atezo/Bev response. Further investigation in larger prospective cohorts is warranted to validated the ABRPS for Atezo/Bev in HCC.

ACKNOWLEDGMENTS

This work was supported by grants from the National Natural Science Foundation of China (No. 82372834 and 82173129), Jiangsu Outstanding Youth Foundation (BK20240119).

FOOTNOTES

Authors’ contribution
All authors contributed to this study at different levels. All authors read and approved the final version.
Study concept and design (Binghua Li, Decai Yu); acquisition of data (Binghua Li, Jingyuan Wen, Zhu Xu, Bing Han, Peng Yan); statistical analysis and interpretation of data (Binghua Li, Jingyuan Wen, Zhu Xu, Bing Han, Decai Yu); drafting of the manuscript (Binghua Li); critical revision of the manuscript for important intellectual content (Binghua Li, Jingyuan Wen, Zhu Xu, Bing Han, Decai Yu).
Conflicts of Interest
The authors have no conflicts to disclose.

SUPPLEMENTAL MATERIAL

Supplementary material is available at Clinical and Molecular Hepatology website (http://www.e-cmh.org).
Supplementary Figure 1.
Comparison of the 23 previously curated immunotherapy response predictive signatures using Upset plot. The right-side barplot displays the gene sizes of each signature, while the top barplot illustrates the overlaps among the signatures.
cmh-2024-0628-Supplementary-Fig-1.pdf
Supplementary Figure 2.
The correlations between 23 previously curated immunotherapy response predictive signatures and the aggressive score, as represented by a correlation matrix (A) and a heatmap (B). In panel (A), the numbers denote Pearson’s correlation coefficients (r values), while the asterisks indicate statistical significance: *P<0.05; **P<0.01; ***P<0.001. In panel (B), four clusters were identified.
cmh-2024-0628-Supplementary-Fig-2.pdf
Supplementary Figure 3.
Kaplan–Meier plots comparing progression-free survival (PFS) and overall survival (OS) of patients treated with Atezolizumab plus Bevacizumab combination therapy versus sorafenib. The comparisons are based on high and low signature scores of the 23 curated immunotherapy response predictive signatures, divided by the median. The signatures are ordered in the same sequence as in Figure 1C.
cmh-2024-0628-Supplementary-Fig-3.pdf
Supplementary Table 1.
The detailed information of the 23 reported transcriptomic immunotherapy response predictive signatures (IRPS)
cmh-2024-0628-Supplementary-Table-1.pdf
Supplementary Table 2.
Contingency table depicting the association between 23 predictors with the clinical response to atezolizumab and bevacizumab in IMbrave150 cohort
cmh-2024-0628-Supplementary-Table-2.pdf
Supplementary Table 3.
Contingency table depicting the association between 23 predictors with the clinical response to Sorafenib therapy in IMbrave150 cohort
cmh-2024-0628-Supplementary-Table-3.pdf

Figure 1.
The predictive value of the 23 curated gene signatures for predicting response to Atezolizumab plus Bevacizumab immunotherapy in hepatocellular carcinoma (HCC). (A, B) Forest plot (left panel) and Bubble plot (right panel) display the hazard ratios (HRs) and Log-rank P-values, respectively, for progression-free survival (PFS) and overall survival (OS) of HCC patients treated with the atezolizumab- bevacizumab combination compared to sorafenib, based on high signature score subtype (A), and low signature sore subtype (B). Highlighted signatures are statistically significant according to both Cox’s regression model and the Log-rank test as calculated using both OS and PFS data, with red indicating a positive correlation and blue indicating a negative correlation to Atezo/Bev response. (C) The predictive performance of the 23 immunotherapy response predictive signatures is evaluated using the HR score, calculated from both OS and PFS data. The HR score is defined as the absolute difference of the HR values between high and low signature subtype, with a higher HR score indicating superior predictive performance. The full names and detailed descriptions of the signatures can be found in Supplementary Table 1.

cmh-2024-0628f1.jpg

Abbreviations

ABRPS
Atezolizumab plus Bevacizumab response predictive signatures
Atezo/Bev
Atezolizumab plus Bevacizumab
FAD
fatty acid degradation
HCC
hepatocellular carcinoma
ICIs
immune checkpoint inhibitors
IRPS
immunotherapy response predictive signatures
ISS
immune signature score
ORR
objective response rate
OS
overall survival
PFS
progression-free survival
TACE
transarterial chemoembolization
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. Shiravand Y, Khodadadi F, Kashani SMA, Hosseini-Fard SR, Hosseini S, Sadeghirad H, et al. Immune checkpoint inhibitors in cancer therapy. Curr Oncol 2022;29:3044-3060.
crossref pmid pmc
3. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894-1905.
crossref pmid
4. Llovet JM, Castet F, Heikenwalder M, Maini MK, Mazzaferro V, Pinato DJ, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol 2022;19:151-172.
crossref pmid pdf
5. Shum B, Larkin J, Turajlic S. Predictive biomarkers for response to immune checkpoint inhibition. Semin Cancer Biol 2022;79:4-17.
crossref pmid
6. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 2019;19:133-150.
crossref pmid pmc pdf
7. Addala V, Newell F, Pearson JV, Redwood A, Robinson BW, Creaney J, et al. Computational immunogenomic approaches to predict response to cancer immunotherapies. Nat Rev Clin Oncol 2024;21:28-46.
crossref pmid pdf
8. Li B, Li Y, Zhou H, Xu Y, Cao Y, Cheng C, et al. Multiomics identifies metabolic subtypes based on fatty acid degradation allocating personalized treatment in hepatocellular carcinoma. Hepatology 2024;79:289-306.
crossref pmid pmc
9. Zhu AX, Abbas AR, de Galarreta MR, Guan Y, Lu S, Koeppen H, et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat Med 2022;28:1599-1611.
crossref pmid pdf
10. 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 pmid pmc
11. Li B, Cao Y, Li Y, Cheng C, Yu D. Letter to the editor: the inflamed subclass predicts immunotherapy response - external validations. Gut 2022 Jul 16;doi: 10.1136/gutjnl-2022-328130.
crossref pmid

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 : 5567
TOTAL : 2392590
Close layer