Skip to main content

Advertisement

Log in

A novel risk score system based on immune subtypes for identifying optimal mRNA vaccination population in hepatocellular carcinoma

  • Research
  • Published:
Cellular Oncology Aims and scope Submit manuscript

Abstract

Purpose

Although mRNA vaccines have shown certain clinical benefits in multiple malignancies, their therapeutic efficacies against hepatocellular carcinoma (HCC) remains uncertain. This study focused on establishing a novel risk score system based on immune subtypes so as to identify optimal HCC mRNA vaccination population.

Methods

GEPIA, cBioPortal and TIMER databases were utilized to identify candidate genes for mRNA vaccination in HCC. Subsequently, immune subtypes were constructed based on the candidate genes. According to the differential expressed genes among various immune subtypes, a risk score system was established using machine learning algorithm. Besides, multi-color immunofluorescence of tumor tissues from 72 HCC patients were applied to validate the feasibility and efficiency of the risk score system.

Results

Twelve overexpressed and mutated genes associated with poor survival and APCs infiltration were identified as potential candidate targets for mRNA vaccination. Three immune subtypes (e.g. IS1, IS2 and IS3) with distinct clinicopathological and molecular profiles were constructed according to the 12 candidate genes. Based on the immune subtype, a risk score system was developed, and according to the risk score from low to high, HCC patients were classified into four subgroups on average (e.g. RS1, RS2, RS3 and RS4). RS4 mainly overlapped with IS3, RS1 with IS2, and RS2+RS3 with IS1. ROC analysis also suggested the significant capacity of the risk score to distinguish between the three immune subtypes. Higher risk score exhibited robustly predictive ability for worse survival, which was further independently proved by multi-color immunofluorescence of HCC samples. Notably, RS4 tumors exhibited an increased immunosuppressive phenotype, higher expression of the twelve potential candidate targets and increased genome altered fraction, and therefore might benefit more from vaccination.

Conclusions

This novel risk score system based on immune subtypes enabled the identification of RS4 tumor that, due to its highly immunosuppressive microenvironment, may benefit from HCC mRNA vaccination.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data availability

All data generated and described in this article are available from the corresponding web servers, and are freely available to any scientist wishing to use them for noncommercial purposes, without breaching participant confidentiality. Further information is available on reasonable request from the corresponding author.

References

  1. A. Villanueva, Hepatocellular carcinoma. N. Engl. J. Med. 380, 1450–1462 (2019)

    Article  CAS  PubMed  Google Scholar 

  2. J.C. Nault, A. Villanueva, Biomarkers for hepatobiliary cancers. Hepatology 73(Suppl 1), 115–127 (2021)

    Article  PubMed  Google Scholar 

  3. U. Harkus et al., Immune checkpoint inhibitors in HCC: cellular, molecular and systemic data. Semin. Cancer Biol. 86, 799–815 (2022)

  4. S.M. Kalasekar, I. Garrido-Laguna, K.J. Evason, Immune checkpoint inhibitors in combinations for hepatocellular carcinoma. Hepatology 73, 2591–2593 (2021)

    Article  PubMed  Google Scholar 

  5. A. Huang, et al., Targeted therapy for hepatocellular carcinoma. Signal Transduct. Target Ther. 5, 146 (2020)

    Article  MathSciNet  PubMed  PubMed Central  Google Scholar 

  6. M. Pinter, B. Scheiner, M. Peck-Radosavljevic, Immunotherapy for advanced hepatocellular carcinoma: a focus on special subgroups. Gut 70, 204–214 (2021)

    Article  CAS  PubMed  Google Scholar 

  7. E.N. De Toni, Immune checkpoint inhibitors: use them early, combined and instead of TACE? Gut 69, 1887–1888 (2020)

    Article  PubMed  Google Scholar 

  8. T.F. Greten, et al., Targeted and immune-based therapies for hepatocellular carcinoma. Gastroenterology 156, 510–524 (2019)

    Article  PubMed  Google Scholar 

  9. K. Shigeta, et al., Dual programmed death receptor-1 and vascular endothelial growth factor receptor-2 blockade promotes vascular normalization and enhances antitumor immune responses in hepatocellular carcinoma. Hepatology 71, 1247–1261 (2020)

    Article  CAS  PubMed  Google Scholar 

  10. K. Shigeta, et al., Regorafenib combined with PD1 blockade increases CD8 T-cell infiltration by inducing CXCL10 expression in hepatocellular carcinoma. J. Immunother. Cancer 8, e001435 (2020)

  11. S.J. Casak, et al., FDA approval summary: atezolizumab plus bevacizumab for the treatment of patients with advanced unresectable or metastatic hepatocellular carcinoma. Clin. Cancer. Res. 27, 1836–1841 (2021)

    Article  CAS  PubMed  Google Scholar 

  12. S. Qin, et al., Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. J. Hematol. Oncol. 12, 27 (2019)

    Article  PubMed  PubMed Central  Google Scholar 

  13. M.E. Couch, et al., Alteration of cellular and humoral immunity by mutant p53 protein and processed mutant peptide in head and neck cancer. Clin. Cancer. Res. 13, 7199–7206 (2007)

    Article  CAS  PubMed  Google Scholar 

  14. E. Dai, et al., Epigenetic modulation of antitumor immunity for improved cancer immunotherapy. Mol. Cancer 20, 171 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. X.M. Shao, et al., HLA class II immunogenic mutation burden predicts response to immune checkpoint blockade. Ann. Oncol. 33, 728–738 (2022)

  16. S.E. Lee, et al., Improvement of STING-mediated cancer immunotherapy using immune checkpoint inhibitors as a game-changer. Cancer Immunol. Immunother. 71, 1–14 (2022)

  17. E.B. Ellingsen, et al., Durable and dynamic hTERT immune responses following vaccination with the long-peptide cancer vaccine UV1: long-term follow-up of three phase I clinical trials. J. Immunother. Cancer 10, e004345 (2022)

  18. A. Rooney, et al., Risk of SARS-CoV-2 breakthrough infection in vaccinated cancer patients: a retrospective cohort study. J. Hematol. Oncol. 15, 67 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. E. Fang, et al., Advances in COVID-19 mRNA vaccine development. Signal Transduct. Target Ther. 7, 94 (2022)

  20. S. Qin, et al., mRNA-based therapeutics: powerful and versatile tools to combat diseases. Signal Transduct. Target Ther. 7, 166 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. W. Mey, et al., RNA in cancer immunotherapy: unlocking the potential of the immune system. Clin. Cancer. Res. 28, 3929–3939 (2022)

  22. J. Wei, A.M. Hui, The paradigm shift in treatment from Covid-19 to oncology with mRNA vaccines. Cancer Treat. Rev. 107, 102405 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. A.J. Barbier, et al., The clinical progress of mRNA vaccines and immunotherapies. Nat. Biotechnol. 40, 840–854 (2022)

  24. T. Huang, et al., Lipid nanoparticle-based mRNA vaccines in cancers: current advances and future prospects. Front. Immunol. 13, 922301 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. C.L. Lorentzen, et al., Clinical advances and ongoing trials on mRNA vaccines for cancer treatment. Lancet Oncol. 23, e450–e458 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. N. Pardi, et al., mRNA vaccines - a new era in vaccinology. Nat. Rev. Drug Discov. 17, 261–279 (2018)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. M. Sebastian, et al., Phase Ib study evaluating a self-adjuvanted mRNA cancer vaccine (RNActive(R)) combined with local radiation as consolidation and maintenance treatment for patients with stage IV non-small cell lung cancer. BMC Cancer 14, 748 (2014)

    Article  PubMed  PubMed Central  Google Scholar 

  28. K.A. Batich, et al., Long-term survival in glioblastoma with cytomegalovirus pp65-targeted vaccination. Clin. Cancer. Res. 23, 1898–1909 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. J. Li, et al., Messenger RNA vaccine based on recombinant MS2 virus-like particles against prostate cancer. Int. J. Cancer 134, 1683–1694 (2014)

    Article  CAS  PubMed  Google Scholar 

  30. H. Kubler, et al., Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/IIa study. J. Immunother. Cancer 3, 26 (2015)

  31. Z. Tang, et al., GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic. Acids Res. 47, W556–W560 (2019)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. T. Li, et al., TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 77, e108–e110 (2017)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. S. Hanzelmann, R. Castelo, J. Guinney, GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinf. 14, 7 (2013)

  34. G. Bindea, et al., Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39, 782–795 (2013)

    Article  CAS  PubMed  Google Scholar 

  35. P. Charoentong, et al., Pan-cancer Immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 18, 248–262 (2017)

    Article  CAS  PubMed  Google Scholar 

  36. K. Yoshihara, et al., Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 4, 2612 (2013)

    Article  ADS  PubMed  Google Scholar 

  37. D.M. Wolf, et al., Gene co-expression modules as clinically relevant hallmarks of breast cancer diversity. PLoS One 9, e88309 (2014)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  38. P. Langfelder, S. Horvath, WGCNA: an R package for weighted correlation network analysis. BMC Bioinf. 9, 559 (2008)

    Article  Google Scholar 

  39. A. Kamburov, et al., The consensusPathDB interaction database: 2013 update. Nucleic. Acids Res. 41, D793–800 (2013)

    Article  CAS  PubMed  Google Scholar 

  40. X. Huang, et al., Identification of tumor antigens and immune subtypes of pancreatic adenocarcinoma for mRNA vaccine development. Mol. Cancer 20, 44 (2021)

  41. R.L. Camp, M. Dolled-Filhart, D.L. Rimm, X-tile: a new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin. Cancer. Res. 10, 7252–7259 (2004)

    Article  CAS  PubMed  Google Scholar 

  42. Y.R. Miao, et al., ImmuCellAI: a unique method for comprehensive T-cell subsets abundance prediction and its application in cancer immunotherapy. Adv. Sci. (Weinh) 7, 1902880 (2020)

    Article  CAS  PubMed  Google Scholar 

  43. G. Zhang, et al., mRNA vaccines in disease prevention and treatment. Signal Transduct. Target Ther. 8, 365 (2023)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. X. Huang, et al., Identification of tumor antigens and immune subtypes of cholangiocarcinoma for mRNA vaccine development. Mol. Cancer 20, 50 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. X. Huang, et al., Personalized pancreatic cancer therapy: from the perspective of mRNA vaccine. Mil. Med. Res. 9, 53 (2022)

  46. T.Y. Tang, et al., mRNA vaccine development for cholangiocarcinoma: a precise pipeline. Mil. Med. Res. 9, 40 (2022)

  47. X. Huang, G. Zhang, T. Liang, Subtyping for pancreatic cancer precision therapy. Trends Pharmacol. Sci. 43, 482–494 (2022)

    Article  CAS  PubMed  Google Scholar 

  48. J. Seo, et al., Fatty-acid-induced FABP5/HIF-1 reprograms lipid metabolism and enhances the proliferation of liver cancer cells. Commun. Biol. 3, 638 (2020)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. V.W. Rebecca, et al., PPT1 promotes tumor growth and is the molecular target of chloroquine derivatives in cancer. Cancer Discov. 9, 220–229 (2019)

    Article  CAS  PubMed  Google Scholar 

  50. J. Xu, et al., High PPT1 expression predicts poor clinical outcome and PPT1 inhibitor DC661 enhances sorafenib sensitivity in hepatocellular carcinoma. Cancer Cell Int. 22, 115 (2022)

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  51. G. Sharma, et al., PPT1 inhibition enhances the antitumor activity of anti-PD-1 antibody in melanoma. JCI Insight 5, e133225 (2020)

  52. S. He, X. Wang, RIP kinases as modulators of inflammation and immunity. Nat. Immunol. 19, 912–922 (2018)

    Article  CAS  PubMed  Google Scholar 

  53. Y. Yan, et al., Receptor-interacting protein kinase 2 (RIPK2) stabilizes c-Myc and is a therapeutic target in prostate cancer metastasis. Nat. Commun. 13, 669 (2022)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. Y. Zhou, et al., Hepatic NOD2 promotes hepatocarcinogenesis via a RIP2-mediated proinflammatory response and a novel nuclear autophagy-mediated DNA damage mechanism. J. Hematol. Oncol. 14, 9 (2021)

  55. M.J. Gough, et al., OX40 agonist therapy enhances CD8 infiltration and decreases immune suppression in the tumor. Cancer Res. 68, 5206–5215 (2008)

    Article  CAS  PubMed  Google Scholar 

  56. S.M. Jensen, et al., Signaling through OX40 enhances antitumor immunity. Semin. Oncol. 37, 524–532 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. A.D. Weinberg, et al., Science gone translational: the OX40 agonist story. Immunol. Rev. 244, 218–231 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. R.B. Bell, et al., OX40 signaling in head and neck squamous cell carcinoma: overcoming immunosuppression in the tumor microenvironment. Oral. Oncol. 52, 1–10 (2016)

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  59. R. Duhen, et al., Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat. Commun. 12, 1047 (2021)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  60. Q. Song, et al., Reinforcing the combinational immuno-oncotherapy of switching “cold” tumor to “hot” by responsive penetrating nanogels. ACS Appl. Mater. Interf. 13, 36824–36838 (2021)

    Article  CAS  Google Scholar 

  61. W.X. Hong, et al., Neoadjuvant intratumoral immunotherapy with TLR9 activation and anti-OX40 antibody eradicates metastatic cancer. Cancer Res. 82, 1396–1408 (2022)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. A.M. Boutelle, L.D. Attardi, p53 and tumor suppression: it takes a network. Trends. Cell Biol. 31, 298–310 (2021)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. N. Raj, L.D. Attardi, Tumor suppression: p53 alters immune surveillance to restrain liver cancer. Curr. Biol. 23, R527–530 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. S.L. Highfill, et al., Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci. Transl. Med. 6, 237ra267 (2014)

    Article  Google Scholar 

  65. C. Lu, et al., Current perspectives on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol. Cancer 18, 130 (2019)

    Article  PubMed  PubMed Central  Google Scholar 

  66. S.J. Yu, et al., Targeting the crosstalk between cytokine-induced killer cells and myeloid-derived suppressor cells in hepatocellular carcinoma. J. Hepatol. 70, 449–457 (2019)

    Article  CAS  PubMed  Google Scholar 

  67. C. Groth, et al., Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 120, 16–25 (2019)

    Article  CAS  PubMed  Google Scholar 

  68. S. Kalathil, et al., Higher frequencies of GARP(+)CTLA-4(+)Foxp3(+) T regulatory cells and myeloid-derived suppressor cells in hepatocellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res. 73, 2435–2444 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. B. Hoechst, et al., Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology 50, 799–807 (2009)

    Article  CAS  PubMed  Google Scholar 

  70. R. Derynck, S.J. Turley, R.J. Akhurst, TGFbeta biology in cancer progression and immunotherapy. Nat. Rev. Clin. Oncol. 18, 9–34 (2021)

    Article  PubMed  Google Scholar 

  71. A. de Gramont, S. Faivre, E. Raymond, Novel TGF-beta inhibitors ready for prime time in onco-immunology. Oncoimmunology 6, e1257453 (2017)

    Article  PubMed  Google Scholar 

  72. J. Cullis, S. Das, D. Bar-Sagi, Kras and tumor immunity: friend or foe? Cold Spring Harb. Perspect. Med. 8, a031849 (2018)

  73. J. Chen, J.A. Gingold, X. Su, Immunomodulatory TGF-beta signaling in hepatocellular carcinoma. Trends Mol. Med. 25, 1010–1023 (2019)

  74. S.C. Casey, et al., MYC regulates the antitumor immune response through CD47 and PD-L1. Science 352, 227–231 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This study was supported by National Natural Science Foundation of China (project NO.: 81972263, 82072714 and 82103221), the program of Guangdong Provincial Clinical Research Center for Digestive Diseases (2020B1111170004), China Postdoctoral Science Foundation (2020M683094), the Science and Technology Program of Guangzhou (202201011427), the Excellent Young Talent Program of Guangdong Provincial People’s Hospital (KY012021190) and the High-level Hospital Construction Project (DFJH201921).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Z.H.K., C.B., C.Y.J., and S.C.Z.; Methodology: Z.H.K., T.C.W., L.H., and Z.Z.D.; Investigation: Z.H.K., and T.C.W.; Writing—Original Draft: Z.H.K., T.C.W., L.H., and Z.Z.D.; Writing—Review & Editing: Z.H.K., C.X.M., T.C.W., W.W.T., T.W.L., Y.L., X.Z.Q., W.B.K. and W.Q.B.; Visualization: Z.H.K.; Supervision: Z.H.K., C.B., C.Y.J., and S.C.Z.; Funding Acquisition: C.B., C.Y.J., S.C.Z., and T.W.L.

Corresponding authors

Correspondence to Bo Chen, Changzhen Shang or Yajin Chen.

Ethics declarations

Ethics approval and consent to participate

The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. All datasets are freely available as public resources. Therefore, local ethics approval was not required. Two tumor arrays with tumor and adjacent tumor samples from 90 HCC patients (HLivH180Su09-T-001 and HLivH180Su09-T-002) were purchased from Shanghai Outdo Biotech (Co. Ltd., Shanghai, China.). All tissues were collected according to the ethical standards from Shanghai Outdo Biotech (No.: SHYJS-CP-1607007). And informed consent was obtained from all participants.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhuang, H., Tang, C., Lin, H. et al. A novel risk score system based on immune subtypes for identifying optimal mRNA vaccination population in hepatocellular carcinoma. Cell Oncol. (2024). https://doi.org/10.1007/s13402-024-00921-1

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s13402-024-00921-1

Keywords

Navigation