Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Presence of onco-fetal neighborhoods in hepatocellular carcinoma is associated with relapse and response to immunotherapy

Abstract

Onco-fetal reprogramming of the tumor ecosystem induces fetal developmental signatures in the tumor microenvironment, leading to immunosuppressive features. Here, we employed single-cell RNA sequencing, spatial transcriptomics and bulk RNA sequencing to delineate specific cell subsets involved in hepatocellular carcinoma (HCC) relapse and response to immunotherapy. We identified POSTN+ extracellular matrix cancer-associated fibroblasts (EM CAFs) as a prominent onco-fetal interacting hub, promoting tumor progression. Cell–cell communication and spatial transcriptomics analysis revealed crosstalk and co-localization of onco-fetal cells, including POSTN+ CAFs, FOLR2+ macrophages and PLVAP+ endothelial cells. Further analyses suggest an association between onco-fetal reprogramming and epithelial–mesenchymal transition (EMT), tumor cell proliferation and recruitment of Treg cells, ultimately influencing early relapse and response to immunotherapy. In summary, our study identifies POSTN+ CAFs as part of the HCC onco-fetal niche and highlights its potential influence in EMT, relapse and immunotherapy response, paving the way for the use of onco-fetal signatures for therapeutic stratification.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Construct a single-cell atlas of HCC.
Fig. 2: Characterization of onco-fetal CAFs in HCC and fetal liver.
Fig. 3: Predicted cell–cell interactions of onco-fetal reprogramming in HCC.
Fig. 4: Spatial mapping of onco-fetal cell co-localization.
Fig. 5: Association between onco-fetal cells and relapse in HCC patients.
Fig. 6: Single-cell and spatial validation of onco-fetal cells enriched in relapsed HCC patients.
Fig. 7: Connections between onco-fetal ecosystem and combination immunotherapy.

Similar content being viewed by others

Data availability

The HCC01 and HCC03 Stereo-seq data have been deposited in the China National GeneBank Sequence Archive (CNSA) under accession code CNP0004497. The M311 and M316 Stereo-seq data have been deposited in the Sequence Read Archive (SRA) under accession code PRJNA1005053. The original data used to construct the HCC atlas are available with accession codes GSE156337, GSE149614, GSE151530, GSE212046 (GEO), HRA000069 (Genome Sequence Archive) and CNP0000650 (CNSA). The original data of the PLANET cohort and GO30140 and IMbrave150 trials are available with accession codes EGAS00001003813 and EGAS00001005503 (European Genome-phenome Archive). The TCGA LIHC datasets were downloaded from UCSC Xena (http://xena.ucsc.edu). All processed expression data have been uploaded to Figshare, which can be visited through the following links: HCC Atlas, https://doi.org/10.6084/m9.figshare.22332568; onco-fetal CAF identification, https://doi.org/10.6084/m9.figshare.22332655; HCC01 and HCC03 Stereo-seq data, https://doi.org/10.6084/m9.figshare.22332223; NanoString CosMx data, https://doi.org/10.6084/m9.figshare.23972991; PLANET cohort data, https://doi.org/10.6084/m9.figshare.23732370; HCC and fetal liver Nanostring DSP data, https://doi.org/10.6084/m9.figshare.22332397; HCC immunotherapy Stereo-seq data, https://doi.org/10.6084/m9.figshare.22332352. Source data for Figs. 17 and Extended Data Figs. 110 have been provided as Source Data files. All other data supporting the findings of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Code availability

All codes generated for analysis are available at https://github.com/liziyie/Onco-fetal-CAF-in-HCC.

References

  1. Forner, A., Reig, M. & Bruix, J. Hepatocellular carcinoma. Lancet 391, 1301–1314 (2018).

    Article  PubMed  Google Scholar 

  2. Finn, R. S. et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 382, 1894–1905 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Bassez, A. et al. A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat. Med. 27, 820–832 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Shani, O. et al. Fibroblast-derived IL33 facilitates breast cancer metastasis by modifying the immune microenvironment and driving Type 2 immunity. Cancer Res. 80, 5317–5329 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kieffer, Y. et al. Single-cell analysis reveals fibroblast clusters linked to immunotherapy resistance in cancer. Cancer Discov. 10, 1330–1351 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Davidson, S. et al. Fibroblasts as immune regulators in infection, inflammation and cancer. Nat. Rev. Immunol. 21, 704–717 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Foster, D. S. et al. Multiomic analysis reveals conservation of cancer-associated fibroblast phenotypes across species and tissue of origin. Cancer Cell 40, 1392–1406.e7 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hanahan, D. Hallmarks of cancer: new dimensions. Cancer Discov. 12, 31–46 (2022).

    Article  CAS  PubMed  Google Scholar 

  9. Yong, K. J. et al. Oncofetal gene SALL4 in aggressive hepatocellular carcinoma. N. Engl. J. Med. 368, 2266–2276 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yamauchi, N. et al. The glypican 3 oncofetal protein is a promising diagnostic marker for hepatocellular carcinoma. Mod. Pathol. 18, 1591–1598 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Sharma, A. et al. Onco-fetal reprogramming of endothelial cells drives immunosuppressive macrophages in hepatocellular carcinoma. Cell 183, 377–394.e321 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Sharma, A., Bleriot, C., Currenti, J. & Ginhoux, F. Oncofetal reprogramming in tumour development and progression. Nat. Rev. Cancer 22, 593–602 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. Zhang, L. et al. Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell 181, 442–459.e29 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Leung, C. S. et al. Cancer-associated fibroblasts regulate endothelial adhesion protein LPP to promote ovarian cancer chemoresistance. J. Clin. Invest. 128, 589–606 (2018).

    Article  PubMed  Google Scholar 

  15. Zhang, Q. et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829–845.e20 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Sun, Y. et al. Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell 184, 404–421.e16 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Filliol, A. et al. Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis. Nature 610, 356–365 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ma, L. et al. Single-cell atlas of tumor cell evolution in response to therapy in hepatocellular carcinoma and intrahepatic cholangiocarcinoma. J. Hepatol. 75, 1397–1408 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lu, Y. et al. A single-cell atlas of the multicellular ecosystem of primary and metastatic hepatocellular carcinoma. Nat. Commun. 13, 4594 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhai, W. et al. Dynamic phenotypic heterogeneity and the evolution of multiple RNA subtypes in hepatocellular carcinoma: the PLANET study. Natl Sci. Rev. 9, nwab192 (2022).

  21. Zhu, A. X. et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat. Med. 28, 1599–1611 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Yin, Z. et al. Heterogeneity of cancer-associated fibroblasts and roles in the progression, prognosis, and therapy of hepatocellular carcinoma. J. Hematol. Oncol. 12, 101 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ramachandran, P. et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Buechler, M. B. et al. Cross-tissue organization of the fibroblast lineage. Nature 593, 575–579 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Wu, R. et al. Comprehensive analysis of spatial architecture in primary liver cancer. Sci. Adv. 7, eabg3750 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. True, L. D. et al. CD90/THY1 is overexpressed in prostate cancer-associated fibroblasts and could serve as a cancer biomarker. Mod. Pathol. 23, 1346–1356 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schliekelman, M. J. et al. Thy-1+ cancer-associated fibroblasts adversely impact lung cancer prognosis. Sci Rep. 7, 6478 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Hutton, C. et al. Single-cell analysis defines a pancreatic fibroblast lineage that supports anti-tumor immunity. Cancer Cell 39, 1227–1244.e20 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim, W. et al. RUNX1 is essential for mesenchymal stem cell proliferation and myofibroblast differentiation. Proc. Natl Acad. Sci. USA 111, 16389–16394 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wei, Y. et al. Liver homeostasis is maintained by midlobular zone 2 hepatocytes. Science 371, eabb1625 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Stadler, M. et al. Stromal fibroblasts shape the myeloid phenotype in normal colon and colorectal cancer and induce CD163 and CCL2 expression in macrophages. Cancer Lett. 520, 184–200 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Orimo, A. et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121, 335–348 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Strickland, L. A. et al. Plasmalemmal vesicle-associated protein (PLVAP) is expressed by tumour endothelium and is upregulated by vascular endothelial growth factor-A (VEGF). J. Pathol. 206, 466–475 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Horwitz, E. et al. Human and mouse VEGFA-amplified hepatocellular carcinomas are highly sensitive to sorafenib treatment. Cancer Discov. 4, 730–743 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Albonici, L., Giganti, M. G., Modesti, A., Manzari, V. & Bei, R. Multifaceted role of the placental growth factor (PIGF) in the antitumor immune response and cancer progression. Int. J. Mol. Sci. 20, 2970 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mani, S. A. et al. The epithelial–mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Krishnamurty, A. T. et al. LRRC15+ myofibroblasts dictate the stromal setpoint to suppress tumour immunity. Nature 611, 148–154 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jin, S. et al. Inference and analysis of cell–cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Oldham, K. A. et al. T lymphocyte recruitment into renal cell carcinoma tissue: a role for chemokine receptors CXCR3, CXCR6, CCR5, and CCR6. Eur. Urol. 61, 385–394 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Loges, S. et al. Malignant cells fuel tumor growth by educating infiltrating leukocytes to produce the mitogen Gas6. Blood 115, 2264–2273 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Lindau, R. et al. Interleukin-34 is present at the fetal–maternal interface and induces immunoregulatory macrophages of a decidual phenotype in vitro. Hum. Reprod. 33, 588–599 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Baghdadi, M. et al. Chemotherapy-induced IL34 enhances immunosuppression by tumor-associated macrophages and mediates survival of chemoresistant lung cancer cells. Cancer Res. 76, 6030–6042 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Chen, A. et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell 185, 1777–1792.e21 (2022).

    Article  CAS  PubMed  Google Scholar 

  46. He, S. et al. High-plex imaging of RNA and proteins at subcellular resolution in fixed tissue by spatial molecular imaging. Nat. Biotechnol. 40, 1794–1806 (2022).

    Article  CAS  PubMed  Google Scholar 

  47. Newman, A. M. et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 37, 773–782 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lim, K. C. et al. Microvascular invasion is a better predictor of tumor recurrence and overall survival following surgical resection for hepatocellular carcinoma compared to the Milan criteria. Ann. Surg. 254, 108–113 (2011).

    Article  PubMed  Google Scholar 

  49. Barkley, D. et al. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat. Genet. 54, 1192–1201 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Canellas-Socias, A. et al. Metastatic recurrence in colorectal cancer arises from residual EMP1+ cells. Nature 611, 603–613 (2022).

    Article  CAS  PubMed  Google Scholar 

  51. Jiang, Y., Leng, J., Lin, Q. & Zhou, F. Epithelial–mesenchymal transition related genes in unruptured aneurysms identified through weighted gene coexpression network analysis. Sci. Rep. 12, 225 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Basu, M. et al. Invasion of ovarian cancer cells is induced by PITX2-mediated activation of TGF-β and Activin-A. Mol. Cancer 14, 162 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Costa, A. et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 33, 463–479.e10 (2018).

    Article  CAS  PubMed  Google Scholar 

  54. Chen, I. X. et al. Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc. Natl Acad. Sci. USA 116, 4558–4566 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Affo, S., Yu, L. X. & Schwabe, R. F. The role of cancer-associated fibroblasts and fibrosis in liver cancer. Annu. Rev. Pathol. 12, 153–186 (2017).

    Article  CAS  PubMed  Google Scholar 

  56. Ohlund, D. et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 214, 579–596 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Elyada, E. et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 9, 1102–1123 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Puram, S. V. et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171, 1611–1624.e24 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Casanova-Acebes, M. et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature 595, 578–584 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Mulder, K. et al. Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity 54, 1883–1900.e5 (2021).

    Article  CAS  PubMed  Google Scholar 

  61. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Haghverdi, L., Lun, A. T. L., Morgan, M. D. & Marioni, J. C. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat. Biotechnol. 36, 421–427 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Welch, J. D. et al. Single-cell multi-omic integration compares and contrasts features of brain cell identity. Cell 177, 1873–1887.e17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hie, B., Bryson, B. & Berger, B. Efficient integration of heterogeneous single-cell transcriptomes using Scanorama. Nat. Biotechnol. 37, 685–691 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Qiu, X. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  CAS  PubMed  Google Scholar 

  69. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, C. et al. SciBet as a portable and fast single cell type identifier. Nat. Commun. 11, 1818 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Kinker, G. S. et al. Pan-cancer single-cell RNA-seq identifies recurring programs of cellular heterogeneity. Nat. Genet. 52, 1208–1218 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Wang, X., Park, J., Susztak, K., Zhang, N. R. & Li, M. Bulk tissue cell type deconvolution with multi-subject single-cell expression reference. Nat. Commun. 10, 380 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all patients and their families for participating in this study. We also thank J. Li, C. Bleriot and B. Su for discussions; J. Li from Renji Hospital for clinical sample preparation; Y. Ye for providing the computing platform; and the BGI STOmics team for Stereo-seq studies. Z.Y.L. was supported by the National Natural Science Foundation of China (NSFC) (32100722). P.K.-H.C. is supported by the National Medical Research Council (NMRC) OFLCG21jun-0001 and TCR/015-NCC/2016. A. Sharmaʼs laboratory is supported by funding from the National Health and Medical Research Council (NHMRC) Ideas Grant [2021/GNT2010795], Medical Research Future Fund (MRFF) Early to Mid-Career Researchers (EMCR) grant [2022/MRF2016215], Perkins-Curtin start-up fellowship and Cancer Research Trust (CRT) Programme Grant to the Liver Cancer Collaborative. F.G. is supported by the Shanghai Institute of Immunology funding, the Biomedical Research Council (BMRC) use-inspired basic research (UIBR) award, and the Singapore National Research Foundation Senior Investigatorship (NRFI) NRF2016NRF-NRFI001-02.

Author information

Authors and Affiliations

Authors

Contributions

A. Sharma, F.G. and P.K.-H.C. conceptualized the study and supervised the project; Z.Y.L., R.P., J.C., W.G., L.Q.L., Y.Q.B., B.C.Y., A.M. and K.Y. conducted the experiments; Z.Y.L., R.P., S.G., J.C., W.G., A. Sharma and F.G. analyzed the data; A.D.B, H.F., Z.J.Z., Z.Z.L., A. Singh, M.W., Q.X., J.C., J.G. and P.K.-H.C. collected clinical samples; Z.Y.L., R.P., S.G., J.C., W.G. and Y.J.Y. generated figures; Z.Y.L., A. Sharma and F.G. wrote the manuscript, with all the authors contributing to writing and providing feedback.

Corresponding authors

Correspondence to Pierce Kah-Hoe Chow, Florent Ginhoux or Ankur Sharma.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Florian Greten, Mara Sherman and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Characteristics of CAF subsets from HCC atlas.

a, Stacked bar plots showing proportions of cells from each study among different mesenchymal subsets. b, Bubble heatmap showing expression of marker genes across 9 mesenchymal subsets in HCC atlas. Dot size indicates the fraction of expressing cells, colored according to z-score normalized expression levels. c, Violin plots showing the specific high expression levels of PI16, CD34 and DPT in cluster F6. d, Volcano plot showing differentially expressed genes between VSMC-like cells and EM CAFs. P-value < 0.05, Two-sided unpaired limma-moderated t test; log2(FC) > 0.5. e, Violin plots showing the expression of myHSC and cyHSC signature score in each cluster. For a-e, n = 6,163 mesenchymal cells. f, Spatial visualization of selected marker genes of CAF subpopulations in VISIUM_1 (left) and VISIUM_2 (right) slides. In VISIUM 1, Normal, n = 2,956 spots; Leading-edge, n = 2,791 spots; Tumor, n = 3,184 spots. In VISIUM 2, Normal, n = 4,628 spots; Leading-edge, n = 4,672 spots; Tumor, n = 4,733 spots.

Source data

Extended Data Fig. 2 Characteristics of fibroblast subsets from HCC.

a, UMAP projection of fibroblast subsets identified in HCC tumors (left) and adjacent normal tissues (right). b, Bubble heatmap showing expression of marker genes across 9 fibroblast clusters. Dot size indicates the fraction of expressing cells, colored according to z-score normalized expression levels. c, Projection of 9 fibroblast clusters generated from Sharma data back onto the mesenchymal cells from HCC atlas. d, UMAP plot showing the developmental trajectories (black skeleton lines) of fibroblasts inferred by Monocle3. Points are colored by clusters. e, UMAP plot showing the developmental trajectories of fibroblasts inferred by RNA velocity. Points are colored by Monocle3 pseudotime. f, Boxplot showing the Monocle3 pseudotime distribution of fibroblasts in the two major paths to POSTN+ CAF or MYH11+ CAF. Center line indicates the median value, lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. g, Scatter plot showing the specificity scores of regulons of POSTN+ CAF and MYH11+ CAF. h, Volcano plot showing differentially expressed genes between POSTN+ CAF and MYH11+ CAF. P-value < 0.05, Two-sided unpaired limma-moderated t test; log2(FC) > 0.5. i, FACs analyses showing a much higher expression of CD105 (ENG) in POSTN+ CAF compared to MYH11+ CAF. For a-f, ABCA8+ Fib, n = 25 cells; Fib, n = 341 cells; MT1M+ Fib, n = 83 cells; CAF, n = 384 cells; HSP+ CAF, n = 190 cells; MYH11+ CAF, n = 213 cells; SDC2+ CAF, n = 283 cells; POSTN+ CAF, n = 97 cells; APOA2+ CAF, n = 96 cells; In total, n = 1,712 cells.

Source data

Extended Data Fig. 3 Characteristics of fetal liver fibroblast subsets and identification of onco-fetal fibroblasts.

a, Heatmap showing the expression patterns of selected genes across fetal liver fibroblast clusters. b, UMAP plots displaying the developmental trajectories (black skeleton lines) of fetal liver fibroblasts inferred by Monocle3. Points are colored by clusters (left) and pseudotime (right). c, Boxplot showing the pseudotime distribution of fetal liver fibroblasts in the two major paths to COL1A1+ Fib or ITM2C+ Fib. Center line indicates the median value, lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. d, Fetal age preference of each cluster estimated by Ro/e score. +++, Ro/e > 1; ++, 0.8 < Ro/e ≤ 1; +, 0.2 ≤ Ro/e ≤ 0.8; +/-, 0 < Ro/e < 0.2. e, UMAP plot of integrated analyses showing 12 clusters of fibroblasts from healthy livers, fetal livers, HCC tumors and HCC adjacent normal tissues. f, UMAP plot showing the developmental trajectories (black skeleton lines) of joint clusters by Monocle 3. g, Bar plot showing tissue distribution of each joint cluster. h, Similarity analysis of fibroblast clusters from human fetal liver and HCC. i, Heatmap depicting expression levels of selected genes across fibroblast clusters identified from human fetal liver and HCC. j,k, Bar plot showing enriched GO terms of genes upregulated in both ITM2C+ Fib and POSTN+ CAF (j) and of POSTN+ CAF-specific upregulated genes (k). Hypergeometric test. For a-d, IGFBP3+ Fib, n = 810 cells; BGN+ Fib, n = 250 cells; PLEK+ Fib, n = 66 cells; ACTA2+ Fib, n = 333 cells; COL1A1+ Fib, n = 310 cells; APOC3+ Fib, n = 150 cells; ITM2C+ Fib, n = 210 cells; In total, n = 2,130 cells. For e-i, healthy liver, n = 1,564 cells; fetal liver, n = 2,130 cells; HCC tumor, n = 1,440 cells; adjacent normal liver, n = 272 cells; In total, n = 5,406 cells.

Source data

Extended Data Fig. 4 Onco-fetal interactions in HCC.

a, Heatmaps depicting the expression of potential ligands regulating the onco-fetal cell types. b, Lollipop plot showing number of significant interaction events between onco-fetal cell types with other clusters. c, Heatmap depicting the expression of shared potential ligands driving onco-fetal reprogramming. d, Bubble heatmap showing the cell-cell communications from malignant hepatocytes to onco-fetal cell types. Dot size indicates P-value, colored by communication probability. e, Bubble heatmap showing expression of selected POSTN+ CAF functional genes across fibroblasts in fetal liver. Dot size indicates the fraction of expressing cells, colored according to z-score normalized expression levels. For a-e, n = 72,330 cells were used for cell-cell communication analyses.

Source data

Extended Data Fig. 5 Spatial transcriptomic profiles of two HCC samples by Stereo-seq.

a, The number of genes captured by Stereo-seq at 10–200 bin resolutions. b, The number of transcripts captured by Stereo-seq at 10–200 bin resolutions. c, The DAPI fluorescence images (left), H&E staining images (middle) and number of genes detected at 100 bin resolution (right) of HCC01 (upper) and HCC03 (bottom). No replicates for these micrographs. d, Spatial visualization of onco-fetal markers FOLR2, PLVAP and POSTN in HCC01 analyzed by Stereo-seq at bin 100 resolution. e, Spatial visualization of onco-fetal cells in HCC01. The bins enriched with onco-fetal cells are highlighted with red. f, Spatial visualization of expression of indicated genes in HCC01 (upper) and HCC03 (bottom) at bin 100 resolution. g, Boxplot showing the expression of POSTN+ CAF signature in bins with different enrichment patterns of PLVAP+ EC or FOLR2+ TAM1 in HCC01. PLVAP+ EC low, n = 29,964 bins; PLVAP+ EC high, n = 6,812 bins; FOLR2+ TAM1 low, n = 29,649 bins; FOLR2+ TAM1 high, n = 7,127 bins. h, Bubble heatmap showing expression of POSTN+, POSTN- and panCAF signature genes in different CAF subsets. Dot size indicates the fraction of expressing cells, colored according to z-score normalized expression levels. i, Spatial visualization of selected cluster signatures in HCC03 analyzed by Stereo-seq at bin 100 resolution. j, Boxplot showing the expression of POSTN- and panCAF signature in bins with different enrichment patterns of PLVAP+ EC or FOLR2+ TAM1 in HCC03. PLVAP+ EC low, n = 19,905 bins; PLVAP+ EC high, n = 4,036 bins; FOLR2+ TAM1 low, n = 20,013 bins; FOLR2+ TAM1 high, n = 3,928 bins. For c-f and i, HCC01, n = 36,776 bins; HCC03, n = 23,941 bins. For g and j, Center line indicates the median value, lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. *P < 0.05, **P < 0.01, ***P < 0.001. Two-sided Wilcoxon test. Spatial cross-correlation test.

Source data

Extended Data Fig. 6 Colocalization of onco-fetal cells in Stereo-seq data.

a, Spatial visualization of onco-fetal niche (upper) and core-ligands score (bottom) in HCC03 analyzed by Stereo-seq at bin 100 resolution. b, Boxplot showing the expression of 10 core ligands driving onco-fetal reprogramming in bins with different enrichment patterns of onco-fetal niche. c, Spatial visualization of selected core-ligands expression in HCC03 analyzed by Stereo-seq at bin 100 resolution. d-g, Spatial visualization of CXCL12-CXCR4 and DLL4-NOTCH3 ligand-receptor pairs expression. h, Boxplot showing the distance quantification between onco-fetal cells and either onco-fetal cells or other non-onco-fetal (Non-OF) cells. A distance of 1000 units is 0.16454 mm. POSTN+ CAF, n = 103 cells; PLVAP+ EC, n = 90 cells; FOLR2+ TAM, n = 28 cells; Non-OF Fibroblast, n = 472 cells; Non-OF EC, n = 103 cells; Non-OF Myeloid, n = 198 cells. For a-g, HCC01, n = 36,776 bins; HCC03, n = 23,941 bins. For b and h, Center line indicates the median value, lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. *P < 0.05, **P < 0.01, ***P < 0.001. Two-sided Wilcoxon test. For c-g, *P < 0.05, **P < 0.01, ***P < 0.001. Spatial cross-correlation test.

Extended Data Fig. 7 Bulk RNA-seq profiles of 55 HCC patients.

a, Schema for analysis of onco-fetal ecosystem in n = 250 bulk HCC specimens. b, Framework for evaluating CIBERSORTx performance. c, Boxplots showing Pearson correlation coefficients between cell fractions estimated by CIBERSORTx and ground truth simulated proportions. n = 5 replicates. d, Boxplot showing the cosine similarity of cell fractions estimated by CIBERSORTx and MuSiC in n = 198 specimens. e, Boxplots showing the inferred proportions of FOLR2+ TAM1 and POSTN+ CAF in specimens with high (n = 99) versus low (n = 99) proportions of PLVAP+ EC. f, Histogram showing the number of patients with Simpsons’ index of diversity measuring the diversity of onco-fetal status of specimens from each patient. n = 54 patients. g, Contingency tables showing relapse (left) and HBV viral status (right) with patients with high onco-fetal score versus low onco-fetal score in TCGA LIHC dataset (left, n = 291 patients) and PLANET cohort (right, n = 54 patients), respectively. Fisher’s exact test. h, Onco-fetal score showing the largest odds ratio (5.99) associated with relapse. Among individual cell types, POSTN+ CAF showed the highest odds ratio (5.28). i, TNM stage preference of n = 124 relapsed specimens with high or low onco-fetal score estimated by Ro/e. j, Boxplots showing the inferred proportions of onco-fetal cell types in different TNM tumor stages. Stage I, n = 87 specimens; Stage II, n = 63 specimens; Stage III, n = 48 specimens. k, The Kaplan-Meier curves of time-to-recurrence stratified by onco-fetal score in n = 29 relapsed patients. l, Volcano plot showing differentially expressed genes between relapsed specimens with low or high onco-fetal score. P-value < 0.05, Two-sided unpaired limma-moderated t test; log2(FC) > 1. m, Heatmap depicting the expression of differentially expressed genes between relapsed HCC samples with high versus low onco-fetal score across tumor cells from scRNA-seq. n, The Kaplan-Meier curves of overall survival (OS) stratified by EMT-like malignant cell signatures in n = 364 patients. o, Boxplot showing the expression of cell cycle, cEMT and epiHR signatures in relapsed specimens with low or high onco-fetal score. Non-relapsed, n = 71 specimens; OFlow relapsed, n = 93 specimens; OFhi relapsed, n = 31 specimens. p, Spatial visualization of non-EMT and EMT-like malignant cell signatures in VISIUM_1 (upper) and VISIUM_2 (bottom) tumor and adjacent normal liver slides. In VISIUM 1, Normal, n = 2,956 spots; Tumor, n = 3,184 spots. In VISIUM 2, Normal, n = 4,628 spots; Tumor, n = 4,733 spots. q, Ligand activities (left) and expression levels across three onco-fetal cell types (right). Prioritized ligands are identified by NicheNet analysis based on upregulated genes in relapsed specimens with high onco-fetal score. Dot size indicates the fraction of expressing cells, colored according to expression levels. r, Charactering Stereo-seq bins by neighborhood index. In the upper-left schematic, grey bins indicate the presence of interested cells, red bins indicate the targets, and dashed lines frame their surrounding neighborhood bins. Other panels showing the neighborhood index of three onco-fetal cell types at bin 100 resolution in HCC03. s, Boxplot depicting the changes in proportions of onco-fetal high bins with the increase of distance to EMT-like tumor cells. Grey boxes indicate the randomly sampled bins (n = 10 replicates) as control, and red dots indicate the bins with EMT-like tumor cells, which exhibited a significant enrichment of onco-fetal high bins surrounding the EMT-like tumor cells. For c-e, j, o and s, center line indicates the median value, lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. For e, j and o, *P < 0.05, **P < 0.01, ***P < 0.001. Two-sided Wilcoxon test. For k and n, HR, hazard ratio. P-value was determined by Kaplan–Meier survival curves and log-rank test. *P < 0.05, **P < 0.01, ***P < 0.001.

Source data

Extended Data Fig. 8 Intercellular communication analysis in relapsed versus non-relapsed patients.

a, Expression of onco-fetal ligands in onco-fetal ligand producing cells from HCC scRNA-seq data. b, Bar plots showing the total number of interactions and interaction strength of the inferred cell-cell communication networks from non-relapsed and relapsed patients by CellChat analysis. c, Scatter plots showing the outgoing and incoming interaction strength in non-relapsed and relapsed patients, respectively. The onco-fetal cell types are highlighted with magenta, while blue points denote other clusters. d, Heatmap depicting the differential expression strength of the inferred cell-cell communication networks. Red (or blue) color indicates the increased (or decreased) signaling in relapse. The top and right colored bar plots represent the sum of column (incoming signaling) and row (outgoing signaling), respectively. e, Bar plot showing the overall information flow of indicated signaling pathways in relapsed and non-relapsed patients. f, Bubble heatmap showing the significantly different cell-cell communication in no-relapsed and relapsed patients. Dot size indicates P-value, colored by communication probability. g, Bubble heatmap showing the expression of prioritized ligands regulating EMT-like tumor cells across onco-fetal cells. Dot size indicates the fraction of expressing cells, colored according to expression levels. For a-g, n = 21,997 cells from relapsed patients and n = 34,232 cells from non-relapsed patients in tumor tissues were used for cell-cell communication analyses.

Source data

Extended Data Fig. 9 NanoString WTA of HCC and fetal liver specimens.

a, Boxplots showing the inferred proportions of POSTN+ CAF in FOLR2+ TAM1 and PLVAP+ EC high (n = 11) or low (n = 11) ROIs. b, PCA analysis of n = 48 ROIs from adjacent normal and tumor tissues. Orange (or skyblue) points indicate the ROIs from tumor (or adjacent normal) tissues. Circle (or triangle) points indicate the ROIs from non-relapsed B015 (or relapsed B017). c, Boxplots showing the expression of onco-fetal cell signatures in B015 and B017. B015 normal, n = 14 ROIs; B017 normal, n = 12 ROIs; B015 tumor, n = 10 ROIs; B017 tumor, n = 12 ROIs. d, Volcano plot showing differentially expressed genes between ROIs in cluster C1 and C2. P-value < 0.05, Two-sided unpaired limma-moderated t test; log2(FC) > 0.5. e, UMAP plots displaying the cluster (left) and AUC score of C1/C2 signature (right) in n = 72,330 cells from scRNA-seq dataset. f, Heatmap showing expression levels of selected genes across n = 72 ROIs. g, PCA analysis of n = 72 ROIs from adjacent normal, tumor and fetal liver tissues, colored by tissue source (upper) and cluster (bottom). h, Stacked bar plot depicting the proportions of tissue source in each cluster. i, Boxplots showing the expression levels of onco-fetal high relapse-related tumor cell signatures between ROIs in cluster C1 (n = 12 ROIs) and C2 (n = 8 ROIs). For a, c and i, center line indicates the median value, lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. *P < 0.05, **P < 0.01, ***P < 0.001. Two-sided Wilcoxon test.

Source data

Extended Data Fig. 10 Associations among onco-fetal ecosystem, ABRS genes and Tregs.

a, Boxplots showing the expression of ABRS genes in scRNA-seq data (left), whose expression patterns are also shown in heatmap (right). b, Boxplots showing the expression of the Treg signature in scRNA-seq data (left), whose expression patterns are also shown in heatmap (right). c, The correlation between ABRS score and Treg signature in GO30140 and IMbrave150 cohorts. Linear regression and Pearson’s correlation. d, The Kaplan–Meier curves of PFS in GO30140 stratified by median expression score of the Treg signature. Multivariate Cox regression. HR, hazard ratio. P-value was determined by Kaplan–Meier survival curves and log-rank test. *P < 0.05, **P < 0.01, ***P < 0.001. e, Boxplots showing the onco-fetal score in patients with high or low levels of ABRS. f, The correlation between ABRS score and onco-fetal cell signatures in GO30140 and IMbrave150 cohorts. Linear regression and Pearson’s correlation. g, Boxplots showing the expression levels of onco-fetal cell signatures in patients with high or low levels of the Treg signature. h, Boxplots showing the onco-fetal score in patients with high or low levels of Treg signature. i, Boxplots showing the expression levels of EMT-like and non-EMT signatures in patients with high or low levels of ABRS score and the Treg signature. j, Schematic of the complete treatment process and gadolinium enhanced MRI images of M311 responder in M0, M11 and M16. M refers to month. k, Spatial visualization of Treg, EMT-like and non-EMT signatures using neighborhood index (NI) in M311 (responder) and M316 (non-responder). For a,b, n = 72,330 cells. For c-i, In GO30140, ABRS low, n = 43 patients; ABRS high, n = 43 patients; Treg low, n = 43 patients; Treg high, n = 43 patients. In IMbave150, ABRS low, n = 60 patients; ABRS high, n = 59 patients; Treg low, n = 60 patients; Treg high, n = 59 patients. For e and g-i, center line indicates the median value, lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. *P < 0.05, **P < 0.01, ***P < 0.001. Two-sided Wilcoxon test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–6.

Reporting Summary

Supplementary Tables 1–12

Supplementary Tables 1–12.

Supplementary Data

Source data for supplementary figures.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig./Table 1

Statistical source data.

Source Data Extended Data Fig./Table 2

Statistical source data.

Source Data Extended Data Fig./Table 3

Statistical source data.

Source Data Extended Data Fig./Table 4

Statistical source data.

Source Data Extended Data Fig./Table 5

Statistical source data.

Source Data Extended Data Fig./Table 7

Statistical source data.

Source Data Extended Data Fig./Table 8

Statistical source data.

Source Data Extended Data Fig./Table 9

Statistical source data.

Source Data Extended Data Fig./Table 10

Statistical source data.

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

Li, Z., Pai, R., Gupta, S. et al. Presence of onco-fetal neighborhoods in hepatocellular carcinoma is associated with relapse and response to immunotherapy. Nat Cancer 5, 167–186 (2024). https://doi.org/10.1038/s43018-023-00672-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00672-2

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer