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:

Liver tumour immune microenvironment subtypes and neutrophil heterogeneity

Abstract

The heterogeneity of the tumour immune microenvironment (TIME), organized by various immune and stromal cells, is a major contributing factor of tumour metastasis, relapse and drug resistance1,2,3, but how different TIME subtypes are connected to the clinical relevance in liver cancer remains unclear. Here we performed single-cell RNA-sequencing (scRNA-seq) analysis of 189 samples collected from 124 patients and 8 mice with liver cancer. With more than 1 million cells analysed, we stratified patients into five TIME subtypes, including immune activation, immune suppression mediated by myeloid or stromal cells, immune exclusion and immune residence phenotypes. Different TIME subtypes were spatially organized and associated with chemokine networks and genomic features. Notably, tumour-associated neutrophil (TAN) populations enriched in the myeloid-cell-enriched subtype were associated with an unfavourable prognosis. Through in vitro induction of TANs and ex vivo analyses of patient TANs, we showed that CCL4+ TANs can recruit macrophages and that PD-L1+ TANs can suppress T cell cytotoxicity. Furthermore, scRNA-seq analysis of mouse neutrophil subsets revealed that they are largely conserved with those of humans. In vivo neutrophil depletion in mouse models attenuated tumour progression, confirming the pro-tumour phenotypes of TANs. With this detailed cellular heterogeneity landscape of liver cancer, our study illustrates diverse TIME subtypes, highlights immunosuppressive functions of TANs and sheds light on potential immunotherapies targeting TANs.

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: The single-cell landscape of 124 patients with liver cancer.
Fig. 2: Five TIME subtypes of PLC.
Fig. 3: Neutrophil heterogeneity and functional validation in humans.
Fig. 4: Neutrophil heterogeneity and depletion in mouse models.

Similar content being viewed by others

Data availability

Raw sequencing data reported in this paper have been deposited at the Genome Sequence Archive at the National Genomics Data Center (Beijing, China) under the BioProject ID PRJCA007744. The data deposited and made public are compliant with the regulations of the Ministry of Science and Technology of China. To facilitate the use of our data by the wider research community, we developed an interactive web-based tool (http://meta-cancer.cn:3838/scPLC) for analysing and visualizing our single-cell data. Other public data used in this study include reference genomes for human (https://asia.ensembl.org/, GRCh38.p13) and mouse (https://asia.ensembl.org/, GRCm39) and TCGA datasets (https://portal.gdc.cancer.gov/). Source data are provided with this paper.

Code availability

Codes used in this study are available at GitHub (https://github.com/meta-cancer/scPLC).

References

  1. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Thorsson, V. et al. The immune landscape of cancer. Immunity 48, 812–830 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Li, X. et al. The immunological and metabolic landscape in primary and metastatic liver cancer. Nat. Rev. Cancer 21, 541–557 (2021).

    Article  CAS  PubMed  Google Scholar 

  4. Marquardt, J. U., Andersen, J. B. & Thorgeirsson, S. S. Functional and genetic deconstruction of the cellular origin in liver cancer. Nat. Rev. Cancer 15, 653–667 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. 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 

  6. Coffelt, S. B., Wellenstein, M. D. & de Visser, K. E. Neutrophils in cancer: neutral no more. Nat. Rev. Cancer 16, 431–446 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Jaillon, S. et al. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 20, 485–503 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Shaul, M. E. & Fridlender, Z. G. Tumour-associated neutrophils in patients with cancer. Nat. Rev. Clin. Oncol. 16, 601–620 (2019).

    Article  PubMed  Google Scholar 

  9. Ng, L. G., Ostuni, R. & Hidalgo, A. Heterogeneity of neutrophils. Nat. Rev. Immunol. 19, 255–265 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Quail, D. F. et al. Neutrophil phenotypes and functions in cancer: a consensus statement. J. Exp. Med. 219, e20220011 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cui, C. et al. Neutrophil elastase selectively kills cancer cells and attenuates tumorigenesis. Cell 184, 3163–3177 (2021).

    Article  CAS  PubMed  Google Scholar 

  12. Ponzetta, A. et al. Neutrophils driving unconventional T cells mediate resistance against murine sarcomas and selected human tumors. Cell 178, 346–360 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yang, L. et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature 583, 133–138 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Szczerba, B. M. et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature 566, 553–557 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  17. 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 

  18. Ma, L. et al. Tumor cell biodiversity drives microenvironmental reprogramming in liver cancer. Cancer Cell 36, 418–430 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. Zheng, C. et al. Landscape of infiltrating t cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Zhang, M. et al. Single-cell transcriptomic architecture and intercellular crosstalk of human intrahepatic cholangiocarcinoma. J. Hepatol. 73, 1118–1130 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Aizarani, N. et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Xie, X. et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat. Immunol. 21, 1119–1133 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ballesteros, I. et al. Co-option of neutrophil fates by tissue environments. Cell 183, 1282–1297 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Bagaev, A. et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 39, 845–865 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  28. Kiss, M. et al. IL1β promotes immune suppression in the tumor microenvironment independent of the inflammasome and gasdermin D. Cancer Immunol. Res. 9, 309–323 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Feig, C. et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl Acad. Sci. USA 110, 20212–20217 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 169, 1327–1341 (2017).

    Article  Google Scholar 

  31. Farshidfar, F. et al. Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH-mutant molecular profiles. Cell Rep. 18, 2780–2794 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Xue, R. et al. Genomic and transcriptomic profiling of combined hepatocellular and intrahepatic cholangiocarcinoma reveals distinct molecular subtypes. Cancer Cell 35, 932–947 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Xiang, X. et al. IDH mutation subgroup status associates with intratumor heterogeneity and the tumor microenvironment in intrahepatic cholangiocarcinoma. Adv. Sci. 8, e2101230 (2021).

    Article  Google Scholar 

  35. Wu, S. Z. et al. A single-cell and spatially resolved atlas of human breast cancers. Nat. Genet. 53, 1334–1347 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ramachandran, P., Matchett, K. P., Dobie, R., Wilson-Kanamori, J. R. & Henderson, N. C. Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis. Nat. Rev. Gastroenterol. Hepatol. 17, 457–472 (2020).

    Article  PubMed  Google Scholar 

  37. Wculek, S. K. & Malanchi, I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 528, 413–417 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Boivin, G. et al. Durable and controlled depletion of neutrophils in mice. Nat. Commun. 11, 2762 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Remmerie, A. et al. Osteopontin expression identifies a subset of recruited macrophages distinct from kupffer cells in the fatty liver. Immunity 53, 641–657 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Guilliams, M. et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell 185, 379–396 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Seehawer, M. et al. Necroptosis microenvironment directs lineage commitment in liver cancer. Nature 562, 69–75 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Muhl, L. et al. Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination. Nat. Commun. 11, 3953 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal-fetal interface in humans. Nature 563, 347–353 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Qiu, X. et al. Single-cell mRNA quantification and differential analysis with Census. Nat. Methods 14, 309–315 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lange, M. et al. CellRank for directed single-cell fate mapping. Nat. Methods 19, 159–170 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Schürch, C. M. et al. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 182, 1341–1359 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Fan, Y. et al. Targeting multiple cell death pathways extends the shelf life and preserves the function of human and mouse neutrophils for transfusion. Sci. Transl. Med. 13, eabb1069 (2021).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Guo, C. Shan and J. Ren from National Center for Protein Sciences at Peking University for FACS and CODEX assistance. This work is jointly supported by National Natural Science Foundation of China (81988101, 82173035, 82030079, 81972656, 81802813, 81902401, 81972735 and 81872508), the National Science and Technology Major Project of China (2018ZX10723204), Beijing Natural Science Foundation (7212108), Changping Laboratory, the Michigan Medicine and PKU-HSC JI for Translational and Clinical Research (BMU2020JI005) and Sino-Russian Math Center in PKU.

Author information

Authors and Affiliations

Authors

Contributions

R.X., Z.Z. and N.Z. conceived and designed the project. R.X., X.X., Z.L. and J.Z. collected the human samples and clinical information. X.X., Z.L. and J.Z. performed pathological examination. R.X. and X.X. performed the scRNA-seq experiments. Q.C., Q. Zhang and R.X. performed bioinformatic analyses. Q. Zhang, R.X. and Q.C. performed IHC, mIHC and CODEX experiments. R.K., R.X., M.F. and F.W. performed functional experiments of neutrophils. R.K. and R.X. constructed the mouse models and analysed the in vivo data. R.X., Q. Zhang, Q.C., R.K., X.X., H.L., Q. Zhan, M.D., J.Z., Z.Z. and N.Z. discussed and interpreted the data. Q.C., R.X. and J.C. built the online website. R.X., Q. Zhang, Q.C. and R.K. wrote the manuscript with help from Z.Z. and N.Z.; Z.Z., J.Z. and N.Z. supervised the project.

Corresponding authors

Correspondence to Jiye Zhu, Zemin Zhang or Ning Zhang.

Ethics declarations

Competing interests

Z.Z. is a founder of Analytical BioSciences and is a consultant for InnoCare Pharma and ArsenalBio. N.Z. is the CSO of Yunnan Baiyao Group. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Andres Hidalgo, Alexander Swarbrick 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 figures and tables

Extended Data Fig. 1 Patient cohort and cluster information.

a, Pie charts showing the composition of cancer types in our cohort. HCC, hepatocellular carcinoma; ICC, intrahepatic cholangiocarcinoma; CHC, combined hepatocellular and cholangiocarcinoma; HH, hepatic hemangioma; ASC, adenosquamous carcinoma; SAR, sarcomatoid carcinoma; SLC, secondary liver cancer. CRC_M, liver metastasis from colorectal cancer, PAN_M, liver metastasis from pancreatic cancer, LYM_M, liver metastasis from lymphoma, GAS_M, liver metastasis from gastric cancer, BRC_M, liver metastasis from breast cancer. b, UMAP plots showing the distribution of patients, cancer types, viruses and liver cirrhosis states. Dots represent individual cells. PB, peripheral blood; AL, adjacent liver; HBV, hepatitis B virus, HCV, hepatitis C virus, NBNC, double negative of HBV and HCV. c, UMAP plots showing expression of canonical marker genes of major cell populations including T cells (CD3D, CD8A, FOXP3), NK cells (NKG7), B cells (CD79A), macrophages (CD68), neutrophils (CSF3R), dendritic cells (CLEC10A), mast cells (TPSAB1), fibroblasts (COL1A1), endothelial cells (VWF), and epithelial cells (EPCAM). d, Stacked barplot showing the distribution of major cell types in each sample. e, UMAP plots showing the distribution of cell identities for tumour cells and TIME cells. Tumour cells were further coloured by patient, cancer type, virus, and cirrhosis. f, CNV profiles inferred from scRNA-seq data for each cell and from matched bulk exome data in the sample A014_HCC. g, Boxplots showing hepatic scores and biliary epithelial scores in tumour (n = 193,877 cells) and TIME cells (n = 898,295 cells). Cells are from 124 patients. h, Boxplots showing hepatic scores and biliary epithelial scores in tumour cells of different PLC subtypes (HCC, n = 96,211 cells from 79 cases, ICC, n = 52,345 cells from 25 cases, CHC, n = 15,493 cells from 7 cases). Cells are from 111 patients. i, Pie charts showing the patient number (top) and cell number (bottom) of our study and published single cell studies for PLC. Colours represent different studies. j, Stacked barplot showing proportions of major cell populations among different studies. Colours represent major cell populations. In g-h, n denotes individual cells. Two-sided Wilcoxon rank-sum test is used. For boxplots, centre line shows median, box limits indicate upper and lower quartiles, and whiskers extend 1.5 times the interquartile range, while data beyond the end of the whiskers are outlying points that are plotted individually. ***, P < 0.001.

Extended Data Fig. 2 Gene expression and tissue preference of 89 TIME cell clusters.

a, UMAP plots showing the expression of canonical marker genes for clusters in each major cell population. Normalized expression level was abbreviated as Exp. b, Heatmap showing tissue preferences of clusters in each major cell population revealed by Ro/e. c, Boxplots showing proportions of several tumour-enriched TIME clusters divided by PLC subtypes. *, P < 0.05; **, P < 0.01; ***, P < 0.001. (HCC, n = 79 cases, ICC, n = 25 cases, CHC, n = 7 cases). d, Boxplots showing proportions of several cell clusters associated with virus or cirrhosis. (HBV, n = 57 cases, HCV, n = 6 cases, NBNC, n = 50 cases; cirrhosis, n = 46 cases, non-cirrhosis, n = 67 cases). In c-d, n denotes biologically independent samples. Two-sided Wilcoxon rank-sum test is used. For boxplots, centre line shows median, box limits indicate upper and lower quartiles, and whiskers extend 1.5 times the interquartile range, while data beyond the end of the whiskers are outlying points that are plotted individually.

Extended Data Fig. 3 Clusters, signatures, and prognosis of five TIMELASER subtypes.

a, Heatmap showing frequencies of TIME cell clusters in 5 CMs. b, Forest plot showing the clinical relevance of clusters in each CM revealed by log10(hazard ratio) based on PFS. Cox regression. Log-rank test. c, Dot heatmap showing enriched pathways across TIMELASER subtypes. Benjamini–Hochberg-adjusted hypergeometric test. d, Boxplots showing the expression of given signatures in different TIMELASER subtypes. Signature scores of TIMELASER subtypes with overhead asterisk are significantly higher than that of subtypes with corresponding asterisk colour. Wilcoxon rank-sum test, two-sided. (TIME-IA, n = 18 cases, TIME-ISM, n = 8 cases, TIME-ISS, n = 12 cases, TIME-IE, n = 42 cases, TIME-IR, n = 31 cases, n denotes biologically independent patients). For boxplots, centre line shows median, box limits indicate upper and lower quartiles, and whiskers extend 1.5 times the interquartile range, while data beyond the end of the whiskers are outlying points that are plotted individually. e, Overall survival (OS) with each patient assigned to a single CM. Log-rank test. f, OS of cases stratified by each TIMELASER module. Log-rank test. In b and d, *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Extended Data Fig. 4 Validation of five TIMELASER subtypes.

a, Boxplots showing the percentage of TIMELASER modules across 3 PLC subtypes. (HCC, n = 79 cases, ICC, n = 25 cases, CHC, n = 7 cases). b, Heatmap showing the percentage of CM1–5 across tumours in our cohort and three published scRNA-seq cohorts. c, Expression of signature genes for the five TIMELASER subtypes in 453 published liver cancer bulk RNA-seq data. d, Boxplot showing z-scores of signature genes for five TIMELASER subtypes in different cancer types. Colours represents HCC (orange, n = 369 cases), ICC (green, n = 33 cases) and CHC (purple, n = 51 cases). e, Pie charts showing the proportion of TIMELASER subtypes in c. f, Representative CODEX results showing four different TIMELASER subtypes. For each sample, only six representative antibodies staining are displayed in the figure along with DAPI. Scale bar, 500 μm. g, Validation of TIMELASER by a published spatial transcriptomic study of liver cancer. H&E staining and the corresponding spatial feature plots of different marker genes of cell types are shown in different samples. In a and d, n denotes biologically independent samples. Two-sided Wilcoxon rank-sum test is used. For boxplots, centre line shows median, box limits indicate upper and lower quartiles, and whiskers extend 1.5 times the interquartile range, while data beyond the end of the whiskers are outlying points that are plotted individually. **, P < 0.01; ***, P < 0.001.

Extended Data Fig. 5 L-R networks and feature summary of five TIMELASER subtypes.

a, Heatmap showing Ro/e enrichment values of TIMELASER-specific L-R pairs. b, Chord diagrams showing the interactions within each TIMELASER subtype mediated by specific L-R pairs. Line width is proportional to interaction intensity and coloured by TIMELASER subtypes. c, Barplots showing the number of ligand-receptor pairs significantly enriched in TIME-IA and TIME-ISM modules. d, Heatmap showing the expression of chemokines and the corresponding receptors in TIME-IA and TIME-ISM patients. Exp, normalized mean expression. e, Summary of key features across TIMELASER subtypes. f, Schematic for five TIMELASER subtypes. Selected cell populations are shown for each TIMELASER subtype with tumour cells as background. Tex, exhausted T cell; NK, nature killer cell; TAN, tumour-associated neutrophil; TAM, tumour-associated macrophage; DC, dendritic cell.

Extended Data Fig. 6 Mutational landscape and GMs of malignant cells.

a, Heatmap showing frequencies of five TIMELASER subtypes across 111 PLC patient samples. Detailed clinical and molecular attributes of individual tumour samples are annotated. P values to the right indicate significant non-random distributions for each attribute. Chi-square test is used for categorical variables. Two-way ANOVA test is used for continuous variables. b, Stacked barplots showing the distribution of cancer types, virus and cirrhosis state across TIMELASER subtypes. Chi-square test. c, Boxplots showing the distribution of tumour purity, CNA and TMB inferred by WES data across TIMELASER subtypes. Two-way ANOVA test is used for comparison of multiple groups. Two-sided Wilcoxon rank-sum test is used for comparison between any two groups. d, Heatmap showing the mutational rate of somatic mutations enriched in different TIMELASER subtypes. e, Barplots showing mutational frequencies of TP53, KRAS, IDH1, and CTNNB1 in different TIMELASER subtypes. Colours represent different TIMELASER subtypes. One-sided Fisher’s exact test. Tests are performed between the denoted TIME subtype (P value colour coded) and a combination of all others. f, Heatmaps showing the eight common gene modules (GMs) extracted from tumour cells. g, Boxplots showing the distributions of signature scores of GMs across tumours stratified into five TIMELASER subtypes. Overhead asterisk is significantly higher than that of subtypes with corresponding asterisk colour. Wilcoxon rank-sum test, two-sided. In c and e, (TIME-IA, n = 13 cases, TIME-ISM, n = 7 cases, TIME-ISS, n = 7 cases, TIME-IE, n = 32 cases, TIME-IR, n = 20 cases). In g, (TIME-IA, n = 18 cases, TIME-ISM, n = 8 cases, TIME-ISS, n = 12 cases, TIME-IE, n = 42 cases, TIME-IR, n = 31 cases). In c, e, and g, n denotes biologically independent patients. For boxplots, centre line shows median, box limits indicate upper and lower quartiles, and whiskers extend 1.5 times the interquartile range, while data beyond the end of the whiskers are outlying points that are plotted individually. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Extended Data Fig. 7 Neutrophil heterogeneity in human PLC.

a, H&E and IHC plots showing the neutrophil frequencies in HCC and ICC patients. Scale bar, 20 μm. Boxplot to the right is the quantitative result. Student’s t-test, two sided. (HCC, n = 5 cases, ICC, n = 8 cases, n denotes biologically independent samples.) In the boxplot, centre line shows median, box limits indicate upper and lower quartiles, and whiskers extend 1.5 times the interquartile range, while data beyond the end of the whiskers are outlying points that are plotted individually. b, Dot heatmap showing the row-scaled expression of marker genes for neutrophil clusters. c, UMAP plots showing the expression of typical marker genes for neutrophil subsets. Exp, normalized expression. d, Distribution of neutrophil clusters by patient. e, Monocle trajectories of neutrophils coloured by tissues (left), cluster identities (middle) and CytoTRACE scores (right). Each dot represents a single cell. Cell orders are inferred based on the expression of the most variable genes across neutrophil clusters. f, Heatmap showing similarity scores of peripheral blood neutrophil clusters from Xie et al. and lung cancer neutrophil clusters from Zillionis et al. compared with liver cancer neutrophil clusters inferred by singleR. g, OS and PFS of patients stratified by the proportion of all neutrophils and neutrophil clusters (Neu_09/10/11) in TIME-ISM. Log-rank test. h, Average expression of classic neutrophil scores and TAN-specific gene scores in neutrophil clusters. i, Gene ontology analysis showing the enrichment of specific pathways in neutrophil clusters. Benjamini–Hochberg-adjusted hypergeometric test.

Source data

Extended Data Fig. 8 Transcription factors, mIHC and in vitro validation of neutrophil clusters.

a, UMAP plots showing regulon activities of five representative transcription factors for specific neutrophil clusters. Binding motifs of these transcription factors are shown on the top. b, Normalized ATAC-seq sequencing tracks of selected transcription factor loci in matched PBN, ALN, and TAN isolated from the same patient. ATAC peaks detected by MACS3 are denoted with the grey box above the gene body and highlighted with light red shading. c, Workflow of co-culture experiments of PBNs with or without cell line (liver cancer cell line HepG2, HCCLM3, and MHCC97H, control cell line HEK293T). d, Survival curve of PBN in culture condition (n = 3, n denotes biologically independent samples). Data are presented as mean values ± SEM. e, Expression of TAN-related signatures in PBNs co-cultured with or without different cell lines for 0 h, 18 h, 24 h, and 30 h. f, Expression of gene signatures of different neutrophil subsets in PBNs co-cultured with or without different cell lines. g, White arrows mark CCL4+CD66b+ neutrophils, with one cell highlighted by the four enlarged panels on the right. Middle panels show another representative CCL4CD66b+ neutrophil while right panels show a representative CCL4− CD66b+ neutrophil. Scale bars are 20 μm and 2 μm. h, Expression of selected genes in PBNs co-cultured with or without different cell lines for 0 h, 18 h, 24 h, and 30 h. i, Chord diagrams showing interactions between neutrophils and other cell types mediated by CCL3-CCR1 and CCL4-CCR5. Line width is proportional to interaction intensity, coloured by cell types with receptors. j, Crystal violet staining of migrated monocytes co-cultured with matched TAN or non-TAN. Scale bar, 100 μm. k, FACS analysis showing the PD-L1 expression of PBNs co-cultured with or without different cell lines for 24 h. Two-way ANOVA test is used for e and h.

Source data

Extended Data Fig. 9 Co-culture experiment of cell line-PBN-CD8+ T cells and analyses of two IFIT+ neutrophil subsets.

a, Experimental workflow. b, Gating strategy separating neutrophils from CD8+ T cells in the bottom chamber of co-culture system in a. c, FACS analysis showing the expression of CD25 (n = 3), CD69 (n = 3), and IFNγ (n = 4) in PBNs co-cultured with different cell lines. Student’s t-test, one-sided. Data are presented as mean values ± SEM. d, FACS analysis showing the expression of IFNγ in CD8+ T cells when anti-PD-L1 or the IgG control is added to the co-culture system. e, FACS analysis showing the expression of CD25, IFNγ, GZMB, and PRF1 in CD8+ T cells co-cultured with matched TAN or non-TAN isolated from patients with liver cancer. f, Volcano plot showing differentially expressed genes between Neu_03_ISG15 and Neu_09_IFIT1. Benjamini-Hochberg adjusted Wilcoxon rank-sum test, two-sided. g, Heatmap showing the predicted ligand activity by NicheNet on genes highly expressed in Neu_09_IFIT1. Pearson correlation indicates the ability of each ligand to predict the target genes, and better predictive ligands are thus ranked higher. h, Dot heatmap showing the selected ligand-receptor pairs between different cell populations and Neu_09_IFIT1. Benjamini-Hochberg adjusted permutation test. i, Boxplots showing the proportion of two IFNG+ populations between patients with or without Neu_09_IFIT1. Wilcoxon rank-sum test, two-sided. (Yes, n = 33 cases, No, n = 78 cases). For boxplots, centre line shows median, box limits indicate upper and lower quartiles, and whiskers extend 1.5 times the interquartile range, while data beyond the end of the whiskers are outlying points that are plotted individually. j, Pearson correlation between the expression of CD274 and IFNG in TIME cells in this study (left) or in the collected bulk RNA-seq datasets (right). In c and i, n denotes biologically independent samples.

Source data

Extended Data Fig. 10 scRNA-seq and functional analyses of mouse models.

a, Schematic of liver cancer mouse models. Intrahepatic delivery of the transposable vectors pTMC (encoding Myc and ∆90Ctnnb1) or pTMK (encoding Myc and KrasG12D) via HDTV in Alb-Cre × Trp53fl/fl mice. b, Representative photos, H&E, and IHC staining of HCC and ICC mouse models. Rulers in the photo show a minimum unit of mm. Scale bar on the staining slides is 20 μm. c, Survival curve of liver cancer mouse model. Log-rank test. d, UMAP plot showing major cell types of mice with liver cancer. Dots represent individual cells, and colours represent the major cell populations. mILC: innate lymphoid cells, mNeu: neutrophils, mMph: macrophages, mMono: monocytes, mEC: endothelial cells; mFb: fibroblasts, mEpithelial: hepatocytes, biliary cells and progenitors; the first letter m indicates mouse clusters. The two small UMAP plots show the distribution of mouse models (left) and tissue types (right). e, UMAP plot showing myeloid clusters including 5 DC, 2 monocyte and 7 macrophage clusters for liver cancer mouse models. f, Dot heatmap showing the row-scaled expression of typical marker genes for neutrophil clusters in mice. g, Stacked barplot showing the fraction of 12 mouse neutrophil subsets across PB, AL, and tumour. h, The trajectory path of mouse neutrophil clusters inferred by Monocle2. Each dot represents a single cell. Cell orders are inferred from the expression of the most variable genes. The trajectory direction is determined by biological prior. i, Heatmap showing Pearson’s correlations across neutrophil clusters in human and mouse. j, UMAP plots showing the integration of mouse and human neutrophil clusters. k, Sankey plot showing the similarities of the joint clusters, mouse tissue isolated neutrophil clusters, and human sample isolated neutrophil clusters. l, FACS analysis on neutrophil, macrophage, and CD8+ T cell populations in isotype and anti-Ly6G groups. The right barplot shows the decreased neutrophil number in anti-Ly6G group (n = 10). m, FACS analyses and coloured histogram showing reduced PD-L1 expression in TANs and reduced PD-1 and TIM3 expression in tumour-infiltrated CD8+ T cells of the anti-Ly6G group compared with isotype control. The left barplot shows the decreased PD-L1 expression of neutrophils in anti-Ly6G group (n = 8). n, IHC of CD68 in tumour regions of mice treated with isotype control or anti-Ly6G antibody (n = 6). o, FACS analysis showing the expression of surface and intracellular Ly6G in the isotype control and anti-Ly6G treatment groups. p, Bar plot showing the statistical analysis of FACS results (n = 3). In l-p, n denotes biologically independent samples, data are presented as mean values ± SEM, and two-sided Student’s t-test is used.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–5, Supplementary Notes. 1–9 and Supplementary Discussion.

Reporting Summary

Supplementary Table 1

Clinical information of the patients and library information of the samples.

Supplementary Table 2

Mutations and CNVs detected from WES.

Supplementary Table 3

Statistics of 89 cell clusters and 5 TIMELASER subtypes.

Supplementary Table 4

Statistics of cell clusters in mouse liver tumours.

Supplementary Table 5

Summary of antibodies and primers.

Supplementary Data

Source Data for Supplementary Fig. 3

Supplementary Data

Source Data for Supplementary Fig. 4

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

Xue, R., Zhang, Q., Cao, Q. et al. Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature 612, 141–147 (2022). https://doi.org/10.1038/s41586-022-05400-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05400-x

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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