Play all audios:
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. Access through your institution Buy or subscribe This is a preview of subscription content, access via your
institution ACCESS OPTIONS Access through your institution Access Nature and 54 other Nature Portfolio journals Get Nature+, our best-value online-access subscription $29.99 / 30 days cancel
any time Learn more Subscribe to this journal Receive 51 print issues and online access $199.00 per year only $3.90 per issue Learn more Buy this article * Purchase on SpringerLink *
Instant access to full article PDF Buy now Prices may be subject to local taxes which are calculated during checkout ADDITIONAL ACCESS OPTIONS: * Log in * Learn about institutional
subscriptions * Read our FAQs * Contact customer support SIMILAR CONTENT BEING VIEWED BY OTHERS SINGLE-CELL PROFILING REVEALS ALTERED IMMUNE LANDSCAPE AND IMPAIRED NK CELL FUNCTION IN
GASTRIC CANCER LIVER METASTASIS Article 26 July 2024 SINGLE-CELL RNA SEQUENCING REVEALED POTENTIAL TARGETS FOR IMMUNOTHERAPY STUDIES IN HEPATOCELLULAR CARCINOMA Article Open access 01
November 2023 THE HETEROGENEOUS IMMUNE LANDSCAPE BETWEEN LUNG ADENOCARCINOMA AND SQUAMOUS CARCINOMA REVEALED BY SINGLE-CELL RNA SEQUENCING Article Open access 26 August 2022 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 * 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 * Thorsson, V. et al. The immune landscape of cancer.
_Immunity_ 48, 812–830 (2018). Article CAS PubMed PubMed Central Google Scholar * 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 * 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 * 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 * 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 * 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 * 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 * Ng, L. G., Ostuni, R. & Hidalgo, A. Heterogeneity of neutrophils. _Nat. Rev. Immunol._ 19, 255–265 (2019). Article CAS PubMed Google Scholar
* 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 *
Cui, C. et al. Neutrophil elastase selectively kills cancer cells and attenuates tumorigenesis. _Cell_ 184, 3163–3177 (2021). Article CAS PubMed Google Scholar * 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 * 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 * 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 * Zhang, Q. et al.
Landscape and dynamics of single immune cells in hepatocellular carcinoma. _Cell_ 179, 829–845(2019). Article CAS PubMed Google Scholar * 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 * 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 * 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 * 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 * 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 * 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 * 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 * 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
* 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 * Ballesteros, I. et al. Co-option of neutrophil fates by tissue environments. _Cell_ 183, 1282–1297 (2020). Article CAS PubMed
Google Scholar * Bagaev, A. et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. _Cancer Cell_ 39, 845–865 (2021). Article CAS PubMed Google Scholar
* 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 * 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
* 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 * Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of
hepatocellular carcinoma. _Cell_ 169, 1327–1341 (2017). Article Google Scholar * 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 * 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 * 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 * 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 * 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 * 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 * 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 * 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 * 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 * 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 * 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 *
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 * 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 * 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 * 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 * 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 * 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 * 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 * 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 * 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 * Lange, M. et al. CellRank for directed single-cell fate
mapping. _Nat. Methods_ 19, 159–170 (2022). Article CAS PubMed PubMed Central Google Scholar * 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 * 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 * 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 * 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 Author notes * These authors contributed equally: Ruidong Xue, Qiming Zhang, Qi Cao, Ruirui Kong, Xiao Xiang * These authors jointly
supervised this work: Jiye Zhu, Zemin Zhang, Ning Zhang AUTHORS AND AFFILIATIONS * Translational Cancer Research Center, Peking University First Hospital, Beijing, China Ruidong Xue, Qi Cao,
Ruirui Kong, Hengkang Liu, Mei Feng, Fangyanni Wang, Jinghui Cheng & Ning Zhang * BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University,
Beijing, China Qiming Zhang & Zemin Zhang * Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Department of Hepatobiliary Surgery, Peking University
People’s Hospital, Beijing, China Xiao Xiang, Zhao Li & Jiye Zhu * International Cancer Institute, Peking University Health Science Center, Beijing, China Qimin Zhan, Mi Deng & Ning
Zhang * Changping Laboratory, Beijing, China Zemin Zhang * Yunnan Baiyao Group, Kunming, China Ning Zhang Authors * Ruidong Xue View author publications You can also search for this author
inPubMed Google Scholar * Qiming Zhang View author publications You can also search for this author inPubMed Google Scholar * Qi Cao View author publications You can also search for this
author inPubMed Google Scholar * Ruirui Kong View author publications You can also search for this author inPubMed Google Scholar * Xiao Xiang View author publications You can also search
for this author inPubMed Google Scholar * Hengkang Liu View author publications You can also search for this author inPubMed Google Scholar * Mei Feng View author publications You can also
search for this author inPubMed Google Scholar * Fangyanni Wang View author publications You can also search for this author inPubMed Google Scholar * Jinghui Cheng View author publications
You can also search for this author inPubMed Google Scholar * Zhao Li View author publications You can also search for this author inPubMed Google Scholar * Qimin Zhan View author
publications You can also search for this author inPubMed Google Scholar * Mi Deng View author publications You can also search for this author inPubMed Google Scholar * Jiye Zhu View author
publications You can also search for this author inPubMed Google Scholar * Zemin Zhang View author publications You can also search for this author inPubMed Google Scholar * Ning Zhang View
author publications You can also search for this author inPubMed Google Scholar 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 _R_o/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 _R_o/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
CCL4+ CD66b+ 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 _Kras__G12D_) via HDTV in _Alb-Cre_ × _Trp53__fl/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 SOURCE DATA FIG. 3
SOURCE DATA FIG. 4 SOURCE DATA EXTENDED DATA FIG. 7 SOURCE DATA EXTENDED DATA FIG. 8 SOURCE DATA EXTENDED DATA FIG. 9 SOURCE DATA EXTENDED DATA FIG. 10 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 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: 07 September 2021 * Accepted: 30 September 2022 * Published: 09 November 2022 * Issue Date: 01 December 2022 * DOI: https://doi.org/10.1038/s41586-022-05400-x
SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to
clipboard Provided by the Springer Nature SharedIt content-sharing initiative