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:

Single-cell dissection of intratumoral heterogeneity and lineage diversity in metastatic gastric adenocarcinoma

Abstract

Intratumoral heterogeneity (ITH) is a fundamental property of cancer; however, the origins of ITH remain poorly understood. We performed single-cell transcriptome profiling of peritoneal carcinomatosis (PC) from 15 patients with gastric adenocarcinoma (GAC), constructed a map of 45,048 PC cells, profiled the transcriptome states of tumor cell populations, incisively explored ITH of malignant PC cells and identified significant correlates with patient survival. The links between tumor cell lineage/state compositions and ITH were illustrated at transcriptomic, genotypic, molecular and phenotypic levels. We uncovered the diversity in tumor cell lineage/state compositions in PC specimens and defined it as a key contributor to ITH. Single-cell analysis of ITH classified PC specimens into two subtypes that were prognostically independent of clinical variables, and a 12-gene prognostic signature was derived and validated in multiple large-scale GAC cohorts. The prognostic signature appears fundamental to GAC carcinogenesis and progression and could be practical for patient 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: A single-cell transcriptome map of PC and the inferred tumor cell lineages.
Fig. 2: The diversity in tumor cell lineage compositions links to ITH at transcriptomic, genotypic and molecular levels.
Fig. 3: 17q copy number gain is prevalent in cells of stomach origin and significantly associated with inferior survival.
Fig. 4: Molecular pathway-based dissection of the transcriptomic ITH and correlation with tumor cell lineage and patient survival.
Fig. 5: Identification and validation of the 12-gene prognostic signature.

Similar content being viewed by others

Data availability

All single-cell RNA-sequencing data generated by this study have been be deposited in the European Genome-Phenome Archive (EGA, https://ega-archive.org/). The data can be accessed under the accession number EGAS00001004443. Bulk mRNA-seq expression data (normalized) generated by The Cancer Genome Atlas (TCGA) on primary stomach adenocarcinoma were downloaded from NCI Cancer Genomic Data Commons (NCI-GDC: https://gdc.cancer.gov). Three large-scale primary GAC datasets (GSE62254 (ref. 28) and GSE15459 (refs. 27,71), GSE84437 (ref. 72)) were downloaded from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/).

References

  1. Bray, F. et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68, 394–424 (2018).

    Article  PubMed  Google Scholar 

  2. Ikoma, N. et al. Preoperative chemoradiation therapy induces primary-tumor complete response more frequently than chemotherapy alone in gastric cancer: analyses of the National Cancer Database 2006–2014 using propensity score matching. Gastric Cancer 21, 1004–1013 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Mizrak Kaya, D. et al. Risk of peritoneal metastases in patients who had negative peritoneal staging and received therapy for localized gastric adenocarcinoma. J. Surg. Oncol. 117, 678–684 (2018).

    Article  PubMed  Google Scholar 

  4. Shiozaki, H. et al. Prognosis of gastric adenocarcinoma patients with various burdens of peritoneal metastases. J. Surg. Oncol. 113, 29–35 (2016).

    Article  PubMed  Google Scholar 

  5. Chen, C. et al. Efficacy and safety of immune checkpoint inhibitors in advanced gastric or gastroesophageal junction cancer: a systematic review and meta-analysis. Oncoimmunology 8, e1581547 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Taieb, J. et al. Evolution of checkpoint inhibitors for the treatment of metastatic gastric cancers: current status and future perspectives. Cancer Treat. Rev. 66, 104–113 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Bartley, A. N. et al. HER2 testing and clinical decision making in gastroesophageal adenocarcinoma: guideline from the College of American Pathologists, American Society for Clinical Pathology, and the American Society of Clinical Oncology. J. Clin. Oncol. 35, 446–464 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513, 202–209 (2014).

    Article  Google Scholar 

  9. Wang, R. et al. Multiplex profiling of peritoneal metastases from gastric adenocarcinoma identified novel targets and molecular subtypes that predict treatment response. Gut 69, 18–31 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Han, X. et al. Construction of a human cell landscape at single-cell level. Nature 581, 303–309 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Buttner, M., Miao, Z., Wolf, F. A., Teichmann, S. A. & Theis, F. J. A test metric for assessing single-cell RNA-seq batch correction. Nat. Methods 16, 43–49 (2019).

    Article  PubMed  Google Scholar 

  13. McInnes, L., Healy, J., Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at https://arxiv.org/abs/1802.03426 (2018).

  14. Smillie, C. S. et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell 178, 714–730.e22 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kiselev, V. Y. et al. SC3: consensus clustering of single-cell RNA-seq data. Nat. Methods 14, 483–486 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997.e24 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Stevens, C. E. & Leblond, C. P. Renewal of the mucous cells in the gastric mucosa of the rat. Anat. Rec. 115, 231–245 (1953).

    Article  CAS  PubMed  Google Scholar 

  22. Karam, S. M. A focus on parietal cells as a renewing cell population. World J. Gastroenterol. 16, 538–546 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Merzel, J. & Leblond, C. P. Origin and renewal of goblet cells in the epithelium of the mouse small intestine. Am. J. Anat. 124, 281–305 (1969).

    Article  CAS  PubMed  Google Scholar 

  24. Blanpain, C., Horsley, V. & Fuchs, E. Epithelial stem cells: turning over new leaves. Cell 128, 445–458 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Coker, E. A. et al. canSAR: update to the cancer translational research and drug discovery knowledgebase. Nucleic Acids Res. 47, D917–D922 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Kim, H. K. et al. A gene expression signature of acquired chemoresistance to cisplatin and fluorouracil combination chemotherapy in gastric cancer patients. PLoS ONE 6, e16694 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ooi, C. H. et al. Oncogenic pathway combinations predict clinical prognosis in gastric cancer. PLoS Genet. 5, e1000676 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Cristescu, R. et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 21, 449–456 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Mizrak Kaya, D. et al. Advanced gastric adenocarcinoma: optimizing therapy options. Expert Rev. Clin. Pharmacol. 10, 263–271 (2017).

    CAS  PubMed  Google Scholar 

  30. Dagogo-Jack, I. & Shaw, A. T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 15, 81–94 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Hudler, P. Challenges of deciphering gastric cancer heterogeneity. World J. Gastroenterol. 21, 10510–10527 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gullo, I., Carneiro, F., Oliveira, C. & Almeida, G. M. Heterogeneity in gastric cancer: from pure morphology to molecular classifications. Pathobiology 85, 50–63 (2018).

    Article  PubMed  Google Scholar 

  33. Oh, S. C. et al. Clinical and genomic landscape of gastric cancer with a mesenchymal phenotype. Nat. Commun. 9, 1777 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Merlo, L. M., Pepper, J. W., Reid, B. J. & Maley, C. C. Cancer as an evolutionary and ecological process. Nat. Rev. Cancer 6, 924–935 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Michor, F. & Polyak, K. The origins and implications of intratumor heterogeneity. Cancer Prev. Res. (Phila.) 3, 1361–1364 (2010).

    Article  Google Scholar 

  36. Barros, R., Freund, J. N., David, L. & Almeida, R. Gastric intestinal metaplasia revisited: function and regulation of CDX2. Trends Mol. Med. 18, 555–563 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Moghimi-Dehkordi, B., Safaee, A. & Zali, M. R. Comparison of colorectal and gastric cancer: survival and prognostic factors. Saudi J. Gastroenterol. 15, 18–23 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Qiu, M. Z. et al. Clinicopathological characteristics and prognostic analysis of Lauren classification in gastric adenocarcinoma in China. J. Transl. Med. 11, 58 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Petrelli, F. et al. Prognostic value of diffuse versus intestinal histotype in patients with gastric cancer: a systematic review and meta-analysis. J. Gastrointest. Oncol. 8, 148–163 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Petitprez, F. et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 577, 556–560 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Cabrita, R. et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 577, 561–565 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Najafi, M. et al. Macrophage polarity in cancer: a review. J. Cell. Biochem. 120, 2756–2765 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Kaneda, M. M. et al. PI3Kγ is a molecular switch that controls immune suppression. Nature 539, 437–442 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Calon, A. et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat. Genet. 47, 320–329 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Fiori, M. E. et al. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol. Cancer 18, 70 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Amin, M. B. et al. AJCC Cancer Staging Manual 8th edn (Springer, 2017).

  49. Amin, M. B. et al. The Eighth Edition AJCC Cancer staging manual: continuing to build a bridge from a population-based to a more ‘personalized’ approach to cancer staging. CA Cancer J. Clin. 67, 93–99 (2017).

    Article  PubMed  Google Scholar 

  50. Niu, B. et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 30, 1015–1016 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Savas, P. et al. Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis. Nat. Med. 24, 986–993 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Galili, T. dendextend: an R package for visualizing, adjusting and comparing trees of hierarchical clustering. Bioinformatics 31, 3718–3720 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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 

  54. Lambrechts, D. et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 24, 1277–1289 (2018).

    Article  CAS  PubMed  Google Scholar 

  55. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Nilsen, G. et al. Copynumber: efficient algorithms for single- and multi-track copy number segmentation. BMC Genomics 13, 591 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cillo, A. R. et al. Immune landscape of viral- and carcinogen-driven head and neck cancer. Immunity 52, 183–199.e9 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e27 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hicks, S. C., Townes, F. W., Teng, M. & Irizarry, R. A. Missing data and technical variability in single-cell RNA-sequencing experiments. Biostatistics 19, 562–578 (2018).

    Article  PubMed  Google Scholar 

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

    Article  Google Scholar 

  62. Cline, M. S. et al. Integration of biological networks and gene expression data using Cytoscape. Nat. Protoc. 2, 2366–2382 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Harrell, F. E. Jr., Lee, K. L. & Mark, D. B. Multivariable prognostic models: issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat. Med. 15, 361–387 (1996).

    Article  PubMed  Google Scholar 

  64. Peng, F. et al. Differential expression analysis at the individual level reveals a lncRNA prognostic signature for lung adenocarcinoma. Mol. Cancer 16, 98 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Lau, S. K. et al. Three-gene prognostic classifier for early-stage non small-cell lung cancer. J. Clin. Oncol. 25, 5562–5569 (2007).

    Article  PubMed  Google Scholar 

  66. Kang, J., D’Andrea, A. D. & Kozono, D. A DNA repair pathway–focused score for prediction of outcomes in ovarian cancer treated with platinum-based chemotherapy. J. Natl Cancer Inst. 104, 670–681 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Newman, A. M. et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12, 453–457 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Becht, E. et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 17, 218 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Liu, J. et al. An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell 173, 400–416.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lei, Z. et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology 145, 554–565 (2013).

    Article  CAS  PubMed  Google Scholar 

  72. Yoon, S. J. et al. Deconvolution of diffuse gastric cancer and the suppression of CD34 on the BALB/c nude mice model. BMC Cancer 20, 314 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Irizarry, R. A. et al. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 31, e15 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by the start-up research fund provided to L.W. by the UT MD Anderson Cancer Center (MDACC); the Andrew Sabin Family Fellowship Program to L.W. by the Andrew Sabin Family Foundation; the DOD grants no. CA150334 and no. CA160445 to J.A.A.; the DOD grants no. CA160433 and no. CA170906 to S.S.; and the generous support from the Caporella, Dallas, Sultan, Park, Smith, Frazier, Oaks, Vanstekelenberg, Planjery, McNeil, Hyland and Cantu families; as well as from the Schecter Private Foundation, the Rivercreek Foundation, the Kevin Fund, the Myer Fund, the Stupid Strong Foundation, the V. Foundation, the Dio Fund, the Milrod Fund and the MDACC multidisciplinary grant programs. This study was also supported by the NIH grant no. 1S10OD024977-01 Award to the Advanced Technology Genomics Core (ATGC) and the Core grant no. CA016672 (ATGC). We thank E. J. Thompson and D. P. Pollock from the ATGC for their excellent technical assistance. We thank all of the patients who participated in this study.

Author information

Authors and Affiliations

Authors

Contributions

L.W. and J.A.A. conceived and jointly supervised the study. S.S., K.H., M.P.P., M.Z., G.T., N.S., A.A.F.A., B.D.B. and M.B.M. contributed to sample collection and processing and collection of patient clinical information. A.J.L., J.S.E. and S.R.-C. contributed to pathology review. Y.L. reviewed the CT images. L.W. supervised the bioinformatics data analysis, data integration and interpretation. R.W. contributed to sequencing data processing, integrative analyses and generation of figures and tables for the manuscript. M.D., G.H., F.W., S. Zhang., D.H., S. Zhao., Y.W., X.S., Y.C., J.Z., M.L. and K.C. assisted with data processing and analysis. L.W., J.A.A. and R.W. wrote the manuscript. L.W., J.A.A., R.W., A.J.L., A.F., S.H., G.A.C. and G.P. revised the manuscript.

Corresponding authors

Correspondence to Jaffer A. Ajani or Linghua Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Javier Carmona was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 A single cell transcriptome map of PC.

a, t-SNE (t-distributed stochastic neighbor embedding) plots showing unbiased clustering analysis of 45,048 single cells that passed quality control in this study. Each dot represents a single cell. Cells are color coded for (left to right): the associated cell types, cell clusters, the corresponding patient origins, and survival status. b, t-SNE as in a, showing expression of canonical marker genes used for cell types assignment.

Extended Data Fig. 2 Relationships between tumor cell clusters and correlation with patient survival.

a, the UMAP (uniform manifold approximation and projection) plot of PC tumor cells, showing the global data structure. Tumor cell clusters from short-term survivors appeared closer to each other on the UMAP plot than to cell clusters from long-term survivors. b, the dendrogram showing relationships between tumor cell clusters. c, the Bhattacharyya pairwise distance between tumor cell clusters from samples of long and short-term survivors. Overall, the pairwise distance between clusters of long and short survivors was significantly larger than that within the clusters of Short or Random, indicating distinct transcriptomic profiles associated with survival. Each dot represents one sampling, in totally 100 times. Box, median ± interquartile range. Whiskers, the minimum and maximum values. P values were calculated by a two-sided Wilcoxon rank sum test with Benjamini-Hochberg correction. P < 2.2e-16 represents a P value approaching 0.

Extended Data Fig. 3 Cell lineage assignment was not confounded by differences in cell cycle states.

The histograms showing tumor cell lineage compositions before (top) and after (bottom) regressing out cell cycle-related genes, respectively.

Extended Data Fig. 4 Unsupervised clustering analysis revealed inter-patient and intra-tumoral transcriptome heterogeneity in PC tumor cells.

The UMAP plots showing unsupervised clustering analysis of tumor cells (using Seurat) from 14 samples underwent HCL mapping and cell lineage inference as in Fig. 1g. Cells are colored by their corresponding cluster IDs (left) and sample origins (right). Dashed circles highlight samples that formed two or more tumor cell clusters (related to Fig. 1g).

Extended Data Fig. 5 SC3 unsupervised clustering analysis of PC tumor cells by patient.

SC3 results of 3 representative patients are shown. Each column represents a cell. The lineage annotation is shown in the top annotation track. The fractions of intestinal cells (IP-067, IP-073) or stomach pit cells (IP-009) in each SC3 defined cell clusters are labelled at the top. Some of the representative marker genes of intestine and stomach origins are labelled on the right. Two-sided proportion tests were performed between C1 and C4 (IP-067), C1 and C3 (IP-073), and C1 and C2 (IP-009), and all are significant (P < 2.2e-16).

Extended Data Fig. 6 The Bhattacharyya distance between and within inferred cell lineages.

The Bhattacharyya pairwise distance between different tumor cell lineages was computed as previously described (see Methods). Only the major lineages that had 500 or more cells were included in the analysis. The Bhattacharyya distance between cells of the same lineage and the Bhattacharyya distance between cells randomly sampled independent of lineage annotation (Random) was also computed to provide background distributions for statistical comparison. Each dot represents one sampling, in total 100 times. Box, median ± interquartile range. Whiskers, the minimum and maximum values. P values were calculated by a two-sided Wilcoxon rank-sum test with Benjamini-Hochberg correction. P < 2.2e-16 represents a P value approaching 0.

Extended Data Fig. 7 Representative examples of somatic variants identified on 3’UTR using scRNA-seq data.

Integrative Genomics Viewer (IGV) was used for visualization of the QC-passed somatic variants. The Bam files of Monocle defined cell clusters C1, C2, C3 of sample IP-067 were loaded to IGV and snapshots of 3’UTR mutations are shown for representative events: somatic mutations shared by PC tumor cells from all three clusters (top); mutations shared by only two of the three clusters (bottom left and middle), and mutations that were unique to one of the three clusters (bottom right) are shown. For each representative mutation across Monocle cell clusters, the gene name, chromosome, start position, base change, total read coverage, and tumor variant allele fraction (TAF) are shown. Total_dp: total read depth.

Extended Data Fig. 8 Prognostic significance of 12-gene signature in TCGA primary gastric cancer cohort and correlation with molecular subtypes and clinical variables.

a, Disease-specific survival (DSS, left) and progression-free interval (PFI, right) of patients whose PCs were in the GI-mixed and gastric- dominant groups defined by expression of the 12-gene signature. The analyses were performed with the Kaplan–Meier estimates and two-sided log-rank tests. Twenty-five out of 411 patients whose DSS information were not available were excluded from survival analysis. b, the alluvial plots display relationships between the PC subtypes defined by the 12-gene signature (left strip) and the molecular subtypes defined by TCGA multi-omic analysis (left), tumor stages (middle), histology types (right), and presence of local recurrence and/or distant metastasis (c). N.S., not statistically significant. P value for alluvial plots were calculated by a two-sided Fisher’s Exact test.

Extended Data Fig. 9 Validation of the 12-gene signature in a large-scale localized GAC cohort from Cristescu R, et al.

a, The multivariate Cox proportional hazard model analysis. The 12-gene signature, clinical and histopathological variables as well as the molecular signatures defined by the original study were included. For each variable, the reference level is the first one. Block in center of error bars represent the weighted mean. Whiskers of error bars represent the 95% confidence interval. b, (left) Alluvial plot shows the relationships between the PC subtypes (left strip) and the molecular signatures (right strip). The two-sided Fisher’s Exact test was used to calculate the P values and asterisks indicate significant enrichment events. (right) The 12-gene signature scores were calculated and compared across the four molecular groups defined by the original the study. Box, median ± interquartile range. Whiskers, 1.5X interquartile range. P value was calculated by one-way Kruskal-Wallis rank-sum test.

Extended Data Fig. 10 Validation of the 12-gene signature in a large-scale localized GAC cohort from Ooi CH, et al.

a, The multivariate Cox proportional hazard model analysis. The 12-gene signature, clinical and histopathological variables as well as the molecular signatures defined by the original study were included. For each variable, the reference level is the first one. Block in center of error bars represent the weighted mean. Whiskers of error bars represent the 95% confidence interval. b, (left) Alluvial plot shows the relationships between the PC subtypes (left strip) and the molecular signatures (right strip). The two-sided Fisher’s Exact test was used to calculate the P values and asterisks indicate significant enrichment events. (right) The 12-gene signature scores were calculated and compared across the four molecular groups defined by the original the study. Box, median ± interquartile range. Whiskers, 1.5X interquartile range. P value was calculated by one-way Kruskal-Wallis rank-sum test.

Supplementary information

Supplementary Information

Supplementary Figs. 1–25 and Tables 1–8.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, R., Dang, M., Harada, K. et al. Single-cell dissection of intratumoral heterogeneity and lineage diversity in metastatic gastric adenocarcinoma. Nat Med 27, 141–151 (2021). https://doi.org/10.1038/s41591-020-1125-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-1125-8

This article is cited by

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