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:

Opposing immune and genetic mechanisms shape oncogenic programs in synovial sarcoma

Abstract

Synovial sarcoma (SyS) is an aggressive neoplasm driven by the SS18–SSX fusion, and is characterized by low T cell infiltration. Here, we studied the cancer–immune interplay in SyS using an integrative approach that combines single-cell RNA sequencing (scRNA-seq), spatial profiling and genetic and pharmacological perturbations. scRNA-seq of 16,872 cells from 12 human SyS tumors uncovered a malignant subpopulation that marks immune-deprived niches in situ and is predictive of poor clinical outcomes in two independent cohorts. Functional analyses revealed that this malignant cell state is controlled by the SS18–SSX fusion, is repressed by cytokines secreted by macrophages and T cells, and can be synergistically targeted with a combination of HDAC and CDK4/CDK6 inhibitors. This drug combination enhanced malignant-cell immunogenicity in SyS models, leading to induced T cell reactivity and T cell–mediated killing. Our study provides a blueprint for investigating heterogeneity in fusion-driven malignancies and demonstrates an interplay between immune evasion and oncogenic processes that can be co-targeted in SyS and potentially in other malignancies.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Single-cell map of the cellular ecosystem of SyS tumors.
Fig. 2: Cellular plasticity and a core oncogenic program characterize SyS cells.
Fig. 3: The core oncogenic program is associated with poor prognosis and aggressive disease.
Fig. 4: Limited immune infiltration and features of antitumor immunity in SyS tumors.
Fig. 5: Impact of the genetic driver and immune cells on SyS malignant cells.
Fig. 6: HDAC and CDK4/6 inhibitors repress the core oncogenic program in SyS cells and sensitize them to T cell–mediated killing.

Similar content being viewed by others

Data availability

Processed scRNA-seq data is available via the Gene Expression Omnibus (GEO), accession number GSE131309 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE131309). Processed scRNA-seq data and interactive plots generated for this study are also provided through the Single Cell Portal at https://portals.broadinstitute.org/single_cell/study/synovial-sarcoma. Raw scRNA-Seq data are deposited in the controlled access repository DUOS (https://duos.broadinstitute.org/#/hom) accession: DUOS-000123 (via the data catalog https://duos.broadinstitute.org/dataset_catalog).

References

  1. Trujillo, J. A., Sweis, R. F., Bao, R. & Luke, J. J. T cell–inflamed versus non-T cell–inflamed tumors: a conceptual framework for cancer immunotherapy drug development and combination therapy selection. Cancer Immunol. Res. 6, 990 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Fridman, W. H., Pagès, F., Sautès-Fridman, C. & Galon, J. The immune contexture in human tumours: impact on clinical outcome. Nat. Rev. Cancer 12, 298–306 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Pollack, S. M. et al. T-cell infiltration and clonality correlate with programmed cell death protein 1 and programmed death-ligand 1 expression in patients with soft tissue sarcomas. Cancer 123, 3291–3304 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Nielsen, T. O., Poulin, N. M. & Ladanyi, M. Synovial sarcoma: recent discoveries as a roadmap to new avenues for therapy. Cancer Discov. 5, 124–134 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Pollack, S. M. The potential of the CMB305 vaccine regimen to target NY-ESO-1 and improve outcomes for synovial sarcoma and myxoid/round cell liposarcoma patients. Expert Rev. Vaccines 17, 107–114 (2018).

    CAS  PubMed  Google Scholar 

  6. Iura, K. et al. Cancer-testis antigen expression in synovial sarcoma: NY-ESO-1, PRAME, MAGEA4, and MAGEA1. Hum. Pathol. 61, 130–139 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Zhou, Y. et al. Evaluation of expression of cancer stem cell markers and fusion gene in synovial sarcoma: insights into histogenesis and pathogenesis. Oncol. Rep. 37, 3351–3360 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Naka, N. et al. Synovial sarcoma is a stem cell malignancy. Stem Cells Dayt. Ohio 28, 1119–1131 (2010).

    Article  CAS  Google Scholar 

  9. Kadoch, C. & Crabtree, G. R. Reversible disruption of mSWI/SNF (BAF) complexes by the SS18–SSX oncogenic fusion in synovial sarcoma. Cell 153, 71–85 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pan, D. et al. A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 359, 770–775 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Miao, D. et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 359, 801–806 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ayyoub, M. et al. CD4+ T cell responses to SSX-4 in melanoma patients. J. Immunol. 174, 5092 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Ayyoub, M. et al. Tumor-reactive, SSX-2-specific CD8+ T cells are selectively expanded during immune responses to antigen-expressing tumors in melanoma patients. Cancer Res. 63, 5601 (2003).

    CAS  PubMed  Google Scholar 

  14. Smith, H. A. & McNeel, D. G. The SSX family of cancer-testis antigens as target proteins for tumor therapy. Clin. Dev. Immunol. 2010, 18 (2010).

    Article  Google Scholar 

  15. Smith, H. A. & McNeel, D. G. Vaccines targeting the cancer-testis antigen SSX-2 elicit HLA-A2 epitope-specific cytolytic T cells. J. Immunother. 34, 569–580 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McBride, M. J. et al. The SS18–SSX fusion oncoprotein hijacks baf complex targeting and function to drive synovial sarcoma. Cancer Cell 33, 1128–1141 (2018).

  17. Banito, A. et al. The SS18–SSX oncoprotein hijacks KDM2B-PRC1.1 to drive synovial sarcoma. Cancer Cell 33, 527–541.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Su, L. et al. Deconstruction of the SS18–SSX fusion oncoprotein complex: insights into disease etiology and therapeutics. Cancer Cell 21, 333–347 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nakayama, R. et al. Gene expression profiling of synovial sarcoma: distinct signature of poorly differentiated type. Am. J. Surg. Pathol. 34, 1599–1607 (2010).

    Article  PubMed  Google Scholar 

  20. Lagarde, P. et al. Chromosome instability accounts for reverse metastatic outcomes of pediatric and adult synovial sarcomas. J. Am. Soc. Clin. Oncol. 31, 608–615 (2013).

    Article  CAS  Google Scholar 

  21. Comprehensive and integrated genomic characterization of adult soft tissue sarcomas. Cell 171, 950–965 (2017).

  22. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Haas, B. et al. STAR-Fusion: fast and accurate fusion transcript detection from RNA-seq. Preprint at bioRxiv https://doi.org/10.1101/120295 (2017).

  25. 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 

  26. 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 

  27. 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 

  28. Venteicher, A. S. et al. Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science 355, eaai8478 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Tirosh, I. et al. Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma. Nature 539, 309–313 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. Taube, J. H. et al. Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc. Natl Acad. Sci. USA 107, 15449–15454 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gröger, C. J., Grubinger, M., Waldhör, T., Vierlinger, K. & Mikulits, W. Meta-analysis of gene expression signatures defining the epithelial to mesenchymal transition during cancer progression. PLoS ONE 7, e51136 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Fan, J. et al. Characterizing transcriptional heterogeneity through pathway and gene set overdispersion analysis. Nat. Methods 13, 241–244 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Newman, M. E. J. & Girvan, M. Finding and evaluating community structure in networks. Phys. Rev. E Stat. Nonlin. Soft Matter Phys. 69, 026113 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yang, Z. & Michailidis, G. A non-negative matrix factorization method for detecting modules in heterogeneous omics multi-modal data. Bioinforma. Oxf. Engl. 32, 1–8 (2016).

    Google Scholar 

  37. Przybyl, J. et al. Metastatic potential is determined early in synovial sarcoma development and reflected by tumor molecular features. Int. J. Biochem. Cell Biol. 53, 505–513 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Stubbington, M. J. T. et al. T cell fate and clonality inference from single-cell transcriptomes. Nat. Methods 13, 329–332 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  39. 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 

  40. 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 

  41. Böttcher, J. P. et al. Functional classification of memory CD8+ T cells by CX3CR1 expression. Nat. Commun. 6, 8306 (2015).

    Article  PubMed  Google Scholar 

  42. Martinez, F. O., Gordon, S., Locati, M. & Mantovani, A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J. Immunol. 177, 7303–7311 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Merritt, C. R. et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol. 38, 586–599 (2020).

    Article  CAS  PubMed  Google Scholar 

  44. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Lin, J.-R. et al. Highly multiplexed immunofluorescence imaging of human tissues and tumors using t-CyCIF and conventional optical microscopes. eLife 7, e31657 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Bovolenta, L. A., Acencio, M. L. & Lemke, N. HTRIdb: an open-access database for experimentally verified human transcriptional regulation interactions. BMC Genomics 13, 405 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Han, H. et al. TRRUST: a reference database of human transcriptional regulatory interactions. Sci. Rep. 5, 11432 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Khan, A. et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46, D260–D266 (2018).

    Article  CAS  PubMed  Google Scholar 

  50. Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Brin, S. & Page, L. in Seventh International World-Wide Web Conference (1998).

  52. Herrmann, C., Van de Sande, B., Potier, D. & Aerts, S. i-cisTarget: an integrative genomics method for the prediction of regulatory features and cis-regulatory modules. Nucleic Acids Res. 40, e114 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Janky, R. et al. iRegulon: from a gene list to a gene regulatory network using large motif and track collections. PLoS Comput. Biol. 10, e1003731 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Balogh, K. N., Templeton, D. J. & Cross, J. V. Macrophage migration inhibitory factor protects cancer cells from immunogenic cell death and impairs anti-tumor immune responses. PLoS OBE 13, e0197702 (2018).

    Article  Google Scholar 

  55. Jones, K. B. et al. Of mice and men: opportunities to use genetically engineered mouse models of synovial sarcoma for preclinical cancer therapeutic evaluation. Cancer Control 18, 196–203 (2011).

    Article  PubMed  Google Scholar 

  56. Martín-Broto, J., Moura, D. S. & Van Tine, B. A. Facts and hopes in immunotherapy of soft-tissue sarcomas. Clin. Cancer Res. 26, 5801 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Robbins, P. F. et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J. Clin. Oncol. 29, 917–924 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Baird, K. et al. Gene expression profiling of human sarcomas: insights into sarcoma biology. Cancer Res. 65, 9226–9235 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Fisher, S. et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 12, R1 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Merritt, C. R. et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol. 38, 586–599 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinf. 12, 323 (2011).

    Article  CAS  Google Scholar 

  63. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinforma. Oxf. Engl. 29, 15–21 (2013).

    Article  CAS  Google Scholar 

  64. Filbin, M. G. et al. Developmental and oncogenic programs in H3K27M gliomas dissected by single-cell. Science 360, 331–335 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Cibulskis, K. et al. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Bioinforma. Oxf. Engl. 27, 2601–2602 (2011).

    Article  CAS  Google Scholar 

  66. 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 

  67. Saunders, C. T. et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinforma. Oxf. Engl. 28, 1811–1817 (2012).

    Article  CAS  Google Scholar 

  68. Costello, M. et al. Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation. Nucleic Acids Res. 41, e67 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Van Allen, E. M. et al. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nat. Med. 20, 682–688 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Cohen, O. et al. Abstract S1-01: whole exome and transcriptome sequencing of resistant ER+ metastatic breast cancer. Cancer Res. 77, S1–S01 (2017).

    Article  Google Scholar 

  71. Ramos, A. H. et al. Oncotator: cancer variant annotation tool. Hum. Mutat. 36, E2423–E2429 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Olshen, A. B., Venkatraman, E. S., Lucito, R. & Wigler, M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostat. Oxf. Engl. 5, 557–572 (2004).

    Google Scholar 

  73. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Waltman, L. & Jan van Eck, N. A smart local moving algorithm for large-scale modularity-based community detection. Eur. Phys. J. B 86, 471 (2013).

  75. McDavid, A. et al. Data exploration, quality control and testing in single-cell qPCR-based gene expression experiments. Bioinforma. Oxf. Engl. 29, 461–467 (2013).

    Article  CAS  Google Scholar 

  76. Haghverdi, L., Buettner, F. & Theis, F. J. Diffusion maps for high-dimensional single-cell analysis of differentiation data. Bioinforma. Oxf. Engl. 31, 2989–2998 (2015).

    Article  CAS  Google Scholar 

  77. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Methodol. 57, 289–300 (1995).

    Google Scholar 

  78. Ramilowski, J. A. et al. A draft network of ligand–receptor-mediated multicellular signalling in human. Nat. Commun. 6, 7866 (2015).

    Article  CAS  PubMed  Google Scholar 

  79. Robbins, P. F. et al. Single and dual amino acid substitutions in TCR CDRs can enhance antigen-specific T cell functions. J. Immunol. 180, 6116–6131 (2008).

    Article  CAS  PubMed  Google Scholar 

  80. Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. 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 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank L. Gaffney and A. Hupalowska for help with artwork and L. Gaffney for help in figure preparation. We thank K. Itoh, N. Naka and S. Takenaka (Osaka University, Japan) for providing the Aska cell lines, and Akira Kawai (National Cancer Center Hospital, Japan) for providing the SYO1 cell line. We thank M. Brown for help with CNA visualization. L.J.-A. is a Chan Zuckerberg Biohub investigator and holds a Career Award at the Scientific Interface from BWF. L.J.-A. was a fellow of the Eric and Wendy Schmidt postdoctoral program and a CRI Irvington Fellow supported by the CRI. A.R. is an HHMI Investigator. Work was supported by the Klarman Cell Observatory, STARR cancer consortium, NCI grants 1U24CA180922, R33-CA202820, the Koch Institute NCI Support (core) grant P30-CA14051, Ludwig Centers at Harvard and MIT, AMRF and the Broad Institute (A.R.). Work was also supported by grants from the Howard Goodman Fellowship at MGH (M.L.S.), the Merkin Institute Fellowship at the Broad Institute of MIT and Harvard (M.L.S.), R37CA245523 (M.L.S.), the Swiss National Science Foundation Sinergia grant CRSII5_177266 (M.L.S. and I.S.). Imaging CyCIF work was supported by a grant (CA225088) from the Center for Cancer Systems Pharmacology at Harvard Medical School (P.K.S.), K08CA222663 (B.I.), Burroughs Wellcome Fund Career Award for Medical Scientists (B.I.), Louis V. Gerstner, Jr. Scholars Program (B.I.) and the Velocity Fellow Program (B.I.). N.R. is supported by the Swiss National Science Foundation Professorship grant (PP00P3-157468/1 and PP00P3_183724), the Swiss Cancer League grant KFS-3973-08-2016, the Fond’Action Contre le Cancer grant and the FORCE grant. N.D.M. was supported by a postdoctoral fellowship from the American Cancer Society (PF-17-042-01-LIB) and the NIH education loan repayment program funded by the NCI (L30 CA231679-01). M.N.R. is supported by the Thomas and Diana Ryan MGH Research Scholar Award. Processed scRNA-seq data are available at https://portals.broadinstitute.org/single_cell/study/synovial-sarcoma and GEO GSE131309. Raw scRNA-seq data is deposited in the controlled access repository DUOS (https://duos.broadinstitute.org/#/hom) accession: DUOS-000123 (via the data catalog https://duos.broadinstitute.org/dataset_catalog).

Author information

Authors and Affiliations

Authors

Contributions

L.J.-A., C.N., N.R., M.L.S. and A.R. conceived the project, designed the study and interpreted results. A.R., O.R.-R., N.R. and M.L.S. obtained funding for the study. L.J.-A. performed computational analyses. C.N., M.E.S., H.R.W., A.R.R., G.B. and A.V. collected synovial sarcoma samples and generated single-cell RNA-seq data. A.S., M.S., B.I., D.R.Z., N.O. and J.M.B. performed tissue spatial analyses. B.H. provided support for single-cell genetic analyses. C.C.L. and R.M. provided flow cytometry expertise. M.J.M. and C.K. provided data and support for chromatin analysis and performed the fusion KD experiments. M.M., G.P.N., I.C., G.M.C., E.C., S.C., P.K.S., A.B.H., J.T.M. and T.N.-N. obtained consent from patients for the study and provided clinical data. J.E.B.-B., I.L., L.C., L.C.B, J.M.G., L.N., S.M., J.C.M., C.G., J.B., S.G., M.S.C., D.L., I.S., M.N.R. and O.R.R. provided experimental support. O.C. and N.W. provided support with mutation calling. N.D.M. performed all the co-culture experiments, with K.W.W.’s supervision. M.L.S., N.R. and A.R. jointly supervised this work. L.J.-A, N.R., M.L.S. and A.R. wrote the manuscript with feedback from all authors.

Corresponding authors

Correspondence to Mario L. Suvà, Nicolò Riggi or Aviv Regev.

Ethics declarations

Competing interests

A.R. is a founder of and equity holder in Celsius Therapeutics, an equity holder in Immunitas Therapeutics, and was a scientific advisory board member for Thermo Fisher Scientific, Syros Pharmaceuticals and Neogene Therapeutics until 1 August 2020. From 1 August 2020, A.R. is an employee of Genentech. M.L.S. is an equity holder, scientific cofounder and advisory board member of Immunitas Therapeutics. K.W.W. serves on the scientific advisory board of TCR2 Therapeutics, T-Scan Therapeutics, SQZ Biotech, Nextechinvest and receives sponsored research funding from Novartis. He is a cofounder of Immunitas Therapeutics. L.J.A, N.R., M.L.S. and A.R. are coinventors on US patent application filed by the Broad Institute relating to synovial sarcoma. O.R.-R. is an employee of Genentech and a coinventor on patent applications filed by the Broad Institute for inventions relating to single cell genomics, such as in PCT/US2018/060860 and US provisional application no. 62/745,259. D.R.Z., N.O. and J.M.B. are employees of Nanostring which developed GeoMx. C.K. is the scientific founder, fiduciary board of directors member, scientific advisory board member, shareholder and consultant for foghorn therapeutics. E.C. reports support paid to his institution for the conduct of clinical trials from Amgen, Astra Zeneca, Novartis, Bayer, Merck, Exelixis, GSK, Adaptimmune and Iterion. G.M.C. reports advisory board fees and support paid to his institution for the conduct of clinical trials from Agios, Epizyme, PharmaMar, Eisai; support paid to his institution for the conduct of clinical trials from Macrogenics, Boston Biomedical, Plexxicon, Merck KGaA / EMD Serono Research and Development Institute, CBA, SpringWorks Therapeutics, Bavarian-Nordic; compound for preclinical research and support paid to his institution for the conduct of clinical trials from Bayer. P.K.S. is a member of the SAB or Board of Directors of Applied Biomath, Glencoe Software and RareCyte and has equity in these companies. In the last five years the Sorger lab has received research funding from Novartis and Merck. The authors declare that these activities are not related to the research reported in this publication and have not influenced the conclusions in this manuscript. B.I. is a consultant for Merck and Volastra Therapeutics. N.W. is an equity holder and scientific advisory board member of Relay Therapeutics, a paid advisor to Eli Lilly and Co, and receives grant support from Puma Biotechnology. N.D.M. serves as a scientific advisor to Immunitas Therapeutics. C.N., M.E.S., H.R.W, M.J.M, B.H., B.I, A.V, G.B., L.C., A.R.Ri, L.C.B., J.M.G., C.C.L, R.M., L.N., S.M., J.C.M., C.G., O.C., J.E.B.-B., A.S., M.S., M.S.C, D.L., S.G., G. P.N., I.C., T.N.-N, M.M., E.C., I.L., S.C., A.B.H., J.T.M., I.S. and M.N.R declare no competing interests.

Additional information

Peer review information Saheli Sadanand 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 Consistent classification of cells based on expression and genetic features.

a, Converging assignments of cell identity. tSNE of single-cell profiles (dots), colored by (1) tumor sample, (2) inferred cell type, (3) SS18-SSX1/2 and MEOX2-AGMO fusion detection, (4) SSX1/2 gene detection (mRNA level > 0), (5) MEOX2 and AGMO gene detection (mRNA level > 0), (6-12) overall expression of well-established cell type markers (Supplementary Table 2). b, Droplet based scRNA-Seq of SyS. tSNE of single cells (dots), profiled with droplet-based scRNA-seq23, colored according to tumor sample (left) and inferred cell type (right). c, Differential similarity to SyS compared to other sarcomas (Methods) distinguishes malignant (n = 4,371) from non-malignant (n = 2,375) cells. Differential similarity (y axis) to SyS shown for cells in each cell subset (x axis). d, The SyS program distinguishes between SyS and non-SyS cancer types. Distribution of the SyS program Overall Expression (y axis) across BAF driven tumors (left, x axis) and in TCGA (right, x axis; n = 9,128; 253; and 10, for other, sarcoma, and SyS tumors). In (c-d) middle line: median; box edges: 25th and 75th percentiles, whiskers: most extreme points that do not exceed ±IQR*1.5; further outliers are marked individually; P-values: one-sided Wilcoxon-ranksum test; AUC: Area Under the receiver operating characteristic Curve.

Extended Data Fig. 2 Characterizing mesenchymal, epithelial and poorly differentiates malignant cells.

a, Epithelial and mesenchymal program genes. The expression of the top epithelial and mesenchymal program genes (rows) across the malignant cells (columns), with cells sorted according to the difference in epithelial minus mesenchymal OE scores (bottom plot). Topmost Color bar: epithelial vs. non-epithelial cell status, and sample. Canonical markers and immune-related genes are in red and blue, respectively. b, Cell cycle signature. Overall Expression of the G2/M (y axis) and G1/S (x axis) phase signatures in each malignant cell, colored by their cycling status. c, Cycling cells are less differentiated. The distribution of differentiation scores of cycling (red) and non-cycling (grey) malignant cells, across all tumors (top) and within each tumor (bottom; only tumors with at least 10 cycling cells are shown); p-value: mixed-effects test.

Extended Data Fig. 3 The core oncogenic program is detected using different approaches and datasets.

a, Agreement between the core oncogenic program detected by a PCA and an iNMF approach36. Overall Expression (OE) of the core oncogenic program across malignant SyS cells, as identified in the PCA-based approach33 (x axis) and in the iNMF approach36 (y axis) (Methods). (b-c) Program Overall Expression captures inter-tumor variation and the MYC-high cluster in 64 SyS tumors from an independent RNA-Seq cohort16. The tumors were previously classified into two transcriptionally distinct clusters16, denoted here as MYC-high and MYC-low. b, For each tumor (dots), shown is the Overall Expression (OE) of the core oncogenic program (y axis) vs. the projection on the second Principle Component (PC2) of the data. c, Normalized expression (centered log-transformed RPKM) of the core oncogenic program genes (columns) most correlated with PC2 across the tumors (columns). Tumors are sorted by their PC2 projection (bottom bar). d, The fraction of TLE1+LGALS1+ cells out of TLE1+ ones based on ISH of tumors SyS5 and SyS13; Data are presented as mean values ± SD, such that each dot corresponds to one high power field (HPF), with a total of 10 HPF per sample; TLE1 is a SyS cell marker and LGALS1 is a positive marker of the core oncogenic program.

Extended Data Fig. 4 Antitumor immunity and immune evasion in SyS.

a, CD8 T cell clones, stratified based on clone size (x axis) and tumor (color). b, Overall expression of the T cell expansion program in CD8 T cells with a reconstructed TCR (TCR+), when stratified based on clonality (Clone+ and Clone, denoting clone size greater or equal to 1, respectively). c, The cancer testis antigens CTAG1A, CTAG1B (encoding for NY-ESO-1), and PRAME are exclusively expressed by SyS malignant (n = 4,371) cells compared to non-malignant ones (n = 2,375). Log-transformed TPM (y axis) in different cell subsets (x axis); p-values: one-sided Mann-Whitney test. d, tSNE of macrophage profiles, colored by M1/M2 polarization scores, according to signatures defined here (Supplementary Table 4). e, M1/M2 polarization scores (y axis) according to previously defined signatures42 in macrophages in our datasets partitioned to M1-like and M2-like subgroups (p-value: two-sided t-test). f, Spearman correlation coefficient (color bar) between each pair of genes from M1 and M2 signatures defined here (top, Supplementary Table 4) or previously42 (bottom) across macrophages in SyS (left) and melanoma30 (right). g, Overall Expression of the immune cell signatures (y axis, Methods) in SyS tumors (orange) and other cancer types (green); p-value: one-sided t-test. (c) and (g) middle line: median; box edges: 25th and 75th percentiles, whiskers: most extreme points that do not exceed ±IQR*1.5; further outliers are marked individually. h, Prognostic value of T cell levels in different tumor types. Kaplan-Meier (KM) curves of survival in melanoma (left; TCGA), sarcoma (middle)21, and SyS20 (right), stratified by high (top 25%, red), low (bottom 25%, blue), or intermediate (remainder, green) levels of inferred T cell infiltration levels; P: COX regression. (i) Protein expression (CyCIF) of core oncogenic program markers in immune-enriched and deprived niches.

Extended Data Fig. 5 Characterizing the transcriptional impact of SS18-SSX inhibition and tumor microenvironment cytokines on SyS cells.

a, The fusion KD induces cell autonomous immune programs. Distribution of Overall Expression scores (y axis) in the pathways most differentially expressed between SyS cells with SS18-SSX (shSSX, grey) vs. control (shCt, blue) shRNA, shown separately for non-cycling and cycling cells (x axis). b, Co-embedding (using PCA and canonical correlation analyses81, Methods) of Aska (top) and SYO1 (bottom) cell profiles (dots), colored by (from left to right): perturbation; or the Overall Expression (colorbar) of the cell cycle, core oncogenic, or mesenchymal differentiation31,32 programs. c, Biological processes regulated in the SS18-SSX program. Gene sets (columns) most enriched (-log10(P-value), hypergeometric test, x axis) in the induced (left) and repressed (right) SS18-SSX program genes, which are either direct (black bars) or indirect (grey bars) targets of SS18-SSX based on ChIP-Seq data16,17 and genetic perturbation. Vertical line denotes statistical significance following multiple hypotheses correction. d, The SS18-SSX program distinguishes SyS from other cancer types and other sarcomas. Overall Expression of the SS18-SSX program (y axis) in either TCGA samples (n = 9,391, top), stratified by cancer types (x axis), or in another independent cohort of sarcoma tumors (n = 164, bottom)58. Middle line: median; box edges: 25th and 75th percentiles, whiskers: most extreme points that do not exceed ±IQR*1.5; further outliers are marked individually. **P < 0.01, ***P < 1*10−3, ****P < 1*10−4, one-sided t-test. e, Repression of the core oncogenic and SS18-SSX programs by short term TNF treatment is not sustained long term. Distribution of Overall Expression scores (y axis) of the core oncogenic program and the direct and indirect SS18-SSX programs (x axis) in control cells (blue) and cells treated with TNF for 4-6 hours (left) or more than 24 hours (right).

Extended Data Fig. 6 HDAC and CDK4/6 inhibitors synergistically repress the core oncogenic program and induce cell autonomous immune responses.

a, The fraction of viable, necrotic, and apoptotic cells, showing four different SyS cell lines. (b-d) Distribution of the expression (y axis) of core oncogenic genes (b), as well as the Overall Expression of TNF (c) and IFN (d) signaling pathways in SyS cells and MSCs (x axis) under different treatments (color legend; n = no. of SYO1, HSSYII, and MSC cells). Middle line: median; box edges: 25th and 75th percentiles, whiskers: most extreme points that do not exceed ±IQR*1.5; further outliers are marked individually. **P < 0.01, ***P < 1*10−3, ****P < 1*10−4, one-sided t-test. e, Workflow of the co-culture CME-1-T-cell experiment. f, HLA-A2 and HLA-E protein levels on the cell surface of CME-1 cells under different treatments. g, Standard, FSC vs. SSC gating was performed followed by strict FSC-width vs. FSC-area criteria to discriminate doublets and gate only single cells. Top: Singlets were gated upon the CD3- population to clearly identify the tumor cell population. The percentage of Zombie-UV+ cells were determined on the CD3- population. Bottom: Singlets were gated upon the Zombie-UV- (live) CD3+ population to identify the viable T cell population.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jerby-Arnon, L., Neftel, C., Shore, M.E. et al. Opposing immune and genetic mechanisms shape oncogenic programs in synovial sarcoma. Nat Med 27, 289–300 (2021). https://doi.org/10.1038/s41591-020-01212-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-01212-6

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