Mature tertiary lymphoid structures are key niches of tumour-specific immune responses in pancreatic ductal adenocarcinomas

Objective To better understand the immune microenvironment of pancreatic ductal adenocarcinomas (PDACs), here we explored the relevance of T and B cell compartmentalisation into tertiary lymphoid structures (TLSs) for the generation of local antitumour immunity. Design We characterised the functional states and spatial organisation of PDAC-infiltrating T and B cells using single-cell RNA sequencing (scRNA-seq), flow cytometry, multicolour immunofluorescence, gene expression profiling of microdissected TLSs, as well as in vitro assays. In addition, we performed a pan-cancer analysis of tumour-infiltrating T cells using scRNA-seq and sc T cell receptor sequencing datasets from eight cancer types. To evaluate the clinical relevance of our findings, we used PDAC bulk RNA-seq data from The Cancer Genome Atlas and the PRINCE chemoimmunotherapy trial. Results We found that a subset of PDACs harbours fully developed TLSs where B cells proliferate and differentiate into plasma cells. These mature TLSs also support T cell activity and are enriched with tumour-reactive T cells. Importantly, we showed that chronically activated, tumour-reactive T cells exposed to fibroblast-derived TGF-β may act as TLS organisers by producing the B cell chemoattractant CXCL13. Identification of highly similar subsets of clonally expanded CXCL13+ tumour-infiltrating T cells across multiple cancer types further indicated a conserved link between tumour-antigen recognition and the allocation of B cells within sheltered hubs in the tumour microenvironment. Finally, we showed that the expression of a gene signature reflecting mature TLSs was enriched in pretreatment biopsies from PDAC patients with longer survival after receiving different chemoimmunotherapy regimens. Conclusion We provided a framework for understanding the biological role of PDAC-associated TLSs and revealed their potential to guide the selection of patients for future immunotherapy trials.

[1]  Seung Il Kim,et al.  CD39+ tissue-resident memory CD8+ T cells with a clonal overlap across compartments mediate antitumor immunity in breast cancer , 2022, Science Immunology.

[2]  Christopher R. Cabanski,et al.  Sotigalimab and/or nivolumab with chemotherapy in first-line metastatic pancreatic cancer: clinical and immunologic analyses from the randomized phase 2 PRINCE trial , 2022, Nature Medicine.

[3]  J. Gartner,et al.  A phenotypic signature that identifies neoantigen-reactive T cells in fresh human lung cancers. , 2022, Cancer cell.

[4]  Brady Bernard,et al.  Transcriptomic profiles of neoantigen-reactive T cells in human gastrointestinal cancers. , 2022, Cancer cell.

[5]  J. Gartner,et al.  Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers , 2022, Science.

[6]  B. Nabet,et al.  Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. , 2022, Cancer cell.

[7]  P. Laurent-Puig,et al.  Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. , 2022, Immunity.

[8]  T. Schumacher,et al.  Tertiary lymphoid structures in cancer , 2022, Science.

[9]  James M. McFarland,et al.  Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer , 2021, Cell.

[10]  I. Soubeyran,et al.  Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression , 2021, Nature Cancer.

[11]  G. Kinker,et al.  B Cell Orchestration of Anti-tumor Immune Responses: A Matter of Cell Localization and Communication , 2021, Frontiers in Cell and Developmental Biology.

[12]  V. Engelhard,et al.  Heterogeneity in tertiary lymphoid structure B-cells correlates with patient survival in metastatic melanoma , 2021, Journal for ImmunoTherapy of Cancer.

[13]  X. Liu,et al.  TRUST4: immune repertoire reconstruction from bulk and single-cell RNA-seq data , 2021, Nature Methods.

[14]  E. Jaffee,et al.  Leukocyte Heterogeneity in Pancreatic Ductal Adenocarcinoma: Phenotypic and Spatial Features Associated with Clinical Outcome. , 2021, Cancer discovery.

[15]  T. J,et al.  Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies , 2021, Oncoimmunology.

[16]  C. Dubay,et al.  Germinal center reactions in tertiary lymphoid structures associate with neoantigen burden, humoral immunity and long-term survivorship in pancreatic cancer , 2021, Oncoimmunology.

[17]  P. Rochaix,et al.  PD-1 blockade restores helper activity of tumor-infiltrating, exhausted PD-1hiCD39+ CD4 T cells , 2020, JCI insight.

[18]  Jessica S. Yu,et al.  Multimodal mapping of the tumor and peripheral blood immune landscape in human pancreatic cancer , 2020, Nature Cancer.

[19]  P. Sharma,et al.  Neoadjuvant PD-L1 plus CTLA-4 blockade in patients with cisplatin-ineligible operable high-risk urothelial carcinoma , 2020, Nature Medicine.

[20]  Joon-Oh Park,et al.  Single-cell transcriptome analysis of tumor and stromal compartments of pancreatic ductal adenocarcinoma primary tumors and metastatic lesions , 2020, Genome medicine.

[21]  M. Heller,et al.  B cell zone reticular cell microenvironments shape CXCL13 gradient formation , 2020, Nature Communications.

[22]  M. Veldhoen,et al.  Type 1 Treg cells promote the generation of CD8+ tissue-resident memory T cells , 2020, Nature Immunology.

[23]  V. Engelhard,et al.  Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts , 2020, Cell reports.

[24]  Thomas D. Wu,et al.  Peripheral T cell expansion predicts tumour infiltration and clinical response , 2020, Nature.

[25]  Mirjana Efremova,et al.  CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes , 2020, Nature Protocols.

[26]  Jeffrey E. Lee,et al.  B cells and tertiary lymphoid structures promote immunotherapy response , 2020, Nature.

[27]  J. Wargo,et al.  B cells are associated with survival and immunotherapy response in sarcoma , 2020, Nature.

[28]  D. Schadendorf,et al.  Tertiary lymphoid structures improve immunotherapy and survival in melanoma , 2020, Nature.

[29]  Yun-Gui Yang,et al.  Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma , 2019, Cell Research.

[30]  Howard Y. Chang,et al.  Clonal replacement of tumor-specific T cells following PD-1 blockade , 2019, Nature Medicine.

[31]  E. Karamitopoulou Tumour microenvironment of pancreatic cancer: immune landscape is dictated by molecular and histopathological features , 2019, British Journal of Cancer.

[32]  T. Bosse,et al.  A Transcriptionally Distinct CXCL13+CD103+CD8+ T-cell Population Is Associated with B-cell Recruitment and Neoantigen Load in Human Cancer , 2019, Cancer Immunology Research.

[33]  Paul J. Hoover,et al.  Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma , 2018, Cell.

[34]  Xueda Hu,et al.  Lineage tracking reveals dynamic relationships of T cells in colorectal cancer , 2018, Nature.

[35]  M. Linterman,et al.  Regulation of the Germinal Center Response , 2018, Front. Immunol..

[36]  S. Matsuda,et al.  Human Sox4 facilitates the development of CXCL13-producing helper T cells in inflammatory environments , 2018, Nature Communications.

[37]  B. Nadel,et al.  Human germinal center transcriptional programs are de-synchronized in B cell lymphoma , 2018, Nature Immunology.

[38]  Helge G. Roider,et al.  Publisher Correction: Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing , 2018, Nature Medicine.

[39]  B. Fox,et al.  Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors , 2018, Nature Communications.

[40]  C. Klein,et al.  A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small cell lung cancer treated with PD-1 blockade , 2018, Nature Medicine.

[41]  R. Stupp,et al.  Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer , 2017, Oncoimmunology.

[42]  Hannah A. Pliner,et al.  Reversed graph embedding resolves complex single-cell trajectories , 2017, Nature Methods.

[43]  Steven J. M. Jones,et al.  Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. , 2017, Cancer cell.

[44]  Boxi Kang,et al.  Landscape of Infiltrating T Cells in Liver Cancer Revealed by Single-Cell Sequencing , 2017, Cell.

[45]  P. Laurent-Puig,et al.  Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression , 2016, Genome Biology.

[46]  C. Sautès-Fridman,et al.  Tertiary lymphoid structures, drivers of the anti‐tumor responses in human cancers , 2016, Immunological reviews.

[47]  Fabio Grizzi,et al.  Spatial distribution of B cells predicts prognosis in human pancreatic adenocarcinoma , 2016, Oncoimmunology.

[48]  J. Ito,et al.  TGF‐β induces the differentiation of human CXCL13‐producing CD4+ T cells , 2015, European journal of immunology.

[49]  Y. Kanai,et al.  Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer , 2015, British Journal of Cancer.

[50]  C. Sautès-Fridman,et al.  Tertiary lymphoid structures in cancer and beyond. , 2014, Trends in immunology.

[51]  M. Nussenzweig,et al.  Identification of human germinal center light and dark zone cells and their relationship to human B-cell lymphomas. , 2012, Blood.

[52]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[53]  Tibor Schuster,et al.  Preoperative/Neoadjuvant Therapy in Pancreatic Cancer: A Systematic Review and Meta-analysis of Response and Resection Percentages , 2010, PLoS medicine.

[54]  A. Jemal,et al.  Cancer statistics, 2018 , 2018, CA: a cancer journal for clinicians.

[55]  Holger Moch,et al.  Germinal Centers Determine the Prognostic Relevance of Tertiary Lymphoid Structures and Are Impaired by Corticosteroids in Lung Squamous Cell Carcinoma. , 2018, Cancer research.