B cells and tertiary lymphoid structures promote immunotherapy response

Treatment with immune checkpoint blockade (ICB) has revolutionized cancer therapy. Until now, predictive biomarkers 1 – 10 and strategies to augment clinical response have largely focused on the T cell compartment. However, other immune subsets may also contribute to anti-tumour immunity 11 – 15 , although these have been less well-studied in ICB treatment 16 . A previously conducted neoadjuvant ICB trial in patients with melanoma showed via targeted expression profiling 17 that B cell signatures were enriched in the tumours of patients who respond to treatment versus non-responding patients. To build on this, here we performed bulk RNA sequencing and found that B cell markers were the most differentially expressed genes in the tumours of responders versus non-responders. Our findings were corroborated using a computational method (MCP-counter 18 ) to estimate the immune and stromal composition in this and two other ICB-treated cohorts (patients with melanoma and renal cell carcinoma). Histological evaluation highlighted the localization of B cells within tertiary lymphoid structures. We assessed the potential functional contributions of B cells via bulk and single-cell RNA sequencing, which demonstrate clonal expansion and unique functional states of B cells in responders. Mass cytometry showed that switched memory B cells were enriched in the tumours of responders. Together, these data provide insights into the potential role of B cells and tertiary lymphoid structures in the response to ICB treatment, with implications for the development of biomarkers and therapeutic targets. Multiomic profiling of several cohorts of patients treated with immune checkpoint blockade highlights the presence and potential role of B cells and tertiary lymphoid structures in promoting therapy response.

[1]  Y. Benjamini,et al.  Controlling the false discovery rate in behavior genetics research , 2001, Behavioural Brain Research.

[2]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[3]  J. Tímár,et al.  Density of DC-LAMP+ mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor , 2007, Cancer Immunology, Immunotherapy.

[4]  Trey Ideker,et al.  Cytoscape 2.8: new features for data integration and network visualization , 2010, Bioinform..

[5]  J. Tímár,et al.  Prognostic impact of B-cell density in cutaneous melanoma , 2011, Cancer Immunology, Immunotherapy.

[6]  J. J. van den Oord,et al.  Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. , 2012, Cancer research.

[7]  Chris Williams,et al.  RNA-SeQC: RNA-seq metrics for quality control and process optimization , 2012, Bioinform..

[8]  C. Slingluff,et al.  Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. , 2012, Cancer research.

[9]  V. Sondak,et al.  12-Chemokine Gene Signature Identifies Lymph Node-like Structures in Melanoma: Potential for Patient Selection for Immunotherapy? , 2012, Scientific Reports.

[10]  P. Rochaix,et al.  High endothelial venules (HEVs) in human melanoma lesions , 2012, Oncoimmunology.

[11]  Steven J. M. Jones,et al.  Comprehensive molecular characterization of clear cell renal cell carcinoma , 2013, Nature.

[12]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[13]  C. Monteagudo,et al.  The density and type of MECA‐79‐positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma , 2013, Histopathology.

[14]  The Cancer Genome Atlas Research Network COMPREHENSIVE MOLECULAR CHARACTERIZATION OF CLEAR CELL RENAL CELL CARCINOMA , 2013, Nature.

[15]  Guanming Wu,et al.  ReactomeFIViz : a Cytoscape app for pathway and network-based data analysis , 2022 .

[16]  N. Ruddle,et al.  Lymphatic vessels and tertiary lymphoid organs. , 2014, The Journal of clinical investigation.

[17]  P. Validire,et al.  Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. , 2014, American journal of respiratory and critical care medicine.

[18]  Pierre Validire,et al.  Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. , 2014, Cancer research.

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

[20]  B. Barnes,et al.  Role of Tertiary Lymphoid Structures (TLS) in Anti-Tumor Immunity: Potential Tumor-Induced Cytokines/Chemokines that Regulate TLS Formation in Epithelial-Derived Cancers , 2014, Cancers.

[21]  J. Taube,et al.  Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.

[22]  Steven J. M. Jones,et al.  Genomic Classification of Cutaneous Melanoma , 2015, Cell.

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

[24]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[25]  Mikhail Pogorelyy,et al.  tcR: an R package for T cell receptor repertoire advanced data analysis , 2015, BMC Bioinformatics.

[26]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[27]  Wolfgang Huber,et al.  RNA-Seq workflow: gene-level exploratory analysis and differential expression , 2015, F1000Research.

[28]  L. Chin,et al.  Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. , 2016, Cancer discovery.

[29]  S. Pillai,et al.  B lymphocytes and cancer: a love-hate relationship. , 2016, Trends in cancer.

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

[31]  K. Mertz,et al.  Tumor-associated B cells in cutaneous primary melanoma and improved clinical outcome. , 2016, Human pathology.

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

[33]  R. Emerson,et al.  Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities , 2016, Proceedings of the National Academy of Sciences.

[34]  S. Hanash,et al.  The Emerging Role of B Cells in Tumor Immunity. , 2016, Cancer research.

[35]  C. Perou,et al.  Genomic Analysis of Immune Cell Infiltrates Across 11 Tumor Types. , 2016, Journal of the National Cancer Institute.

[36]  H. Lee,et al.  Predictive Value of Tertiary Lymphoid Structures Assessed by High Endothelial Venule Counts in the Neoadjuvant Setting of Triple-Negative Breast Cancer , 2016, Cancer research and treatment : official journal of Korean Cancer Association.

[37]  J. Lunceford,et al.  IFN-&ggr;–related mRNA profile predicts clinical response to PD-1 blockade , 2017, The Journal of clinical investigation.

[38]  Kristina M. Ilieva,et al.  B cells and the humoral response in melanoma: The overlooked players of the tumor microenvironment , 2017, Oncoimmunology.

[39]  E. Jaffee,et al.  Tumor Mutational Burden and Response Rate to PD-1 Inhibition. , 2017, The New England journal of medicine.

[40]  Laurence Zitvogel,et al.  The immune contexture in cancer prognosis and treatment , 2017, Nature Reviews Clinical Oncology.

[41]  J. Madrigal,et al.  B cell regulation in cancer and anti-tumor immunity , 2017, Cellular &Molecular Immunology.

[42]  N. Hacohen,et al.  Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors , 2017, Science.

[43]  A. Gallimore,et al.  Tertiary Lymphoid Structures in Cancer: Drivers of Antitumor Immunity, Immunosuppression, or Bystander Sentinels in Disease? , 2017, Front. Immunol..

[44]  David M. Woods,et al.  Predictors of responses to immune checkpoint blockade in advanced melanoma , 2017, Nature Communications.

[45]  Laurence Zitvogel,et al.  Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors , 2018, Science.

[46]  Adrian V. Lee,et al.  An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics , 2018, Cell.

[47]  Merrick I Ross,et al.  Neoadjuvant Immune Checkpoint Blockade in High-Risk Resectable Melanoma , 2018, Nature Medicine.

[48]  A. Broeks,et al.  Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma , 2018, Nature Medicine.

[49]  J. Taube,et al.  PD-L1 and Emerging Biomarkers in Immune Checkpoint Blockade Therapy , 2018, Cancer journal.

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

[51]  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.

[52]  E. Le Chatelier,et al.  Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients , 2018, Science.

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

[54]  Jeffrey E. Lee,et al.  Neoadjuvant plus adjuvant dabrafenib and trametinib versus standard of care in patients with high-risk, surgically resectable melanoma: a single-centre, open-label, randomised, phase 2 trial. , 2018, The Lancet. Oncology.

[55]  Riyue Bao,et al.  The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients , 2018, Science.

[56]  Johannes Griss,et al.  B cells sustain inflammation and predict response to immune checkpoint blockade in human melanoma , 2019, Nature Communications.

[57]  W. Woodward,et al.  Poor Response to Neoadjuvant Chemotherapy Correlates with Mast Cell Infiltration in Inflammatory Breast Cancer , 2019, Cancer Immunology Research.

[58]  Lisle E. Mose,et al.  Prognostic value of B cells in cutaneous melanoma , 2019, Genome Medicine.

[59]  C. Sautès-Fridman,et al.  Tertiary lymphoid structures in the era of cancer immunotherapy , 2019, Nature Reviews Cancer.

[60]  Jun S. Liu,et al.  Landscape of B cell immunity and related immune evasion in human cancers , 2018, Nature Genetics.

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

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