5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Molecular Signature of Tumor-Associated High Endothelial Venules That Can Predict Breast Cancer Survival

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The authors identify a molecular signature of breast tumor–associated high endothelial venules, which correlate with presence of tertiary lymphoid structures and survival, suggesting the signature may provide information for treatment selection and determining prognosis.

          Abstract

          High endothelial venules (HEV) are specialized post-capillary venules that recruit naïve lymphocytes to lymph nodes. HEVs are essential for the development of adaptive immunity. HEVs can also develop in tumors where they are thought to be important for recruiting naïve T cells and B cells into the tumors and locally enhancing antitumor immunity by supporting the formation of tertiary lymphoid structures. Herein, we used comparative transcriptome analysis of human breast cancer to investigate genes differentially expressed between tumor-associated HEVs and the rest of the tumor vasculature. Tumor vessels highly expressing HEV-upregulated genes, such as the homeobox gene MEOX2 and the tetraspanin gene TSPAN7, were associated with extensive infiltration of T and B cells and the occurrence of tertiary lymphoid structures, which is known to predict therapeutic responses to immune-checkpoint inhibitors. Moreover, high transcript counts of these genes in clinical tumor specimens were associated with a significant survival benefit in advanced breast cancer. The molecular signature of HEVs identified herein may be useful for guiding immunotherapies and provides a new direction for investigating tumor-associated HEVs and their clinical significance.

          See related Spotlight by Gallimore, [Related article:]p. 371 .

          Related collections

          Most cited references68

          • Record: found
          • Abstract: found
          • Article: not found

          B cells and tertiary lymphoid structures promote immunotherapy response

          Treatment with immune checkpoint blockade (ICB) has revolutionized cancer therapy. Until now, predictive biomarkers1-10 and strategies to augment clinical response have largely focused on the T cell compartment. However, other immune subsets may also contribute to anti-tumour immunity11-15, although these have been less well-studied in ICB treatment16. A previously conducted neoadjuvant ICB trial in patients with melanoma showed via targeted expression profiling17 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-counter18) 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.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A pathology atlas of the human cancer transcriptome

            Cancer is one of the leading causes of death, and there is great interest in understanding the underlying molecular mechanisms involved in the pathogenesis and progression of individual tumors. We used systems-level approaches to analyze the genome-wide transcriptome of the protein-coding genes of 17 major cancer types with respect to clinical outcome. A general pattern emerged: Shorter patient survival was associated with up-regulation of genes involved in cell growth and with down-regulation of genes involved in cellular differentiation. Using genome-scale metabolic models, we show that cancer patients have widespread metabolic heterogeneity, highlighting the need for precise and personalized medicine for cancer treatment. All data are presented in an interactive open-access database (www.proteinatlas.org/pathology) to allow genome-wide exploration of the impact of individual proteins on clinical outcomes.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Tertiary lymphoid structures improve immunotherapy and survival in melanoma

              Checkpoint blockade therapies that reactivate tumour-associated T cells can induce durable tumour control and result in the long-term survival of patients with advanced cancers1. Current predictive biomarkers for therapy response include high levels of intratumour immunological activity, a high tumour mutational burden and specific characteristics of the gut microbiota2,3. Although the role of T cells in antitumour responses has thoroughly been studied, other immune cells remain insufficiently explored. Here we use clinical samples of metastatic melanomas to investigate the role of B cells in antitumour responses, and find that the co-occurrence of tumour-associated CD8+ T cells and CD20+ B cells is associated with improved survival, independently of other clinical variables. Immunofluorescence staining of CXCR5 and CXCL13 in combination with CD20 reveals the formation of tertiary lymphoid structures in these CD8+CD20+ tumours. We derived a gene signature associated with tertiary lymphoid structures, which predicted clinical outcomes in cohorts of patients treated with immune checkpoint blockade. Furthermore, B-cell-rich tumours were accompanied by increased levels of TCF7+ naive and/or memory T cells. This was corroborated by digital spatial-profiling data, in which T cells in tumours without tertiary lymphoid structures had a dysfunctional molecular phenotype. Our results indicate that tertiary lymphoid structures have a key role in the immune microenvironment in melanoma, by conferring distinct T cell phenotypes. Therapeutic strategies to induce the formation of tertiary lymphoid structures should be explored to improve responses to cancer immunotherapy.
                Bookmark

                Author and article information

                Journal
                Cancer Immunol Res
                Cancer Immunol Res
                Cancer Immunology Research
                American Association for Cancer Research
                2326-6066
                2326-6074
                01 April 2022
                21 February 2022
                : 10
                : 4
                : 468-481
                Affiliations
                [1 ]Cancer and Blood Disorders Institute and Department of Surgery, Johns Hopkins All Children's Hospital, St. Petersburg, Florida.
                [2 ]Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
                [3 ]Division of Pathology and Clinical Laboratories, National Cancer Center Hospital/Division of Molecular Pathology, Analytical Pathology, National Cancer Center Research Institute, Tokyo, Japan.
                [4 ]Department of Health Informatics, Johns Hopkins All Children's Hospital, St. Petersburg, Florida.
                [5 ]Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan.
                Author notes
                [* ] Corresponding Author: Masanobu Komatsu, Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, 600 5th Street S, St. Petersburg, FL 33701. Phone: 727-767-3459; E-mail: mkomats1@ 123456jhmi.edu
                Author information
                https://orcid.org/0000-0001-9100-7037
                https://orcid.org/0000-0003-4215-4385
                https://orcid.org/0000-0002-6599-7329
                Article
                CIR-21-0369
                10.1158/2326-6066.CIR-21-0369
                8976767
                35201289
                5fb3f4cd-00a2-46d6-a08a-082a05a7a211
                ©2022 The Authors; Published by the American Association for Cancer Research

                This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.

                History
                : 13 May 2021
                : 11 October 2021
                : 18 February 2022
                Page count
                Pages: 14
                Funding
                Funded by: National Cancer Institute (NCI), https://doi.org/10.13039/100000054;
                Award ID: R01CA125255
                Award Recipient :
                Funded by: Florida Department of Health, https://doi.org/10.13039/100006827;
                Award ID: 20B01
                Award Recipient :
                Funded by: Florida Breast Cancer Foundation (FBCF), https://doi.org/10.13039/100014237;
                Award Recipient :
                Categories
                Research Articles

                Comments

                Comment on this article