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

      Prediction of immunotherapy response of bladder cancer with a pyroptosis-related signature indicating tumor immune microenvironment

      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

          Background

          Although prognostic models based on pyroptosis-related genes (PRGs) have been constructed in bladder cancer (BLCA), the comprehensive impact of these genes on tumor microenvironment (TME) and immunotherapeutic response has yet to be investigated.

          Methods

          Based on expression profiles of 52 PRGs, we utilized the unsupervised clustering algorithm to identify PRGs subtypes and ssGSEA to quantify immune cells and hallmark pathways. Moreover, we screened feature genes of distinct PRGs subtypes and validated the associations with immune infiltrations in tissue using the multiplex immunofluorescence. Univariate, LASSO, and multivariate Cox regression analyses were employed to construct the scoring scheme.

          Results

          Four PRGs clusters were identified, samples in cluster C1 were infiltrated with more immune cells than those in others, implying a favorable response to immunotherapy. While the cluster C2, which shows an extremely low level of most immune cells, do not respond to immunotherapy. CXCL9/CXCL10 and SPINK1/DHSR2 were identified as feature genes of cluster C1 and C2, and the specimen with high CXCL9/CXCL10 was characterized by more CD8 + T cells, macrophages and less Tregs. Based on differentially expressed genes (DEGs) among PRGs subtypes, a predictive model (termed as PRGs score) including five genes (CACNA1D, PTK2B, APOL6, CDK6, ANXA2) was built. Survival probability of patients with low-PRGs score was significantly higher than those with high-PRGs score. Moreover, patients with low-PRGs score were more likely to benefit from anti-PD1/PD-L1 regimens.

          Conclusion

          PRGs are closely associated with TME and oncogenic pathways. PRGs score is a promising indicator for predicting clinical outcome and immunotherapy response.

          Related collections

          Most cited references52

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

          The Tumor Microenvironment Innately Modulates Cancer Progression

          Cancer development and progression occurs in concert with alterations in the surrounding stroma. Cancer cells can functionally sculpt their microenvironment through the secretion of various cytokines, chemokines, and other factors. This results in a reprogramming of the surrounding cells, enabling them to play a determinative role in tumor survival and progression. Immune cells are important constituents of the tumor stroma and critically take part in this process. Growing evidence suggests that the innate immune cells (macrophages, neutrophils, dendritic cells, innate lymphoid cells, myeloid-derived suppressor cells, and NK cells) as well as adaptive immune cells (T cells and B cells) contribute to tumor progression when present in the tumor microenvironment (TME). Crosstalk between cancer cells and the proximal immune cells ultimately results in an environment that fosters tumor growth and metastasis. Understanding the nature of this dialog will allow for improved therapeutics that simultaneously target multiple components of the TME, increasing the likelihood of favorable patient outcomes.
            Bookmark
            • 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

              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

                Contributors
                URI : https://loop.frontiersin.org/people/1638817/overviewRole: Role: Role: Role: Role: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1831283/overviewRole: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/2239730/overviewRole: Role: Role: Role: Role: Role:
                Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1289414/overviewRole: Role: Role: Role:
                Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/2053018/overviewRole: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/596972/overviewRole: Role: Role:
                URI : https://loop.frontiersin.org/people/533518/overviewRole: Role: Role: Role: Role:
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                25 June 2024
                2024
                : 15
                : 1387647
                Affiliations
                [1] 1 Institute for Breast Health Medicine , State Key Laboratory of Biotherapy , West China Hospital , Sichuan University and Collaborative Innovation Center , Chengdu, Sichuan, China
                [2] 2 Department of Medical Oncology , Cancer Center , West China Hospital , Sichuan University , Chengdu, Sichuan, China
                [3] 3 Department of Biotherapy , West China Hospital and State Key Laboratory of Biotherapy , Sichuan University , Chengdu, Sichuan, China
                Author notes

                Edited by: Hailin Tang, Sun Yat-Sen University Cancer Center (SYSUCC), China

                Reviewed by: Xiangmei Zhang, Fourth Hospital of Hebei Medical University, China

                Qianrong Huang, Guangxi Medical University Cancer Hospital, China

                *Correspondence: Xuelei Ma, drmaxuelei@ 123456gmail.com ; Hubing Shi, shihb@ 123456scu.edu.cn
                [ † ]

                These authors have contributed equally to this work and share first authorship

                Article
                1387647
                10.3389/fphar.2024.1387647
                11231188
                38983908
                69a1af8a-c4b9-4ed5-a926-d84cd0497cee
                Copyright © 2024 Xu, Zhao, Zhang, Liu, Song, Chen, Xiao, Ma and Shi.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 18 February 2024
                : 04 June 2024
                Funding
                Funded by: China Postdoctoral Science Foundation , doi 10.13039/501100002858;
                Award ID: 2023M742486
                Funded by: National Key Research and Development Program of China , doi 10.13039/501100012166;
                Award ID: 2022YFA1207300
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: 82172634
                The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by 1) the National Key Research and Development Program of China [2022YFA1207300 (2022YFA1207303)]; 2) National Natural Science Foundation of China (No. 82172634); 3) Key Program of the Science and Technology Bureau of Sichuan (No. 2021YFSY0007); 4) 1.3.5 project for disciplines of excellence, West China Hospital, Sichuan University (No. ZYGD23028); 5) The China Postdoctoral Science Foundation (2023M742486).
                Categories
                Pharmacology
                Original Research
                Custom metadata
                Pharmacology of Anti-Cancer Drugs

                Pharmacology & Pharmaceutical medicine
                pyroptosis,bladder cancer,tumor microenvironment,immunotherapy,predictive model

                Comments

                Comment on this article