1
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2025; 33:295-315. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
2
|
Pirhoushiaran M, Heidarzadehpilehrood R, Mokhtarinejad M, Hesami S, Rezaei N, Farahani AS. Upregulated PCAT-1 predicts poor prognosis and reduced immune cell infiltration in head and neck squamous cell carcinoma through the miR-145-5p / FSCN-1 axis. Mol Biol Rep 2025; 52:121. [PMID: 39806246 DOI: 10.1007/s11033-024-10208-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND LncRNA PCAT-1 is known to promote cancer proliferation, invasion, and metastasis. However, its significance in HNSCC is not fully understood. This research investigates how the PCAT-1 / miR-145-5p / FSCN-1 axis promote HNSCC. METHODS AND RESULTS We analyzed the gene expression patterns on 238 fresh-frozen samples, comparing tumors with their normal adjacent tissues (NATs). HNSCC samples showed higher PCAT-1 and FSCN-1 expression compared to NATs (p < 0.001 and p < 0.001, respectively). In contrast, miR-145-5p expression was markedly downregulated compared to NATs (p < 0.001). Notably, ROC curve analysis revealed exceptional diagnostic power, with an AUC of 0.83 for PCAT-1, 0.95 for miR-145-5p, and 0.91 for FSCN-1. Pearson correlation analysis unveiled a significant positive correlation between PCAT-1 and FSCN-1 expression levels (r = 0.084, p < 0.001) and negative correlations between FSCN-1 and miR-145-5p (r = -0.710, p < 0.001) as well as between PCAT-1 and miR-145-5p (r = -0.759, p < 0.001). Distinct molecular profiles were observed in the levels of PCAT-1, miR-145-5p, and FSCN-1 between HPV (-) and HPV ( +) 16 and 18 genotypes (p = 0.007, p = 0.027, and p = 0.002). MiR-145-5p expression showed significant differences between HPV (-) and HPV ( +) other genotypes (p = 0.035). FSCN-1 expression showed notable distinctions between HPV ( +) 18 & 16 and HPV ( +) other genotypes (p = 0.031). CONCLUSIONS Elevated levels of lncRNA PCAT-1 promote HNSCC through the miR-145-5p/FSCN-1 axis and are associated with poor prognosis and reduced immune cell infiltration levels.
Collapse
Affiliation(s)
- Maryam Pirhoushiaran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, 1417613151, Iran
| | - Roozbeh Heidarzadehpilehrood
- Department of Obstetrics & Gynaecology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mahnoosh Mokhtarinejad
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, 1417613151, Iran
| | - Sara Hesami
- Medical Genetic Ward, Faculty of Medicine, Imam Khomeini Hospital Complex, IKHC, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden
| | - Abbas Shakoori Farahani
- Medical Genetic Ward, Faculty of Medicine, Imam Khomeini Hospital Complex, IKHC, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Gulubova MV, Valkanov SP, Ignatova MMK, Minkov GA. Tertiary lymphoid structures in colorectal cancer - organization and immune cell interactions. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:236-245. [PMID: 39839346 PMCID: PMC11744347 DOI: 10.62347/gryy2849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/12/2024] [Indexed: 01/23/2025]
Abstract
Tertiary lymphoid structures (TLS), formerly recognized as Crohn's-like structures, serve as crucial biomarkers for evaluating the progression of colorectal cancer (CRC). Understanding their spatial distribution, cellular composition, and interactions within CRC is paramount for comprehending the immune response in the tumor microenvironment (TME). TLS are comprised of a T-cellular compartment and a B-cellular compartment, the latter encompassing follicular dendritic cells (FDCs), high endothelial venules (HEVs), and lymphatic vessels. While T helper cells predominate in cancer TLS, the specific functions of their subpopulations remain inadequately understood. Notably, T follicular helper (Tfh) cells play a central role in the activation of CD8+ T cells, and both Tfh cells and Tfh-associated genes have been linked to enhanced CRC survival. In stage II CRC TLS, an escalation in the number of FoxP3+ T regulatory cells (Tregs) is regarded as a negative prognostic factor. Moreover, within TLS, T lymphocytes shield B lymphocytes from the immunosuppressive effects of the TME. B lymphocyte activation is succeeded by class recombination (CSR) and somatic hypermutation (SHM). Dendritic cells (DCs) constitute a vital cellular component of the TLS T compartment. During steady state and early stages of CRC, specialized antigen-presenting cells such as DCs migrate to regional lymph nodes through afferent lymphatics. They deliver MHC antigen-derived peptide complexes (tumor antigens) to naïve CD4+ and CD8+ T cells, which subsequently infiltrate the tumor site as antigen-specific T cells. Key DC markers studied in TLS include CD83 and DC-LAMP. Research has indicated that the DC-LAMP gene signature in tumor TLS reflects Th1 cell targeting, cytotoxicity, and T cell activation. This review comprehensively outlines the functions performed by distinct cell subsets within tertiary lymphoid structures (TLS) in tumors.
Collapse
Affiliation(s)
- Maya Vladova Gulubova
- Clinics of Pathology, University Hospital “Prof. Dr. Stoyan Kirkovich”Stara Zagora, Bulgria
- Department of Anatomy, Histology, Embryology and Pathology, Medical Faculty, “Asen Zlatarov University Bourgas”Bourgas, Bulgaria
| | - Stefan P Valkanov
- Clinics of Neurosurgery, University Hospital “Prof. Dr. Stoyan Kirkovich”Stara Zagora, Bulgria
- Department of Surgery, Medical Faculty, Trakia UniversityStara Zagora, Bulgria
| | | | - Georgi A Minkov
- Department of Surgery, Medical Faculty, Trakia UniversityStara Zagora, Bulgria
- Clinics of Surgery, University Hospital “Prof. Dr. Stoyan Kirkovich”Stara Zagora, Bulgria
| |
Collapse
|
4
|
Petrov EA, Malabuiok DM, Zheng H, Mokrushina YA, Abrikosova VA, Kuzmin YB, Tzarapaev PV, Kochkina SO, Eltsov IV, Knorre VD, Smirnov IV, Terekhov SS, Mamedli Z, Kushlinskii NE, Rogozhin DV, Matveev VB, Kononets PV, Stilidi IS, Zhang H, Gabibov AG. Abundance of Tumor-Infiltrating B Cells in Human Epithelial Malignancies. Acta Naturae 2024; 16:67-73. [PMID: 39555171 PMCID: PMC11569841 DOI: 10.32607/actanaturae.27353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/28/2024] [Indexed: 11/19/2024] Open
Abstract
Cancer is a major global health problem. The type of malignant neoplasm and the potency of the immune response against tumors are two of the key factors influencing the outcome of the disease. The degree of tumor infiltration by lymphocytes plays an important role in antitumor response development, generally correlating with a favorable prognosis of treatment for certain cancers. We analyzed the abundance of tumor-infiltrating B cells (TIBs) in solid tumors of different cancers. TIBs were shown to be more abundant in colon and sigmoid colon cancer samples compared with cecal, rectal, and kidney cancer samples. The median and interquartile range of the TIB fraction were 11.5% and 4-20% in colon cancer, 6% and 3-11% in sigmoid colon cancer, 2.7% and 0.7-3.7% in cecal cancer, 2.5% and 0.9-3.6% in rectal cancer, 1.4% and 1.0-2.3% in kidney cancer, and 3.0% and 1.8-12% in lung cancer, respectively. However, there were no significant differences in the abundance of TIBs among samples at different stages of the cancer. Hence, investigation of the B cell response in colon cancer is of particular interest, since increased quantities of TIBs may indicate the existence of immunogenic tumor markers or the cell-cell interactions involved in disease progression. We believe that studying the diversity of TIBs in colon cancer will increaseour understanding of the mechanisms of the disease, contributing to the identification of new molecular targets for targeted oncotherapy.
Collapse
Affiliation(s)
- E. A. Petrov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - D. M. Malabuiok
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - H. Zheng
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Yu. A. Mokrushina
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - V. A. Abrikosova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - Yu. B. Kuzmin
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - P. V. Tzarapaev
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - S. O. Kochkina
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - I. V. Eltsov
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - V. D. Knorre
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - I. V. Smirnov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Endocrinology Research Center, Moscow, 117036 Russian Federation
| | - S. S. Terekhov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - Z. Mamedli
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - N. E. Kushlinskii
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - D. V. Rogozhin
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - V. B. Matveev
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - P. V. Kononets
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - I. S. Stilidi
- Blokhin National Medical Research Center of Oncology, Moscow, 115522 Russian Federation
| | - H. Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - A. G. Gabibov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| |
Collapse
|
5
|
Sun X, He W, Lin B, Huang W, Ye D. Defining three ferroptosis-based molecular subtypes and developing a prognostic risk model for high-grade serous ovarian cancer. Aging (Albany NY) 2024; 16:9106-9126. [PMID: 38795391 PMCID: PMC11164503 DOI: 10.18632/aging.205857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/08/2024] [Indexed: 05/27/2024]
Abstract
BACKGROUND As a newly defined regulated cell death, ferroptosis is a potential biomarker in ovarian cancer (OV). However, its underlying mechanism in tumor microenvironment (TME) and clinical prediction significance in OV remained to be elucidated. METHODS The transcriptome data of high-grade serous OV from The Cancer Genome Atlas (TCGA) database were downloaded. Molecular subtypes were classified based on ferroptosis-correlated genes from the FerrDb database by performing consensus clustering analysis. The associations between the subtypes and clinicopathologic characteristics, mutation, regulatory pathways and immune landscape were assessed. A ferroptosis-related prognostic model was constructed and verified using International Cancer Genome Consortium (ICGC) cohort and GSE70769. RESULTS Three molecular subtypes of OV were defined. Patients in subtype C3 tended to have the most favorable prognosis, while subtype C1 showing more mesenchymal cells, increased immune infiltration of Macrophages_M2, lower tumor purity, and epithelial-to-mesenchymal transition (EMT) features had the poorest prognosis. A ferroptosis-related risk model was constructed using 8 genes (PDP1, FCGBP, EPHA4, GAS1, SLC7A11, BLOC1S1, SPOCK2, and CXCL9) and manifested a strong prediction performance. High-risk patients had enriched EMT pathways, more Macrophages_M2, less plasma cells and CD8 cell infiltration, greater tendency of immune escape and worse prognosis. The risk score has negatively correlated relation with LAG3, TIGIT, CTLA4, IDO1, CD27, ICOS, and IL2RB but positively correlated with PVR, CD276, and CD28. Moreover, low-risk patients were more sensitive to Cisplatin and Gefitinib, Gemcitabine. CONCLUSIONS Our results could improve the understanding of ferroptosis in OV, providing promising insights for the clinical targeted therapy for the cancer.
Collapse
Affiliation(s)
- Xiang Sun
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Wenbin He
- Department of Otolaryngology, Lanzhou University Second Hospital, Gansu 730030, China
| | - Baohua Lin
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Weiming Huang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Danping Ye
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| |
Collapse
|
6
|
Sun XY, Wang CQ, Mao Y, Zhang ZQ, Cui J, Dong XN, Wang HB. Prognostic value and distribution pattern of tumor infiltrating lymphocytes and their subsets in distant metastases of advanced breast cancer. Clin Breast Cancer 2024; 24:e167-e176. [PMID: 38212189 DOI: 10.1016/j.clbc.2023.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/09/2023] [Accepted: 12/29/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND There are significant correlations between the levels of tumor infiltrating lymphocytes (TILs) and the prognosis of primary breast cancer. While little is known about immunological mechanisms in the distant metastasis of advanced breast cancer. PATIENTS AND METHODS A total of 106 patients with advanced metastatic breast cancer were enrolled in this study between 2016 and 2022. Hematoxylin and eosin staining and immunohistochemistry were used to assess the densities of stromal TILs (sTILs), intratumoral TILs (iTILs) and invasive marginal TILs (imTILs) and CD4+, CD8+, CD20+, FOXP3+ TILs in the primary tumor and metastasis (bone, lung, liver, and distant lymph node) of advanced breast cancer. RESULTS Higher levels of sTILs at metastatic sites were associated with better progression-free survival (PFS), postmetastasis survival (PMS) and overall survival (OS) (p = .026, .001 and .005, respectively). The levels of iTILs were significantly lower than those of sTILs and imTILs in both primary tumor (p< .001, both) and metastasis (p< .001, both). The level of CD4+ T cells was higher than those of CD8+ T cells and CD20+ B cells in both primary tumor (p < .001) and metastasis (p < .001). The levels of sTILs (p=0. 001) and imTILs (p< .001) in the primary tumor were generally higher than those in the metastasis. CONCLUSION The levels of TILs and their subsets can predict the survival and prognosis of patients with advanced breast cancer. The distributions of TILs and their subsets are similar between the primary tumor and metastasis. The metastases have a lower degree of lymphocytes infiltration than its corresponding primary tumor.
Collapse
Affiliation(s)
- Xin-Yi Sun
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, PR China
| | - Cheng-Qin Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, PR China
| | - Yan Mao
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, PR China
| | - Zhen-Qi Zhang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, PR China
| | - Jian Cui
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, PR China
| | - Xian-Ning Dong
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, PR China
| | - Hai-Bo Wang
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, PR China.
| |
Collapse
|
7
|
Chandnani N, Gupta I, Mandal A, Sarkar K. Participation of B cell in immunotherapy of cancer. Pathol Res Pract 2024; 255:155169. [PMID: 38330617 DOI: 10.1016/j.prp.2024.155169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
Even though their effector roles extend beyond conventional humoral immunity, B and plasma cells may exhibit antitumor effects through antibody-dependent cell cytotoxicity (ADCC) and activation of the complement cascade. Depending on whether they are positioned in immature or mature compartments termed tertiary lymphoid structures (TLS), which include T cells, B cells are believed to play numerous functions in modulating the immune system's capacity to destroy cancer cells. These formations represent a process of lymphoid neogenesis that takes place in peripheral tissues in response to prolonged exposure to inflammatory signals. Activated in the germinal centres of tertiary lymphoid structures, B cells may directly present tumor-associated antigens to T cells, make antibodies that enhance antigen presentation to T cells, or kill tumour cells, resulting in a favourable therapeutic effect. Immune complexes may also enhance inflammation, angiogenesis, and immunosuppression via the activation of macrophages and complement, resulting in detrimental effects. The functional variety of B-cell subsets includes professional antigen-presenting cells, regulatory cells, memory populations, and plasma cells that produce antibodies. Importantly, antibodies may independently generate innate immune responses and the cancer immunity cycle. B cells and B-cell-mediated antibody responses constitute the largely underestimated second arm of the adaptive immune system and unquestionably need more consideration in cancer. This article reviews the known roles of B lymphocytes in the tumour microenvironment, their contribution to anticancer activity of immunotherapies, and their significance in overall survival of cancer patients. In addition to producing antibodies, B cells regulate the immune system and serve as effective antigen-presenting cells.
Collapse
Affiliation(s)
- Nikhil Chandnani
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ishika Gupta
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ayush Mandal
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
8
|
Chen C, Zhang Y, Wu X, Shen J. The role of tertiary lymphoid structure and B cells in nasopharyngeal carcinoma: Based on bioinformatics and experimental verification. Transl Oncol 2024; 41:101885. [PMID: 38295746 PMCID: PMC10846412 DOI: 10.1016/j.tranon.2024.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/02/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE Transcriptomic characteristics and prognosis of tertiary lymphoid structures (TLS) and infiltrating B cells in nasopharyngeal carcinoma (NPC) remain unclear. Here, NPC transcriptomic data and clinical samples were used to investigate the role of infiltrating B cells and TLS in NPC. METHODS We investigated the gene expression and infiltrating immune cells of NPC patients and further investigated the clinical relevance of B cell and TLS signatures. Transcriptional features of infiltrating B cell subsets were revealed by single-cell RNA sequencing (scRNA-seq) analysis. Immunohistochemical (IHC) and HE staining were performed to validate the clinical relevance of infiltrating B cells and TLS in NPC samples. RESULTS 27 differentially expressed immune-related genes (IRGs) associated with prognosis were identified, including B cell marker genes CD19 and CD79B. The higher B cells and TLS signature scores were associated with better outcomes and early pathological staging in 88 NPC patients. ScRNA-seq identified five distinct B cell subsets in NPC, including the BC-4 cluster associated with poor outcomes and the BC-0 cluster associated with better outcomes. EBV infection was positively associated with the formation of TLS. Furthermore, experimental results showed that the infiltration of B cells in NPC tissues was higher than that of normal tissues, and the density of TLS in an early stage of NPC was higher than that in advanced-stage TLS. CONCLUSION Our findings demonstrate the functional importance of distinct B cell subsets in the prognosis of NPC. Additionally, we confirmed that B cells and TLS may serve as prognostic biomarkers of survival for NPC patients.
Collapse
Affiliation(s)
- Chujun Chen
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Yan Zhang
- Pathology Dept., The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaoting Wu
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Juan Shen
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, PR China.
| |
Collapse
|
9
|
Chandnani N, Mandal A, Gupta I, Mukherjee O, Rakshit S, Shanmugam G, George M, Sarkar K. Association of Wiskott-Aldrich syndrome protein (WASp) in epigenetic regulation of B cell differentiation in non-small-cell lung cancer (NSCLC). Med Oncol 2023; 41:28. [PMID: 38146020 DOI: 10.1007/s12032-023-02264-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/21/2023] [Indexed: 12/27/2023]
Abstract
Non-small-cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer which is the deadliest type of cancer for both men and women. Previous studies already showed that cell-intrinsic loss of WASp causes B cell tolerance and WASp deficiency in T helper (TH) cells is linked to negative effects on cytokine gene transcription necessary for TH1 differentiation. In the current study, we investigated the molecular mechanisms involved in WASp-mediated epigenetic regulation of B cell differentiation during NSCLC. Our ChIP-qPCR data suggest the less percentage enrichment of the B cell differentiating factors (Ikaros, Pax5, PU.1, BATF) and WASp across the WAS gene in the B cells of NSCLC patients in comparison with normal healthy donors and overexpression of WASp showed the reverse effects. WASp-depleted B cells while co-culturing with respective PBMCs isolated from normal healthy donors and NSCLC patients, we observed upregulation of TH2-, TH17-, and Treg-specific cytokines (IL4, ILI7A, IL10) & transcription factors (GATA3, RORC, FOXP3) and downregulation of TH1-specific cytokine (IFNγ) & transcription factor (TBX21). Our study showed that the overexpression of WASp resulted into upregulation of B cell differentiating factors, tumor suppressor protein (p53), histone methylation marker (H3K4me3) with concomitant downregulation of tumor-promoting factors (Notch 1, β-Catenin, DNAPKcs) and histone deacetylation marker (HDAC2) and increase in percentage cytotoxicity of NSCLC-specific cells (A549). Successful overexpression of WASp not only helps in epigenetic regulation of B cell differentiation but also supports tumor suppression in NSCLC. Thus, WASp can be targeted for therapeutic intervention of NSCLC.
Collapse
Affiliation(s)
- Nikhil Chandnani
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Ayush Mandal
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Ishika Gupta
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Oishi Mukherjee
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Sudeshna Rakshit
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Melvin George
- Department of Clinical Pharmacology, SRM Medical College Hospital and Research Centre, Kattankulathur, Tamil Nadu, 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| |
Collapse
|
10
|
Koeck S, Amann A, Kern J, Zwierzina M, Lorenz E, Sopper S, Zwierzina H, Mildner F, Sykora M, Sprung S, Hackl H, Augustin F, Maier HT, Pircher A, Pall G, Wolf D, Gamerith G. Whole stromal fibroblast signature is linked to specific chemokine and immune infiltration patterns and to improved survival in NSCLC. Oncoimmunology 2023; 12:2274130. [PMID: 38126028 PMCID: PMC10732611 DOI: 10.1080/2162402x.2023.2274130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 10/18/2023] [Indexed: 12/23/2023] Open
Abstract
Cancer associated fibroblasts (CAF) are known to orchestrate multiple components of the tumor microenvironment, whereas the influence of the whole stromal-fibroblast compartment is less understood. Here, an extended stromal fibroblast signature was investigated to define its impact on immune cell infiltration. The lung cancer adenocarcinoma (LUAD) data set of the cancer genome atlas (TCGA) was used to test whole stroma signatures and cancer-associated fibroblast signatures for their impact on prognosis. 3D cell cultures of the NSCLC cancer cell line A549 together with the fibroblast cell line SV80 were used in combination with infiltrating peripheral blood mononuclear cells (PBMC) for in-vitro investigations. Immune cell infiltration was assessed via flow cytometry, chemokines were analyzed by immunoassays and RNA microarrays. Results were confirmed in specimens from NSCLC patients by flow cytometry or immunohistochemistry as well as in the TCGA data set. The TCGA analyses correlated the whole stromal-fibroblast signature with an improved outcome, whereas no effect was found for the CAF signatures. In 3D microtumors, the presence of fibroblasts induced infiltration of B cells and CD69+CD4+ T cells, which was linked to an increased expression of CCL13 and CXCL16. The stroma/lymphocyte interaction was confirmed in NSCLC patients, as stroma-rich tumors displayed an elevated B cell count and survival in the local cohort and the TCGA data set. A whole stromal fibroblast signature was associated with an improved clinical outcome in lung adenocarcinoma and in vitro and in vivo experiments suggest that this signature increases B and T cell recruitment via induction of chemokines.
Collapse
Affiliation(s)
- Stefan Koeck
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Arno Amann
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Johan Kern
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
- Department of Otorhinolaryngology, Head and Neck Surgery, Mannheim Medical Faculty of Heidelberg University, Mannheim, Germany
| | - Marit Zwierzina
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Edith Lorenz
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Sieghart Sopper
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Heinz Zwierzina
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Finn Mildner
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Martina Sykora
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Susanne Sprung
- Department of Pathology, Neuropathology, and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Florian Augustin
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Hubert T. Maier
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Andreas Pircher
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Georg Pall
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Dominik Wolf
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Gabriele Gamerith
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
- Tyrolean Cancer Research Institute, Innsbruck, Austria
| |
Collapse
|
11
|
Al Zein M, Boukhdoud M, Shammaa H, Mouslem H, El Ayoubi LM, Iratni R, Issa K, Khachab M, Assi HI, Sahebkar A, Eid AH. Immunotherapy and immunoevasion of colorectal cancer. Drug Discov Today 2023; 28:103669. [PMID: 37328052 DOI: 10.1016/j.drudis.2023.103669] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/20/2023] [Accepted: 06/08/2023] [Indexed: 06/18/2023]
Abstract
The tremendous success of immunotherapy in clinical trials has led to its establishment as a new pillar of cancer therapy. However, little clinical efficacy has been achieved in microsatellite stable colorectal cancer (MSS-CRC), which constitutes most CRC tumors. Here, we discuss the molecular and genetic heterogeneity of CRC. We review the immune escape mechanisms, and focus on the latest advances in immunotherapy as a treatment modality for CRC. By providing a better understanding of the tumor microenvironment (TME) and the molecular mechanisms underlying immunoevasion, this review offers an insight into developing therapeutic strategies that are effective for patients with various subsets of CRC.
Collapse
Affiliation(s)
- Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Mona Boukhdoud
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Hadi Shammaa
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Hadi Mouslem
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | | | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, UAE
| | - Khodr Issa
- University of Lille, Proteomics, Inflammatory Response, Mass Spectrometry, INSERM U-1192, Lille, France
| | - Maha Khachab
- Faculty of Medicine, University of Balamand, Lebanon
| | - Hazem I Assi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University, QU Health, Doha, Qatar.
| |
Collapse
|
12
|
Gylstorff S, Wilke V, Kraft D, Bertrand J, Pech M, Haag F, Relja B. Selective Internal Radiotherapy Alters the Profiles of Systemic Extracellular Vesicles in Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:12512. [PMID: 37569887 PMCID: PMC10419408 DOI: 10.3390/ijms241512512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Incidence of hepatocellular carcinoma (HCC) is increasing globally. Radioembolization (RE)/selective internal radiotherapy (SIRT) is a promising treatment for inoperable HCC. RE triggers an immune response, involving extracellular vesicles (EVs) which are crucial for cell communication and tumor development. This study explores EV immune profiles and origins in patients with inoperable HCC before and after SIRT/RE. Blood samples from 50 HCC-patients treated with SIRT/RE were collected before and after therapy to determine cytokines and isolate EVs using size exclusion chromatography. The dynamic range and EV quality required for detecting variations in surface markers were assessed. Thirty-seven EV surface markers were analyzed using flow cytometry and correlated with clinical parameters. Several immunological markers (CD4, CD2, CD40, CD45, CD49e, CD69, CD209-EVs) were present in the circulation of HCC patients. These markers positively correlated with therapy response and survival. Conversely, B cell CD20, endothelial cell CD146, platelet CD49e, and CD41b EV markers negatively correlated with 60-day survival. Elevated levels of IL-6 and IL-8 before therapy correlated negatively with patient survival, coinciding with a positive correlation with CD20-positive EVs. Plasma EVs from HCC patients exhibit immunological, cancer, and coagulation markers, including potential biomarkers (CD4, CD20, CD49e, CD146). These may enhance our understanding of cancer biology and facilitate SIRT therapy monitoring.
Collapse
Affiliation(s)
- Severin Gylstorff
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Ulm, 89081 Ulm, Germany
| | - Vanessa Wilke
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Daniel Kraft
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Maciej Pech
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Florian Haag
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Ulm, 89081 Ulm, Germany
| |
Collapse
|
13
|
Schina A, Sztupinszki Z, Marie Svane I, Szallasi Z, Jönsson G, Donia M. Intratumoral T-cell and B-cell receptor architecture associates with distinct immune tumor microenvironment features and clinical outcomes of anti-PD-1/L1 immunotherapy. J Immunother Cancer 2023; 11:e006941. [PMID: 37604641 PMCID: PMC10445359 DOI: 10.1136/jitc-2023-006941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Effective cooperation between B-cells and T-cells within the tumor microenvironment may lead to the regression of established tumors. B-cells and T-cells can recognize tumor antigens with exquisite specificity via their receptor complexes. Nevertheless, whether a diverse intratumoral B-cells and T-cell receptor (BCR, TCR) repertoire affects the tumor immune microenvironment (TIME) and clinical outcomes in patients treated with immunotherapy is unclear. METHODS We extracted information on BCR and TCR repertoire diversity from large clinical datasets and measured the association between immune receptor diversity features, the TIME, and clinical outcomes of patients treated with anti-PD-1/PD-L1 immunotherapy. RESULTS In multiple tumor types, an increasingly diverse TCR repertoire was strongly associated with a highly activated TIME, while BCR diversity was more associated with antibody responses but not with the overall B-cell infiltration nor with measures related to intratumoral CD8+T cell activity. Neither TCR nor BCR diversity was independent prognostic biomarkers of survival across multiple cancer types. However, both TCR and BCR diversity improved the performance of predictive models combined with established biomarkers of response to immunotherapy. CONCLUSION Overall, these data indicate a currently unexplored immunological role of intratumoral B-cells associated with BCR diversity and antibody responses but independent of classical anticancer T-cells intratumoral activities.
Collapse
Affiliation(s)
- Aimilia Schina
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| | | | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| | | | - Göran Jönsson
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| |
Collapse
|
14
|
Kajihara N, Tanaka Y, Takeuchi R, Kobayashi T, Tanji M, Ataka T, Nakano S, Yamada T, Takaoka A, Hasegawa Y, Seino KI, Wada H. Augmented interferon regulatory factor 7 axis in whole tumor cell vaccines prevents tumor recurrence by inducing interferon gamma-secreting B cells. Oncoimmunology 2023; 12:2213132. [PMID: 37235066 PMCID: PMC10208210 DOI: 10.1080/2162402x.2023.2213132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/17/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Among cancer immunotherapy, which has received great attention in recent years, cancer vaccines can potentially prevent recurrent tumors by using the exquisite power and specificity of the immune system. Specifically, whole tumor cell vaccines (WTCVs) based on surgically resected tumors have been considered to elicit robust anti-tumor immune responses by exposing various tumor-associated antigens to host immunity. However, most tumors have little immunogenicity because of immunoediting by continuous interactions with host immunity; thus, preparing WTCVs based on patient-derived non-modified tumors cannot prevent tumor onset. Hence, the immunogenicity of tumor cells must be improved for effective WTCVs. In this study, we indicate the importance of the interferon regulatory factor 7 (Irf7) axis, including Irf7 and its downstream factors, within tumor cells in regulating immunogenicity. Indeed, WTCVs that augmented the Irf7 axis have exerted remarkable recurrence-preventive effects when vaccinated after tumor inactivation by radiation. Most notably, vaccination with murine colon cancer cells that enhanced the Irf7 axis prevented the development of challenged tumors in all mice and resulted in a 100% survival rate during the observation period. Furthermore, the mechanism leading to vaccine effectiveness was mediated by interferon-gamma-producing B cells. This study provides novel insights into how to enhance tumor immunogenicity and use WTCVs as recurrence prophylaxis.
Collapse
Affiliation(s)
- Nabeel Kajihara
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshino Tanaka
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Riko Takeuchi
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Takuto Kobayashi
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Masafumi Tanji
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Tsukasa Ataka
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Shiho Nakano
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Taisho Yamada
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Akinori Takaoka
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshinori Hasegawa
- Laboratory of Gene Sequencing Analysis, Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Ken-Ichiro Seino
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Haruka Wada
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
15
|
Milardi G, Lleo A. Tumor-Infiltrating B Lymphocytes: Promising Immunotherapeutic Targets for Primary Liver Cancer Treatment. Cancers (Basel) 2023; 15:2182. [PMID: 37046842 PMCID: PMC10093314 DOI: 10.3390/cancers15072182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Hepatocellular carcinoma and cholangiocarcinoma are the fourth most lethal primary cancers worldwide. Therefore, there is an urgent need for therapeutic strategies, including immune cell targeting therapies. The heterogeneity of liver cancer is partially explained by the characteristics of the tumor microenvironment (TME), where adaptive and innate immune system cells are the main components. Pioneering studies of primary liver cancers revealed that tumor-infiltrating immune cells and their dynamic interaction with cancer cells significantly impacted carcinogenesis, playing an important role in cancer immune evasion and responses to immunotherapy treatment. In particular, B cells may play a prominent role and have a controversial function in the TME. In this work, we highlight the effect of B lymphocytes as tumor infiltrates in relation to primary liver cancers and their potential prognostic value. We also present the key pathways underlying B-cell interactions within the TME, as well as the way that a comprehensive characterization of B-cell biology can be exploited to develop novel immune-based therapeutic approaches.
Collapse
Affiliation(s)
- Giulia Milardi
- Hepatobiliary Immunopathology Labaratory, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| |
Collapse
|
16
|
Abstract
Cancer is still a serious health problem globally. Conventional therapies have adverse effects, which affect human life quality. Tumor microenvironment (TME), also known as surrounding stroma, has a contributory role in cancer development. Understanding the interaction between TME and cancer progression is a challenge and helps to develop new therapeutic strategies that neutralize the tracks taken by cancer cells to grow, spread, and resist therapy. Therefore, targeting TME components may be effective in improving tumor therapy. Using nanotechnology for drug delivery is of great interest, where it overcomes some obstacles such as solubility and absorption of drugs and delivering them to the appropriate place of action. The main target of nanotechnology for drug delivery is the ability to differentiate between normal and cancer cells. It can be concluded that TME is an important complementary strategy for the development of anticancer drugs. Multitargeted therapy has better efficient potential than individual therapy against cancer.
Collapse
|
17
|
Wauchope BA, Coventry BJ, Roder DM. Increased Early Cancer Diagnosis: Unveiling Immune-Cancer Biology to Explain Clinical "Overdiagnosis". Cancers (Basel) 2023; 15:cancers15041139. [PMID: 36831482 PMCID: PMC9953985 DOI: 10.3390/cancers15041139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/28/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023] Open
Abstract
Even though clinically small 'early' cancers represent many millions of cells biologically, when removed surgically, these often never recur or regrow, nor reduce the individual's lifespan. However, some early cancers remain quiescent and indolent; while others grow and metastasize, threatening life. Distinguishing between these different clinical behaviours using clinical/pathological criteria is currently problematic. It is reported that many suspicious lesions and early cancers are being removed surgically that would not threaten the patient's life. This has been termed 'overdiagnosis', especially in the sphere of cancer screening. Although a controversial and emotive topic, it poses clinical and public health policy challenges. The diagnostic differentiation between 'non-lethal' and 'lethal' tumor forms is generally impossible. One perspective gathering evidential support is that a dynamic balance exists between the immune response and malignant processes governing 'lethality', where many more cancers are produced than become clinically significant due to the immune system preventing their progression. Higher medical screening "diagnosis" rates may reflect lead-time effects, with more 'non-progressing' cancers detected when an early immune-cancer interaction is occurring. We present a model for this immune-cancer interaction and review 'excess' or 'overdiagnosis' claims that accompany increasingly sensitive diagnostic and screening technologies. We consider that immune tools should be incorporated into future research, with potential for immune system modulation for some early cancers.
Collapse
Affiliation(s)
- Bruce A. Wauchope
- Molechecks Australia, 1284 South Road, Clovelly Park 5042, Australia
- Discipline of Surgery, Cancer Immunotherapy Laboratory, University of Adelaide, Royal Adelaide Hospital, Adelaide 5005, Australia
| | - Brendon J. Coventry
- Discipline of Surgery, Cancer Immunotherapy Laboratory, University of Adelaide, Royal Adelaide Hospital, Adelaide 5005, Australia
- Correspondence:
| | - David M. Roder
- Cancer Epidemiology and Population Health, Allied Health and Human Performance, University of South Australia, Adelaide 5000, Australia
| |
Collapse
|
18
|
Saudi A, Banday V, Zirakzadeh AA, Selinger M, Forsberg J, Holmbom M, Henriksson J, Waldén M, Alamdari F, Aljabery F, Winqvist O, Sherif A. Immune-Activated B Cells Are Dominant in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15030920. [PMID: 36765877 PMCID: PMC9913271 DOI: 10.3390/cancers15030920] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/22/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
B cells are multifaceted immune cells responding robustly during immune surveillance against tumor antigens by presentation to T cells and switched immunoglobulin production. However, B cells are unstudied in prostate cancer (PCa). We used flow cytometry to analyze B-cell subpopulations in peripheral blood and lymph nodes from intermediate-high risk PCa patients. B-cell subpopulations were related to clinicopathological factors. B-cell-receptor single-cell sequencing and VDJ analysis identified clonal B-cell expansion in blood and lymph nodes. Pathological staging was pT2 in 16%, pT3a in 48%, and pT3b in 36%. Lymph node metastases occurred in 5/25 patients (20%). Compared to healthy donors, the peripheral blood CD19+ B-cell compartment was significantly decreased in PCa patients and dominated by naïve B cells. The nodal B-cell compartment had significantly increased fractions of CD19+ B cells and switched memory B cells. Plasmablasts were observed in tumor-draining sentinel lymph nodes (SNs). VDJ analysis revealed clonal expansion in lymph nodes. Thus, activated B cells are increased in SNs from PCa patients. The increased fraction of switched memory cells and plasmablasts together with the presence of clonally expanded B cells indicate tumor-specific T-cell-dependent responses from B cells, supporting an important role for B cells in the protection against tumors.
Collapse
Affiliation(s)
- Aws Saudi
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
- Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Viqar Banday
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umea University, 901 85 Umea, Sweden
- Department of Clinical Microbiology, Immunology, Umea University, 901 85 Umeå, Sweden
| | | | - Martin Selinger
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), 901 87 Umeå, Sweden
- Department of Molecular Biology, Umeå Centre for Microbial Research, 6K and 6L, Umeå University, 901 87 Umeå, Sweden
| | - Jon Forsberg
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Martin Holmbom
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Johan Henriksson
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), 901 87 Umeå, Sweden
- Department of Molecular Biology, Umeå Centre for Microbial Research, 6K and 6L, Umeå University, 901 87 Umeå, Sweden
| | - Mauritz Waldén
- Department of Urology, Central Hospital of Karlstad, 652 30 Karlstad, Sweden
| | - Farhood Alamdari
- Department of Urology, Västmanland Hospital, 721 89 Västerås, Sweden
| | - Firas Aljabery
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
- Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Ola Winqvist
- ABClabs, BioClinicum, Campus Solna, 171 76 Stockholm, Sweden
| | - Amir Sherif
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umea University, 901 85 Umea, Sweden
- Correspondence:
| |
Collapse
|
19
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
20
|
Drouillard D, Craig BT, Dwinell MB. Physiology of chemokines in the cancer microenvironment. Am J Physiol Cell Physiol 2023; 324:C167-C182. [PMID: 36317799 PMCID: PMC9829481 DOI: 10.1152/ajpcell.00151.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 01/07/2023]
Abstract
Chemokines are chemotactic cytokines whose canonical functions govern movement of receptor-expressing cells along chemical gradients. Chemokines are a physiological system that is finely tuned by ligand and receptor expression, ligand or receptor oligomerization, redundancy, expression of atypical receptors, and non-GPCR binding partners that cumulatively influence discrete pharmacological signaling responses and cellular functions. In cancer, chemokines play paradoxical roles in both the directed emigration of metastatic, receptor-expressing cancer cells out of the tumor as well as immigration of tumor-infiltrating immune cells that culminate in a tumor-unique immune microenvironment. In the age of precision oncology, strategies to effectively harness the power of immunotherapy requires consideration of chemokine gradients within the unique spatial topography and temporal influences with heterogeneous tumors. In this article, we review current literature on the diversity of chemokine ligands and their cellular receptors that detect and process chemotactic gradients and illustrate how differences between ligand recognition and receptor activation influence the signaling machinery that drives cellular movement into and out of the tumor microenvironment. Facets of chemokine physiology across discrete cancer immune phenotypes are contrasted to existing chemokine-centered therapies in cancer.
Collapse
Affiliation(s)
- Donovan Drouillard
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Brian T Craig
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael B Dwinell
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Center for Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
21
|
Cao H, Gao S, Jogani R, Sugimura R. The Tumor Microenvironment Reprograms Immune Cells. Cell Reprogram 2022; 24:343-352. [PMID: 36301256 DOI: 10.1089/cell.2022.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Tumor tissue comprises a highly complex network of diverse cell types. The tumor microenvironment (TME) can be mainly subdivided into cancer cells and stromal cell compartments, the latter include different types of immune cells, fibroblasts, endothelial cells, and pericytes. Tumor cells reprogram immune cells and other stromal cells in the TME to constrain their antitumor capacity by creating an immunosuppressive milieu and metabolism competition. Moreover, the reprogramming effect on immune cells is localized not only in the tumor but also at the systemic level. With wide application of single-cell sequencing technology, tumor-specific characteristics of immune cells and other stromal cells in the TME have been dissected. In this review, we mainly focus on how tumor cells reprogram immune cells both within the TME and peripheral blood. This information can further help us to improve the efficiency of current immunotherapy as well as bring up new ideas to combat cancer.
Collapse
Affiliation(s)
- Handi Cao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,Centre for Translational Stem Cell Biology, Science Park, Hong Kong
| | - Sanxing Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Ritika Jogani
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,Centre for Translational Stem Cell Biology, Science Park, Hong Kong
| |
Collapse
|
22
|
Anti-mesothelin immunotoxin induces mesothelioma eradication, anti-tumor immunity, and the development of tertiary lymphoid structures. Proc Natl Acad Sci U S A 2022; 119:e2214928119. [PMID: 36409889 PMCID: PMC9860319 DOI: 10.1073/pnas.2214928119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
LMB-100 is a recombinant immunotoxin composed of a Fab linked to a toxin. It kills cells expressing human mesothelin (hMSLN), which is highly expressed on the surface of mesothelioma and many other cancer cells. Clinically, we observed some patients had delayed responses to an anti-hMSLN immunotoxin treatment, suggesting the induction of anti-tumor immunity. We aimed to develop a mouse model to investigate whether immunotoxin alone can induce anti-tumor immunity and to study the mechanism of this immunity. An immunocompetent transgenic mouse was used to grow mouse mesothelioma AB1 cells expressing hMSLN in the peritoneal cavity. Mice were treated with LMB-100, and mice with complete responses (CRs) were rechallenged with tumor cells to determine whether anti-tumor immunity developed. Changes in gene expression profiles were evaluated by Nanostring, and changes in cytokines and chemokines were checked by protein arrays. The distribution of various immune cells was assessed by immunohistochemistry. Our results show that the mice with tumor reached CRs and developed anti-tumor immunity after LMB-100 treatment alone. The primary response requires CD8+ T cells, CD4+ T cells, and B cells. Transcriptional profiling shows that LMB-100 treatment reshapes the tumor immune microenvironment by upregulating chemotaxis signals. LMB-100 treatment upregulates genes associated with tertiary lymphoid structures (TLS) development and induces TLS formation in tumors. In sum, immunotoxin-mediated cell death induces anti-tumor immunity and the development of TLS, which provides insights into how immunotoxins cause tumor regressions.
Collapse
|
23
|
Zhu C, Wang S, Du Y, Dai Y, Huai Q, Li X, Du Y, Dai H, Yuan W, Yin S, Wang H. Tumor microenvironment-related gene selenium-binding protein 1 (SELENBP1) is associated with immunotherapy efficacy and survival in colorectal cancer. BMC Gastroenterol 2022; 22:437. [PMID: 36253721 PMCID: PMC9575293 DOI: 10.1186/s12876-022-02532-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 10/07/2022] [Indexed: 11/29/2022] Open
Abstract
Background Selenium-binding protein 1 (SELENBP1), a member of the selenium-containing protein family, plays an important role in malignant tumorigenesis and progression. However, it is currently lacking research about relationship between SELENBP1 and immunotherapy in colorectal cancer (CRC). Methods We first analyzed the expression levels of SELENBP1 based on the Cancer Genome Atlas (TCGA), Oncomine andUALCAN. Chisq.test, Fisher.test, Wilcoxon-Mann-Whitney test and logistic regression were used to analyze the relationship of clinical characteristics with SELENBP1 expression. Then Gene ontology/ Kyoto encyclopedia of genes and genomes (GO/KEGG), Gene set enrichment analysis (GSEA) enrichment analysis to clarify bio-processes and signaling pathways. The cBioPortal was used to perform analysis of mutation sites, types, etc. of SELENBP1. In addition, the correlation of SELENBP1 gene with tumor immune infiltration and prognosis was analyzed using ssGSEA, ESTIMATE, tumor immune dysfunction and rejection (TIDE) algorithm and Kaplan-Meier (KM) Plotter database. Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were used to validate the expression of SELENBP1 in CRC samples and matched normal tissues. Immunohistochemistry (IHC) was further performed to detect the expression of SELENBP1 in CRC samples and matched normal tissues. Results We found that SELENBP1 expression was lower in CRC compared to normal colorectal tissue and was associated with poor prognosis. The aggressiveness of CRC increased with decreased SELENBP1 expression. Enrichment analysis showed that the SELENBP1 gene was significantly enriched in several pathways, such as programmed death 1 (PD-1) signaling, signaling by interleukins, TCR signaling, collagen degradation, costimulation by the CD28 family. Decreased expression of SELENBP1 was associated with DNA methylation and mutation. Immune infiltration analysis identified that SELENBP1 expression was closely related to various immune cells and immune chemokines/receptors. With increasing SELENBP1 expression, immune and stromal components in the tumor microenvironment were significantly decreased. SELENBP1 expression in CRC patients affects patient prognosis by influencing tumor immune infiltration. Beside this, SELENBP1 expression is closely related to the sensitivity of chemotherapy and immunotherapy. Conclusions Survival analysis as well as enrichment and immunoassay results suggest that SELENBP1 can be considered as a promising prognostic biomarker for CRC. SELENBP1 expression is closely associated with immune infiltration and immunotherapy. Collectively, our study provided useful information on the oncogenic role of SELENBP1, contributing to further exploring the underlying mechanisms. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02532-2.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Siya Wang
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230001, China
| | - Yishan Du
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230001, China
| | - Ying Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Qian Huai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Xiaolei Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Yingying Du
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Hanren Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China
| | - Wenkang Yuan
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230036, China
| | - Shi Yin
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230001, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230036, China.
| |
Collapse
|
24
|
Viveiros N, Flores BC, Lobo J, Martins-Lima C, Cantante M, Lopes P, Deantonio C, Palu C, Sainson RC, Henrique R, Jerónimo C. Detailed bladder cancer immunoprofiling reveals new clues for immunotherapeutic strategies. Clin Transl Immunology 2022; 11:e1402. [PMID: 36092481 PMCID: PMC9440624 DOI: 10.1002/cti2.1402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/22/2022] [Accepted: 06/17/2022] [Indexed: 11/09/2022] Open
Abstract
Objectives Bladder cancer (BlCa) is the tenth most frequent malignancy worldwide and the costliest to treat and monitor. Muscle-invasive BlCa (MIBC) has a dismal prognosis, entailing the need for alternative therapies for the standard radical cystectomy. Checkpoint blockade immunotherapy has been approved for high-grade non-muscle-invasive BlCa (HG NMIBC) and metastatic disease, but its effectiveness in localised MIBC remains under scrutiny. Herein, we sought to characterise and compare the immune infiltrate of HG NMIBC and MIBC samples, including ICOS expression, a targetable immune checkpoint associated with regulatory T cell(Tregs)-mediated immunosuppression. Methods Immunohistochemistry for CD83, CD20, CD68, CD163, CD3, CD8, CD4, FoxP3/ICOS and PD-L1 was performed in HG NMIBC and MIBC samples (n = 206), and positive staining was quantified in the peritumoral and/or intratumoral tissue compartments with QuPath imaging software. Results CD20+ B cells, CD68+ and CD163+ tumor-associated macrophages were significantly increased in MIBCs and associated with poor prognosis. In turn, higher infiltration of T cells was associated with prolonged survival, with exception of the CD4+ helper subset. Intratumoral expression of CD3 and CD8 was independent prognostic factors for increased disease-free survival (DFS) in multivariable analysis. Remarkably, Tregs (FoxP3+/FoxP3+ICOS+) were found differentially distributed between tissue compartments. PD-L1 immunoexpression independently predicted a shorter DFS and associated with higher infiltration of FoxP3+ICOS+ Tregs. Conclusions Immune infiltrates of HG NMIBC and MIBC display significant differences that may help selecting patients for immunotherapies. Considering ICOS immunoexpression results, it might constitute a relevant therapeutic target, eventually in combination with anti-PD-1/PD-L1 therapies, for certain BlCa patient subsets.
Collapse
Affiliation(s)
- Nicole Viveiros
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal
| | - Bianca Ct Flores
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal.,Department of Pathology Portuguese Oncology Institute of Porto (IPO Porto) Porto Portugal.,Department of Pathology and Molecular Immunology School of Medicine and Biomedical Sciences- University of Porto (ICBAS-UP) Porto Portugal
| | - Cláudia Martins-Lima
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal.,Department of Precision Medicine University of Campania "Luigi Vanvitelli" Naples Italy
| | - Mariana Cantante
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal.,Department of Pathology Portuguese Oncology Institute of Porto (IPO Porto) Porto Portugal
| | - Paula Lopes
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal.,Department of Pathology Portuguese Oncology Institute of Porto (IPO Porto) Porto Portugal
| | | | | | | | - Rui Henrique
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal.,Department of Pathology Portuguese Oncology Institute of Porto (IPO Porto) Porto Portugal.,Department of Pathology and Molecular Immunology School of Medicine and Biomedical Sciences- University of Porto (ICBAS-UP) Porto Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC) Porto Portugal.,Department of Pathology and Molecular Immunology School of Medicine and Biomedical Sciences- University of Porto (ICBAS-UP) Porto Portugal
| |
Collapse
|
25
|
A Mathematical Study of the Role of tBregs in Breast Cancer. Bull Math Biol 2022; 84:112. [PMID: 36048369 DOI: 10.1007/s11538-022-01054-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/12/2022] [Indexed: 12/24/2022]
Abstract
A model for the mathematical study of immune response to breast cancer is proposed and studied, both analytically and numerically. It is a simplification of a complex one, recently introduced by two of the present authors. It serves for a compact study of the dynamical role in cancer promotion of a relatively recently described subgroup of regulatory B cells, which are evoked by the tumour.
Collapse
|
26
|
Li Q, Yu J, Zhang H, Meng Y, Liu YF, Jiang H, Zhu M, Li N, Zhou J, Liu F, Fang X, Li J, Feng X, Lu J, Shao C, Bian Y. Prediction of Tumor-Infiltrating CD20 + B-Cells in Patients with Pancreatic Ductal Adenocarcinoma Using a Multilayer Perceptron Network Classifier Based on Non-contrast MRI. Acad Radiol 2022; 29:e167-e177. [PMID: 34922828 DOI: 10.1016/j.acra.2021.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 11/01/2022]
Abstract
RATIONALE AND OBJECTIVES Conventional chemotherapy has limited benefit in pancreatic ductal adenocarcinoma (PDAC), necessitating identification of novel therapeutic targets. Radiomics may enable non-invasive prediction of CD20 expression, a hypothesized therapeutic target in PDAC. To develop a machine learning classifier based on noncontrast magnetic resonance imaging for predicting CD20 expression in PDAC. MATERIALS AND METHODS Retrospective study was conducted on preoperative noncontrast magnetic resonance imaging of 156 patients with pathologically confirmed PDAC from January 2017 to April 2018. For each patient, 1409 radiomics features were selected using minimum absolute contraction and selective operator logistic regression algorithms. CD20 expression was quantified using immunohistochemistry. A multilayer perceptron network classifier was developed using the training and validation set. RESULTS A log-rank test showed that the CD20-high group (22.37 months, 95% CI: 19.10-25.65) had significantly longer survival than the CD20-low group (14.9 months, 95% CI: 10.96-18.84). The predictive model showed good differentiation in training (area under the curve [AUC], 0.79) and validation (AUC, 0.79) sets. Sensitivity, specificity, accuracy, positive predictive value, and negative predictive value were 73.21%, 75.47%, 0.74, 0.76, and 0.73, respectively, for the training set and 69.23%, 80.95%, 0.74, 0.82, and 0.68, respectively, for the validation set. CONCLUSION Multilayer perceptron classifier based on noncontrast magnetic resonance imaging scanning can predict the level of CD20 expression in PDAC patients.
Collapse
Affiliation(s)
- Qi Li
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yinghao Meng
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yan Fang Liu
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mengmeng Zhu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Na Li
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jian Zhou
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xiaochen Feng
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yun Bian
- Department of Radiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
27
|
Masaoutis C, Palamaris K, Kokkali S, Levidou G, Theocharis S. Unraveling the Immune Microenvironment of Thymic Epithelial Tumors: Implications for Autoimmunity and Treatment. Int J Mol Sci 2022; 23:ijms23147864. [PMID: 35887212 PMCID: PMC9323059 DOI: 10.3390/ijms23147864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Thymic Epithelial Tumors (TETs) represent a rare tumor family, originating from the epithelial component of the thymus gland. Clinicopathologically, they are segregated into six major subtypes, associated with distinct histological features and clinical outcomes. Their emergence and evolution are accompanied by the generation of a complex tumor microenvironment (TME), dominated by phenotypically and functionally divergent immune cellular subsets, in different maturation states and in analogies that vary significantly among different subtypes. These heterogenous leukocyte populations exert either immune-permissive and tumor-suppressive functions or vice versa, and the dynamic equilibrium established among them either dictates the tumor immune milieu towards an immune-tolerance state or enables the development of a productive spontaneous tumoricidal response. The immunologically “hot” microenvironment, defining a significant proportion of TETs, makes them a promising candidate for the implementation of immune checkpoint inhibitors (ICIs). A number of phase I and II clinical trials have already demonstrated significant, type-specific clinical efficacy of PD-L1 inhibitors, even though substantial limitations in their utilization derive from their immune-mediated adverse effects. Moreover, the completed clinical studies involved relatively restricted patient samples and an expansion in the enrolled cohorts is required, so that more trustworthy conclusions regarding the benefit from ICIs in TETs can be extracted.
Collapse
Affiliation(s)
- Christos Masaoutis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Str., Bld 10, Goudi, GR11527 Athens, Greece; (C.M.); (K.P.); (G.L.)
| | - Kostas Palamaris
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Str., Bld 10, Goudi, GR11527 Athens, Greece; (C.M.); (K.P.); (G.L.)
| | - Stefania Kokkali
- Oncology Unit, 2nd Department of Medicine, Medical School, National and Kapodistrian University of Athens, Hippocratio General Hospital of Athens, 114, V. Sofias Str., GR11527 Athens, Greece;
| | - Georgia Levidou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Str., Bld 10, Goudi, GR11527 Athens, Greece; (C.M.); (K.P.); (G.L.)
- Second Department of Pathology, Paracelsus Medical University, 90419 Nurenberg, Germany
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Str., Bld 10, Goudi, GR11527 Athens, Greece; (C.M.); (K.P.); (G.L.)
- Correspondence:
| |
Collapse
|
28
|
Chen S, Xie P, Cowan M, Huang H, Cardenas H, Keathley R, Tanner EJ, Fleming GF, Moroney JW, Pant A, Akasha AM, Davuluri RV, Kocherginsky M, Zhang B, Matei D. Epigenetic priming enhances antitumor immunity in platinum-resistant ovarian cancer. J Clin Invest 2022; 132:e158800. [PMID: 35671108 PMCID: PMC9282926 DOI: 10.1172/jci158800] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022] Open
Abstract
BackgroundImmune checkpoint inhibitors (ICIs) have modest activity in ovarian cancer (OC). To augment their activity, we used priming with the hypomethylating agent guadecitabine in a phase II study.MethodsEligible patients had platinum-resistant OC, normal organ function, measurable disease, and received up to 5 prior regimens. The treatment included guadecitabine (30 mg/m2) on days 1-4, and pembrolizumab (200 mg i.v.) on day 5, every 21 days. The primary endpoint was the response rate. Tumor biopsies, plasma, and PBMCs were obtained at baseline and after treatment.ResultsAmong 35 evaluable patients, 3 patients had partial responses (8.6%), and 8 (22.9%) patients had stable disease, resulting in a clinical benefit rate of 31.4% (95% CI: 16.9%-49.3%). The median duration of clinical benefit was 6.8 months. Long-interspersed element 1 (LINE1) was hypomethylated in post-treatment PBMCs, and methylomic and transcriptomic analyses showed activation of antitumor immunity in post-treatment biopsies. High-dimensional immune profiling of PBMCs showed a higher frequency of naive and/or central memory CD4+ T cells and of classical monocytes in patients with a durable clinical benefit or response (CBR). A higher baseline density of CD8+ T cells and CD20+ B cells and the presence of tertiary lymphoid structures in tumors were associated with a durable CBR.ConclusionEpigenetic priming using a hypomethylating agent with an ICI was feasible and resulted in a durable clinical benefit associated with immune responses in selected patients with recurrent OC.Trial registrationClinicalTrials.gov NCT02901899.FundingUS Army Medical Research and Material Command/Congressionally Directed Medical Research Programs (USAMRMC/CDMRP) grant W81XWH-17-0141; the Diana Princess of Wales Endowed Professorship and LCCTRAC funds from the Robert H. Lurie Comprehensive Cancer Center; Walter S. and Lucienne Driskill Immunotherapy Research funds; Astex Pharmaceuticals; Merck & Co.; National Cancer Institute (NCI), NIH grants CCSG P30 CA060553, CCSG P30 CA060553, and CA060553.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Medicine, Hematology/Oncology Division
| | - Ping Xie
- Department of Medicine, Hematology/Oncology Division
| | | | - Hao Huang
- Department of Obstetrics and Gynecology
| | | | - Russell Keathley
- Department of Obstetrics and Gynecology
- Driskill Graduate Training Program in Life Sciences, and
| | - Edward J. Tanner
- Department of Obstetrics and Gynecology
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gini F. Fleming
- Department of Medicine, Hematology/Oncology Division, University of Chicago, Chicago, Illinois, USA
| | - John W. Moroney
- Department of Medicine, Hematology/Oncology Division, University of Chicago, Chicago, Illinois, USA
| | - Alok Pant
- Northwestern Medicine, Lake Forest Hospital, Lake Forest, Illinois, USA
| | - Azza M. Akasha
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ramana V. Davuluri
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, USA
| | - Masha Kocherginsky
- Department of Obstetrics and Gynecology
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Division of Biostatistics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Bin Zhang
- Department of Medicine, Hematology/Oncology Division
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Jesse Brown VA Medical Center, Chicago, Illinois, USA
| |
Collapse
|
29
|
Raj SKS, Routh ED, Chou JW, Votanopoulos KI, Triozzi PL, Miller LD. Prognostic attributes of immune signatures in soft tissue sarcomas show differential dependencies on tumor mutational burden. Cancer 2022; 128:3254-3264. [PMID: 35767280 PMCID: PMC9544607 DOI: 10.1002/cncr.34333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/14/2022] [Accepted: 04/05/2022] [Indexed: 11/11/2022]
Abstract
Background Cellular and intrinsic markers of sarcoma immunogenicity are poorly understood. To gain insight into whether tumor–immune interactions correlate with clinical aggressiveness, the authors examined the prognostic significance of immune gene signatures in combination with tumor mutational burden (TMB) and cancer–testis antigen (CTA) expression. Methods RNA sequencing and clinical data of 259 soft tissue sarcomas from The Cancer Genome Atlas project were used to investigate associations between published immune gene signatures and patient overall survival (OS) in the contexts of TMB, as computed from whole‐exome sequencing data, and CTA gene expression. Multivariate Cox proportional hazards regression models and log‐rank tests were used to assess survival associations. Results Immune signature scores that reflected in part the intratumoral abundance of cytotoxic T cells showed significant positive associations with OS. However, the prognostic power of the T‐cell signatures was highly dependent on TMB‐high status, consistent with protective effects of tumor‐infiltrating T cells in tumors with elevated antigenicity. In TMB‐low tumors, a signature of infiltrating plasma B cells was significantly and positively associated with OS, independent of T‐cell signature status. Although tumor subtypes based on differential expression patterns of CTA genes showed different survival associations within leiomyosarcoma and myxofibrosarcoma histologies, neither CTA nor histologic subtype interacted with the T‐cell–survival association. Conclusions Signatures of T‐cell and plasma B‐cell infiltrates were associated with a survival benefit in soft tissue sarcomas. TMB, but not CTA expression, influenced the prognostic power of T‐cell–associated, but not plasma B‐cell–associated, survival. Lay summary Clinical data and RNA analysis of 259 soft tissue sarcomas from The Cancer Genome Atlas project were used to investigate associations between five published gene immune cell expression signatures and survival in the context of tumor mutations. Activated T cells had a significant positive association with patient survival. Although high tumor mutation burden was associated with good survival, the prognostic power of T‐cell signatures was highly dependent on tumor mutational status, consistent with protective effects of tumor‐infiltrating T cells in tumors with high levels of antigens. In low tumor mutation‐bearing tumors, plasma B cells were positively associated with survival.
Signatures of T‐cell and plasma B‐cell infiltrates are associated with survival benefit in soft tissue sarcomas, and tumor mutational burden (TMB), but not cancer–testis antigen expression, is a tumor‐intrinsic determinant of T‐cell–associated survival, but not plasma B‐cell–associated survival. Although high TMB trended toward an association with good survival, the prognostic power of the T‐cell signatures was highly dependent on TMB‐high status, consistent with the protective effects of tumor‐infiltrating T cells in tumors with elevated antigenicity; however, in TMB‐low tumors, a signature of infiltrating plasma B cells was positively associated with overall survival, independent of T‐cell signature status.
Collapse
Affiliation(s)
- Shailaja K S Raj
- Section of Hematology and Oncology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston Salem, North Carolina, USA
| | - Eric D Routh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jeff W Chou
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Konstantinos I Votanopoulos
- Department of Surgery, Division of Surgical Oncology, Wake Forest Baptist Medical Center, Winston Salem, North Carolina, USA.,Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, North Carolina, USA
| | - Pierre L Triozzi
- Section of Hematology and Oncology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston Salem, North Carolina, USA.,Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, North Carolina, USA
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA.,Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, North Carolina, USA
| |
Collapse
|
30
|
Wang J, Huang F, Zhao J, Huang P, Tan J, Huang M, Ma R, Xiao Y, He S, Wang Z, Shen J, Lu H, Meng L. Tumor-Infiltrated CD8+ T Cell 10-Gene Signature Related to Clear Cell Renal Cell Carcinoma Prognosis. Front Immunol 2022; 13:930921. [PMID: 35812454 PMCID: PMC9263606 DOI: 10.3389/fimmu.2022.930921] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) usually affects multiple organs (e.g., bone and brain), and patient prognosis is usually poor. Although it is known that CD8+ T cell infiltration can potentially alleviate ccRCC progression, few studies have concentrated on the correlation between CD8+ T cell infiltration and ccRCC prognosis. In this study, ten genes expressed by infiltrated CD8+ T cells (i.e., AMD1, CCSER2, CIB1, DRAP1, HMGB2, HMGN1, NPIPB5, PTP4A2, RORA, and SAP18) were suggested as potential ccRCC prognostic biomarkers, by using next-generation sequencing (i.e. bulk sequencing and single-cell sequencing) of ccRCC, papillary renal cell carcinoma (papRCC), and control kidney biopsies. Specifically, we identified four genes (i.e., CCSER2, DRAP1, NPIPB5, and SAP18) as potential novel prognostic biomarkers for ccRCC. It is noteworthy that SAP18 derived from CD8+ T cells negatively correlates to Atg7+ neutrophils in ccRCC, compared with papRCC, indicating a potential decreased neutrophil metabolic function in autophagy and fatty acids. This study elucidated the protective role of infiltrated CD8+ T cells in ccRCC and identified ten candidate genes related to an improved prognosis in patients with ccRCC.
Collapse
Affiliation(s)
- Jie Wang
- Center for Systemic Inflammation Research (CSIR), Youjiang Medical University for Nationalities, Baise, China
- Department of Kidney Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Feifan Huang
- Department of Kidney Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Jingjie Zhao
- Life Science and Clinical Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Peng Huang
- Department of Kidney Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Junhua Tan
- Department of Kidney Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Meiying Huang
- Department of Kidney Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Ruiying Ma
- Department of Kidney Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yu Xiao
- Department of Kidney Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Siyuan He
- Center for Systemic Inflammation Research (CSIR), Youjiang Medical University for Nationalities, Baise, China
| | - Zechen Wang
- Center for Systemic Inflammation Research (CSIR), Youjiang Medical University for Nationalities, Baise, China
| | - Jiajia Shen
- Center for Systemic Inflammation Research (CSIR), Youjiang Medical University for Nationalities, Baise, China
| | - Heming Lu
- Department of Radiation Oncology, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- *Correspondence: Lingzhang Meng, ; Heming Lu,
| | - Lingzhang Meng
- Center for Systemic Inflammation Research (CSIR), Youjiang Medical University for Nationalities, Baise, China
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China
- *Correspondence: Lingzhang Meng, ; Heming Lu,
| |
Collapse
|
31
|
Sagiv-Barfi I, Czerwinski DK, Shree T, Lohmeyer JJK, Levy R. Intratumoral immunotherapy relies on B and T cell collaboration. Sci Immunol 2022; 7:eabn5859. [PMID: 35622903 DOI: 10.1126/sciimmunol.abn5859] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Antitumor T cell responses are the primary mediators of cancer immunotherapy. However, many other components of the immune system are needed for efficient T cell responses to be generated. Here, we developed a combinatorial approach where a Toll-like receptor 9 agonist (CpG) and Fc-fused IL-12 protein were injected together into just one of several tumor sites in a mouse. This combination led to body-wide (abscopal) therapeutic responses in multiple cancer models. These systemic responses were dependent not only on T cells but also on B cells. B cells were activated by the treatment and were required for optimal T cell activation. This cross-talk was dependent on MHC and was tumor antigen specific. The addition of an agonistic antibody against OX40 further enhanced T cell activation and therapeutic responses. Our data suggest that the combination of CpG, anti-OX40, and IL-12Fc may have success in patients with cancer and that B and T cell collaboration is crucial for the efficacy of this combination immunotherapy.
Collapse
Affiliation(s)
- Idit Sagiv-Barfi
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Debra K Czerwinski
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Tanaya Shree
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Julian J K Lohmeyer
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ronald Levy
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
32
|
Lou Y, Shi Q, Zhang Y, Qi Y, Zhang W, Wang H, Lu J, Han B, Zhong H. Multi-Omics Signatures Identification for LUAD Prognosis Prediction Model Based on the Integrative Analysis of Immune and Hypoxia Signals. Front Cell Dev Biol 2022; 10:840466. [PMID: 35359451 PMCID: PMC8960258 DOI: 10.3389/fcell.2022.840466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/21/2022] [Indexed: 11/21/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common histological subtype of lung cancer with heterogeneous outcomes and diverse therapeutic responses. However, the understanding of the potential mechanism behind LUAD initiation and progression remains limited. Increasing evidence shows the clinical significance of the interaction between immune and hypoxia in tumor microenvironment. To mine reliable prognostic signatures related to both immune and hypoxia and provide a more comprehensive landscape of the hypoxia-immune genome map, we investigated the hypoxia-immune-related alteration at the multi-omics level (gene expression, somatic mutation, and DNA methylation). Multiple strategies including lasso regression and multivariate Cox proportional hazards regression were used to screen the signatures with clinical significance and establish an incorporated prognosis prediction model with robust discriminative power on survival status on both the training and test datasets. Finally, combing all the samples, we constructed a robust model comprising 19 signatures for the prognosis prediction of LUAD patients. The results of our study provide a comprehensive landscape of hypoxia-immune related genetic alterations and provide a robust prognosis predictor for LUAD patients.
Collapse
Affiliation(s)
- Yuqing Lou
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Shi
- Department of Oncology, Baoshan Branch of Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanwei Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Qi
- School of Basic Medical Science, Hangzhou Normal University, Hangzhou, China
| | - Wei Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huimin Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Lu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Translational Medical Research Platform for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Bio-bank, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jun Lu, ; Baohui Han, ; Hua Zhong,
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Translational Medical Research Platform for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jun Lu, ; Baohui Han, ; Hua Zhong,
| | - Hua Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Translational Medical Research Platform for Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jun Lu, ; Baohui Han, ; Hua Zhong,
| |
Collapse
|
33
|
Bitsouni V, Tsilidis V. Mathematical modeling of tumor-immune system interactions: the effect of rituximab on breast cancer immune response. J Theor Biol 2022; 539:111001. [PMID: 34998860 DOI: 10.1016/j.jtbi.2021.111001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/25/2021] [Indexed: 12/11/2022]
Abstract
tBregs are a newly discovered subcategory of B regulatory cells, which are generated by breast cancer, resulting in the increase of Tregs and therefore in the death of NK cells. In this study, we use a mathematical and computational approach to investigate the complex interactions between the aforementioned cells as well as CD8+ T cells, CD4+ T cells and B cells. Furthermore, we use data fitting to prove that the functional response regarding the lysis of breast cancer cells by NK cells has a ratio-dependent form. Additionally, we include in our model the concentration of rituximab - a monoclonal antibody that has been suggested as a potential breast cancer therapy - and test its effect, when the standard, as well as experimental dosages, are administered.
Collapse
Affiliation(s)
- Vasiliki Bitsouni
- Department of Mathematics, National and Kapodistrian University of Athens, Panepistimioupolis, GR-15784 Athens, Greece; School of Science and Technology, Hellenic Open University, 18 Parodos Aristotelous Str., GR-26335 Patras, Greece.
| | - Vasilis Tsilidis
- School of Science and Technology, Hellenic Open University, 18 Parodos Aristotelous Str., GR-26335 Patras, Greece.
| |
Collapse
|
34
|
Wang W, Ye LF, Bao H, Hu MT, Han M, Tang HM, Ren C, Wu X, Shao Y, Wang FH, Zhou ZW, Li YH, Xu RH, Wang DS. Heterogeneity and evolution of tumour immune microenvironment in metastatic gastroesophageal adenocarcinoma. Gastric Cancer 2022; 25:1017-1030. [PMID: 35904677 PMCID: PMC9587966 DOI: 10.1007/s10120-022-01324-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 07/16/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tumour immune microenvironment heterogeneity is prevalent in numerous cancers and can negatively impact immunotherapy response. Immune heterogeneity and evolution in gastroesophageal adenocarcinoma (GEA) have not been studied in the past. METHODS Together with a multi-region sampling of normal, primary and metastatic tissues, we performed whole exome sequencing, TCR sequencing as well as immune cell infiltration estimation through deconvolution of gene expression signals. RESULTS We discovered high TCR repertoire and immune cell infiltration heterogeneity among metastatic sites, while they were homogeneous among primary and normal samples. Metastatic sites shared high levels of abundant TCR clonotypes with blood, indicating immune surveillance via blood. Metastatic sites also had low levels of tumour-eliminating immune cells and were undergoing heavy immunomodulation compared to normal and primary tumour tissues. There was co-evolution of neo-antigen and TCR repertoire, but only in patients with late diverging mutational evolution. Co-evolution of TCR repertoire and immune cell infiltration was seen in all except one patient. CONCLUSIONS Our findings revealed immune heterogeneity and co-evolution in GEA, which may inform immunotherapy decision-making.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Liu-Fang Ye
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060 People’s Republic of China ,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong feng, East Road, Guangzhou, 510060 People’s Republic of China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu China
| | - Ming-Tao Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060 People’s Republic of China ,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong feng, East Road, Guangzhou, 510060 People’s Republic of China
| | - Ming Han
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu China
| | - Hai-Meng Tang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu China
| | - Chao Ren
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060 People’s Republic of China ,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong feng, East Road, Guangzhou, 510060 People’s Republic of China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu China ,School of Public Health, Nanjing Medical University, Nanjing, China
| | - Feng-Hua Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060 People’s Republic of China ,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong feng, East Road, Guangzhou, 510060 People’s Republic of China
| | - Zhi-Wei Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Department of Gastric Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 People’s Republic of China
| | - Yu-Hong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060 People’s Republic of China ,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong feng, East Road, Guangzhou, 510060 People’s Republic of China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060 People’s Republic of China ,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong feng, East Road, Guangzhou, 510060 People’s Republic of China
| | - De-Shen Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060 People’s Republic of China ,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060 People’s Republic of China ,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, 651 Dong feng, East Road, Guangzhou, 510060 People’s Republic of China
| |
Collapse
|
35
|
Aggarwal V, Rathod S, Vashishth K, Upadhyay A. Immune Cell Metabolites as Fuel for Cancer Cells. IMMUNO-ONCOLOGY CROSSTALK AND METABOLISM 2022:153-186. [DOI: 10.1007/978-981-16-6226-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Kim MJ, Ha SJ. Differential Role of PD-1 Expressed by Various Immune and Tumor Cells in the Tumor Immune Microenvironment: Expression, Function, Therapeutic Efficacy, and Resistance to Cancer Immunotherapy. Front Cell Dev Biol 2021; 9:767466. [PMID: 34901012 PMCID: PMC8662983 DOI: 10.3389/fcell.2021.767466] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/08/2021] [Indexed: 12/19/2022] Open
Abstract
In the tumor immune microenvironment (TIME), tumor cells interact with various cells and operate various strategies to avoid antitumor immune responses. These immune escape strategies often make the TIME resistant to cancer immunotherapy. Neutralizing immune escape strategies is necessary to overcome resistance to cancer immunotherapy. Immune checkpoint receptors (ICRs) expressed in effector immune cells inhibit their effector function via direct interaction with immune checkpoint ligands (ICLs) expressed in tumor cells. Therefore, blocking ICRs or ICLs has been developed as a promising cancer immunotherapy by reinvigorating the function of effector immune cells. Among the ICRs, programmed cell death 1 (PD-1) has mainly been antagonized to enhance the survival of human patients with cancer by restoring the function of tumor-infiltrating (TI) CD8+ T cells. It has been demonstrated that PD-1 is expressed not only in TI CD8+ T cells, but also in other TI immune cells and even tumor cells. While PD-1 suppresses the function of TI CD8+ T cells, it is controversial whether PD-1 suppresses or amplifies the suppressive function of TI-suppressive immune cells (e.g., regulatory T cells, tumor-associated macrophages, and myeloid cells). There is also controversy regarding the role of tumor-expressing PD-1. Therefore, a precise understanding of the expression pattern and function of PD-1 in each cell subset is important for improving the efficacy of cancer immunotherapy. Here, we review the differential role of PD-1 expressed by various TI immune cells and tumor cells. We focused on how cell-type-specific ablation or blockade of PD-1 affects tumor growth in a murine tumor model. Furthermore, we will also describe how the blockade of PD-1 acts on TI immune cells in human patients with cancer.
Collapse
Affiliation(s)
- Myeong Joon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, South Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, South Korea
| |
Collapse
|
37
|
Understanding and improving cellular immunotherapies against cancer: From cell-manufacturing to tumor-immune models. Adv Drug Deliv Rev 2021; 179:114003. [PMID: 34653533 DOI: 10.1016/j.addr.2021.114003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment (TME) is shaped by dynamic metabolic and immune interactions between precancerous and cancerous tumor cells and stromal cells like epithelial cells, fibroblasts, endothelial cells, and hematopoietically-derived immune cells. The metabolic states of the TME, including the hypoxic and acidic niches, influence the immunosuppressive phenotypes of the stromal and immune cells, which confers resistance to both host-mediated tumor killing and therapeutics. Numerous in vitro TME platforms for studying immunotherapies, including cell therapies, are being developed. However, we do not yet understand which immune and stromal components are most critical and how much model complexity is needed to answer specific questions. In addition, scalable sourcing and quality-control of appropriate TME cells for reproducibly manufacturing these platforms remain challenging. In this regard, lessons from the manufacturing of immunomodulatory cell therapies could provide helpful guidance. Although immune cell therapies have shown unprecedented results in hematological cancers and hold promise in solid tumors, their manufacture poses significant scale, cost, and quality control challenges. This review first provides an overview of the in vivo TME, discussing the most influential cell populations in the tumor-immune landscape. Next, we summarize current approaches for cell therapies against cancers and the relevant manufacturing platforms. We then evaluate current immune-tumor models of the TME and immunotherapies, highlighting the complexity, architecture, function, and cell sources. Finally, we present the technical and fundamental knowledge gaps in both cell manufacturing systems and immune-TME models that must be addressed to elucidate the interactions between endogenous tumor immunity and exogenous engineered immunity.
Collapse
|
38
|
Zinovkin DA, Kose SY, Nadyrov EA, Achinovich SL, Los' DM, Gavrilenko TE, Gavrilenko DI, Yuzugulen J, Pranjol MZI. Potential role of tumor-infiltrating T-, B-lymphocytes, tumor-associated macrophages and IgA-secreting plasma cells in long-term survival in the rectal adenocarcinoma patients. Life Sci 2021; 286:120052. [PMID: 34656554 DOI: 10.1016/j.lfs.2021.120052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/22/2021] [Accepted: 10/11/2021] [Indexed: 11/18/2022]
Abstract
AIMS Many studies investigated the associations between the role of immune cells of rectal cancer microenvironment and survival during the first 5 years post-surgery. This is problematic as this disease has the potential to progress even after 5 years after relapse and infiltrating immune cells could play key roles. Therefore, this retrospective study investigates expression and roles of tumor-infiltrating T-lymphocytes (TIL-T), tumor-infiltrating B-lymphocytes (TILB), IgA+ plasma cells (IgA+ PC) and tumor-associated macrophages (TAM) in patients with or without progression over 5 years survival with rectal adenocarcinoma. MAIN METHODS Here we used immunohistochemical staining of CD3, CD20, IgA, CD68 positive cells and its detection in rectal cancer stroma. Data was analyzed using Mann Whitney U test, ROC, survival and Cox's regression analysis. KEY FINDINGS The number of TIL-T (p = 0.0276), TIL-B (p < 0.0001) and IgA+ PC (p = 0.015) immune cells was significantly higher in rectal cancer stroma of patients with favorable outcome. Univariate Cox's regression analysis revealed a predictive role of TIL-T (HR = 0.482; 95% CI, 0.303 to 0.704; p < 0.0001), TIL-B (HR = 0.301; 95% CI, 0.198 to 0.481; p < 0.0001) and IgA+-PC (HR = 0.488; 95% CI, 0.322 to 0.741; p < 0.0001). Multivariate Cox's regression analysis showed prognostic role of TIL-B (HR = 0.940; 95% CI, 0.914 to 0.968; p < 0.0001) and IgA+-PC (HR = 0.985; 95% CI, 0.975 to 0.996; p = 0.006) play role in long time survival. SIGNIFICANCE CD20+ TIL-B and IgA+ cells have significant associations with long -term survival of patients with rectal cancer, with potential therapeutic intervention in cancer immunotherapy.
Collapse
Affiliation(s)
- Dmitry A Zinovkin
- Department of Pathology, Gomel State Medical University, 246000 Gomel, Belarus.
| | - Suheyla Y Kose
- School of Life Sciences, University of Sussex, Brighton, UK
| | - Eldar A Nadyrov
- Department of Pathology, Gomel State Medical University, 246000 Gomel, Belarus
| | - Sergey L Achinovich
- Department of Anatomical Pathology, Gomel Regional Clinical Oncological Hospital, Gomel, Belarus
| | - Dmitry M Los'
- Department of Anatomical Pathology, Gomel Regional Clinical Oncological Hospital, Gomel, Belarus
| | - Tatyana E Gavrilenko
- Republican Research Center for Radiation Medicine and Human Ecology, Gomel, Belarus
| | - Dmitry I Gavrilenko
- Republican Research Center for Radiation Medicine and Human Ecology, Gomel, Belarus
| | - Jale Yuzugulen
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | | |
Collapse
|
39
|
Targeting Inflammatory Signaling in Prostate Cancer Castration Resistance. J Clin Med 2021; 10:jcm10215000. [PMID: 34768524 PMCID: PMC8584457 DOI: 10.3390/jcm10215000] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/04/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Although castration-resistant prostate cancer (CRPC) as a whole, by its name, refers to the tumors that relapse and/or regrow independently of androgen after androgen deprivation therapy (ADT), untreated tumor, even in early-stage primary prostate cancer (PCa), contains androgen-independent (AI) PCa cells. The transformation of androgen-dependent (AD) PCa to AI PCa under ADT is a forced evolutionary process, in which the small group of AI PCa cells that exist in primary tumors has the unique opportunity to proliferate and expand selectively and dominantly, while some AD PCa cells that have escaped from ADT-induced death acquire the capability to survive in an androgen-depleted environment. The adaptation and reprogramming of both PCa cells and the tumor microenvironment (TME) under ADT make PCa much stronger than primary tumors so that, currently, there are no effective therapeutic methods available for the treatment of CRPC. Many mechanisms have been found to be related to the emergence and maintenance of PCa castration resistance; in this review, we focus on the role of inflammatory signaling in both PCa cells and the TME for the emergence and maintenance of CRPC and summarize the recent advances of therapeutic strategies that target inflammatory signaling for the treatment of CRPC.
Collapse
|
40
|
Liu H, Zhong J, Hu J, Han C, Li R, Yao X, Liu S, Chen P, Liu R, Ling F. Single-cell transcriptomics reveal DHX9 in mature B cell as a dynamic network biomarker before lymph node metastasis in CRC. Mol Ther Oncolytics 2021; 22:495-506. [PMID: 34553035 PMCID: PMC8433066 DOI: 10.1016/j.omto.2021.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence indicates that mature B cells in the adjacent tumor tissue, both as an intermediate state, are vital in advanced colorectal cancer (CRC), which is associated with a low survival rate. Developing predictive biomarkers that detect the tipping point of mature B cells before lymph node metastasis in CRC is critical to prevent irreversible deterioration. We analyzed B cells in the adjacent tissues of CRC samples from different stages using the dynamic network biomarker (DNB) method. Single-cell profiling of 725 CRC-derived B cells revealed the emergence of a mature B cell subtype. Using the DNB method, we identified stage II as a critical period before lymph node metastasis and that reversed difference genes triggered by DNBs were enriched in the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway involving B cell immune capability. DHX9 (DEAH-box helicase 9) was a specific para-cancerous tissue DNB key gene. The dynamic expression levels of DHX9 and its proximate network genes involved in B cell-related pathways were reversed at the network level from stage I to III. In summary, DHX9 in mature B cells of CRC-adjacent tissues may serve as a predictable biomarker and a potential immune target in CRC progression.
Collapse
Affiliation(s)
- Huisheng Liu
- School of Biology and Biological Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, Guangdong 510641, China
| | - JiaYuan Zhong
- School of Mathematics, South China University of Technology, Guangzhou, Guangdong 510641, China
| | - JiaQi Hu
- School of Biology and Biological Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, Guangdong 510641, China
| | - ChongYin Han
- School of Biology and Biological Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, Guangdong 510641, China
| | - Rui Li
- Department of Pathology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong 510515, China
| | - XueQing Yao
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510080, China
| | - ShiPing Liu
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518083, China
| | - Pei Chen
- School of Mathematics, South China University of Technology, Guangzhou, Guangdong 510641, China
| | - Rui Liu
- School of Mathematics, South China University of Technology, Guangzhou, Guangdong 510641, China
- Pazhou Lab, Guangzhou, Guangdong 510330, China
| | - Fei Ling
- School of Biology and Biological Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, Guangdong 510641, China
| |
Collapse
|
41
|
Zheng X, Jin W, Wang S, Ding H. Progression on the Roles and Mechanisms of Tumor-Infiltrating T Lymphocytes in Patients With Hepatocellular Carcinoma. Front Immunol 2021; 12:729705. [PMID: 34566989 PMCID: PMC8462294 DOI: 10.3389/fimmu.2021.729705] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022] Open
Abstract
Primary liver cancer (PLC) is one of the most common malignancies in China, where it ranks second in mortality and fifth in morbidity. Currently, liver transplantation, hepatic tumor resection, radiofrequency ablation, and molecular-targeted agents are the major treatments for hepatocellular carcinoma (HCC). Overall, HCC has a poor survival rate and a high recurrence rate. Tumor-infiltrating lymphocytes (TILs) have been discovered to play essential roles in the development, prognosis, and immunotherapy treatment of HCC. As the major component cells of TILs, T cells are also proved to show antitumor and protumor effects in HCC. Foxp3+, CD8+, CD3+, and CD4+ T lymphocytes are the broadly studied subgroups of TILs. This article reviews the roles and mechanisms of different tumor-infiltrating T lymphocyte subtypes in HCC.
Collapse
Affiliation(s)
- Xiaoqin Zheng
- Department of Gastrointestinal and Hepatology, Beijing You’An Hospital, Capital Medical University, Beijing, China
| | - Wenjie Jin
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zurich, Switzerland
| | - Shanshan Wang
- Beijing Institute of Hepatology, Beijing You’An Hospital, Capital Medical University, Beijing, China
| | - Huiguo Ding
- Department of Gastrointestinal and Hepatology, Beijing You’An Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
42
|
The Interplay between Glioblastoma and Its Microenvironment. Cells 2021; 10:cells10092257. [PMID: 34571905 PMCID: PMC8469987 DOI: 10.3390/cells10092257] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 01/05/2023] Open
Abstract
GBM is the most common primary brain tumor in adults, and the aggressive nature of this tumor contributes to its extremely poor prognosis. Over the years, the heterogeneous and adaptive nature of GBM has been highlighted as a major contributor to the poor efficacy of many treatments including various immunotherapies. The major challenge lies in understanding and manipulating the complex interplay among the different components within the tumor microenvironment (TME). This interplay varies not only by the type of cells interacting but also by their spatial distribution with the TME. This review highlights the various immune and non-immune components of the tumor microenvironment and their consequences f the efficacy of immunotherapies. Understanding the independent and interdependent aspects of the various sub-populations encapsulated by the immune and non-immune components will allow for more targeted therapies. Meanwhile, understanding how the TME creates and responds to different environmental pressures such as hypoxia may allow for other multimodal approaches in the treatment of GBM. Ultimately, a better understanding of the GBM TME will aid in the development and advancement of more effective treatments and in improving patient outcomes.
Collapse
|
43
|
Azwar S, Seow HF, Abdullah M, Faisal Jabar M, Mohtarrudin N. Recent Updates on Mechanisms of Resistance to 5-Fluorouracil and Reversal Strategies in Colon Cancer Treatment. BIOLOGY 2021; 10:854. [PMID: 34571731 PMCID: PMC8466833 DOI: 10.3390/biology10090854] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
5-Fluorouracil (5-FU) plus leucovorin (LV) remain as the mainstay standard adjuvant chemotherapy treatment for early stage colon cancer, and the preferred first-line option for metastatic colon cancer patients in combination with oxaliplatin in FOLFOX, or irinotecan in FOLFIRI regimens. Despite treatment success to a certain extent, the incidence of chemotherapy failure attributed to chemotherapy resistance is still reported in many patients. This resistance, which can be defined by tumor tolerance against chemotherapy, either intrinsic or acquired, is primarily driven by the dysregulation of various components in distinct pathways. In recent years, it has been established that the incidence of 5-FU resistance, akin to multidrug resistance, can be attributed to the alterations in drug transport, evasion of apoptosis, changes in the cell cycle and DNA-damage repair machinery, regulation of autophagy, epithelial-to-mesenchymal transition, cancer stem cell involvement, tumor microenvironment interactions, miRNA dysregulations, epigenetic alterations, as well as redox imbalances. Certain resistance mechanisms that are 5-FU-specific have also been ascertained to include the upregulation of thymidylate synthase, dihydropyrimidine dehydrogenase, methylenetetrahydrofolate reductase, and the downregulation of thymidine phosphorylase. Indeed, the successful modulation of these mechanisms have been the game plan of numerous studies that had employed small molecule inhibitors, plant-based small molecules, and non-coding RNA regulators to effectively reverse 5-FU resistance in colon cancer cells. It is hoped that these studies would provide fundamental knowledge to further our understanding prior developing novel drugs in the near future that would synergistically work with 5-FU to potentiate its antitumor effects and improve the patient's overall survival.
Collapse
Affiliation(s)
- Shamin Azwar
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| | - Heng Fong Seow
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| | - Maha Abdullah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| | - Mohd Faisal Jabar
- Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Norhafizah Mohtarrudin
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| |
Collapse
|
44
|
Miracco C, Toti P, Gelmi MC, Aversa S, Baldino G, Galluzzi P, De Francesco S, Petrelli F, Sorrentino E, Belmonte G, Galimberti D, Bracco S, Hadjistilianou T. Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1- Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy. Invest Ophthalmol Vis Sci 2021; 62:6. [PMID: 33538768 PMCID: PMC7862737 DOI: 10.1167/iovs.62.2.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose To investigate the impact of chemotherapy (CHT) on human retinoblastoma (RB) tumor microenvironment (TME). Cases and Methods Ninety-four RBs were studied, including 44 primary RBs treated by upfront surgery (Group 1) and 50 primary RBs enucleated after CHT (CHT), either intra-arterial (IAC; Group 2, 33 cases) or systemic (S-CHT; Group 3, 17 cases). Conventional and multiplexed immunohistochemistry were performed to make quantitative comparisons among the three groups, for the following parameters: tumor-infiltrating inflammatory cells (TI-ICs); programmed cell death protein 1 (PD-1) positive TI-ICs; Ki67 proliferation index; gliosis; PD-1 ligand (PD-L1) protein expression; vessel number. We also correlated these TME factors with the presence of histological high-risk factors (HHRF+) and RB anaplasia grade (AG). Results After CHT, a decrease in both RB burden and Ki67 positivity was observed. In parallel, most subsets of TI-ICs, PD-1+ TI-ICs, gliosis, and PD-L1 protein expression significantly increased (P < 0.001, P = 0.02, P < 0.001, respectively). Vessel number did not significantly vary. Age, HHRFs+ and AG were significantly different between primary and chemoreduced RBs (P < 0.001, P = 0.006, P = 0.001, respectively) and were correlated with most TME factors. Conclusions CHT modulates host antitumor immunity by reorienting the RB TME from anergic into an active, CD8+, PD-L1+ hot state. Furthermore, some clinicopathological characteristics of RB correlate with several factors of TME. Our study adds data in favor of the possibility of a new therapeutic scenario in human RB.
Collapse
Affiliation(s)
- Clelia Miracco
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Paolo Toti
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Maria Chiara Gelmi
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University Hospital of Siena, Siena, Italy
| | - Sara Aversa
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Gennaro Baldino
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Paolo Galluzzi
- Department of Medicine, Surgery and Neuroscience, Unit of Neuroimaging and Neurointervention, University Hospital of Siena, Siena, Italy
| | - Sonia De Francesco
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University Hospital of Siena, Siena, Italy
| | - Federica Petrelli
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Ester Sorrentino
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Giuseppe Belmonte
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Daniela Galimberti
- Department of Maternal, Newborn and Child Health, Unit of Pediatrics, University Hospital of Siena, Siena, Italy
| | - Sandra Bracco
- Department of Medicine, Surgery and Neuroscience, Unit of Neuroimaging and Neurointervention, University Hospital of Siena, Siena, Italy
| | - Theodora Hadjistilianou
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University Hospital of Siena, Siena, Italy
| |
Collapse
|
45
|
Qin M, Wang D, Fang Y, Zheng Z, Liu X, Wu F, Wang L, Li X, Hui B, Ma S, Tang W, Pan X. Current Perspectives on B Lymphocytes in the Immunobiology of Hepatocellular Carcinoma. Front Oncol 2021; 11:647854. [PMID: 34235074 PMCID: PMC8256159 DOI: 10.3389/fonc.2021.647854] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
Immune cells infiltrating tumors are capable of significantly impacting carcinogenesis through cancer promotion and anticancer responses. There are many aspects of hepatocellular carcinoma (HCC) related T lymphocytes that are undergoing extensive studies, whereas the effect exerted by B lymphocytes remains a less researched area. In this study, the latest research on the effect of B lymphocytes as they infiltrate tumors in relation to HCC is presented. Their prognosis-related importance is analyzed, along with their function in the tumor microenvironment (TME), as well as the way that B cell biology can be employed to help create a B cell therapy strategy for HCC.
Collapse
Affiliation(s)
- Miaomiao Qin
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Danping Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yijiao Fang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhiying Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fan Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liangliang Wang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bingqing Hui
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shijie Ma
- Department of Gastroenterology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Weiwei Tang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, China
| | - Xiongxiong Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
46
|
Wang J, Han Q, Liu H, Luo H, Li L, Liu A, Jiang Y. Identification of Radiotherapy-Associated Genes in Lung Adenocarcinoma by an Integrated Bioinformatics Analysis Approach. Front Mol Biosci 2021; 8:624575. [PMID: 34212001 PMCID: PMC8239180 DOI: 10.3389/fmolb.2021.624575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/31/2021] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy (RT) plays an important role in the prognosis of lung adenocarcinoma (LUAD) patients, but the radioresistance (RR) of LUAD is still a challenge that needs to be overcome. The current study aimed to investigate LUAD patients with RR to illuminate the underlying mechanisms. We utilized gene set variation analysis (GSVA) and The Cancer Immunome Atlas (TCIA) database to characterize the differences in biological functions and neoantigen-coding genes between RR and radiosensitive (RS) patients. Weighted Gene co-expression network analysis (WGCNA) was used to explore the relationship between RT-related traits and hub genes in two modules, i.e., RR and RS; two representative hub genes for RR (MZB1 and DERL3) and two for RS (IFI35 and PSMD3) were found to be related to different RT-related traits. Further analysis of the hub genes with the Lung Cancer Explorer (LCE), PanglaoDB and GSVA resources revealed the differences in gene expression levels, cell types and potential functions. On this basis, the Tumor and Immune System Interaction Database (TISIDB) was used to identify the potential association between RR genes and B cell infiltration. Finally, we used the Computational Analysis of Resistance (CARE) database to identify specific gene-associated drugs for RR patients and found that GSK525762A and nilotinib might be promising candidates for RR treatment. Taken together, these results demonstrate that B cells in TME may have a significant impact on the RT and that these two drug candidates, GSK525762A and nilotinib, might be helpful for the treatment of RR patients.
Collapse
Affiliation(s)
- Junhao Wang
- State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qizheng Han
- State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Huizi Liu
- State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Haihua Luo
- State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lei Li
- State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Aihua Liu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Jiang
- State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Kumar A, Swain CA, Shevde LA. Informing the new developments and future of cancer immunotherapy : Future of cancer immunotherapy. Cancer Metastasis Rev 2021; 40:549-562. [PMID: 34003425 DOI: 10.1007/s10555-021-09967-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/14/2021] [Indexed: 12/19/2022]
Abstract
The application of cancer immunotherapy (CIT) in reinforcing anti-tumor immunity in response to carcinogenesis and metastasis has shown promising advances, along with new therapeutic challenges, in the landscape of cancer care. To promote tumor growth and metastasis, cancer cells aim to manipulate their microenvironment by mediating a crosstalk with various immune cells through the secretion of chemokines, cytokines, and other associated factors. Understanding this crosstalk is the key to discovering the best targets for improved immunotherapies and clinical strategies in cancer treatment. Here, we review the tumor immune crosstalk in cancer growth and metastasis. This review also highlights the development and expansion of CIT over the years. Moreover, we highlight clinical challenges and limitations involving immune-related adverse events, treating cancer patients with pre-existing autoimmune diseases, and the management of immunotherapy-induced treatment resistance. Possible clinical solutions to these current challenges in CIT are also proposed. Altogether, this review can contribute to the formation of pre-clinical and treatment-related strategies that further develop the availability and effectiveness of CIT.
Collapse
Affiliation(s)
- Atul Kumar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Courtney A Swain
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA. .,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
48
|
Trüb M, Zippelius A. Tertiary Lymphoid Structures as a Predictive Biomarker of Response to Cancer Immunotherapies. Front Immunol 2021; 12:674565. [PMID: 34054861 PMCID: PMC8149953 DOI: 10.3389/fimmu.2021.674565] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid formations which are formed under long-lasting inflammatory conditions, including tumours. TLS are composed predominantly of B cells, T cells and dendritic cells, and display various levels of organisation, from locally concentrated aggregates of immune cells, through clearly defined B cell follicles to mature follicles containing germinal centres. Their presence has been strongly associated with improved survival and clinical outcome upon cancer immunotherapies for patients with solid tumours, indicating potential for TLS to be used as a prognostic and predictive factor. Although signals involved in TLS generation and main cellular components of TLS have been extensively characterised, the exact mechanism by which TLS contribute to the anti-tumour response remain unclear. Here, we summarise the most recent development in our understanding of their role in cancer and in particular in the response to cancer immunotherapy. Deciphering the relationship between B cells and T cells found in TLS is a highly exciting field of investigation, with the potential to lead to novel, B-cell focused immunotherapies.
Collapse
Affiliation(s)
- Marta Trüb
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Alfred Zippelius
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland.,Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
49
|
Ciccolella M, Andreone S, Mancini J, Sestili P, Negri D, Pacca AM, D’Urso MT, Macchia D, Canese R, Pang K, SaiYing Ko T, Decadt Y, Schiavoni G, Mattei F, Belardelli F, Aricò E, Bracci L. Anticancer Effects of Sublingual Type I IFN in Combination with Chemotherapy in Implantable and Spontaneous Tumor Models. Cells 2021; 10:845. [PMID: 33917958 PMCID: PMC8068355 DOI: 10.3390/cells10040845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/06/2023] Open
Abstract
Salivary gland tumors are a heterogeneous group of neoplasms representing less than 10% of all head and neck tumors. Among salivary gland tumors, salivary duct carcinoma (SDC) is a rare, but highly aggressive malignant tumor resembling ductal breast carcinoma. Sublingual treatments are promising for SDC due to the induction of both local and systemic biological effects and to reduced systemic toxicity compared to other administration routes. In the present study, we first established that the sublingual administration of type I IFN (IFN-I) is safe and feasible, and exerts antitumor effects both as monotherapy and in combination with chemotherapy in transplantable tumor models, i.e., B16-OVA melanoma and EG.7-OVA lymphoma. Subsequently, we proved that sublingual IFN-I in combination with cyclophosphamide (CTX) induces a long-lasting reduction of tumor mass in NeuT transgenic mice that spontaneously develop SDC. Most importantly, tumor shrinkage in NeuT transgenic micewas accompanied by the emergence of tumor-specific cellular immune responses both in the blood and in the tumor tissue. Altogether, these results provide evidence that sublingual IFN holds promise in combination with chemotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Maria Ciccolella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Jacopo Mancini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Paola Sestili
- National Center for the Control and Evaluation of Medicines, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Anna Maria Pacca
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Maria Teresa D’Urso
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Daniele Macchia
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Rossella Canese
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Ken Pang
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville 3010, Australia
| | - Thomas SaiYing Ko
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Yves Decadt
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Filippo Belardelli
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, 00133 Rome, Italy;
| | - Eleonora Aricò
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| |
Collapse
|
50
|
A novel cancer immunotherapy using tumor-infiltrating B cells in the APCmin/+ mouse model. PLoS One 2021; 16:e0245608. [PMID: 33465122 PMCID: PMC7815094 DOI: 10.1371/journal.pone.0245608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 01/04/2021] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence has suggested a correlation of tumor infiltrating B cells (TiBcs) and a good prognosis of cancer diseases. In some cases, TiBcs appear to have experienced antigen stimulation since they have undergone class-switching and somatic hypermutation and formed tertiary lymphoid structures around tumors together with T cells. Assuming TiBcs include those that recognize some tumor antigens, we sought to investigate their possible usefulness for cell-mediated immunotherapies. To expand usually small number of TiBcs in vitro, we modified our B cell culture system: we transduced B cells with ERT2-Bach2 so that they grow unlimitedly provided with tamoxifen, IL-21 and our original feeder cells. Such cells differentiate into plasma cells and produce antibodies upon withdrawal of tamoxifen, and further by addition of a Bach2-inhibitor in vitro. As a preliminary experiment, thus expanded splenic B cells expressing a transgenic antigen receptor/antibody against hen egg lysozyme were intravenously injected into mice pre-implanted with B16 melanoma cells expressing membrane-bound HEL in the skin, which resulted in suppression of the growth of B16 tumors and prolonged survival of the recipient mice. To test the usefulness of TiBcs for the immunotherapy, we next used APCmin/+ mice as a model that spontaneously develop intestinal tumors. We cultured TiBcs separated from the tumors of APCmin/+ mice as above and confirmed that the antibodies they produce recognize the APCmin/+ tumor. Repeated injection of such TiBcs into adult APCmin/+ mice resulted in suppression of intestinal tumor growth and elongation of the survival of the recipient mice. Serum antibody from the TiBc-recipient mice selectively bound to an antigen expressed in the tumor of APCmin/+ mice. These data suggest a possibility of the novel individualized cancer immunotherapy, in which TiBcs from surgically excised tumor tissues are expanded and infused into the donor patients.
Collapse
|