1
|
Qu F, Wang G, Wen P, Liu X, Zeng X. Knowledge mapping of immunotherapy for breast cancer: A bibliometric analysis from 2013 to 2022. Hum Vaccin Immunother 2024; 20:2335728. [PMID: 38563136 PMCID: PMC10989689 DOI: 10.1080/21645515.2024.2335728] [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: 01/17/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related death among women globally. Immunotherapy has emerged as a major milestone in contemporary oncology. This study aims to conduct a bibliometric analysis in the field of immunotherapy for breast cancer, providing a comprehensive overview of the current research status, identifying trends and hotspots in research topics. We searched and retrieved data from the Web of Science Core Collection, and performed a bibliometric analysis of publications on immunotherapy for breast cancer from 2013 to 2022. Current status and hotspots were evaluated by co-occurrence analysis using VOSviewer. Evolution and bursts of knowledge base were assessed by co-citation analysis using CiteSpace. Thematic evolution by bibliometrix package was used to discover keywords trends. The attribution and collaboration of countries/regions, institutions and authors were also explored. A total of 7,975 publications were included. In co-occurrence analysis of keywords, 6 major clusters were revealed: tumor microenvironment, prognosis biomarker, immune checkpoints, novel drug delivery methods, immune cells and therapeutic approaches. The top three most frequently mentioned keywords were tumor microenvironment, triple-negative breast cancer, and programmed cell death ligand 1. The most productive country, institution and author were the USA (2926 publications), the University of Texas MD Anderson Cancer Center (219 publications), and Sherene Loi (28 publications), respectively. There has been a rapid growth in studies on immunotherapy for breast cancer worldwide. This research area has gained increasing attention from different countries and institutions. With the rising incidence of breast cancer, immunotherapy represents a research field of significant clinical value and potential.
Collapse
Affiliation(s)
- Fanli Qu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Guanwen Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Ping Wen
- School of Medicine, Chongqing University, Chongqing, China
| | - Xiaoyu Liu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
2
|
Conejo-Garcia JR, Lopez-Bailon LU, Anadon CM. Unraveling spontaneous humoral immune responses against human cancer: a road to novel immunotherapies. J Leukoc Biol 2024; 116:919-926. [PMID: 39190797 DOI: 10.1093/jleuko/qiae179] [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: 05/08/2024] [Revised: 07/01/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
In immuno-oncology, the focus has traditionally been on αβ T cells, and immune checkpoint inhibitors that primarily target PD-1 or CTLA4 in these lymphocytes have revolutionized the management of multiple human malignancies. However, recent research highlights the crucial role of B cells and the antibodies they produce in antagonizing malignant progression, offering new avenues for immunotherapy. Our group has demonstrated that dimeric Immunoglobulin A can penetrate tumor cells, neutralize oncogenic drivers in endosomes, and expel them from the cytosol. This mechanistic insight suggests that engineered antibodies targeting this pathway may effectively reach previously inaccessible targets. Investigating antibody production within intratumoral germinal centers and understanding the impact of different immunoglobulins on malignant progression could furnish new tools for the therapeutic arsenal, including the development of tumor-penetrating antibodies. This review aims to elucidate the nature of humoral adaptive immune responses in human cancer and explore how they could herald a new era of immunotherapeutic modalities. By expanding the scope of antitumor immunotherapies, these approaches have the potential to benefit a broader range of cancer patients, particularly through the utilization of tumor cell-penetrating antibodies.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC 27710, United States
- Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, United States
| | - Luis U Lopez-Bailon
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC 27710, United States
- Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, United States
| | - Carmen M Anadon
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC 27710, United States
- Duke Cancer Institute, Duke School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
3
|
Xue D, Hu S, Zheng R, Luo H, Ren X. Tumor-infiltrating B cells: Their dual mechanistic roles in the tumor microenvironment. Biomed Pharmacother 2024; 179:117436. [PMID: 39270540 DOI: 10.1016/j.biopha.2024.117436] [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: 05/21/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024] Open
Abstract
The occurrence and development of tumors are closely associated with abnormalities in the immune system's structure and function, with tumor immunotherapy being intricately linked to the tumor microenvironment (TME). Early studies on lymphocytes within the TME primarily concentrated on T cells. However, as research has advanced, the multifaceted roles of tumor-infiltrating B cells (TIL-Bs) in tumor immunity, encompassing both anti-tumor and pro-tumor effects, have garnered increasing attention. This paper explored the composition of the TME and the biological characteristics of TIL-Bs, investigating the dual roles within the TME to offer new insights and strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Demin Xue
- School of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shaozhen Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Runchen Zheng
- School of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huidan Luo
- Department of Pulmonology, Hechi Hospital of Traditional Chinese Medicine, Guangxi 547000, China
| | - Xi Ren
- Department of Oncology II, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
4
|
Xi Y, Liu R, Zhang X, Guo Q, Zhang X, Yang Z, Zheng H, Song Q, Hua B. A Bibliometric Analysis of Metabolic Reprogramming in the Tumor Microenvironment From 2003 to 2022. Cancer Rep (Hoboken) 2024; 7:e2146. [PMID: 39158178 PMCID: PMC11331499 DOI: 10.1002/cnr2.2146] [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: 11/01/2023] [Revised: 06/23/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Despite considerable progress in cancer immunotherapy, it is not available for many patients. Resistance to immune checkpoint blockers arises from the intricate interactions between cancer and its microenvironment. Metabolic reprogramming in tumor and immune cells in the tumor microenvironment (TME) influences anti-tumor immune responses by remodeling the immune microenvironment. Metabolic reprogramming has emerged as an important hallmark of tumorigenesis. However, few studies have focused on the TME and metabolic reprogramming. Therefore, we aimed to explore the current research status and popular topics in TME-related metabolic reprogramming over a 20 years using a bibliometric approach. METHODS Studies focusing on metabolic reprogramming and TME were searched using the Web of Science Core Collection database. Bibliometric and visual analyses of the articles and reviews were performed using Bibliometrix, VOSviewer, and CiteSpace. RESULTS In total, 4726 articles published between 2003 and 2022 were selected. The number of publications and citations has increased annually. Cooperation network analysis indicated that the United States holds the foremost position in metabolic reprogramming and TME research with the highest volume of publications and citations, thus exerting the greatest influence. Among these institutions, Fudan University displayed the highest level of productivity. Frontiers in Immunology showed the highest degree of productivity in this field. Ho Ping-Chih made the most article contributions, and Pearce Edward J. was the most co-cited author. Four clusters were obtained after a cluster analysis of the authors' keywords: TME, metabolic reprogramming, immunometabolism, and immunity. Immunometabolism, glycolysis, immune cells, and tumor-associated macrophages are relatively recent keywords that have attracted increasing attention. CONCLUSIONS A comprehensive landscape of advancements in metabolic reprogramming and the TME was evaluated, which provided crucial information for scholars to further advance this promising field. Further research should explore new topics related to immunometabolism in the TME using a transdisciplinary approach.
Collapse
Affiliation(s)
- Yupeng Xi
- Department of General Internal Medicine, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Rui Liu
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Xing Zhang
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Qiujun Guo
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Xiwen Zhang
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Zizhen Yang
- Department of General Internal MedicineXi'an Fifth HospitalXi'anShanxiChina
| | - Honggang Zheng
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Baojin Hua
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
5
|
Mandal G, Pradhan S. B cell responses and antibody-based therapeutic perspectives in human cancers. Cancer Rep (Hoboken) 2024; 7:e2056. [PMID: 38522010 PMCID: PMC10961090 DOI: 10.1002/cnr2.2056] [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: 11/30/2023] [Revised: 02/26/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Immuno-oncology has been focused on T cell-centric approaches until the field recently started appreciating the importance of tumor-reactive antibody production by tumor-infiltrating plasma B cells, and the necessity of developing novel therapeutic antibodies for the treatment of different cancers. RECENT FINDINGS B lymphocytes often infiltrate solid tumors and the extent of B cell infiltration normally correlates with stronger T cell responses while generating humoral responses against malignant progression by producing tumor antigens-reactive antibodies that bind and coat the tumor cells and promote cytotoxic effector mechanisms, reiterating the fact that the adaptive immune system works by coordinated humoral and cellular immune responses. Isotypes, magnitude, and the effector functions of antibodies produced by the B cells within the tumor environment differ among cancer types. Interestingly, apart from binding with specific tumor antigens, antibodies produced by tumor-infiltrating B cells could bind to some non-specific receptors, peculiarly expressed by cancer cells. Antibody-based immunotherapies have revolutionized the modalities of cancer treatment across the world but are still limited against hematological malignancies and a few types of solid tumor cancers with a restricted number of targets, which necessitates the expansion of the field to have newer effective targeted antibody therapeutics. CONCLUSION Here, we discuss about recent understanding of the protective spontaneous antitumor humoral responses in human cancers, with an emphasis on the advancement and future perspectives of antibody-based immunotherapies in cancer.
Collapse
Affiliation(s)
- Gunjan Mandal
- Division of Cancer BiologyDBT‐Institute of Life SciencesBhubaneswarIndia
| | - Suchismita Pradhan
- Division of Cancer BiologyDBT‐Institute of Life SciencesBhubaneswarIndia
| |
Collapse
|
6
|
Lin X, Zhao X, Chen Y, Yang R, Dai Z, Li W, Lin C, Cao W. CXC ligand 13 orchestrates an immunoactive microenvironment and enhances immunotherapy response in head and neck squamous cell carcinoma. Int J Immunopathol Pharmacol 2024; 38:3946320241227312. [PMID: 38252495 PMCID: PMC10807398 DOI: 10.1177/03946320241227312] [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: 06/15/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Objectives: This study aims to systematically explore the role of chemokine CXC ligand 13 (CXCL13) in head and neck squamous cell carcinoma (HNSCC). Methods: The Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases provided the RNA-seq data for cancer and normal tissues, respectively. Gene set enrichment analysis was applied to search the cancer hallmarks associated with CXCL13 expression. TIMER2.0 was the main platform used to investigate the immune cell infiltration related to CXCL13. Immunohistochemistry was applied to explore the relationship between CXCL13 and patients' prognosis and the relationship between CXCL13 and tertiary lymphoid structures (TLSs). Results: The expression of CXCL13 was upregulated in most tumors, including HNSCC. The higher expression of CXCL13 was closely related to the positive prognosis of HNSCC. CXCL13 was mainly expressed in B cells and CD8 + T cells, revealing the relationship between its expression and immune activation in the tumor microenvironment. Furthermore, immunohistochemistry and multiple fluorescence staining analysis of HNSCC samples showed a powerful correlation between CXCL13 expression, TLSs formation, and positive prognosis. Finally, CXCL13 significantly increased the response to cancer immunotherapy. Conclusions: CXCL13 may function as a potential biomarker for predicting prognosis and immunotherapy response and associate with TLSs in HNSCC.
Collapse
Affiliation(s)
- Xiaohu Lin
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xiaomei Zhao
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yiming Chen
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Rong Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhenlin Dai
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wei Li
- Xuchang Central Hospital, Henan, China
| | - Chengzhong Lin
- Department of Oral Maxillofacial Surgery, Zhongshan Hospital, Fu Dan University, Shanghai, China
| | - Wei Cao
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
7
|
Huang Q, Mo J, Yang H, Ji Y, Huang R, Liu Y, Pan Y. Analysis of m7G-Related signatures in the tumour immune microenvironment and identification of clinical prognostic regulators in breast cancer. BMC Cancer 2023; 23:583. [PMID: 37353728 DOI: 10.1186/s12885-023-11012-z] [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: 11/19/2022] [Accepted: 05/25/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Breast cancer is a malignant tumour that seriously threatens women's life and health and exhibits high inter-individual heterogeneity, emphasising the need for more in-depth research on its pathogenesis. While internal 7-methylguanosine (m7G) modifications affect RNA processing and function and are believed to be involved in human diseases, little is currently known about the role of m7G modification in breast cancer. METHODS AND RESULTS We elucidated the expression, copy number variation incidence and prognostic value of 24 m7G-related genes (m7GRGs) in breast cancer. Subsequently, based on the expression of these 24 m7GRGs, consensus clustering was used to divide tumour samples from the TCGA-BRCA dataset into four subtypes based on significant differences in their immune cell infiltration and stromal scores. Differentially expressed genes between subtypes were mainly enriched in immune-related pathways such as 'Ribosome', 'TNF signalling pathway' and 'Salmonella infection'. Support vector machines and multivariate Cox regression analysis were applied based on these 24 m7GRGs, and four m7GRGs-AGO2, EIF4E3, DPCS and EIF4E-were identified for constructing the prediction model. An ROC curve indicated that a nomogram model based on the risk model and clinical factors had strong ability to predict the prognosis of breast cancer. The prognoses of patients in the high- and low-TMB groups were significantly different (p = 0.03). Moreover, the four-gene signature was able to predict the response to chemotherapy. CONCLUSIONS In conclusion, we identified four different subtypes of breast cancer with significant differences in the immune microenvironment and pathways. We elucidated prognostic biomarkers associated with breast cancer and constructed a prognostic model involving four m7GRGs. In addition, we predicted the candidate drugs related to breast cancer based on the prognosis model.
Collapse
Affiliation(s)
- Qinghua Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, China
- Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, P.R. China
| | - Jianlan Mo
- Department of Anesthesiology, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Huawei Yang
- Department of Breast Surgery, Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, China
- Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, P.R. China
| | - Yinan Ji
- Department of Breast Surgery, Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, China
- Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, P.R. China
| | - Rong Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, China
- Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, P.R. China
| | - Yan Liu
- Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, P.R. China.
- Department of BreastBone and Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530000, China.
| | - You Pan
- Department of Breast Surgery, Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, China.
- Key Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning, 530000, P.R. China.
| |
Collapse
|
8
|
Abstract
Immuno-oncology has traditionally focused on the cellular arm of the adaptive immune response, while attributing tumor-promoting activity to humoral responses in tumor-bearing hosts. This view stems from mouse models that do not necessarily recapitulate the antibody response process consistently observed in most human cancers. In recent years, the field has reconsidered the coordinated action of T and B cell responses in the context of anti-tumor immunity, as in any other immune response. Thus, recent studies in human cancer identify B cell responses with better outcome, typically in association with superior T cell responses. An area of particular interest is tertiary lymphoid structures, where germinal centers produce isotype switched antibodies and B cells and T lymphocytes interact with other immune cell types. The presence of these lymphoid structures is associated with better immunotherapeutic responses and remain poorly understood. Here, we discuss recent discoveries on how coordination between humoral and cellular responses is required for effective immune pressure against malignant progression, providing a perspective on the role of tertiary lymphoid structures and interventions to elicit their formation in unresectable tumors.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ricardo Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
9
|
Abstract
Exosomes are a type of extracellular vesicles secreted by cells in normal or pathological conditions for cell-cell communication. With immunomodulatory characteristics and potential therapeutic properties, immune-cell-derived exosomes play an important role in cancer therapy. They express various antigens on their surface, which can be employed for antigen presentation, immunological activation, and metabolic regulation, leading to the killing of cancerous cells. In addition, immune-cell-derived exosomes have received extensive attention as a drug delivery platform in effective antitumor therapy due to their excellent biocompatibility, low immunogenicity, and high loading capacity. In this review, the biological and therapeutic characteristics of immune-cell-derived exosomes are comprehensively outlined. The antitumor mechanism of exosomes secreted by immune cells, including macrophages, dendritic cells, T cells, B cells, and natural killer cells, are systematically summarized. Moreover, the applications of immune-cell-derived exosomes as nanocarriers to transport antitumor agents (chemotherapeutic drugs, genes, proteins, etc.) are discussed. More importantly, the existing challenges of immune-cell-derived exosomes are pointed out, and their antitumor potentials are also discussed.
Collapse
Affiliation(s)
- Yongmei Zhao
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales 2145, Australia
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong 226019, China
| |
Collapse
|
10
|
Larsson AM, Nordström O, Johansson A, Rydén L, Leandersson K, Bergenfelz C. Peripheral Blood Mononuclear Cell Populations Correlate with Outcome in Patients with Metastatic Breast Cancer. Cells 2022; 11:1639. [PMID: 35626676 PMCID: PMC9139201 DOI: 10.3390/cells11101639] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 01/04/2023] Open
Abstract
Local tumor-associated immune cells hold prognostic and predictive value in various forms of malignancy. The role of systemic, circulating leukocytes is, however, not well-characterized. In this prospective and explorative study, we aim to delineate the clinical relevance of a broad panel of circulating immune cells in 32 patients with newly diagnosed metastatic breast cancer (MBC) before the start of systemic treatment. Freshly isolated peripheral blood mononuclear cells (PBMCs) were analyzed by flow cytometry and evaluated for potential associations to clinicopathological variables and patient outcome. We show that the levels of specific circulating leukocyte populations are associated with clinical parameters such as hormone receptor status, histological subtype, number of circulating tumor cells (CTCs) and metastatic burden. Importantly, high levels of CD8+ cytotoxic T lymphocytes (CTLs) are significantly linked to improved overall survival (OS). In patients with estrogen receptor (ER)-positive primary tumors, high levels of circulating CTLs and non-classical (CD14+CD16++) monocytes were associated with improved OS, whereas in patients with ER-negative tumors low levels of circulating natural killer (NK) cells potentially associate with improved OS. We propose that the levels of specific circulating immune cell populations, such as CD8+ CTLs, may be used to predict clinical outcomes in MBC patients. Thus, larger studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Anna-Maria Larsson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, SE-223 81 Lund, Sweden; (A.-M.L.); (A.J.); (L.R.)
| | - Olle Nordström
- Experimental Infection Medicine, Department of Translational Medicine, Lund University, SE-214 28 Malmö, Sweden;
| | - Alexandra Johansson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, SE-223 81 Lund, Sweden; (A.-M.L.); (A.J.); (L.R.)
| | - Lisa Rydén
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, SE-223 81 Lund, Sweden; (A.-M.L.); (A.J.); (L.R.)
- Department of Surgery, Skåne University Hospital, SE-223 81 Lund, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department of Translational Medicine, Lund University, SE-214 28 Malmö, Sweden;
| | - Caroline Bergenfelz
- Experimental Infection Medicine, Department of Translational Medicine, Lund University, SE-214 28 Malmö, Sweden;
| |
Collapse
|
11
|
Guan L, Zhang Z, Gao T, Fu S, Mu W, Liang S, Liu Y, Chu Q, Fang Y, Liu Y, Zhang N. Depleting Tumor Infiltrating B Cells to Boost Antitumor Immunity with Tumor Immune-Microenvironment Reshaped Hybrid Nanocage. ACS NANO 2022; 16:4263-4277. [PMID: 35179349 DOI: 10.1021/acsnano.1c10283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor infiltrating B cells (TIBs)-dependent immunotherapy has emerged as a promising method for tumor treatment. Depleting TIBs to boost antitumor immunity is a highly desirable yet challenging approach to TIBs-dependent immunotherapy. Herein, a tumor immune-microenvironment reshaped hybrid nanocage CPN-NLI/MLD coloaded with the Bruton's tyrosine kinase inhibitor ibrutinib, and cytotoxic drug docetaxel was developed for stepwise targeting TIBs and tumor cells, respectively. The tumor microenvironment responsive CPN-NLI/MLD promoted charge reversal and size reduction under acidic conditions (pH < 6.5). The accumulation of CPN-NLI/MLD in tumor tissues was achieved through CD13 targeting, and cellular uptake was increased due to the differ-targeting delivery. Targeting of docetaxel to tumor cells was achieved by the interaction of α-MSH modified on inner docetaxel-particle MLD and melanocortin-1 receptor on the surface of tumor cells. Targeting of ibrutinib to TIBs was achieved by the interaction of Neu5Ac modified on inner ibrutinib-particle NLI and CD22 on the surface of TIBs. The boosted antitumor immunity was achieved mainly by the inhibition of Bruton's tyrosine kinase activation mediated by ibrutinib, which reduced the proportion of TIBs, enhanced infiltration of CD8+ and CD4+ T cells, increased the secretion of immunogenic cytokines including IL-2 and IFN-γ, and inhibited the proliferation of regulatory T cells and secretion of immunosuppressive cytokines including IL-10, IL-4, and TGF-β. Furthermore, CPN-NLI/MLD improved the antitumor efficiency of chemoimmunotherapy by reshaping tumor immune-microenvironment by TIBs depletion. Taken together, CPN-NLI/MLD represents a promising method for effective tumor treatment and combination therapy by TIBs-dependent immunotherapy.
Collapse
Affiliation(s)
- Li Guan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Zipeng Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Tong Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Yang Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Qihui Chu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Yuxiao Fang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, China
| |
Collapse
|
12
|
Worsley CM, Veale RB, Mayne ES. The acidic tumour microenvironment: Manipulating the immune response to elicit escape. Hum Immunol 2022; 83:399-408. [PMID: 35216847 DOI: 10.1016/j.humimm.2022.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/18/2022]
Abstract
The success of cancer treatment relies on the composition of the tumour microenvironment which is comprised of tumour cells, blood vessels, stromal cells, immune cells, and extracellular matrix components. Barriers to effective cancer treatment need to be overcome, and the acidic microenvironment of the tumour provides a key target for treatment. This review intends to provide an overview of the effects that low extracellular pH has on components of the tumour microenvironment and how they contribute to immune escape. Further, potential therapeutic targets will be discussed.
Collapse
Affiliation(s)
- Catherine M Worsley
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, South Africa; Department of Haematology and Molecular Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa; National Health Laboratory Service, South Africa.
| | - Rob B Veale
- School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, South Africa
| | - Elizabeth S Mayne
- Department of Haematology and Molecular Medicine, Faculty of Health Sciences, University of the Witwatersrand, South Africa; Department of Immunology Faculty of Health Sciences, University of the Witwatersrand, South Africa; Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, South Africa
| |
Collapse
|
13
|
Liu K, Hoover AR, Krawic JR, DeVette CI, Sun XH, Hildebrand WH, Lang ML, Axtell RC, Chen WR. Antigen presentation and interferon signatures in B cells driven by localized ablative cancer immunotherapy correlate with extended survival. Am J Cancer Res 2022; 12:639-656. [PMID: 34976205 PMCID: PMC8692917 DOI: 10.7150/thno.65773] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/11/2021] [Indexed: 12/14/2022] Open
Abstract
Rationale: B cells have emerged as key regulators in protective cancer immunity. However, the activation pathways induced in B cells during effective immunotherapy are not well understood. Methods: We used a novel localized ablative immunotherapy (LAIT), combining photothermal therapy (PTT) with intra-tumor delivery of the immunostimulant N-dihydrogalactochitosan (GC), to treat mice bearing mouse mammary tumor virus-polyoma middle tumor-antigen (MMTV-PyMT). We used single-cell RNA sequencing to compare the transcriptional changes induced by PTT, GC and PTT+GC in B cells within the tumor microenvironment (TME). Results: LAIT significantly increased survival in the tumor-bearing mice, compared to the treatment by PTT and GC alone. We found that PTT, GC and PTT+GC increased the proportion of tumor-infiltrating B cells and induced gene expression signatures associated with B cell activation. Both GC and PTT+GC elevated gene expression associated with antigen presentation, whereas GC elevated transcripts that regulate B cell activation and GTPase function and PTT+GC induced interferon response genes. Trajectory analysis, where B cells were organized according to pseudotime progression, revealed that both GC and PTT+GC induced the differentiation of B cells from a resting state towards an effector phenotype. The analyses confirmed upregulated interferon signatures in the differentiated tumor-infiltrating B cells following treatment by PTT+GC but not by GC. We also observed that breast cancer patients had significantly longer survival time if they had elevated expression of genes in B cells that were induced by PTT+GC therapy in the mouse tumors. Conclusion: Our findings show that the combination of local ablation and local application of immunostimulant initiates the activation of interferon signatures and antigen-presentation in B cells which is associated with positive clinical outcomes for breast cancer. These findings broaden our understanding of LAIT's regulatory roles in remodeling TME and shed light on the potentials of B cell activation in clinical applications.
Collapse
|
14
|
Yan Q. The Yin-Yang Dynamics in Cancer Pharmacogenomics and Personalized Medicine. Methods Mol Biol 2022; 2547:141-163. [PMID: 36068463 DOI: 10.1007/978-1-0716-2573-6_6] [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] [Indexed: 06/15/2023]
Abstract
The enormous heterogeneity of cancer systems has made it very challenging to overcome drug resistance and adverse reactions to achieve personalized therapies. Recent developments in systems biology, especially the perception of cancer as the complex adaptive system (CAS), may help meet the challenges by deciphering the interactions at various levels from the molecular, cellular, tissue-organ, to the whole organism. The ubiquitous Yin-Yang interactions among the coevolving components, including the genes and proteins, decide their spatiotemporal features at various stages from cancer initiation to metastasis. The Yin-Yang imbalances across different systems levels, from genetic mutations to tumor cells adaptation, have been related to the intra- and inter-tumoral heterogeneity in the micro- and macro-environments. At the molecular and cellular levels, dysfunctional Yin-Yang dynamics in the cytokine networks, mitochondrial activities, redox systems, apoptosis, and metabolism can contribute to tumor cell growth and escape of immune surveillance. Up to the organism and system levels, the Yin-Yang imbalances in the cancer microenvironments can lead to different phenotypes from breast cancer to leukemia. These factors may be considered the systems-based biomarkers and treatment targets. The features of adaptation and nonlinearity in Yin-Yang dynamical interactions should be addressed by individualized drug combinations, dosages, intensities, timing, and frequencies at different cancer stages. The comprehensive "Yin-Yang dynamics" framework would enable powerful approaches for personalized and systems medicine strategies.
Collapse
|
15
|
CXCL13 in Cancer and Other Diseases: Biological Functions, Clinical Significance, and Therapeutic Opportunities. Life (Basel) 2021; 11:life11121282. [PMID: 34947813 PMCID: PMC8708574 DOI: 10.3390/life11121282] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/31/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
The development of cancer is a multistep and complex process involving interactions between tumor cells and the tumor microenvironment (TME). C-X-C chemokine ligand 13 (CXCL13) and its receptor, CXCR5, make crucial contributions to this process by triggering intracellular signaling cascades in malignant cells and modulating the sophisticated TME in an autocrine or paracrine fashion. The CXCL13/CXCR5 axis has a dominant role in B cell recruitment and tertiary lymphoid structure formation, which activate immune responses against some tumors. In most cancer types, the CXCL13/CXCR5 axis mediates pro-neoplastic immune reactions by recruiting suppressive immune cells into tumor tissues. Tobacco smoke and haze (smohaze) and the carcinogen benzo(a)pyrene induce the secretion of CXCL13 by lung epithelial cells, which contributes to environmental lung carcinogenesis. Interestingly, the knockout of CXCL13 inhibits benzo(a)pyrene-induced lung cancer and azoxymethane/dextran sodium sulfate-induced colorectal cancer in mice. Thus, a better understanding of the context-dependent functions of the CXCL13/CXCR5 axis in tumor tissue and the TME is required to design an efficient immune-based therapy. In this review, we summarize the molecular events and TME alterations caused by CXCL13/CXCR5 and briefly discuss the potentials of agents targeting this axis in different malignant tumors.
Collapse
|
16
|
Trivedi T, Pagnotti GM, Guise TA, Mohammad KS. The Role of TGF-β in Bone Metastases. Biomolecules 2021; 11:1643. [PMID: 34827641 PMCID: PMC8615596 DOI: 10.3390/biom11111643] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Complications associated with advanced cancer are a major clinical challenge and, if associated with bone metastases, worsen the prognosis and compromise the survival of the patients. Breast and prostate cancer cells exhibit a high propensity to metastasize to bone. The bone microenvironment is unique, providing fertile soil for cancer cell propagation, while mineralized bone matrices store potent growth factors and cytokines. Biologically active transforming growth factor β (TGF-β), one of the most abundant growth factors, is released following tumor-induced osteoclastic bone resorption. TGF-β promotes tumor cell secretion of factors that accelerate bone loss and fuel tumor cells to colonize. Thus, TGF-β is critical for driving the feed-forward vicious cycle of tumor growth in bone. Further, TGF-β promotes epithelial-mesenchymal transition (EMT), increasing cell invasiveness, angiogenesis, and metastatic progression. Emerging evidence shows TGF-β suppresses immune responses, enabling opportunistic cancer cells to escape immune checkpoints and promote bone metastases. Blocking TGF-β signaling pathways could disrupt the vicious cycle, revert EMT, and enhance immune response. However, TGF-β's dual role as both tumor suppressor and enhancer presents a significant challenge in developing therapeutics that target TGF-β signaling. This review presents TGF-β's role in cancer progression and bone metastases, while highlighting current perspectives on the therapeutic potential of targeting TGF-β pathways.
Collapse
Affiliation(s)
- Trupti Trivedi
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Gabriel M. Pagnotti
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Theresa A. Guise
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
| | - Khalid S. Mohammad
- Department of Endocrine Neoplasia and Hormonal Disorders, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.T.); (G.M.P.); (T.A.G.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
17
|
Cai Z, Teng Y, Chen Y. The Effect of Shenyi Capsule on Non-Small-Cell Lung Cancer Combined with Chemotherapy from the Yin-Yang Perspective. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:1653750. [PMID: 34512772 PMCID: PMC8426066 DOI: 10.1155/2021/1653750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/26/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
As an example of Shenyi capsule on non-small-cell lung cancer combined with chemotherapy, this review discusses the synergistic effect and mechanism of natural drugs in oncotherapy from the yin-yang perspective in ancient Chinese philosophy, so as to reflect the therapeutic principle of natural drugs for tumor more comprehensively. The major focuses of this review are on the philosophical thinking of yin-yang as a tool which can not only explain the effect of Shenyi capsule in NSCLC combined with chemotherapy but also explore the mechanism of Shenyi capsule at the cellular and molecular level. Learning from the "yin-yang" thinking of ancient Chinese philosophy will bring more enlightenment to the research and development of traditional Chinese drugs in the future.
Collapse
Affiliation(s)
- Zhixing Cai
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Yue Teng
- Outpatient Department of Clinic Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai 200437, China
| | - Yue Chen
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| |
Collapse
|
18
|
Diagnostic and prognostic value of tumor-infiltrating B cells in lymph node metastases of papillary thyroid carcinoma. Virchows Arch 2021; 479:947-959. [PMID: 34148127 DOI: 10.1007/s00428-021-03137-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/18/2021] [Accepted: 06/03/2021] [Indexed: 10/21/2022]
Abstract
Lymph node metastases are strongly associated with unfavorable prognosis in papillary thyroid carcinoma (PTC) patients. However, there are few sensitive or specific indicators that can diagnose or predict lymph node metastases in PTC. The objective of our study was to identify reliable indicators for the diagnosis and prediction of lymph node metastases of PTC. The PTC data set was obtained from The Cancer Genome Atlas (TCGA) cohort. Information on tumor-infiltrating immune cells in PTC was acquired using single-sample gene set enrichment analysis (ssGSEA). Then, the progression-free survival (PFS) rates of PTC patients were evaluated by Kaplan-Meier curves. A tissue microarray including 58 normal thyroid tissues and 57 PTC tissues was processed for CD19 immunohistochemistry staining. Finally, evaluation of phenotype permutations was performed using gene set enrichment analysis (GSEA). There was an appreciable association between immune infiltration and lymph node metastases in PTC. Among those immune cells, B cells and cytotoxic cells showed significant predictive accuracy for lymph node metastases in PTC. Tumor-infiltrating B cells and NK cells were associated with favorable prognosis, while tumor-associated NK CD56bright cells were correlated with poor prognosis in PTC patients. IHC analyses of PTC further confirmed a notably negative correlation between B cell infiltration and lymph node metastases in PTC. Additionally, mutations in BRAF, a dominant cause of tumor mutation burden (TMB), were positively correlated with reduced B cell infiltration and lymph node metastases in PTC. GSEA revealed that epithelial-mesenchymal transition, IL-6/JAK/STAT3 signaling, the inflammatory response, and TNF-α signaling via the NFκB pathway were remarkably suppressed pathways in patients with BRAF mutations. Tumor-associated lymphocytic infiltration, especially B cell infiltration, provides diagnostic and prognostic value for lymph node metastases in PTC.
Collapse
|
19
|
Qin Y, Peng F, Ai L, Mu S, Li Y, Yang C, Hu Y. Tumor-infiltrating B cells as a favorable prognostic biomarker in breast cancer: a systematic review and meta-analysis. Cancer Cell Int 2021; 21:310. [PMID: 34118931 PMCID: PMC8199375 DOI: 10.1186/s12935-021-02004-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 06/01/2021] [Indexed: 01/01/2023] Open
Abstract
Background Tumor-infiltrating B lymphocytes (TIL-Bs) is a heterogeneous population of lymphocytes. The prognostic value of TIL-Bs in patients with breast cancer remains controversial. Here we conducted this meta-analysis to clarify the association of TIL-Bs with outcomes of patients with breast cancer. Methods We searched PubMed, Embase, and Web of Science to identify relevant studies assessing the prognostic significance of TIL-Bs in patients with breast cancer. Fixed- or random-effects models were used to evaluate the pooled hazard ratios (HRs) for overall survival (OS), breast cancer-specific survival (BCSS), disease-free survival (DFS), and relapse-free survival (RFS) in breast cancer. Results
A total of 8 studies including 2628 patients were included in our study. Pooled analyses revealed that high level of TIL-Bs was associated with longer OS (pooled HR = 0.42, 95% CI 0.24–0.60), BCSS (pooled HR = 0.66, 95% CI 0.47–0.85), and DFS/RFS (pooled HR = 0.41, 95% CI 0.27–0.55). Conclusions This meta-analysis suggests that TIL-Bs could be a promising prognostic marker for breast cancer. Novel therapeutic strategies for breast cancer treatment could be developed by enhancement of B cell-mediated antitumor immunity. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02004-9.
Collapse
Affiliation(s)
- You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fei Peng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China
| | - Lisha Ai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China.
| | - Shidai Mu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China
| | - Yuting Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chensu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China.
| |
Collapse
|
20
|
Ren Y, Liang S, Zheng Y, Deng X, Lei L, Ai J, Li Y, Zhang T, Chen L, Mei Z, Cheng YC, He C. Investigation on the function tropism of Tiaoqin and Kuqin (different specification of Scutellaria baicalensis) by comparing their curative effect on different febrile disease model. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113596. [PMID: 33221498 DOI: 10.1016/j.jep.2020.113596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 11/13/2020] [Accepted: 11/14/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellaria baicalensis (S. baicalensis) is the root of S. baicalensis Georgi. In traditional Chinese medicine it is divided into Tiaoqin (TQ, 1-3 years old) and Kuqin (KQ, more than 3 years old). However, the differences in TQ and KQ efficacy and their exact mechanisms are still unclear. AIM OF THE STUDY This study aimed to clarify the difference in the efficacy of TQ and KQ in relation to different fever types (damp heat and hyperpyrexia) by using rat models, as well as to determine the primary molecular mechanism. MATERIALS AND METHODS This study compared the compositional content of TQ and KQ by UPLC-MS/MS. Then, rat models of hyperpyrexia (HP, LPS) and damp heat (DH, high-fat and high-sugar diet feeding + fumigation in artificial climate chamber + E. coli injection) were established and their clinical symptoms, blood biochemistry, histopathological sections, cell cytokines and protein expression were compared following treatment with TQ or KQ. Finally, the mechanisms underpinning the differences observed for TQ and KQ were determined by measuring the components of these treatments in different target organs. RESULTS This study identified 31 compounds in the water extracts of both TQ and KQ, which differed significantly in their relative content. TQ and KQ showed different functional tropism in HP and DH model rats. Baicalin, wogonoside, oroxin A, baicalein, wogonin and oroxylin A appeared to be the basic functional components responsible for the functional tropism hypothesis, while the remaining compounds appeared to be the efficacy-oriented components. In addition, the difference in pharmacodynamics between TQ and KQ may be related to their absorption in vivo, which was consistent with the hypothesis of functional tropism proposed in this work. CONCLUSION In this study we adopted TQ and KQ-different specifications of Scutellaria baicalensis with similar chemical components-as a case study to systematically reveal the functional tropism of Chinese herbal medicine (CHM). The results showed that TQ and KQ contain the basic functional components to enable the basic function of 'clearing heat', while the variation in compositional content may result in their different therapeutic effects. A greater understanding and utilisation of the functional tropism of CHM would enormously improve the accuracy and scientific basis for the application of CHM medication, as well as in promoting the multi-function mechanism of CHM and guiding new drug development of CHM.
Collapse
Affiliation(s)
- Yongshen Ren
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China; School of Medicine, Yale University, New Haven, CT, 06511, USA.
| | - Shuai Liang
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Yao Zheng
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Xin Deng
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Lei Lei
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Jiao Ai
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Yanqiu Li
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Tianpei Zhang
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Linlin Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhinan Mei
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| | - Yung-Chi Cheng
- School of Medicine, Yale University, New Haven, CT, 06511, USA.
| | - Caijing He
- School of Pharmaceutical Science, South-central University for Nationalities, Wuhan, 430074, China
| |
Collapse
|
21
|
Schnellhardt S, Erber R, Büttner-Herold M, Rosahl MC, Ott OJ, Strnad V, Beckmann MW, King L, Hartmann A, Fietkau R, Distel L. Tumour-Infiltrating Inflammatory Cells in Early Breast Cancer: An Underrated Prognostic and Predictive Factor? Int J Mol Sci 2020; 21:ijms21218238. [PMID: 33153211 PMCID: PMC7663093 DOI: 10.3390/ijms21218238] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
The role of tumour-infiltrating inflammatory cells (TIICs) in the disease progression of hormone-receptor-positive breast cancer (HR+ BC) is largely unclear since it is generally regarded as the least immunogenic BC subtype. This study investigated the prognostic significance of CD1a+ dendritic cells, CD20+ B cells, CD45RO+ memory T cells and CD4+ T-helper cells in HR+ BC. One hundred and forty-six patients were treated for early stage, distant-metastases-free HR+ BC in an accelerated partial breast irradiation (APBI) phase II trial. Immunohistochemistry was used to double-stain two adjoining sets of tissue microarrays from pre-RT (radiotherapy) tumour resection samples for CD1a/CD20 and CD45RO/CD4. Cell densities of CD1a+, CD20+, CD45RO+ and CD4+ TIICs in the stromal and intraepithelial compartment were registered semiautomatically. High densities of CD20+ and CD4+ TIICs were strongly associated with reduced disease-free survival (DFS), while high stromal CD45RO+ TIIC densities were indicators of subsequent successful treatment. An immunoscore based on CD20+ and CD45RO+ TIIC densities identified three different risk groups (p < 0.001). Thus, contrary to current assumptions, intratumoural immune cell composition might be an important prognostic indicator and a possible contributing factor in the outcome of HR+ BC and should be the subject of further research. Specifically, B-cell infiltration entailed an increased relapse rate and could play an important role in disease progression.
Collapse
Affiliation(s)
- Sören Schnellhardt
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (S.S.); (M.-C.R.); (O.J.O.); (V.S.); (R.F.)
| | - Ramona Erber
- Institute of Pathology, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (R.E.); (A.H.)
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany;
| | - Marie-Charlotte Rosahl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (S.S.); (M.-C.R.); (O.J.O.); (V.S.); (R.F.)
| | - Oliver J. Ott
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (S.S.); (M.-C.R.); (O.J.O.); (V.S.); (R.F.)
| | - Vratislav Strnad
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (S.S.); (M.-C.R.); (O.J.O.); (V.S.); (R.F.)
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany;
| | - Lillian King
- Intensive Care Unit, QEII Jubilee Hospital, Brisbane, Queensland 4108, Australia;
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (R.E.); (A.H.)
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (S.S.); (M.-C.R.); (O.J.O.); (V.S.); (R.F.)
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (S.S.); (M.-C.R.); (O.J.O.); (V.S.); (R.F.)
- Correspondence: ; Tel.: +49-9131-853-2312; Fax: +49-9131-853-9335
| |
Collapse
|
22
|
He W, Zhang D, Liu H, Chen T, Xie J, Peng L, Zheng X, Xu B, Li Q, Jiang J. The High Level of Tertiary Lymphoid Structure Is Correlated With Superior Survival in Patients With Advanced Gastric Cancer. Front Oncol 2020; 10:980. [PMID: 32733793 PMCID: PMC7358602 DOI: 10.3389/fonc.2020.00980] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/18/2020] [Indexed: 01/05/2023] Open
Abstract
Background: A tertiary lymphoid structure (TLS) is a crucial component of the tumor microenvironment, which reflects the anti-tumor immune response in the host. The aim of the present study was to carry out a histopathological evaluation for TLS and assess its prognostic value in gastric cancer (GC). Methods: A total of 1,033 cases that have received a gastrectomy were reviewed, including 914 in the primary cohort and 119 in the validation cohort. TLS was assessed by optical microscopy and verified by immunohistochemistry. A total of five histopathological evaluation methods were compared in the primary cohort and validated in the validation cohort. In addition, MECA-79 and CD21 were used to verify the accuracy of the histopathological scoring system for TLS. The association among TLS, clinicopathological parameters, and patient prognosis was analyzed. Results: TLS as assessed by morphology and immunohistochemistry were significantly correlated and consistent. The morphological evaluation of TLS was accurate. Typically, the high level of TLS was significantly correlated with tumor size (P = 0.047), histological grade (P = 0.039), pTN stage (P = 0.044), and WHO subtype (P < 0.001). In addition, TLShi was a positive indicator of overall survival, as determined by Kaplan–Meier survival (P = 0.038) and multivariate Cox regression analyses (hazard ratio = 0.794, 95% CI: 0.668–0.942, P = 0.008). According to the results, TLShi had a positive effect on the primary cohort patients with pTN stages II and III (P = 0.027, P = 0.042). Conclusions: The histopathological evaluation of TLS was accurate. Diagnosis based solely on hematoxylin and eosin staining of the sections did not easily distinguish tumor-associated TLS. The density of TLS in the center of the tumor was found to be more indicative of patient prognosis than TLS in the invasive margin, with the levels of total TLS shown to best correlate with overall survival in patients with advanced-stage GC.
Collapse
Affiliation(s)
- Wenting He
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dachuan Zhang
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hong Liu
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Tongbing Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Xie
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lei Peng
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Qing Li
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| |
Collapse
|
23
|
Conejo-Garcia JR, Biswas S, Chaurio R. Humoral immune responses: Unsung heroes of the war on cancer. Semin Immunol 2020; 49:101419. [PMID: 33183950 PMCID: PMC7738315 DOI: 10.1016/j.smim.2020.101419] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/23/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
Solid cancers progress from primordial lesions through complex interactions between tumor-promoting and anti-tumor immune cell types, ultimately leading to the orchestration of humoral and T cell adaptive immune responses, albeit in an immunosuppressive environment. B cells infiltrating most established tumors have been associated with a dual role: Some studies have associated antibodies produced by tumor-associated B cells with the promotion of regulatory activities on myeloid cells, and also with direct immunosuppression through the production of IL-10, IL-35 or TGF-β. In contrast, recent studies in multiple human malignancies identify B cell responses with delayed malignant progression and coordinated T cell protective responses. This includes the elusive role of Tertiary Lymphoid Structures identified in many human tumors, where the function of B cells remains unknown. Here, we discuss emerging data on the dual role of B cell responses in the pathophysiology of human cancer, providing a perspective on future directions and possible novel interventions to restore the coordinated action of both branches of the adaptive immune response, with the goal of maximizing immunotherapeutic effectiveness.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ricardo Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
24
|
Liang S, Deng X, Lei L, Zheng Y, Ai J, Chen L, Xiong H, Mei Z, Cheng YC, Ren Y. The Comparative Study of the Therapeutic Effects and Mechanism of Baicalin, Baicalein, and Their Combination on Ulcerative Colitis Rat. Front Pharmacol 2019; 10:1466. [PMID: 31920656 PMCID: PMC6923254 DOI: 10.3389/fphar.2019.01466] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: Ulcerative colitis (UC) is an inflammatory bowel disease with a high incidence rate and a difficult treatment regimen. Recently, significant advances in the treatment of intestinal diseases, particularly UC, have been made with the use of the drugs baicalin and baicalein, separately or in combination. However, the therapeutic efficacy and mechanism of action of baicalin, baicalein, and their combination therapy, in the treatment of UC has not been fully elucidated. Materials and Methods: we constructed a UC rat model that encompassed a variety of complex factors, including a high-sugar and high-fat diet, a high temperature and humidity environment (HTHE), excess drinking, and infection of Escherichia coli. Model rats were then treated with baicalin, baicalein, or a combination of the two. Results: The results showed significant differences in the therapeutic effects of baicalin, baicalein, and the combination therapy, in the treatment of UC, as well as differences in the inhibition of inflammation via the nuclear factor-κB and MAPK pathways. The rat model of UC was established as described above. Then, the rats were treated for 7 days with baicalin (100 mg kg-1), baicalein (100 mg kg-1), or both (100 mg kg-1, baicalin: baicalein = 4:1/1:1). Clinical symptoms and signs, body temperature, organ indices, histopathology, blood biochemistry, and metabolites were examined to compare treatment effects and indicators of UC. Baicalin, YSR (Young Scutellaria baicalensis ratio of baicalin and baicalein), baicalein, and WSR (Withered Scutellaria baicalensis ratio of baicalin and baicalein) had significantly different effects in terms of clinical symptoms and signs, body temperature, organ indices, serum inflammatory cytokine levels, blood biochemistry, and histopathology changes in the main organs; YSR exhibited the best treatment effects. LC-MS/MS was used to detect the conversion of baicalin, baicalein, or both, into the six types of metabolites: baicalin, wogonoside, oroxin A, baicalein, wogonin, and oroxylin A. The levels of the six metabolites under the different treatment conditions were significantly different in the large intestine, small intestine, and lungs, but not in the blood. The levels of the six metabolites were significantly different in the large intestine, small intestine, and lung, but not in the serum. Conclusion: All these results indicate that baicalin and baicalein should be used more accurately in specific diseases, especially baicalin or high content of baicalin in Scutellaria baicalensis (Tiaoqin) should be preferred in treatment of UC.
Collapse
Affiliation(s)
- Shuai Liang
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Xin Deng
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Lei Lei
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Yao Zheng
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Jiao Ai
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Linlin Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Hui Xiong
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Zhinan Mei
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China
| | - Yung-Chi Cheng
- School of Medicine, Yale University, New Haven, CT, United States
| | - Yongshen Ren
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan, China.,School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
25
|
Schiano C, Franzese M, Pane K, Garbino N, Soricelli A, Salvatore M, de Nigris F, Napoli C. Hybrid 18F-FDG-PET/MRI Measurement of Standardized Uptake Value Coupled with Yin Yang 1 Signature in Metastatic Breast Cancer. A Preliminary Study. Cancers (Basel) 2019; 11:cancers11101444. [PMID: 31561604 PMCID: PMC6827137 DOI: 10.3390/cancers11101444] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose: Detection of breast cancer (BC) metastasis at the early stage is important for the assessment of BC progression status. Image analysis represents a valuable tool for the management of oncological patients. Our preliminary study combined imaging parameters from hybrid 18F-FDG-PET/MRI and the expression level of the transcriptional factor Yin Yang 1 (YY1) for the detection of early metastases. Methods: The study enrolled suspected n = 217 BC patients that underwent 18F-FDG-PET/MRI scans. The analysis retrospectively included n = 55 subjects. n = 40 were BC patients and n = 15 imaging-negative female individuals were healthy subjects (HS). Standard radiomics parameters were extracted from PET/MRI image. RNA was obtained from peripheral blood mononuclear cells and YY1 expression level was evaluated by real time reverse transcription polymerase chain reactions (qRT-PCR). An enzyme-linked immuosorbent assay (ELISA) was used to determine the amount of YY1 serum protein. Statistical comparison between subgroups was evaluated by Mann-Whitney U and Spearman’s tests. Results: Radiomics showed a significant positive correlation between Greg-level co-occurrence matrix (GLCM) and standardized uptake value maximum (SUVmax) (r = 0.8 and r = 0.8 respectively) in BC patients. YY1 level was significant overexpressed in estrogen receptor (ER)-positive/progesteron receptor-positive/human epidermal growth factor receptor2-negative (ER+/PR+/HER2-) subtype of BC patients with synchronous metastasis (SM) at primary diagnosis compared to metachronous metastasis (MM) and HS (p < 0.001) and correlating significantly with 18F-FDG-uptake parameter (SUVmax) (r = 0.48). Conclusions: The combination of functional 18F-FDG-PET/MRI parameters and molecular determination of YY1 could represent a novel integrated approach to predict synchronous metastatic disease with more accuracy than 18F-FDG-PET/MRI alone.
Collapse
Affiliation(s)
| | | | | | | | - Andrea Soricelli
- IRCCS SDN, 80134 Naples, Italy
- Department of Motor Sciences and Healthiness, University of Naples Parthenope, 80134 Naples, Italy
| | | | - Filomena de Nigris
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Claudio Napoli
- IRCCS SDN, 80134 Naples, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
26
|
Chakraborty D, Pati S, Bose S, Dhar S, Dutta S, Sa G. Cancer immunotherapy: present scenarios and the future of immunotherapy. THE NUCLEUS 2019. [DOI: 10.1007/s13237-019-00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
27
|
Liu M, Sun Q, Wang J, Wei F, Yang L, Ren X. A new perspective: Exploring future therapeutic strategies for cancer by understanding the dual role of B lymphocytes in tumor immunity. Int J Cancer 2018; 144:2909-2917. [PMID: 30183084 DOI: 10.1002/ijc.31850] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/23/2018] [Accepted: 08/29/2018] [Indexed: 12/16/2022]
Abstract
Our previous understanding of the role of B lymphocytes in tumor immunity is its antitumor effects. However, further evidence indicates B lymphocytes can also promote tumorigenesis by modulating immune responses. Therefore, the increasingly complex role of B lymphocytes in tumor immunity may become an important factor in tumor immunotherapy. In this review, we describe the development of B cells in tumor microenvironments. We then focus on the most controversial issues of the biological functions of B lymphocytes. Finally, we nominate B cells as therapeutic targets, which should open broad perspectives for the development of their clinical applications.
Collapse
Affiliation(s)
- Min Liu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
28
|
Lin Z, Liu L, Xia Y, Chen X, Xiong Y, Qu Y, Wang J, Bai Q, Guo J, Xu J. Tumor infiltrating CD19 + B lymphocytes predict prognostic and therapeutic benefits in metastatic renal cell carcinoma patients treated with tyrosine kinase inhibitors. Oncoimmunology 2018; 7:e1477461. [PMID: 30288343 DOI: 10.1080/2162402x.2018.1477461] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/28/2018] [Accepted: 05/13/2018] [Indexed: 12/31/2022] Open
Abstract
The objective response rate (ORR) of tyrosine kinase inhibitors (TKIs) therapy in metastatic renal cell cancer (mRCC) patients was not satisfactory. Effective indicator of mRCC patient selection for TKI therapy is urgently needed. The function of tumor infiltrating B lymphocytes (TIBs) in tumor immune elimination is still unclear. We aim to investigate the prognostic and predictive value of TIBs for TKI therapy in mRCC patients in this study. 108 eligible patients treated with TKI were enrolled in this study. TIBs was estimated by immunohistochemical staining of CD19 in the resected tumor, and its relationship with clinicopathological features, clinical outcomes and CD8+ tumor infiltrating T lymphocytes (CD8+ TILs) were evaluated. Associations between the expression level of CD19 and CD8+ TILs associated cytotoxic effectors were also assessed in public databases. Results showed TIBs positive infiltration predicted better therapeutic response to sunitinib (p = 0.006), longer overall survival (OS) (p < 0.001) and progression-free survival (PFS) (p = 0.028) in mRCC patients. Combining TIBs and International Metastatic Renal-Cell Carcinoma Database Consortium (IMDC) model showed a better predict value of OS in TKI-treated mRCC patients than IMDC model alone. We also found a positive correlation between TIBs and CD8+ TILs (p < 0.001). Patients with both cells high infiltration showed markedly better OS compared with those infiltrated by CD8+ T cells alone (p = 0.015). To conclude, TIBs density was not only an independent prognostic factor for mRCC patients, but also a predictive marker for TKI therapy response. It may potently enhance the antitumor effect by recruiting and activating CD8+ TILs in mRCC.
Collapse
Affiliation(s)
- Zhiyuan Lin
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Yu Xia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Xiang Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Ying Xiong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Yang Qu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Jiajun Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Qi Bai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai China
| |
Collapse
|
29
|
Wei S. Yin-yang regulating effects of cancer-associated genes, proteins, and cells: An ancient Chinese concept in vogue in modern cancer research. Biosci Trends 2017; 11:612-618. [PMID: 29238002 DOI: 10.5582/bst.2017.01259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Great achievements have been made in human cancer research, but most of this research is focused on conditions at the microscopic rather than the systemic level. Recent studies have increasingly cited the ancient Chinese theory of yin-yang in an effort to expand beyond the microscopic level. Various cancer-associated genes and proteins such as mitogen-activated protein kinase (MAPK), p38, p53, c-Myc, tumor necrosis factor (TNF)-α, NF-κB, Cyclin D1, and cyclin-dependent kinase (CDK) and cells such as T cells, B cells, macrophages, neutrophils, and fibroblasts have been reported to regulate various types of cancers in a yin-yang manner. These studies have brought the theory of yin-yang into vogue in cancer research worldwide.
Collapse
Affiliation(s)
- Shuyong Wei
- College of Animal Science, Southwest University
| |
Collapse
|
30
|
The roles and applications of autoantibodies in progression, diagnosis, treatment and prognosis of human malignant tumours. Autoimmun Rev 2017; 16:1270-1281. [PMID: 29042252 DOI: 10.1016/j.autrev.2017.10.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023]
Abstract
The existence of autoantibodies towards an individual's own proteins or nucleic acids has been established for more than 100years, and for a long period, these autoantibodies have been believed to be closely associated with autoimmune diseases. However, in recent years, researchers have become more interested in the role and application of autoantibodies in progression, diagnosis, treatment and prognosis of human malignant tumours. Over the past few decades, numerous epidemiological studies have shown that the risk of certain cancers is significantly altered (increased or decreased) in patients with autoimmune diseases, which suggests that autoantibodies may play either promoting or suppressing roles in cancer progression. The idea that autoantibodies are directly involved in tumour progression gains special support by the findings that some antibodies secreted by a variety of cancer cells can promote their proliferation and metastasis. Because the cancer cells generate cell antigenic changes (neoantigens), which trigger the immune system to produce autoantibodies, serum autoantibodies against tumour-associated antigens have been established as a novel type of cancer biomarkers and have been extensively studied in different types of cancer. The autoantibodies as biomarkers in cancer diagnosis are not only more sensitive and specific than antigens, but also could appear before clinical evidences of the tumours, thus disclosing them. The observations that cancer risk is lower in patients with some autoimmune diseases suggest that certain autoantibodies may be protective from certain cancers. Moreover, the presence of autoantibodies in healthy individuals implies that it could be safe to employ autoantibodies to treat cancer. Of note, an autoantibodies derived from lupus murine model received much attention due to their selective cytotoxicity for malignant tumour cell without harming normal ones. These studies showed the therapeutic value of autoantibodies in cancer. In this review, we revisited the pathological or protective role of autoantibodies in cancer progression, summarize the application of autoantibodies in cancer diagnosis and prognosis, and discuss the value of autoantibodies in cancer therapy. The studies established to date suggest that autoantibodies not only regulate cancer progression but also promise to be valuable instruments in oncological diagnosis and therapy.
Collapse
|