1
|
Li S, Yao ZC, Wang H, Ecker JA, Omotoso MO, Lee J, Kong J, Feng H, Chaisawangwong W, Kang SS, Shannon SR, Livingston NK, Bieler JG, Singh S, Zhang ML, O’Neal P, Ariail E, Biggs B, Hickey JW, Mao HQ, Schneck JP. Ex vivo expansion and hydrogel-mediated in vivo delivery of tissue-resident memory T cells for immunotherapy. SCIENCE ADVANCES 2024; 10:eadm7928. [PMID: 39671478 PMCID: PMC11641059 DOI: 10.1126/sciadv.adm7928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/06/2024] [Indexed: 12/15/2024]
Abstract
Tissue-resident memory T (TRM) cells preferentially reside in peripheral tissues, serving as key players in tumor immunity and immunotherapy. The lack of effective approaches for expanding TRM cells and delivering these cells in vivo hinders the exploration of TRM cell-mediated cancer immunotherapy. Here, we report a nanoparticle artificial antigen-presenting cell (nano-aAPC) ex vivo expansion approach and an in vivo delivery system for TRM cells. Using the nano-aAPC platform, we expanded functional antigen-specific murine and human TRM-like CD8+ T cells ex vivo. We also developed an injectable macroporous hyaluronic acid (HA) hydrogel to deliver TRM-like cells. TRM-like cells delivered in the optimized HA hydrogel trigger robust local and systemic antitumor immunity and show synergistic effects with anti-PD-1 treatment. Our findings suggest that nano-aAPC-induced TRM-like cells, coupled with a hydrogel delivery system, offer an efficient way to advance the understanding of TRM cell-mediated cancer therapy.
Collapse
Affiliation(s)
- Shuyi Li
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Translational Immunoengineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Zhi-Cheng Yao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hanzhi Wang
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan A. Ecker
- Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Mary O. Omotoso
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jaechan Lee
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jiayuan Kong
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hexiang Feng
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | - Si-Sim Kang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sydney R. Shannon
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Natalie K. Livingston
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Joan G. Bieler
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shweta Singh
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Maya L. Zhang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Pilar O’Neal
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Emily Ariail
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Benjamin Biggs
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - John W. Hickey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Translational Immunoengineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan P. Schneck
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Johns Hopkins Translational Immunoengineering Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Yang C, Zhang Y, Wang R, Cheng B, Wu Y, Fu X. IL-10 +CD19 + regulatory B cells induce CD4 +Foxp3 +regulatory T cells in serum of cervical cancer patients. Autoimmunity 2024; 57:2290909. [PMID: 38084896 DOI: 10.1080/08916934.2023.2290909] [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: 11/01/2022] [Accepted: 11/27/2023] [Indexed: 12/18/2023]
Abstract
Increase of regulatory T cells (Tregs) in the tumour microenvironment predicts worse survival of patients with various types of cancer. Recently, B cells play a significant role in the maintenance of Treg cells. However, the relevance of regulatory B cells (Bregs) to tumour immunity in humans remains elusive. Flow cytometry analysis was used to detect the Bregs and Tregs. Double staining results illustrated that the proportion of Bregs and Tregs were prominently higher in cervical cancer than normal tissues. Increase of Bregs and Tregs in cervical cancer microenvironment was associated with poor survival. Furthermore, Bregs cocultured with cervical cancer cell lines increased and induced Tregs. To sum up, the increased expression of Bregs contributes to the differentiation of CD4+ T cells into Tregs in the cervical cancer.
Collapse
Affiliation(s)
- Chunfeng Yang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen, China
| | - Yuanyuan Zhang
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Rui Wang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bing Cheng
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - You Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Xi Fu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory for Major Obstetric Diseases; Guangdong Province Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macoa Greater Bay Area Higher Educaiton Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Shen A, Garrett A, Chao CC, Liu D, Cheng C, Wang Z, Qian C, Zhu Y, Mai J, Jiang C. A comprehensive meta-analysis of tissue resident memory T cells and their roles in shaping immune microenvironment and patient prognosis in non-small cell lung cancer. Front Immunol 2024; 15:1416751. [PMID: 39040095 PMCID: PMC11260734 DOI: 10.3389/fimmu.2024.1416751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Tissue-resident memory T cells (TRM) are a specialized subset of long-lived memory T cells that reside in peripheral tissues. However, the impact of TRM-related immunosurveillance on the tumor-immune microenvironment (TIME) and tumor progression across various non-small-cell lung cancer (NSCLC) patient populations is yet to be elucidated. Our comprehensive analysis of multiple independent single-cell and bulk RNA-seq datasets of patient NSCLC samples generated reliable, unique TRM signatures, through which we inferred the abundance of TRM in NSCLC. We discovered that TRM abundance is consistently positively correlated with CD4+ T helper 1 cells, M1 macrophages, and resting dendritic cells in the TIME. In addition, TRM signatures are strongly associated with immune checkpoint and stimulatory genes and the prognosis of NSCLC patients. A TRM-based machine learning model to predict patient survival was validated and an 18-gene risk score was further developed to effectively stratify patients into low-risk and high-risk categories, wherein patients with high-risk scores had significantly lower overall survival than patients with low-risk. The prognostic value of the risk score was independently validated by the Cancer Genome Atlas Program (TCGA) dataset and multiple independent NSCLC patient datasets. Notably, low-risk NSCLC patients with higher TRM infiltration exhibited enhanced T-cell immunity, nature killer cell activation, and other TIME immune responses related pathways, indicating a more active immune profile benefitting from immunotherapy. However, the TRM signature revealed low TRM abundance and a lack of prognostic association among lung squamous cell carcinoma patients in contrast to adenocarcinoma, indicating that the two NSCLC subtypes are driven by distinct TIMEs. Altogether, this study provides valuable insights into the complex interactions between TRM and TIME and their impact on NSCLC patient prognosis. The development of a simplified 18-gene risk score provides a practical prognostic marker for risk stratification.
Collapse
Affiliation(s)
- Aidan Shen
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Aliesha Garrett
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Cheng-Chi Chao
- Department of Pipeline Development, Biomap, Inc., San Francisco, CA, United States
| | - Dongliang Liu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Zhaohui Wang
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Chen Qian
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yangzhi Zhu
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Chongming Jiang
- Department of Precision Medicine, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States
| |
Collapse
|
4
|
Liu S, Wang P, Wang P, Zhao Z, Zhang X, Pan Y, Pan J. Tissue-resident memory CD103+CD8+ T cells in colorectal cancer: its implication as a prognostic and predictive liver metastasis biomarker. Cancer Immunol Immunother 2024; 73:176. [PMID: 38954030 PMCID: PMC11219596 DOI: 10.1007/s00262-024-03709-2] [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: 02/08/2024] [Accepted: 04/19/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Tissue-resident memory CD103+CD8+ T cells (CD103+CD8+ TRMs) are important components of anti-tumor immunity. However, the significance of CD103+CD8+ TRMs in colorectal cancer (CRC) and their advantages remain unclear. METHODS Clinical data and specimens were used to evaluate the significance of CD103+CD8+ TRMs in CRC. A mouse subcutaneous tumorigenesis model and colony-formation assay were conducted to evaluate the anti-tumor effects of CD103+CD8+ TRMs. Finally, the infiltration density and function of CD103+CD8+ TRMs in the tumors were evaluated using flow cytometry. RESULTS In this study, we showed that highly infiltrated CD103+CD8+ TRMs were associated with earlier clinical stage and negative VEGF expression in CRC patients and predicted a favorable prognosis for CRC/CRC liver metastases patients. Interestingly, we also found that CD103+CD8+ TRMs may have predictive potential for whether CRC develops liver metastasis in CRC. In addition, we found a positive correlation between the ratio of the number of α-SMA+ vessels to the sum of the number of α-SMA+ and CD31+ vessels in CRC, and the infiltration level of CD103+CD8+ TRMs. In addition, anti-angiogenic therapy promoted infiltration of CD103+CD8+ TRMs and enhanced their ability to secrete interferon (IFN)-γ, thus further improving the anti-tumor effect. Moreover, in vivo experiments showed that compared with peripheral blood CD8+ T cells, CD103+CD8+ TRMs infused back into the body could also further promote CD8+ T cells to infiltrate the tumor, and they had a stronger ability to secrete IFN-γ, which resulted in better anti-tumor effects. CONCLUSION We demonstrated that CD103+CD8+ TRMs have the potential for clinical applications and provide new ideas for combined anti-tumor therapeutic strategies, such as anti-tumor angiogenesis therapy and CAR-T combined immunotherapy.
Collapse
Affiliation(s)
- Shijin Liu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Penglin Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Peize Wang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Zhan Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Xiaolin Zhang
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Jinan University, Heyuan, 517000, China.
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou, 510632, China.
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
5
|
Cai L, Lai W, Yao D, Gu Y, Liang C, Liu L, Lai J, Yu Z, Zha X, Yu X, Wu X, Chen S, Luo OJ, Li Y, Wang C, Qin P, Huang X, Xu L. High percentage of bone marrow CD8 + tissue-resident-like memory T cells predicts inferior survival in patients with acute myeloid leukemia. BLOOD SCIENCE 2024; 6:e00194. [PMID: 38854481 PMCID: PMC11161300 DOI: 10.1097/bs9.0000000000000194] [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: 04/11/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
Tissue-resident memory T (TRM) cells infiltrating solid tumors could influence tumor progression and the response to immune therapies. However, the proportion and prognostic value of TRM cells in the bone marrow (BM) of patients with acute myeloid leukemia (AML) are unclear. In this study, we used flow cytometry to assay the phenotype of 49 BM samples from patients newly diagnosed with AML (ND-AML). We found that the BM CD8+ effector memory (TEM) cells highly expressed CD69 (CD8+ TRM-like T cells), and their percentage was significantly increased in patients with ND-AML compared with that in healthy individuals (HI). The high percentage of CD8+ TRM-like subset was associated with poor overall survival in our ND-AML cohort. The Kaplan-Meier Plotter database verified a significantly reduced survival rate among patients with high expression of CD8+ TRM-like T cell characteristic genes (CD8A, CD69, and TOX), especially the M4 and M5 subtypes. Phenotypic analysis revealed that the BM CD8+ TRM-like subpopulation exhibited exhausted T cell characteristics, but its high expression of CD27 and CD28 and low expression of CD57 suggested its high proliferative potential. The single-cell proteogenomic dataset confirmed the existence of TRM-like CD8+ T cells in the BM of patients with AML and verified the high expression of immune checkpoints and costimulatory molecules. In conclusion, we found that the accumulation of BM CD8+ TRM-like cells could be an immune-related survival prediction marker for patients with AML.
Collapse
Affiliation(s)
- Letong Cai
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Wenpu Lai
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Danlin Yao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Yinfeng Gu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Chaofeng Liang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Lian Liu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Jing Lai
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Zhi Yu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Xibao Yu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Xiuli Wu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Shaohua Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Chunyan Wang
- Department of Hematology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pengfei Qin
- Department of Hematology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Huang
- Department of Hematology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Department of Hematology, The First Affiliated Hospital Jinan University, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| |
Collapse
|
6
|
Sato H, Meng S, Hara T, Tsuji Y, Arao Y, Sasaki K, Kobayashi S, di Luccio E, Hirotsu T, Satoh T, Doki Y, Eguchi H, Ishii H. Tissue-Resident Memory T Cells in Gastrointestinal Cancers: Prognostic Significance and Therapeutic Implications. Biomedicines 2024; 12:1342. [PMID: 38927549 PMCID: PMC11202222 DOI: 10.3390/biomedicines12061342] [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: 04/26/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Gastrointestinal cancers, which include a variety of esophageal and colorectal malignancies, present a global health challenge and require effective treatment strategies. In the evolving field of cancer immunotherapy, tissue-resident memory T cells (Trm cells) have emerged as important players in the immune response within nonlymphoid tissues. In this review, we summarize the characteristics and functions of Trm cells and discuss their profound implications for patient outcomes in gastrointestinal cancers. Positioned strategically in peripheral tissues, Trm cells have functions beyond immune surveillance, affecting tumor progression, prognosis, and response to immunotherapy. Studies indicate that Trm cells are prognostic markers and correlate positively with enhanced survival. Their presence in the tumor microenvironment has sparked interest in their therapeutic potential, particularly with respect to immune checkpoint inhibitors, which may improve cancer treatment. Understanding how Trm cells work will not only help to prevent cancer spread through effective treatment but will also contribute to disease prevention at early stages as well as vaccine development. The role of Trm cells goes beyond just cancer, and they have potential applications in infectious and autoimmune diseases. This review provides a thorough analysis of Trm cells in gastrointestinal cancers, which may lead to personalized and effective cancer therapies.
Collapse
Affiliation(s)
- Hiromichi Sato
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan; (H.S.)
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan
| | - Sikun Meng
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan; (H.S.)
| | - Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan; (H.S.)
| | - Yoshiko Tsuji
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan; (H.S.)
| | - Yasuko Arao
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan; (H.S.)
| | - Kazuki Sasaki
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan; (H.S.)
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan
| | - Eric di Luccio
- Hirotsu Bio Science Inc., Chiyoda-Ku, Tokyo 102-0094, Japan
| | | | - Taroh Satoh
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita 565-0871, Japan; (H.S.)
| |
Collapse
|
7
|
Ruocco MR, Gisonna A, Acampora V, D’Agostino A, Carrese B, Santoro J, Venuta A, Nasso R, Rocco N, Russo D, Cavaliere A, Altobelli GG, Masone S, Avagliano A, Arcucci A, Fiume G. Guardians and Mediators of Metastasis: Exploring T Lymphocytes, Myeloid-Derived Suppressor Cells, and Tumor-Associated Macrophages in the Breast Cancer Microenvironment. Int J Mol Sci 2024; 25:6224. [PMID: 38892411 PMCID: PMC11172575 DOI: 10.3390/ijms25116224] [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: 02/20/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancers (BCs) are solid tumors composed of heterogeneous tissues consisting of cancer cells and an ever-changing tumor microenvironment (TME). The TME includes, among other non-cancer cell types, immune cells influencing the immune context of cancer tissues. In particular, the cross talk of immune cells and their interactions with cancer cells dramatically influence BC dissemination, immunoediting, and the outcomes of cancer therapies. Tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) represent prominent immune cell populations of breast TMEs, and they have important roles in cancer immunoescape and dissemination. Therefore, in this article we review the features of TILs, TAMs, and MDSCs in BCs. Moreover, we highlight the mechanisms by which these immune cells remodel the immune TME and lead to breast cancer metastasis.
Collapse
Affiliation(s)
- Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Armando Gisonna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Vittoria Acampora
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Anna D’Agostino
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Barbara Carrese
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Jessie Santoro
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Alessandro Venuta
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Rosarita Nasso
- Department of Movement Sciences and Wellness, University of Naples “Parthenope”, 80133 Naples, Italy;
| | - Nicola Rocco
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Daniela Russo
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | | | - Giovanna Giuseppina Altobelli
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
| |
Collapse
|
8
|
Liu Q, Zhang X, Song Y, Si J, Li Z, Dong Q. Construction and analysis of a reliable five-gene prognostic signature for colon adenocarcinoma associated with the wild-type allelic state of the COL6A6 gene. Transl Cancer Res 2024; 13:2475-2496. [PMID: 38881933 PMCID: PMC11170513 DOI: 10.21037/tcr-23-463] [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: 03/18/2023] [Accepted: 11/29/2023] [Indexed: 06/18/2024]
Abstract
BACKGROUND Tumors emerge by acquiring a number of mutations over time. The first mutation provides a selective growth advantage compared to adjacent epithelial cells, allowing the cell to create a clone that can outgrow the cells that surround it. Subsequent mutations determine the risk of the tumor progressing to metastatic cancer. Some secondary mutations may inhibit the aggressiveness of the tumor while still increasing the survival of the clone. Meaningful mutations in genes may provide a strong molecular foundation for developing novel therapeutic strategies for cancer. METHODS The somatic mutation and prognosis in colon adenocarcinoma (COAD) were analyzed. The copy number variation (CNV) and differentially expressed genes (DEGs) between the collagen type VI alpha 6 chain (COL6A6) mutation (COL6A6-MUT) and the COL6A6 wild-type (COL6A6-WT) subgroups were evaluated. The independent prognostic signatures based on COL6A6-allelic state were determined to construct a Cox model. The biological characteristics and the immune microenvironment between the two risk groups were compared. RESULTS COL6A6 was found to be highly mutated in COAD at a frequency of 9%. Patients with COL6A6-MUT had a good overall survival (OS) compared to those with COL6A6-WT, who had a different CNV pattern. Significant differences in gene expression were established for 593 genes between the COL6A6-MUT and COL6A6-WT samples. Among them, MUC16, ASNSP1, PRR18, PEG10, and RPL26P8 were determined to be independent prognostic factors. The internally validated prognostic risk model, constructed using these five genes, demonstrated its value by revealing a significant difference in patient prognosis between the high-risk and low-risk groups. Specifically, patients in the high-risk group exhibited a considerably worse prognosis than did those in the low-risk group. The high-risk group had a significantly higher proportion of patients over 60 years of age and patients in stage III. Moreover, the tumor immune dysfunction and exclusion (TIDE) score and the expression of human leukocyte antigen (HLA) family genes were all higher in the high-risk group than that in the low-risk group. CONCLUSIONS The allelic state of COL6A6 and the five associated DEGs were identified as novel biomarkers for the diagnosis and prognosis of COAD and may be therapeutic targets in COAD.
Collapse
Affiliation(s)
- Qun Liu
- Second Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Xiaohua Zhang
- Gastroenterology Center, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Yan Song
- Outpatient Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Junli Si
- Second Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Zhaoshui Li
- Qingdao University, Qingdao Medical College, Qingdao, China
| | - Quanjiang Dong
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| |
Collapse
|
9
|
Sun W, Xie S, Liu SF, Hu X, Xing D. Evolving Tumor Characteristics and Smart Nanodrugs for Tumor Immunotherapy. Int J Nanomedicine 2024; 19:3919-3942. [PMID: 38708176 PMCID: PMC11070166 DOI: 10.2147/ijn.s453265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Typical physiological characteristics of tumors, such as weak acidity, low oxygen content, and upregulation of certain enzymes in the tumor microenvironment (TME), provide survival advantages when exposed to targeted attacks by drugs and responsive nanomedicines. Consequently, cancer treatment has significantly progressed in recent years. However, the evolution and adaptation of tumor characteristics still pose many challenges for current treatment methods. Therefore, efficient and precise cancer treatments require an understanding of the heterogeneity degree of various factors in cancer cells during tumor evolution to exploit the typical TME characteristics and manage the mutation process. The highly heterogeneous tumor and infiltrating stromal cells, immune cells, and extracellular components collectively form a unique TME, which plays a crucial role in tumor malignancy, including proliferation, invasion, metastasis, and immune escape. Therefore, the development of new treatment methods that can adapt to the evolutionary characteristics of tumors has become an intense focus in current cancer treatment research. This paper explores the latest understanding of cancer evolution, focusing on how tumors use new antigens to shape their "new faces"; how immune system cells, such as cytotoxic T cells, regulatory T cells, macrophages, and natural killer cells, help tumors become "invisible", that is, immune escape; whether the diverse cancer-associated fibroblasts provide support and coordination for tumors; and whether it is possible to attack tumors in reverse. This paper discusses the limitations of targeted therapy driven by tumor evolution factors and explores future strategies and the potential of intelligent nanomedicines, including the systematic coordination of tumor evolution factors and adaptive methods, to meet this therapeutic challenge.
Collapse
Affiliation(s)
- Wenshe Sun
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Shaowei Xie
- Department of Ultrasound, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Shi Feng Liu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Xiaokun Hu
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China
- Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
10
|
Gou Z, Li J, Liu J, Yang N. The hidden messengers: cancer associated fibroblasts-derived exosomal miRNAs as key regulators of cancer malignancy. Front Cell Dev Biol 2024; 12:1378302. [PMID: 38694824 PMCID: PMC11061421 DOI: 10.3389/fcell.2024.1378302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/08/2024] [Indexed: 05/04/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs), a class of stromal cells in the tumor microenvironment (TME), play a key role in controlling cancer cell invasion and metastasis, immune evasion, angiogenesis, and resistance to chemotherapy. CAFs mediate their activities by secreting soluble chemicals, releasing exosomes, and altering the extracellular matrix (ECM). Exosomes contain various biomolecules, such as nucleic acids, lipids, and proteins. microRNA (miRNA), a 22-26 nucleotide non-coding RNA, can regulate the cellular transcription processes. Studies have shown that miRNA-loaded exosomes secreted by CAFs engage in various regulatory communication networks with other TME constituents. This study focused on the roles of CAF-derived exosomal miRNAs in generating cancer malignant characteristics, including immune modulation, tumor growth, migration and invasion, epithelial-mesenchymal transition (EMT), and treatment resistance. This study thoroughly examines miRNA's dual regulatory roles in promoting and suppressing cancer. Thus, changes in the CAF-derived exosomal miRNAs can be used as biomarkers for the diagnosis and prognosis of patients, and their specificity can be used to develop newer therapies. This review also discusses the pressing problems that require immediate attention, aiming to inspire researchers to explore more novel avenues in this field.
Collapse
Affiliation(s)
- Zixuan Gou
- Bethune First Clinical School of Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jianming Liu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Na Yang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Jiang C, Chao CC, Li J, Ge X, Shen A, Jucaud V, Cheng C, Shen X. Tissue-resident memory T cell signatures from single-cell analysis associated with better melanoma prognosis. iScience 2024; 27:109277. [PMID: 38455971 PMCID: PMC10918229 DOI: 10.1016/j.isci.2024.109277] [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: 08/28/2023] [Revised: 01/05/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024] Open
Abstract
Tissue-resident memory T cells (TRM) are a specialized T cell population residing in peripheral tissues. The presence and potential impact of TRM in the tumor immune microenvironment (TIME) remain to be elucidated. Here, we systematically investigated the relationship between TRM and melanoma TIME based on multiple clinical single-cell RNA-seq datasets and developed signatures indicative of TRM infiltration. TRM infiltration is associated with longer overall survival and abundance of T cells, NK cells, M1 macrophages, and memory B cells in the TIME. A 22-gene TRM-derived risk score was further developed to effectively classify patients into low- and high-risk categories, distinguishing overall survival and immune activation, particularly in T cell-mediated responses. Altogether, our analysis suggests that TRM abundance is associated with melanoma TIME activation and patient survival, and the TRM-based machine learning model can potentially predict prognosis in melanoma patients.
Collapse
Affiliation(s)
- Chongming Jiang
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Cheng-Chi Chao
- Department of Pipeline Development, Biomap, Inc, San Francisco, CA, USA
| | - Jianrong Li
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xin Ge
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Aidan Shen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Chao Cheng
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiling Shen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
- Xilis, Inc., Durham, NC 27713, USA
| |
Collapse
|
12
|
Guo J, Zhou M, Li J, Yang Y, Hu Y, Tang T, Quan Y. The Prognosis and Immunotherapy Prediction Model of Ovarian Serous Cystadenocarcinoma Patient was Constructed Based on Cuproptosis-Related LncRNA. TOHOKU J EXP MED 2024; 262:63-74. [PMID: 37438122 DOI: 10.1620/tjem.2023.j056] [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: 07/14/2023]
Abstract
Cuproptosis can serve as potential prognostic predictors in patients with cancer. However, the role of this relationship in ovarian serous cystadenocarcinoma (OV) remains unclear. 376 OV tumor samples were obtained from the Cancer Genome Atlas (TCGA) database, and long non-coding RNAs (lncRNAs) related to cuproptosis were obtained through correlation analysis. The risk assessment model was further constructed by univariate Cox regression analysis and LASSO Cox regression. Bioinformatics was used to analyze the regulatory effect of relevant risk assessment models on tumor mutational burden (TMB) and immune microenvironment. We obtained 5 lncRNAs (AC025287.2, AC092718.4, AC112721.2, LINC00996, and LINC01639) and incorporated them into the Cox proportional hazards model. Kaplan-Meier (KM) curve analysis of the prognosis found that the high-risk group was associated with a poorer prognosis. The receiver operating characteristic (ROC) curve showed stronger predictive power compared to other clinicopathological features. Immune infiltration analysis showed that high-risk scores were inversely correlated with CD8+ T cells, CD4+ T cells, macrophages, NK cells, and B cells. Functional enrichment analysis found that they may act via the extracellular matrix (ECM)-interacting proteins and other pathways. We successfully constructed a reliable cuproptosis-related lncRNA model for the prognosis of OV.
Collapse
Affiliation(s)
- Junliang Guo
- Department of Obstetrics and Gynaecology, Centre for Reproductive Medicine, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University
- Sichuan Provincial Key Laboratory of Development and Related Diseases of Women and Children
| | - Muchuan Zhou
- Department of Anesthesia, Sichuan Integrative Medicine Hospital, Sichuan Academy of Chinese Medicine Science (SACMS)
- Sichuan Provincial Key Laboratory of Quality of Chinese Medicinal Materials and Research on Innovative Chinese Medicine
| | - Jinhong Li
- Department of Obstetrics and Gynaecology, Centre for Reproductive Medicine, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University
- Sichuan Provincial Key Laboratory of Development and Related Diseases of Women and Children
| | - Yihong Yang
- Department of Obstetrics and Gynaecology, Centre for Reproductive Medicine, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University
- Sichuan Provincial Key Laboratory of Development and Related Diseases of Women and Children
| | - Yang Hu
- West China School of Medicine, Sichuan University
| | - Tian Tang
- Department of Obstetrics and Gynaecology, Centre for Reproductive Medicine, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University
- Sichuan Provincial Key Laboratory of Development and Related Diseases of Women and Children
| | - Yi Quan
- Department of Obstetrics and Gynaecology, Centre for Reproductive Medicine, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University
- Sichuan Provincial Key Laboratory of Development and Related Diseases of Women and Children
| |
Collapse
|
13
|
Fan W, Tang J, Tang S, Lin Z, Li M, Zhang Z, Wu D. Bibliometric analysis of photodynamic therapy and immune response from 1989-2023. Front Pharmacol 2024; 15:1299253. [PMID: 38288443 PMCID: PMC10822948 DOI: 10.3389/fphar.2024.1299253] [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: 09/22/2023] [Accepted: 01/03/2024] [Indexed: 01/31/2024] Open
Abstract
Objective: Photodynamic therapy (PDT) is a minimally invasive treatment approach for precancerous and cancerous lesions, known for its ability to activate the host immune response. This study conducted a bibliometric analysis to identify the research trends and hotspots related to the immune response in PDT. Methods: We analyzed articles and reviews published from 1989 to 2023, retrieved from the Web of Science database. Using Citespace and VOSviewer, we visualized the distribution patterns of these studies in time and space. Results: The analysis revealed a substantial increase in the number of publications on PDT-related immune response since 1989. A total of 1,688 articles from 1,701 institutions were included in this analysis. Among thei nstitutions, the Chinese Academy of Sciences demonstrated exceptional productivity and a willingness to collaborate with others. Additionally, 8,567 authors contributed to the field, with Mladen Korbelik, Michael R. Hamblin, and Wei R. Chen being the most prolific contributors. The current research focus revolves around novel strategies to enhance antitumor immunity in PDT, including PDT-based dendritic cell vaccines, combination therapies with immune checkpoint inhibitors (ICIs), and the use of nanoparticles for photosensitizer delivery. Furthermore, genes such as CD8A, TNF, CD4, IFNG, CD274, IL6, IL10, CALR, HMGB1, and CTLA4 have been evaluated in the context of PDT-related immunity. Conclusion: PDT not only achieves tumor ablation but also stimulates the immune response, bolstering antitumor immunity. This study highlights the emerging hotspots in PDT-related immune response research and provides valuable insights for future investigations aimed at further enhancing antitumor immunity.
Collapse
Affiliation(s)
- Wanting Fan
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Jianming Tang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Su Tang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Zhengshen Lin
- Department of Stomatology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Mohan Li
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Zheng Zhang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Donglei Wu
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| |
Collapse
|
14
|
Burn OK, Dasyam N, Hermans IF. Recruiting Natural Killer T Cells to Improve Vaccination: Lessons from Preclinical and Clinical Studies. Crit Rev Oncog 2024; 29:31-43. [PMID: 38421712 DOI: 10.1615/critrevoncog.2023049407] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The capacity of type I natural killer T (NKT) cells to provide stimulatory signals to antigen-presenting cells has prompted preclinical research into the use of agonists as immune adjuvants, with much of this work focussed on stimulating T cell responses to cancer. In attempting to evaluate this approach in the clinic, our recent dendritic-cell based study failed to show an advantage to adding an agonist to the vaccine. Here we present potential limitations of the study, and suggest why other simpler strategies may be more effective. These include strategies to target antigen-presenting cells in the host, either through promoting efficient transfer from injected cell lines, facilitating uptake of antigen and agonist as injected conjugates, or encapsulating the components into injected nanovectors. While the vaccine landscape has changed with the rapid uptake of mRNA vaccines, we suggest that there is still a role for recruiting NKT cells in altering T cell differentiation programmes, notably the induction of resident memory T cells.
Collapse
Affiliation(s)
- Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
15
|
Xiao Z, Yan R, Liu H, Huang X, Liang Z, An G, Ge Y. Preventive Treatment with PD-1 Antibody Increases Tissue-resident Memory T Cells Infiltration and Delays Esophageal Carcinogenesis. Cancer Prev Res (Phila) 2023; 16:669-679. [PMID: 37857481 PMCID: PMC10690045 DOI: 10.1158/1940-6207.capr-23-0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/10/2023] [Accepted: 10/13/2023] [Indexed: 10/21/2023]
Abstract
Numerous studies and clinical trials have shown that immune checkpoint inhibitors can effectively prevent tumor growth and metastasis in esophageal squamous cell carcinoma (ESCC) patients. In this study, we aimed to evaluate the anti-tumor effects of PD-1 antibody preventive treatment in patients with early stages ESCC as well as patients with high-grade intraepithelial neoplasia (HGIN). We first established an ESCC model using C57BL/6J mice treated with the chemical carcinogen 4- NQO and observed esophageal lesions at different time points. Second, we compared the antitumor efficacy of PD-1 antibody treatment in mice at the ESCC stage and PD-1 antibody preventive treatment in mice at the HGIN stage. The results showed that PD-1 antibody preventive treatment effectively impeded the progression of 4NQO-induced esophageal tumorigenesis. IHC analysis was performed to observe the infiltration of immune cells into the tumor microenvironment. It has been shown that active tissue-resident memory T cells can be induced and resided into the tumor microenvironment for a long period after treatment with PD-1 antibody. Reexposure to the oncogenic environment colonized by CD8+TRM cells can still exert antitumor effects. These results provide new strategies for the treatment of patients with early stage ESCC and HGIN. PREVENTION RELEVANCE Immune checkpoint inhibitors have shown promising results in multiple tumor species. However, there is currently no clinical application to evaluate their therapeutic value in cancer preventive treatment. Prophylactic use of immune checkpoint inhibitors in the early stages of ESCC may provide long-term benefits to patients.
Collapse
Affiliation(s)
- Zeru Xiao
- Beijing Chao-Yang Hospital, Department of Oncology, Capital Medical University, Beijing, China
| | - Rui Yan
- Beijing Chao-Yang Hospital, Department of Oncology, Capital Medical University, Beijing, China
| | - Heshu Liu
- Beijing Tongren Hospital, Department of Oncology, Capital Medical University, Beijing, China
| | - Xuying Huang
- Chinese Institutes for Medical Research, Beijing, China
| | - Ziwei Liang
- Beijing Chao-Yang Hospital, Department of Oncology, Capital Medical University, Beijing, China
| | - Guangyu An
- Beijing Chao-Yang Hospital, Department of Oncology, Capital Medical University, Beijing, China
| | - Yang Ge
- Beijing Chao-Yang Hospital, Department of Oncology, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
ZHANG YUN, TANG SHALING, GAO YUBO, LU ZHONGTING, YANG YUAN, CHEN JING, LI TAO. Application of exosomal miRNA mediated macrophage polarization in colorectal cancer: Current progress and challenges. Oncol Res 2023; 32:61-71. [PMID: 38188683 PMCID: PMC10767244 DOI: 10.32604/or.2023.043481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/06/2023] [Indexed: 01/09/2024] Open
Abstract
Colorectal cancer (CRC) is a major global health problem with high morbidity and mortality rates. Surgical resection is the main treatment for early-stage CRC, but detecting it early is challenging. Therefore, effective therapeutic targets for advanced patients are still lacking. Exosomes, tiny vesicles in body fluids, play a crucial role in tumor metastasis, immune regulation, and drug resistance. Interestingly, they can even serve as a biomarker for cancer diagnosis and prognosis. Studies have shown that exosomes can carry miRNA, mediate the polarization of M1/M2 macrophages, promote the proliferation and metastasis of cancer cells, and affect the prognosis of CRC. Since the gastrointestinal tract has many macrophages, understanding the mechanism behind exosomal miRNA-mediated macrophage polarization in CRC treatment is crucial. This article summarizes recent advancements in the study of exosomal miRNAs in CRC and their potential as diagnostic and prognostic markers.
Collapse
Affiliation(s)
- YUN ZHANG
- Department of Oncology, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - SHALING TANG
- Department of Oncology, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - YUBO GAO
- Department of Oncology, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - ZHONGTING LU
- Department of Oncology, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - YUAN YANG
- Department of Oncology, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - JING CHEN
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - TAO LI
- Department of Surgical Oncology, Tumor Hospital, The General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
17
|
Chen Z, Shi W, Chen K, Lu C, Li X, Li Q. Elucidating the causal association between gut microbiota and intrahepatic cholangiocarcinoma through Mendelian randomization analysis. Front Microbiol 2023; 14:1288525. [PMID: 38033576 PMCID: PMC10682188 DOI: 10.3389/fmicb.2023.1288525] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Background Intrahepatic cholangiocarcinoma (ICC) is an aggressive liver cancer with poor prognosis. The gut microbiota has been linked to ICC, but evidence for causality is lacking. Elucidating causal gut microbiota-ICC links could inform prevention and treatment strategies. Materials and methods We performed a bidirectional two-sample Mendelian randomization (MR) study to investigate causal associations between gut microbiota and ICC risk. Genome-wide significant single nucleotide polymorphisms (SNPs) associated with gut microbiota abundances were utilized as instrumental variables (IVs). Multiple methods assessed causality and sensitivity analyses evaluated result robustness. Bioinformatics analysis of genetic loci linked to gut microbiota and ICC examined potential mechanisms. Results Genetically predicted increases in Veillonellaceae, Alistipes, Enterobacteriales, and Firmicutes were suggestively associated with higher ICC risk, while increases in Anaerostipes, Paraprevotella, Parasutterella, and Verrucomicrobia appeared protective. Bioinformatics analysis revealed differentially expressed genes near gut microbiota-associated loci may influence ICC through regulating pathways and tumor immune microenvironment. Conclusion Our findings provide suggestive evidence for causal links between specific gut microbiota and ICC risk.
Collapse
Affiliation(s)
- Zhitao Chen
- Department of Hepatobiliary Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| | - Weiguang Shi
- Department of Hepatobiliary Surgery, Shulan (Anji) Hospital, Anji, China
| | - Kailei Chen
- School of Medicine, Zhejiang Shuren University, Hangzhou, China
| | - Chicheng Lu
- School of Medicine Zhejiang Chinese Medical University Zhejiang Shuren College, Hangzhou, China
| | - Xinyuan Li
- School of Medicine, Zhejiang Shuren University, Hangzhou, China
| | - Qiyong Li
- Department of Hepatobiliary Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, China
| |
Collapse
|
18
|
Sebastian A, Martin KA, Peran I, Hum NR, Leon NF, Amiri B, Wilson SP, Coleman MA, Wheeler EK, Byers SW, Loots GG. Loss of Cadherin-11 in pancreatic ductal adenocarcinoma alters tumor-immune microenvironment. Front Oncol 2023; 13:1286861. [PMID: 37954069 PMCID: PMC10639148 DOI: 10.3389/fonc.2023.1286861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the top five deadliest forms of cancer with very few treatment options. The 5-year survival rate for PDAC is 10% following diagnosis. Cadherin 11 (Cdh11), a cell-to-cell adhesion molecule, has been suggested to promote tumor growth and immunosuppression in PDAC, and Cdh11 inhibition significantly extended survival in mice with PDAC. However, the mechanisms by which Cdh11 deficiency influences PDAC progression and anti-tumor immune responses have yet to be fully elucidated. To investigate Cdh11-deficiency induced changes in PDAC tumor microenvironment (TME), we crossed p48-Cre; LSL-KrasG12D/+; LSL-Trp53R172H/+ (KPC) mice with Cdh11+/- mice and performed single-cell RNA sequencing (scRNA-seq) of the non-immune (CD45-) and immune (CD45+) compartment of KPC tumor-bearing Cdh11 proficient (KPC-Cdh11+/+) and Cdh11 deficient (KPC-Cdh11+/-) mice. Our analysis showed that Cdh11 is expressed primarily in cancer-associated fibroblasts (CAFs) and at low levels in epithelial cells undergoing epithelial-to-mesenchymal transition (EMT). Cdh11 deficiency altered the molecular profile of CAFs, leading to a decrease in the expression of myofibroblast markers such as Acta2 and Tagln and cytokines such as Il6, Il33 and Midkine (Mdk). We also observed a significant decrease in the presence of monocytes/macrophages and neutrophils in KPC-Cdh11+/- tumors while the proportion of T cells was increased. Additionally, myeloid lineage cells from Cdh11-deficient tumors had reduced expression of immunosuppressive cytokines that have previously been shown to play a role in immune suppression. In summary, our data suggests that Cdh11 deficiency significantly alters the fibroblast and immune microenvironments and contributes to the reduction of immunosuppressive cytokines, leading to an increase in anti-tumor immunity and enhanced survival.
Collapse
Affiliation(s)
- Aimy Sebastian
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Kelly A. Martin
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Ivana Peran
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Nicole F. Leon
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Beheshta Amiri
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Stephen P. Wilson
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Matthew A. Coleman
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Elizabeth K. Wheeler
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Stephen W. Byers
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - Gabriela G. Loots
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
- University of California Davis Health, Department of Orthopaedic Surgery, Sacramento, CA, United States
| |
Collapse
|
19
|
Jiang D, Gao X, Tan R, Liu X, Zhu Y, Zhang L. Euphorbia factor L1 suppresses breast cancer liver metastasis via DDR1-mediated immune infiltration. Aging (Albany NY) 2023; 15:9217-9229. [PMID: 37709489 PMCID: PMC10522367 DOI: 10.18632/aging.205030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
Euphorbia factor L1 (EFL1), a lathyrane-type diterpenoid from the medicinal herb Euphorbia lathyris L., has been documented to possess various pharmacologic actives. However, the function of EFL1 on breast cancer is not clear. In this study, we explored the effect and mechanism of EFL1 on breast cancer liver metastasis. Female BALB/c mice were subjected to breast cancer-surgical hepatic implantation (SHI) to establish breast cancer liver metastasis model in vivo. At 10 days post-surgery, mice were administrated with EFL1 once daily for a total of 2 weeks. Serum AST and ALT activities, abdominal circumference, peritoneal fluid, tumor weight and volume were determined to assess liver and mesenteric re-metastasis of breast cancer. H&E staining was used to observe morphology changes in tumor, liver and small intestine tissues. ELISA was applied to observe inflammatory levels. Tumor DDR1 expression and immune infiltration were determined using western blotting, immunohistochemistry and flow cytometer methods. Our results showed that EFL1 administration improved liver function (AST and ALT activities), ascites, liver metastasis and mesenteric re-metastasis in SHI mice. Also, SHI-induced inflammatory cell infiltration and IL-1β, IL-6, TNF-α generation in ascites were decreased by EFL1 treatment. Mechanism study revealed that EFL1 intervention enhanced the ratios of CD4+ and CD8+ and CD49b+(NK) T lymphocytes and decreased Treg cells through downregulating DDR1 in the tumor of SHI mice. Furthermore, overexpression of DDR1 abolished the anti-liver metastasis effect and pro-immune infiltration action of EFL1 in SHI mice. Together, our findings suggested that EFL1 protects against breast cancer liver metastasis in vivo by targeting DDR1-mediated immune infiltration.
Collapse
Affiliation(s)
- Dongjing Jiang
- Traditional Chinese Medicine and Research Office, Suzhou Health College of Technology, Suzhou 215000, China
| | - XiaoQin Gao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - RuLan Tan
- Traditional Chinese Medicine and Research Office, Suzhou Health College of Technology, Suzhou 215000, China
| | - Xun Liu
- Traditional Chinese Medicine and Research Office, Suzhou Health College of Technology, Suzhou 215000, China
| | - Ye Zhu
- Traditional Chinese Medicine and Research Office, Suzhou Health College of Technology, Suzhou 215000, China
| | - Li Zhang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
20
|
Wang M, Hu Q, Huang J, Zhang F, Yao Z, Shao S, Zhao X, Liang T. In Situ Formed ROS-Responsive Hydrogel with STING Agonist and Gemcitabine to Intensify Immunotherapy against Pancreatic Ductal Adenocarcinoma. Adv Healthc Mater 2023; 12:e2203264. [PMID: 36971070 DOI: 10.1002/adhm.202203264] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/22/2023] [Indexed: 03/29/2023]
Abstract
Immunotherapy, the most revolutionary anticancer strategy, faces major obstacles in yielding desirable outcomes in pancreatic ductal adenocarcinoma (PDAC) due to the highly immunosuppressive tumor microenvironment (TME). Meanwhile, when used alone, the traditional first-line chemotherapeutic agent gemcitabine (GEM) in PDAC treatment is also insufficient to achieve lasting efficacy. In this study, a reactive oxygen species degradable hydrogel system, denoted as GEM-STING@Gel, is engineered to codeliver gemcitabine and the stimulator of interferon genes (STING) agonist DMXAA (5,6-dimethylxanthenone-4-acetic acid) into the tumor site. In this work, the strategy addresses the major challenges of current immunotherapies with a facile platform, which can synergistically activate innate immunity and promote the cytotoxic T lymphocytes infiltration at the tumor site, thereby modulating the immunosuppressive TME. Further, the efficient therapeutic potency of the immunotherapy is confirmed in an orthotopic postsurgical model, unleashing the translational potential to prevent tumor recurrence after surgical resection. This study underscores the advantages of this integrative strategy that combines chemotherapy, immunotherapy, and biomaterial-based hydrogel, including improved therapeutic efficacy, operational convenience, and superior biosafety.
Collapse
Affiliation(s)
- Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Qida Hu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Junming Huang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Fu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhuo Yao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shiyi Shao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Xinyu Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310003, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310003, China
- Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 311121, China
| |
Collapse
|
21
|
Borella R, Paolini A, Aramini B, Gibellini L, Masciale V, Lo Tartaro D, Dominici M, De Biasi S, Cossarizza A. Multiparametric analysis of tumor infiltrating lymphocytes in solid tumors. Methods Cell Biol 2023; 195:39-70. [PMID: 40180454 DOI: 10.1016/bs.mcb.2023.03.006] [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: 04/05/2025]
Abstract
The use of single-cell technologies in characterizing the interactions between immune and cancer cells is in continuous expansion. Indeed, the combination of different single-cell approaches enables the definition of novel phenotypic and functional aspects of the immune cells infiltrating the tumor microenvironment (TME). This approach is promoting the discovery of relevant and reliable predictive biomarkers, along with the development of new promising treatments. In this chapter, we describe the main subsets of tumor-infiltrating lymphocytes from a phenotypical and functional point of view and discuss the use of single-cell technologies used to characterize these cell populations within TME.
Collapse
Affiliation(s)
- Rebecca Borella
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum-University of Bologna and G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy.
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy; Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
| |
Collapse
|
22
|
Zhu X, Li S. Nanomaterials in tumor immunotherapy: new strategies and challenges. Mol Cancer 2023; 22:94. [PMID: 37312116 PMCID: PMC10262535 DOI: 10.1186/s12943-023-01797-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
Tumor immunotherapy exerts its anti-tumor effects by stimulating and enhancing immune responses of the body. It has become another important modality of anti-tumor therapy with significant clinical efficacy and advantages compared to chemotherapy, radiotherapy and targeted therapy. Although various kinds of tumor immunotherapeutic drugs have emerged, the challenges faced in the delivery of these drugs, such as poor tumor permeability and low tumor cell uptake rate, had prevented their widespread application. Recently, nanomaterials had emerged as a means for treatment of different diseases due to their targeting properties, biocompatibility and functionalities. Moreover, nanomaterials possess various characteristics that overcome the defects of traditional tumor immunotherapy, such as large drug loading capacity, precise tumor targeting and easy modification, thus leading to their wide application in tumor immunotherapy. There are two main classes of novel nanoparticles mentioned in this review: organic (polymeric nanomaterials, liposomes and lipid nanoparticles) and inorganic (non-metallic nanomaterials and metallic nanomaterials). Besides, the fabrication method for nanoparticles, Nanoemulsions, was also introduced. In summary, this review article mainly discussed the research progress of tumor immunotherapy based on nanomaterials in the past few years and offers a theoretical basis for exploring novel tumor immunotherapy strategies in the future.
Collapse
Affiliation(s)
- Xudong Zhu
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| |
Collapse
|
23
|
Li Y, Shi Y, Zhang X, Li P, Ma L, Hu P, Xu L, Dai Y, Xia S, Qiu H. FGFR2 upregulates PAI-1 via JAK2/STAT3 signaling to induce M2 polarization of macrophages in colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166665. [PMID: 36781088 DOI: 10.1016/j.bbadis.2023.166665] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/17/2023] [Accepted: 02/07/2023] [Indexed: 02/13/2023]
Abstract
Fibroblast growth factor receptor 2 (FGFR2) is frequently activated by overexpression or mutation, and an abnormal fibroblast growth factor (FGF)/FGFR signaling pathway is associated with the occurrence, development, and poor prognosis of colorectal cancer (CRC). Our preliminary analysis found that plasminogen activator inhibitor-1 (PAI-1) expression may be related to FGF/FGFR signaling, however, their role in the tumor immune microenvironment remains unclear. In this study, we observed markedly higher PAI-1 expression in CRC patients with poor survival rates. PAI-1 is regulated by FGF/FGFR2 in colon cancer cells and is involved in M2 macrophage polarization. Mechanistically, inhibiting the JAK2/STAT3 signaling pathway could cause PAI-1 downregulation. Furthermore, the activation of phosphorylated STAT3 upregulated PAI-1. In vivo, FGFR2 overexpression in tumor-bearing mouse models suggested that a PAI-1 inhibitor could rescue FGFR2/PAI-1 axis-induced M2 macrophage polarization, which leads to effective immune activity and tumor suppression. Moreover, the combination of a PAI-1 inhibitor and anti-PD-1 therapy exhibited superior antitumor activity in mice. These findings offer novel insights into the molecular mechanisms underlying tumor deterioration and provide potential therapeutic targets for CRC treatment.
Collapse
Affiliation(s)
- Yiming Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yongkang Shi
- Department of Biliary and Pancreatic Surgery/Cancer Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiuyuan Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Piao Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Pengbo Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Liang Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yuhong Dai
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Shu Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
24
|
Koch MRA, Gong R, Friedrich V, Engelsberger V, Kretschmer L, Wanisch A, Jarosch S, Ralser A, Lugen B, Quante M, Vieth M, Vasapolli R, Schulz C, Buchholz VR, Busch DH, Mejías-Luque R, Gerhard M. CagA-specific Gastric CD8 + Tissue-Resident T Cells Control Helicobacter pylori During the Early Infection Phase. Gastroenterology 2023; 164:550-566. [PMID: 36587707 DOI: 10.1053/j.gastro.2022.12.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS Infection with Helicobacter pylori strongly affects global health by causing chronic gastritis, ulcer disease, and gastric cancer. Although extensive research into the strong immune response against this persistently colonizing bacterium exists, the specific role of CD8+ T cells remains elusive. METHODS We comprehensively characterize gastric H pylori-specific CD8+ T-cell responses in mice and humans by flow cytometry, RNA-sequencing, immunohistochemistry, and ChipCytometry, applying functional analyses including T-cell depletion, H pylori eradication, and ex vivo restimulation. RESULTS We define CD8+ T-cell populations bearing a tissue-resident memory (TRM) phenotype, which infiltrate the gastric mucosa shortly after infection and mediate pathogen control by executing antigen-specific effector properties. These induced CD8+ tissue-resident memory T cells (TRM cells) show a skewed T-cell receptor beta chain usage and are mostly specific for cytotoxin-associated gene A, the distinctive oncoprotein injected by H pylori into host cells. As the infection progresses, we observe a loss of the TRM phenotype and replacement of CD8+ by CD4+ T cells, indicating a shift in the immune response during the chronic infection phase. CONCLUSIONS Our results point toward a hitherto unknown role of CD8+ T-cell response in this bacterial infection, which may have important clinical implications for treatment and vaccination strategies against H pylori.
Collapse
Affiliation(s)
- Maximilian R A Koch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Ruolan Gong
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Verena Friedrich
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Veronika Engelsberger
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Lorenz Kretschmer
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Andreas Wanisch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Sebastian Jarosch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Anna Ralser
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Bob Lugen
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Michael Quante
- Technical University of Munich (TUM), School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, Munich, Germany; Department of Internal Medicine II, University Hospital Freiburg, University Freiburg, Freiburg, Germany
| | - Michael Vieth
- Institute of Pathology, Hospital Bayreuth, Friedrich Alexander University, Erlangen-Nuremberg, Bayreuth, Germany
| | - Riccardo Vasapolli
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany; Medical Department II, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Christian Schulz
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany; Medical Department II, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Veit R Buchholz
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Dirk H Busch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Raquel Mejías-Luque
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany.
| | - Markus Gerhard
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany.
| |
Collapse
|
25
|
Hao M, Li H, Yi M, Zhu Y, Wang K, Liu Y, Liang X, Ding L. Development of an immune-related gene prognostic risk model and identification of an immune infiltration signature in the tumor microenvironment of colon cancer. BMC Gastroenterol 2023; 23:58. [PMID: 36890467 PMCID: PMC9996977 DOI: 10.1186/s12876-023-02679-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 02/15/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND Colon cancer is a common and highly malignant tumor. Its incidence is increasing rapidly with poor prognosis. At present, immunotherapy is a rapidly developing treatment for colon cancer. The aim of this study was to construct a prognostic risk model based on immune genes for early diagnosis and accurate prognostic prediction of colon cancer. METHODS Transcriptome data and clinical data were downloaded from the cancer Genome Atlas database. Immunity genes were obtained from ImmPort database. The differentially expressed transcription factors (TFs) were obtained from Cistrome database. Differentially expressed (DE) immune genes were identified in 473 cases of colon cancer and 41 cases of normal adjacent tissues. An immune-related prognostic model of colon cancer was established and its clinical applicability was verified. Among 318 tumor-related transcription factors, differentially expressed transcription factors were finally obtained, and a regulatory network was constructed according to the up-down regulatory relationship. RESULTS A total of 477 DE immune genes (180 up-regulated and 297 down-regulated) were detected. We developed and validated twelve immune gene models for colon cancer, including SLC10A2, FABP4, FGF2, CCL28, IGKV1-6, IGLV6-57, ESM1, UCN, UTS2, VIP, IL1RL2, NGFR. The model was proved to be an independent prognostic variable with good prognostic ability. A total of 68 DE TFs (40 up-regulated and 23 down-regulated) were obtained. The regulation network between TF and immune genes was plotted by using TF as source node and immune genes as target node. In addition, Macrophage, Myeloid Dendritic cell and CD4+ T cell increased with the increase of risk score. CONCLUSION We developed and validated twelve immune gene models for colon cancer, including SLC10A2, FABP4, FGF2, CCL28, IGKV1-6, IGLV6-57, ESM1, UCN, UTS2, VIP, IL1RL2, NGFR. This model can be used as a tool variable to predict the prognosis of colon cancer.
Collapse
Affiliation(s)
- Mengdi Hao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
| | - Huimin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
| | - Meng Yi
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
| | - Yubing Zhu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
| | - Kun Wang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
| | - Yin Liu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
| | - Xiaoqing Liang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China
| | - Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, No. 10, Tieyi Road, Haidian District, Beijing, 100038, China.
- Department of Oncology, Ninth School of Clinical Medicine, Peking University, Beijing, 100038, China.
| |
Collapse
|
26
|
Meng L, Yang Y, Hu X, Zhang R, Li X. Prognostic value of the pretreatment systemic immune-inflammation index in patients with prostate cancer: a systematic review and meta-analysis. J Transl Med 2023; 21:79. [PMID: 36739407 PMCID: PMC9898902 DOI: 10.1186/s12967-023-03924-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The systemic immune-inflammation index (SII) is a novel biomarker to predict the prognosis of some malignant tumors based on neutrophil, platelet, and lymphocyte counts. Evidence is scarce about the prognostic value of SII for prostate cancer patients. This systematic review and meta-analysis was conducted to explore the prognostic value of the SII in prostate cancer. METHODS The PubMed, Embase, Web of Science, and Cochrane Library (CENTRAL) databases were searched to determine eligible studies from inception to August 15, 2022. Hazard ratios (HRs) with 95% confidence intervals (CIs) were extracted to pool the results. Statistical analyses were conducted by using Stata 17.0 software. RESULTS A total of 12 studies with 8083 patients were included. The quantitative synthesis showed that a high SII was related to poor overall survival (OS) (HR = 1.44, 95% CI 1.23-1.69, p < 0.001). Furthermore, a subgroup analysis showed that a high SII was associated with poor OS in the groups of any ethnicity, tumor type, and cutoff value. An increased SII was also associated with inferior progression-free survival (PFS) (HR = 1.80, 95% CI 1.27-2.56, p = 0.001). In the subgroup analysis, a high SII value was related to poor PFS in Asian patients (HR = 4.03, 95% CI 1.07-15.17, p = 0.04) and a cutoff value > 580 (HR = 1.19, 95% CI 1.04-1.36, p = 0.01). CONCLUSION Based on the current evidence, a high pretreatment SII may be associated with poor OS and PFS. The SII may serve as an important prognostic indicator in patients with prostate cancer. More rigorously designed studies are needed to explore the SII and the prognosis of prostate cancer.
Collapse
Affiliation(s)
- Linghao Meng
- grid.13291.380000 0001 0807 1581Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041 China ,grid.13291.380000 0001 0807 1581West China School of Medicine, Sichuan University, Chengdu, 610041 China
| | - Yujia Yang
- grid.13291.380000 0001 0807 1581Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041 China ,grid.13291.380000 0001 0807 1581West China School of Medicine, Sichuan University, Chengdu, 610041 China
| | - Xu Hu
- grid.13291.380000 0001 0807 1581Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Ruohan Zhang
- grid.13291.380000 0001 0807 1581West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041 China
| | - Xiang Li
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
27
|
[Advances in the Study of Tissue-resident Memory T Cells in Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:862-869. [PMID: 36617472 PMCID: PMC9845087 DOI: 10.3779/j.issn.1009-3419.2022.102.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have been widely used in the treatment of lung cancer, but the benefit population is limited and there is a lack of effective predictive markers of efficacy. Tissue-resident memory T cells (TRM) reside in tissues and exert anti-tumor effects by expressing the integrins CD103, CD49a or C-type lectin CD69 and immune checkpoint receptors. TRM expressing programmed cell death 1 (PD-1) is enriched with transcriptional products associated with cytotoxicity and enhances T cell (antigen) receptor (TCR)-mediated cytotoxicity. TRM is a promising biomarker for predicting the efficacy and prognosis of immunotherapy in lung cancer patients. This review will describe the progress of TRM research in lung cancer.
.
Collapse
|
28
|
Wang J, Li Z. TREM2 Is a Prognostic Biomarker and Correlated with an Immunosuppressive Microenvironment in Thyroid Cancer. DISEASE MARKERS 2022; 2022:1807386. [PMID: 36438899 PMCID: PMC9683966 DOI: 10.1155/2022/1807386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 03/12/2024]
Abstract
PURPOSES To identify the differentially expressed genes (DEGs) related to the immune microenvironment and elucidate the biological functions of key genes in papillary thyroid cancer (PTC) by analyzing the immune microenvironment. METHODS The relative quantities of immune and matrix components in 507 patients with PTC were calculated from the TCGA database. Analysis of differentially expressed genes in tumor samples throughout the genome, intersection of DEGs obtained from PTC patients, and genome-wide tumor samples and survival analysis were performed. Survival analysis was used for identification of prognostic factor. Immunohistochemical analysis of the TREM2 expression in PTC tissues, flow cytometry, and transwell assays were used to detect the effect of TREM2 on PTC cell proliferation, migration, and invasion. RESULTS There were a total of 1242 upregulated genes with high intersection in the immune score and 124 downregulated genes with low intersection in the stromal score. A total of 1,366 genes in these DEGs may be determinants in the immune microenvironment. GO enrichment and KEGG enrichment analysis revealed that the overall function of DEGs appeared to map onto immune-related activities. Gene intersection and survival analysis showed that there were 435 DEG crosses in PTC patients and genome-wide tumor samples, only CXCL10, CD40LG, KRT14, TRAT1, and TREM2 were associated with patient prognosis, and TCGA showed that only the TREM2 expression was upregulated in PTC. TREM2 knockdown inhibited the cell cycle and cell proliferation, migration, and invasion by PTC cells. TREM2 was associated with the immunosuppressive microenvironment by via NF-κB pathway in PTC. CONCLUSION TREM2 possibly was a potential indicator of altered TME status in PTC, and that TREM2 promoted PTC cell proliferation and cell cycle, migration, and invasion by NF-κB pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, NO.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| | - Zhendong Li
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, NO.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
29
|
Advancements in the characterization of tissue resident memory T cells in skin disease. Clin Immunol 2022; 245:109183. [DOI: 10.1016/j.clim.2022.109183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
|
30
|
Yaping W, Zhe W, Zhuling C, Ruolei L, Pengyu F, Lili G, Cheng J, Bo Z, Liuyin L, Guangdong H, Yaoling W, Niuniu H, Rui L. The soldiers needed to be awakened: Tumor-infiltrating immune cells. Front Genet 2022; 13:988703. [PMID: 36246629 PMCID: PMC9558824 DOI: 10.3389/fgene.2022.988703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
In the tumor microenvironment, tumor-infiltrating immune cells (TIICs) are a key component. Different types of TIICs play distinct roles. CD8+ T cells and natural killer (NK) cells could secrete soluble factors to hinder tumor cell growth, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) release inhibitory factors to promote tumor growth and progression. In the meantime, a growing body of evidence illustrates that the balance between pro- and anti-tumor responses of TIICs is associated with the prognosis in the tumor microenvironment. Therefore, in order to boost anti-tumor response and improve the clinical outcome of tumor patients, a variety of anti-tumor strategies for targeting TIICs based on their respective functions have been developed and obtained good treatment benefits, including mainly immune checkpoint blockade (ICB), adoptive cell therapies (ACT), chimeric antigen receptor (CAR) T cells, and various monoclonal antibodies. In recent years, the tumor-specific features of immune cells are further investigated by various methods, such as using single-cell RNA sequencing (scRNA-seq), and the results indicate that these cells have diverse phenotypes in different types of tumors and emerge inconsistent therapeutic responses. Hence, we concluded the recent advances in tumor-infiltrating immune cells, including functions, prognostic values, and various immunotherapy strategies for each immune cell in different tumors.
Collapse
Affiliation(s)
- Wang Yaping
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Zhe
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Chu Zhuling
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
| | - Li Ruolei
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Fan Pengyu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Guo Lili
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ji Cheng
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhang Bo
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Liu Liuyin
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Hou Guangdong
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wang Yaoling
- Department of Geriatrics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hou Niuniu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of General Surgery, Eastern Theater Air Force Hospital of PLA, Nanjing, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| | - Ling Rui
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Hou Niuniu, ; Ling Rui,
| |
Collapse
|
31
|
Hao L, Chen Q, Chen X, Zhou Q. The Role of Gender-Related Immune Genes in Childhood Acute Myeloid Leukemia. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3235238. [PMID: 36193320 PMCID: PMC9525781 DOI: 10.1155/2022/3235238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/17/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
The study of immune genes and immune cells is highly focused in recent years. To find immunological genes with prognostic value, the current study examines childhood acute myeloid leukemia according to gender. The TARGET database was used to gather the "mRNA expression profile data" and relevant clinical data of children with AML. To normalize processing and find differentially expressed genes (DEG) between male and female subgroups, the limma software package is utilized. We identified prognostic-related genes and built models using LASSO, multivariate Cox, and univariate Cox analysis. The prognostic significance of prognostic genes was then examined through the processing of survival analysis and risk score (RS) calculation. We investigated the connections between immune cells and prognostic genes as well as the connections between prognostic genes and medications. Finally, five immune genes from the TARGET database have been identified. These immune genes are considerably correlated to the prognosis of male patients.
Collapse
Affiliation(s)
- Lu Hao
- Science and Education Department, Shenzhen Baoan Shiyan People's Hospital, Shenzhen, China
| | - Qiuyan Chen
- Science and Education Department, Shenzhen Baoan Shiyan People's Hospital, Shenzhen, China
| | - Xi Chen
- Central Laboratory, The People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qing Zhou
- Central Laboratory, The People's Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
32
|
Lin XH, Zhang DY, Liu ZY, Tang WQ, Chen RX, Li DP, Weng S, Dong L. lncRNA-AC079061.1/VIPR1 axis may suppress the development of hepatocellular carcinoma: a bioinformatics analysis and experimental validation. Lab Invest 2022; 20:379. [PMID: 36038907 PMCID: PMC9422102 DOI: 10.1186/s12967-022-03573-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/04/2022] [Indexed: 11/12/2022]
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most malignant tumors to threaten human life, and the survival rate remains low due to delayed diagnosis. Meanwhile, lncRNAs have great potential for application in tumor prognosis, therefore relevant research in hepatocellular carcinoma is indispensable. Methods Based on the EZH2 expression, the differentially expressed lncRNAs DElncRNAs), miRNAs (DEmiRNAs), and mRNAs (DEmRNAs) were identified in hepatocellular carcinoma by using the TCGA database. Bioinformatics technology was utilized to determine the effect of key genes in HCC progression. The methylation and immune infiltration analyses were performed to explore the underlying function of hub genes. Finally, cellular function experiments were performed to investigate the association between identified genes and biological phenotypes in HCC. Results lncRNA-AC079061.1, hsa-miR-765, and VIPR1 were identified as independent factors that affect the prognosis of hepatocellular carcinoma. The immune infiltration analyses revealed that lncRNA-AC079061.1 can alter the immune microenvironment and thus inhibit the development of HCC by regulating the expression of an immune-related gene (VIPR1). Methylation analyses demonstrated that VIPR1 expression is negatively related to the methylation level in HCC. Experimental results suggested that lncRNA-AC079061.1 and VIPR1 were frequently downregulated in HCC cells, while hsa-miR-765 was significantly upregulated. Moreover, the lncRNA-AC079061.1/VIPR1 axis suppressed the proliferation and invasion of HCC cells. Conclusion The present study identified the lncRNA-AC079061.1/VIPR1 axis as a novel biomarker that inhibited the proliferation and invasion of hepatocellular carcinoma, affecting the ultimate disease outcome. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03573-7.
Collapse
Affiliation(s)
- Xia-Hui Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Institute of Liver Disease, Shanghai, 200032, China
| | - Dan-Ying Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Institute of Liver Disease, Shanghai, 200032, China
| | - Zhi-Yong Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Institute of Liver Disease, Shanghai, 200032, China
| | - Wen-Qing Tang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Institute of Liver Disease, Shanghai, 200032, China
| | - Rong-Xin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Dong-Ping Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Shanghai Institute of Liver Disease, Shanghai, 200032, China
| | - Shuqiang Weng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Liver Disease, Shanghai, 200032, China.
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Liver Disease, Shanghai, 200032, China.
| |
Collapse
|
33
|
Fernández-Arjona MDM, León-Rodríguez A, Grondona JM, López-Ávalos MD. Microbial neuraminidase induces TLR4-dependent long-term immune priming in the brain. Front Cell Neurosci 2022; 16:945229. [PMID: 35966200 PMCID: PMC9366060 DOI: 10.3389/fncel.2022.945229] [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: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Innate immune memory explains the plasticity of immune responses after repeated immune stimulation, leading to either enhanced or suppressed immune responses. This process has been extensively reported in peripheral immune cells and also, although modestly, in the brain. Here we explored two relevant aspects of brain immune priming: its persistence over time and its dependence on TLR receptors. For this purpose, we used an experimental paradigm consisting in applying two inflammatory stimuli three months apart. Wild type, toll-like receptor (TLR) 4 and TLR2 mutant strains were used. The priming stimulus was the intracerebroventricular injection of neuraminidase (an enzyme that is present in various pathogens able to provoke brain infections), which triggers an acute inflammatory process in the brain. The second stimulus was the intraperitoneal injection of lipopolysaccharide (a TLR4 ligand) or Pam3CSK4 (a TLR2 ligand). One day after the second inflammatory challenge the immune response in the brain was examined. In wild type mice, microglial and astroglial density, as well as the expression of 4 out of 5 pro-inflammatory genes studied (TNFα, IL1β, Gal-3, and NLRP3), were increased in mice that received the double stimulus compared to those exposed only to the second one, which were initially injected with saline instead of neuraminidase. Such enhanced response suggests immune training in the brain, which lasts at least 3 months. On the other hand, TLR2 mutants under the same experimental design displayed an enhanced immune response quite similar to that of wild type mice. However, in TLR4 mutant mice the response after the second immune challenge was largely dampened, indicating the pivotal role of this receptor in the establishment of immune priming. Our results demonstrate that neuraminidase-induced inflammation primes an enhanced immune response in the brain to a subsequent immune challenge, immune training that endures and that is largely dependent on TLR4 receptor.
Collapse
Affiliation(s)
- María del Mar Fernández-Arjona
- Laboratorio de Medicina Regenerativa, Grupo de investigación en Neuropsicofarmacología, Hospital Regional Universitario de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Ana León-Rodríguez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Laboratorio de Fisiología Animal, Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Jesús M. Grondona
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Laboratorio de Fisiología Animal, Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - María Dolores López-Ávalos
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Laboratorio de Fisiología Animal, Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- *Correspondence: María Dolores López-Ávalos
| |
Collapse
|
34
|
Chen Y, Yan SM, Pu Z, Feng J, Tan L, Li Y, Hu H, Huang W, Lin Y, Peng Z, He X, Huang F, Zhang H, Zhang Y. Dopamine signaling promotes tissue-resident memory differentiation of CD8+ T cells and antitumor immunity. Cancer Res 2022; 82:3130-3142. [PMID: 35802647 DOI: 10.1158/0008-5472.can-21-4084] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/06/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
Tissue-resident memory CD8+ T (TRM)-cells have been associated with robust protective anti-tumor immune responses and improved prognosis of cancer patients. Therefore, therapeutic strategies that modulate either the production or activity of TRM cells could be effective for treating cancer. Using a high-throughput drug screen, we showed that the neurotransmitter dopamine drives differentiation of CD8+ T cells into CD103+ TRM cells. In murine syngeneic tumor xenograft models and clinical human colon cancer samples, DRD5 served as the major functional dopamine receptor on CD8+ T cells and positively correlated with TRM cell density. DRD5 deficiency led to a failure of CD8+ T cells to accumulate in tissues, resulting in impaired TRM cell formation, reduced effector function, and uncontrolled disease progression. Moreover, dopamine treatment promoted the antitumor activity of CD8+ T cells and suppressed colorectal cancer growth in immunocompentent mouse models, and ex-vivo pre-conditioning with dopamine enhanced the in vivo efficacy of CAR-T cells. Finally, in a colorectal cancer patient cohort, dopamine expression was positively associated with patient survival and CD8+ T cell infiltration. These findings suggest that dopaminergic immunoregulation plays an important role in the differentiation of CD8+ cells into CD103+ TRM cells and thereby modulates TRM-elicited antitumor immunity in colorectal cancer.
Collapse
Affiliation(s)
- Yingshi Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shu-Mei Yan
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zeyu Pu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinzhu Feng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Likai Tan
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yuzhuang Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hongrong Hu
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | | | - Yingtong Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhilin Peng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xin He
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Feng Huang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), China
| | - Hui Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Anadon CM, Yu X, Hänggi K, Biswas S, Chaurio RA, Martin A, Payne KK, Mandal G, Innamarato P, Harro CM, Mine JA, Sprenger KB, Cortina C, Powers JJ, Costich TL, Perez BA, Gatenbee CD, Prabhakaran S, Marchion D, Heemskerk MHM, Curiel TJ, Anderson AR, Wenham RM, Rodriguez PC, Conejo-Garcia JR. Ovarian cancer immunogenicity is governed by a narrow subset of progenitor tissue-resident memory T cells. Cancer Cell 2022; 40:545-557.e13. [PMID: 35427494 PMCID: PMC9096229 DOI: 10.1016/j.ccell.2022.03.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/06/2022] [Accepted: 03/23/2022] [Indexed: 02/05/2023]
Abstract
Despite repeated associations between T cell infiltration and outcome, human ovarian cancer remains poorly responsive to immunotherapy. We report that the hallmarks of tumor recognition in ovarian cancer-infiltrating T cells are primarily restricted to tissue-resident memory (TRM) cells. Single-cell RNA/TCR/ATAC sequencing of 83,454 CD3+CD8+CD103+CD69+ TRM cells and immunohistochemistry of 122 high-grade serous ovarian cancers shows that only progenitor (TCF1low) tissue-resident T cells (TRMstem cells), but not recirculating TCF1+ T cells, predict ovarian cancer outcome. TRMstem cells arise from transitional recirculating T cells, which depends on antigen affinity/persistence, resulting in oligoclonal, trogocytic, effector lymphocytes that eventually become exhausted. Therefore, ovarian cancer is indeed an immunogenic disease, but that depends on ∼13% of CD8+ tumor-infiltrating T cells (∼3% of CD8+ clonotypes), which are primed against high-affinity antigens and maintain waves of effector TRM-like cells. Our results define the signature of relevant tumor-reactive T cells in human ovarian cancer, which could be applicable to other tumors with unideal mutational burden.
Collapse
Affiliation(s)
- Carmen M Anadon
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kay Hänggi
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Ricardo A Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Alexandra Martin
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kyle K Payne
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Gunjan Mandal
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Patrick Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Carly M Harro
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jessica A Mine
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Kimberly B Sprenger
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Carla Cortina
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - John J Powers
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Tara Lee Costich
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Bradford A Perez
- Department of Radiation Therapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Chandler D Gatenbee
- Department of Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Sandhya Prabhakaran
- Department of Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Douglas Marchion
- Department of Tissue Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tyler J Curiel
- Department of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Alexander R Anderson
- Department of Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Robert M Wenham
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA; Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
36
|
Li L, Tang W, Zhang Y, Jia M, Wang L, Li Q, Han Q, Peng X, Xie Y, Wu J, Wang Z, Zhen J, Wang X, Liu M, Sun Y, Zhang C, Yi F. Targeting tissue-resident memory CD8 + T cells in the kidney is a potential therapeutic strategy to ameliorate podocyte injury and glomerulosclerosis. Mol Ther 2022; 30:2746-2759. [PMID: 35514086 PMCID: PMC9372318 DOI: 10.1016/j.ymthe.2022.04.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 12/01/2022] Open
Abstract
Although tissue-resident memory T (TRM) cells, a recently identified non-circulating memory T cell population, play a crucial role in mediating local immune responses and protect against pathogens upon local reinfection, the composition, effector function, and specificity of TRM cells in the kidney and their relevance for chronic kidney disease remain unknown. In this study, we found that renal tissue displayed high abundance of tissue-resident lymphocytes and the proportion of CD8+ TRM cells was significantly increased in the kidney from patients and mice with focal segmental glomerulosclerosis (FSGS), diabetic kidney disease (DKD) and lupus nephritis (LN). Mechanistically, IL-15 significantly promoted CD8+ TRM cell formation and activation, thereby promoting podocyte injury and glomerulosclerosis. Interestingly, Sparsentan, the dual angiotensin II (Ang II) receptor and endothelin Type A receptor antagonist, can also reduce TRM cell responses by intervening IL-15 signaling, exploring its new pharmacological functions. Mechanistically, Sparsentan inhibited Ang II or endothelin-1 (ET-1)-mediated IL-15 signaling, thereby further regulating renal CD8+ TRM cell fates. Collectively, our studies provide direct evidence for the pivotal role of renal CD8+ TRM cells in podocyte injury, and further strengthen that targeting TRM cells represents a novel therapeutic strategy for patients with glomerular diseases.
Collapse
Affiliation(s)
- Liang Li
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yan Zhang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Meng Jia
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Limei Wang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China, 250012
| | - Quanxin Li
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Qingsheng Han
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Xiuping Peng
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yusheng Xie
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Jichao Wu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Ziying Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Junhui Zhen
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Xiaojie Wang
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Min Liu
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Yu Sun
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430022
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China, 250012.
| |
Collapse
|
37
|
Wang X, Zhang J, Hu B, Qian F. High Expression of CSF-1R Predicts Poor Prognosis and CSF-1R high Tumor-Associated Macrophages Inhibit Anti-Tumor Immunity in Colon Adenocarcinoma. Front Oncol 2022; 12:850767. [PMID: 35444953 PMCID: PMC9014714 DOI: 10.3389/fonc.2022.850767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/10/2022] [Indexed: 11/20/2022] Open
Abstract
Background Colony stimulating factor 1 receptor (CSF-1R) is a single channel III transmembrane receptor tyrosine kinase (RTK) and plays an important role in immune regulation and the development of various cancer types. The expression of CSF-1R in colon adenocarcinoma (COAD) and its prognostic value remain incompletely understood. Therefore, we aim to explore the prognostic value of CSF-1R in COAD and its relationship with tumor immunity. Methods CSF-1R expression in a COAD cohort containing 103 patients was examined using immunohistochemistry (IHC). The relationship between CSF-1R expression and clinicopathological parameters and prognosis was evaluated. Dual immunofluorescence staining was conducted to determine the localization of CSF-1R in COAD tissues. Univariate and multivariate Cox regression analysis were performed to evaluate independent prognostic factors. Transcriptomic profiles of CSF-1Rhigh and CSF-1Rlow tumor-associated macrophages (TAMs) were investigated. Gene enrichment analysis was used to explore the signal pathways related to CSF-1R. In addition, the relationship between CSF-1R in tumor microenvironment (TME) and tumor immunity was also studied. Results IHC analysis showed that CSF-1R was overexpressed in COAD, and higher expression was associated with shorter overall survival (OS). Immunofluorescence staining showed that CSF-1R was co-localized with macrophage marker CD68. Univariate and multivariate Cox regression analysis showed that CSF-1R was an independent prognostic factor for COAD. The results of gene enrichment analysis showed that CSF-1R was involved in tumor immune response and regulation of TME. In addition, CSF-1R was significantly correlated with TME, immune cell infiltration, TMB, MSI, Neoantigen, and immune checkpoint molecules. Conclusion CSF-1R can serve as an independent prognostic factor of COAD and promising immunotherapeutic target of COAD.
Collapse
Affiliation(s)
- Xingchao Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jianfeng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Baoying Hu
- Department of Immunology, Medical College, Nantong University, Nantong, China
| | - Fei Qian
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
38
|
Wei W, Ding Y, He J, Wu J. Association of CD103+ T cell infiltration with overall survival in solid tumors of the digestive duct and its potential in anti-PD-1 treatment: A review and meta-analysis. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2022; 166:127-135. [PMID: 35352706 DOI: 10.5507/bp.2022.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
We looked into the most recent studies of digestive tumor patients and performed a meta-analysis to explore the association of CD103+ T cell infiltration with overall survival (OS) in solid tumors of the digestive duct. Major databases were searched. The hazard ratios (HR) and 95% confidence intervals (CI) for overall survival were extracted and pooled. A total of 1915 patients from 11 cohorts were included into the present meta-analysis. The pooled HR was 0.64 (95% CI: 0.42-0.96, P=0.03), suggesting that high CD103+ T cell infiltration is associated with better prognosis. Yet significant heterogeneity was revealed and located in the subgroup of CD4+CD103+ T cells. The pooled result indicated that CD103+ T cell infiltration in solid tumors of the digestive duct may possess predictive value for prognosis. Preclinical studies suggested that CD103+ T cell infiltration could predict response to anti-PD-1/PD-L1 treatment.
Collapse
Affiliation(s)
- Wei Wei
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yun Ding
- Department of Radiotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiajia He
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
39
|
Guan M, Jiao Y, Zhou L. Immune Infiltration Analysis with the CIBERSORT Method in Lung Cancer. DISEASE MARKERS 2022; 2022:3186427. [PMID: 35340416 PMCID: PMC8956442 DOI: 10.1155/2022/3186427] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/08/2022] [Accepted: 01/17/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Immune infiltration of lung cancer (LC) is tightly related to clinical results. Nevertheless, past researches have not elucidated the diversities of functionally different cellular types making up the immunoresponse. METHODS In the present research, on the foundation of a deconvolution algorithm (CIBERSORT) and clinically annotated expression profiles, our team studied the tumor-infiltrating immune cells (TIICs) presenting in 502 LC samples and 49 normal samples in a comprehensive way. The fraction of 22 immunocyte subgroups was assessed to identify the relationship among every cellular type and survival and reaction to chemical therapies. RESULTS Consequently, profiles of immunity infiltration change remarkably between paired tumor and precancerous tissues, and the change can describe the diversity of individuals. Of the cellular subgroups studied, cancers without dendritic resting cells or with a decreased quantity of follicular helper T (Tfh) cells were related to the poor prognosis. Correlation analysis between different stages of LC and 22 immune cell subpopulations revealed that the amount of 14 immune cells in LC was remarkably related to tumor stage. The high expression of resting dendritic cells and follicular helper T cells predicted better prognostic value, and univariate analyses proved that two TIICs were significantly associated with patients' prognosis. CONCLUSIONS To sum up, the data herein reveal that there may be subtle differences in the cell constituents of the immune infiltrate in LC, and those diversities may be vital determinating factors of prognostic results and reactions to therapies.
Collapse
Affiliation(s)
- Meng Guan
- Cancer Center, The First Hospital of Jilin University, Changchun 130031, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130031, China
| | - Lili Zhou
- Department of Radiology, The First Hospital of Jilin University, Changchun 130031, China
| |
Collapse
|
40
|
Mei X, Li H, Zhou X, Cheng M, Cui K. The Emerging Role of Tissue-Resident Memory CD8 + T Lymphocytes in Human Digestive Tract Cancers. Front Oncol 2022; 11:819505. [PMID: 35096624 PMCID: PMC8795735 DOI: 10.3389/fonc.2021.819505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
Malignant digestive tract tumors are a great threat to human public health. In addition to surgery, immunotherapy brings hope for the treatment of these tumors. Tissue-resident memory CD8+ T (Trm) cells are a focus of tumor immunology research and treatment due to their powerful cytotoxic effects, ability to directly kill epithelial-derived tumor cells, and overall impact on maintaining mucosal homeostasis and antitumor function in the digestive tract. They are a group of noncirculating immune cells expressing adhesion and migration molecules such as CD69, CD103, and CD49a that primarily reside on the barrier epithelium of nonlymphoid organs and respond rapidly to both viral and bacterial infection and tumorigenesis. This review highlights new research exploring the role of CD8+ Trm cells in a variety of digestive tract malignant tumors, including esophageal cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma. A summary of CD8+ Trm cell phenotypes and characteristics, tissue distribution, and antitumor functions in different tumor environments is provided, illustrating how these cells may be used in immunotherapies against digestive tract tumors.
Collapse
Affiliation(s)
- Xinyu Mei
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huan Li
- Department of Thoracic Surgery, Anhui Provincial Hospital Affiliated With Anhui Medical University, Hefei, China
| | - Xinpeng Zhou
- Department of Thoracic Surgery, Anhui Provincial Hospital, Wannan Medical College, Hefei, China
| | - Min Cheng
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Cancer Immunotherapy Center, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kele Cui
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Cancer Immunotherapy Center, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
41
|
Zhang J, Song C, Tian Y, Yang X. Single-Cell RNA Sequencing in Lung Cancer: Revealing Phenotype Shaping of Stromal Cells in the Microenvironment. Front Immunol 2022; 12:802080. [PMID: 35126365 PMCID: PMC8807562 DOI: 10.3389/fimmu.2021.802080] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
The lung tumor microenvironment, which is composed of heterogeneous cell populations, plays an important role in the progression of lung cancer and is closely related to therapeutic efficacy. Increasing evidence has shown that stromal components play a key role in regulating tumor invasion, metastasis and drug resistance. Therefore, a better understanding of stromal components in the tumor microenvironment is helpful for the diagnosis and treatment of lung cancer. Rapid advances in technology have brought our understanding of disease into the genetic era, and single-cell RNA sequencing has enabled us to describe gene expression profiles with unprecedented resolution, enabling quantitative analysis of gene expression at the single-cell level to reveal the correlations among heterogeneity, signaling pathways, drug resistance and microenvironment molding in lung cancer, which is important for the treatment of this disease. In this paper, several common single-cell RNA sequencing methods and their advantages and disadvantages are briefly introduced to provide a reference for selection of suitable methods. Furthermore, we review the latest progress of single-cell RNA sequencing in the study of stromal cells in the lung tumor microenvironment.
Collapse
|
42
|
Saluzzo S, Pandey RV, Gail LM, Dingelmaier-Hovorka R, Kleissl L, Shaw L, Reininger B, Atzmüller D, Strobl J, Touzeau-Römer V, Beer A, Staud C, Rieger A, Farlik M, Weninger W, Stingl G, Stary G. Delayed antiretroviral therapy in HIV-infected individuals leads to irreversible depletion of skin- and mucosa-resident memory T cells. Immunity 2021; 54:2842-2858.e5. [PMID: 34813775 DOI: 10.1016/j.immuni.2021.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/20/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023]
Abstract
People living with HIV (PLWH) are at increased risk for developing skin and mucosal malignancies despite systemic reconstitution of CD4+ T cells upon antiretroviral therapy (ART). The underlying mechanism of chronic tissue-related immunodeficiency in HIV is unclear. We found that skin CD4+ tissue-resident memory T (Trm) cells were depleted after HIV infection and replenished only upon early ART initiation. TCR clonal analysis following early ART suggested a systemic origin for reconstituting CD4+ Trm cells. Single-cell RNA sequencing in PLWH that received late ART treatment revealed a loss of CXCR3+ Trm cells and a tolerogenic skin immune environment. Human papilloma virus-induced precancerous lesion biopsies showed reduced CXCR3+ Trm cell frequencies in the mucosa in PLWH versus HIV- individuals. These results reveal an irreversible loss of CXCR3+ Trm cells confined to skin and mucosa in PLWH who received late ART treatment, which may be a precipitating factor in the development of HPV-related cancer.
Collapse
Affiliation(s)
- Simona Saluzzo
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria.
| | - Ram Vinay Pandey
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Laura Marie Gail
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | | | - Lisa Kleissl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
| | - Lisa Shaw
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Bärbel Reininger
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Denise Atzmüller
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | | | - Andrea Beer
- Department of Pathology, Medical University of Vienna, Vienna 1090, Austria
| | - Clement Staud
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna 1090, Austria
| | - Armin Rieger
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Georg Stingl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria; LBI-RUD - Ludwig-Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria.
| |
Collapse
|
43
|
Antimetastatic defense by CD8 + T cells. Trends Cancer 2021; 8:145-157. [PMID: 34815204 DOI: 10.1016/j.trecan.2021.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023]
Abstract
Metastasis is an intricate process whereby tumor cells migrate from the primary tumor, survive in the circulation, seed distal organs, and proliferate to create metastatic foci. CD8+ T cells can detect and eliminate tumor cells. Research on CD8+ T cell-dependent antitumor immunity has classically focused on its role in the primary tumor. There is increasing evidence, however, that CD8+ T cells have unique antimetastatic functions in various steps of the metastatic cascade. Here, we review the mechanisms whereby CD8+ T cells control metastatic lesions. We discuss their role in each step of metastasis, metastatic dormancy, and metastatic clonal evolution as well as the consequent clinical repercussions.
Collapse
|
44
|
Liang Y, Gong H, Li Y, Lu Y, Wu X, Zhang X, Ding D, Tang X, Tang Q. Aggregation-Induced Emission-Based Vaccine Improves Potential Antitumor Immunotherapy. J Biomed Nanotechnol 2021; 17:2053-2061. [PMID: 34706805 DOI: 10.1166/jbn.2021.3174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Recently, immunomodulation based on biomaterials has held great promise for preventing and treating cancer. Tumor vaccination can be considered as one of promising immunotherapies, compared with the vaccines for infectious disease, it still stays in its infant. Herein, we designed a near-infrared-emitting AIEgens (named TPE-Ph-DCM) based vaccine as an adjuvant in enhancing immune response. AIE-based photodynamic vaccine exhibited efficiently enhancement of the DC?s antigen prestation and elicited antigen-specific cytotoxic T lymphocyte functionality, and significantly inhibited B16-OVA tumor growth prophylactically and therapeutically in mice model. This study is expected to provide a scientific basis for developing effective and safe tumor vaccines.
Collapse
Affiliation(s)
- Yong Liang
- Central and Clinical Laboratory, The Affiliated Huaian Hospital of Xuzhou Medical University and Huaian Second Hospital, Huaian, 223002, China
| | - Huanle Gong
- Institute of Blood and Marrow Transplantation, Medical College of Soochow University, Soochow University, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Yan Li
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yinghao Lu
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Xiaoqian Wu
- Central and Clinical Laboratory, The Affiliated Huaian Hospital of Xuzhou Medical University and Huaian Second Hospital, Huaian, 223002, China
| | - Xiaoyan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Tianjin, 300071, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Tianjin, 300071, China
| | - Xiaolong Tang
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of the Ministry of Education, Anhui University of Science & Technology, Huainan, 232001, China
| | - Qiyun Tang
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
45
|
Vyas V, Blythe H, Wood EG, Sandhar B, Sarker SJ, Balmforth D, Ambekar SG, Yap J, Edmondson SJ, Di Salvo C, Wong K, Roberts N, Uppal R, Adams B, Shipolini A, Oo AY, Lawrence D, Kolvekar S, Lall KS, Finlay MC, Longhi MP. Obesity and diabetes are major risk factors for epicardial adipose tissue inflammation. JCI Insight 2021; 6:e145495. [PMID: 34283808 PMCID: PMC8409986 DOI: 10.1172/jci.insight.145495] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 07/14/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Epicardial adipose tissue (EAT) directly overlies the myocardium, with changes in its morphology and volume associated with myriad cardiovascular and metabolic diseases. However, EAT’s immune structure and cellular characterization remain incompletely described. We aimed to define the immune phenotype of EAT in humans and compare such profiles across lean, obese, and diabetic patients. METHODS We recruited 152 patients undergoing open-chest coronary artery bypass grafting (CABG), valve repair/replacement (VR) surgery, or combined CABG/VR. Patients’ clinical and biochemical data and EAT, subcutaneous adipose tissue (SAT), and preoperative blood samples were collected. Immune cell profiling was evaluated by flow cytometry and complemented by gene expression studies of immune mediators. Bulk RNA-Seq was performed in EAT across metabolic profiles to assess whole-transcriptome changes observed in lean, obese, and diabetic groups. RESULTS Flow cytometry analysis demonstrated EAT was highly enriched in adaptive immune (T and B) cells. Although overweight/obese and diabetic patients had similar EAT cellular profiles to lean control patients, the EAT exhibited significantly (P ≤ 0.01) raised expression of immune mediators, including IL-1, IL-6, TNF-α, and IFN-γ. These changes were not observed in SAT or blood. Neither underlying coronary artery disease nor the presence of hypertension significantly altered the immune profiles observed. Bulk RNA-Seq demonstrated significant alterations in metabolic and inflammatory pathways in the EAT of overweight/obese patients compared with lean controls. CONCLUSION Adaptive immune cells are the predominant immune cell constituent in human EAT and SAT. The presence of underlying cardiometabolic conditions, specifically obesity and diabetes, rather than cardiac disease phenotype appears to alter the inflammatory profile of EAT. Obese states markedly alter EAT metabolic and inflammatory signaling genes, underlining the impact of obesity on the EAT transcriptome profile. FUNDING Barts Charity MGU0413, Abbott, Medical Research Council MR/T008059/1, and British Heart Foundation FS/13/49/30421 and PG/16/79/32419.
Collapse
Affiliation(s)
- Vishal Vyas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Cardiology, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Hazel Blythe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Elizabeth G Wood
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Balraj Sandhar
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Shah-Jalal Sarker
- Research Department of Medical Education, UCL Medical School, University College London, London, United Kingdom
| | - Damian Balmforth
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Shirish G Ambekar
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - John Yap
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Stephen J Edmondson
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Carmelo Di Salvo
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Kit Wong
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Neil Roberts
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Rakesh Uppal
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Ben Adams
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Alex Shipolini
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Aung Y Oo
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - David Lawrence
- Research Department of Medical Education, UCL Medical School, University College London, London, United Kingdom.,Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Shyam Kolvekar
- Research Department of Medical Education, UCL Medical School, University College London, London, United Kingdom.,Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Kulvinder S Lall
- Department of Cardiac Surgery, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Malcolm C Finlay
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Cardiology, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - M Paula Longhi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
46
|
Foroutan M, Molania R, Pfefferle A, Behrenbruch C, Scheer S, Kallies A, Speed TP, Cursons J, Huntington ND. The Ratio of Exhausted to Resident Infiltrating Lymphocytes Is Prognostic for Colorectal Cancer Patient Outcome. Cancer Immunol Res 2021; 9:1125-1140. [PMID: 34413087 DOI: 10.1158/2326-6066.cir-21-0137] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/14/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapy success in colorectal cancer is mainly limited to patients whose tumors exhibit high microsatellite instability (MSI). However, there is variability in treatment outcomes within this group, which is in part driven by the frequency and characteristics of tumor-infiltrating immune cells. Indeed, the presence of specific infiltrating immune-cell subsets has been shown to correlate with immunotherapy response and is in many cases prognostic of treatment outcome. Tumor-infiltrating lymphocytes (TIL) can undergo distinct differentiation programs, acquiring features of tissue-residency or exhaustion, a process during which T cells upregulate inhibitory receptors, such as PD-1, and lose functionality. Although residency and exhaustion programs of CD8+ T cells are relatively well studied, these programs have only recently been appreciated in CD4+ T cells and remain largely unknown in tumor-infiltrating natural killer (NK) cells. In this study, we used single-cell RNA sequencing (RNA-seq) data to identify signatures of residency and exhaustion in colorectal cancer-infiltrating lymphocytes, including CD8+, CD4+, and NK cells. We then tested these signatures in independent single-cell data from tumor and normal tissue-infiltrating immune cells. Furthermore, we used versions of these signatures designed for bulk RNA-seq data to explore tumor-intrinsic mutations associated with residency and exhaustion from TCGA data. Finally, using two independent transcriptomic datasets from patients with colon adenocarcinoma, we showed that combinations of these signatures, in particular combinations of NK-cell activity signatures, together with tumor-associated signatures, such as TGFβ signaling, were associated with distinct survival outcomes in patients with colon adenocarcinoma.
Collapse
Affiliation(s)
- Momeneh Foroutan
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
| | - Ramyar Molania
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Aline Pfefferle
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,oNKo-innate Pty Ltd., Moonee Ponds, Victoria, Australia
| | - Corina Behrenbruch
- University of Melbourne Centre for Cancer Research, Parkville, Victoria, Australia
| | - Sebastian Scheer
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | - Terence P Speed
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,School of Mathematics & Statistics, The University of Melbourne, Parkville, Victoria, Australia
| | - Joseph Cursons
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia. .,oNKo-innate Pty Ltd., Moonee Ponds, Victoria, Australia
| | - Nicholas D Huntington
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia. .,oNKo-innate Pty Ltd., Moonee Ponds, Victoria, Australia
| |
Collapse
|
47
|
Burn OK, Pankhurst TE, Painter GF, Connor LM, Hermans IF. Harnessing NKT cells for vaccination. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab013. [PMID: 36845569 PMCID: PMC9914585 DOI: 10.1093/oxfimm/iqab013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 11/14/2022] Open
Abstract
Natural killer T (NKT) cells are innate-like T cells capable of enhancing both innate and adaptive immune responses. When NKT cells are stimulated in close temporal association with co-administered antigens, strong antigen-specific immune responses can be induced, prompting the study of NKT cell agonists as novel immune adjuvants. This activity has been attributed to the capacity of activated NKT cells to act as universal helper cells, with the ability to provide molecular signals to dendritic cells and B cells that facilitate T cell and antibody responses, respectively. These signals can override the requirement for conventional CD4+ T cell help, so that vaccines can be designed without need to consider CD4+ T cell repertoire and major histocompatibility complex Class II diversity. Animal studies have highlighted some drawbacks of the approach, namely, concerns around induction of NKT cell hyporesponsiveness, which may limit vaccine boosting, and potential for toxicity. Here we highlight studies that suggest these obstacles can be overcome by targeted delivery in vivo. We also feature new studies that suggest activating NKT cells can help encourage differentiation of T cells into tissue-resident memory cells that play an important role in prophylaxis against infection, and may be required in cancer therapy.
Collapse
Affiliation(s)
- Olivia K Burn
- Malaghan Institute of Medical Research, PO Box 7060, Wellington 6042, New Zealand
| | - Theresa E Pankhurst
- The School of Biological Sciences, Victoria University of Wellington, PO Box 600, Wellington 6140, New Zealand
| | - Gavin F Painter
- The Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone 5046, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Lisa M Connor
- Malaghan Institute of Medical Research, PO Box 7060, Wellington 6042, New Zealand,The School of Biological Sciences, Victoria University of Wellington, PO Box 600, Wellington 6140, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington 6042, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, New Zealand,Correspondence address. Malaghan Institute of Medical Research, Wellington, New Zealand. Tel: +64 4 4996914; E-mail: (I.F.H.)
| |
Collapse
|
48
|
Toh JWT, Ferguson AL, Spring KJ, Mahajan H, Palendira U. Cytotoxic CD8+ T cells and tissue resident memory cells in colorectal cancer based on microsatellite instability and BRAF status. World J Clin Oncol 2021; 12:238-248. [PMID: 33959477 PMCID: PMC8085513 DOI: 10.5306/wjco.v12.i4.238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/14/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recent studies in non-colorectal malignancy have associated T resident memory (TRM) cells with improved patient survival. It is unknown if TRM plays a role in colorectal cancer (CRC).
AIM To examine the potential role of TRM cells in providing immunogenicity in CRC stratified by microsatellite instability (MSI) and BRAF status.
METHODS Patients with known MSI and BRAF mutation status were eligible for inclusion in this study. CRC tumour sections stained with haematoxylin and eosin were microscopically reviewed and the images scanned prior to assessment for location of invading edge and core of tumour. Sequential sections were prepared for quantitative multiplex immunohistochemistry (IHC) staining. Opal Multiplex IHC staining was performed with appropriate positive and negative controls and imaged using a standard fluorescent microscope fitted with a spectral scanning camera (Mantra) in conjunction with Mantra snap software. Images were unmixed and annotated in inForm 2.2.0. Statistical analysis was performed using Graphpad Prism Version 7 and Stata Version 15.
RESULTS Seventy-two patients with known MSI and BRAF status were included in the study. All patients were assessed for MSI by IHC and high resolution capillary electrophoresis testing and 44 of these patients successfully underwent quantitative multiplex IHC staining. Overall, there was a statistically significant increase in CD8+ TRM cells in the MSI (BRAF mutant and wild type) group over the microsatellite stable (MSS) group. There was a statistically significant difference in CD8+ TRM between high level MSI (MSI-H):BRAF mutant [22.57, 95% confidence interval (CI): 14.31-30.84] vs MSS [8.031 (95%CI: 4.698-11.36)], P = 0.0076 andMSI-H:BRAF wild type [16.18 (95%CI: 10.44-21.93)] vs MSS [8.031 (95%CI: 4.698-11.36)], P = 0.0279. There was no statistically significant difference in CD8 T cells (both CD8+CD103- and CD8+CD103+TRM) between MSI-H: BRAF mutant and wild type CRC.
CONCLUSION This study has shown that CD8+ TRM are found in greater abundance in MSI-H CRC, both BRAF mutant and MSI-H:BRAF wild type, when compared with their MSS counterpart. CD8+ TRM may play a role in the immunogenicity in MSI-H CRC (BRAF mutant and BRAF wild type). Further studies should focus on the potential immunogenic qualities of TRM cells and investigate potential immunotherapeutic approaches to improve treatment and survival associated with CRC.
Collapse
Affiliation(s)
- James Wei Tatt Toh
- Division of Surgery and Anaesthesia, Department of Colorectal Surgery, Westmead Hospital, Westmead Clinical School, The University of Sydney, Ingham Institute for Applied Medical Research, Westmead 2145, NSW, Australia
| | - Angela L Ferguson
- Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, Human Viral & Cancer Immunology, Centenary Institute, Charles Perkin Centre, The University of Sydney, Sydney 2000, NSW, Australia
| | - Kevin J Spring
- Medical Oncology Group, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool Clinical School, University of Western Sydney, South Western Clinical School UNSW, Liverpool 2170, NSW, Australia
| | - Hema Mahajan
- Department of Anatomical Pathology, ICPMR, Westmead Hospital, Westmead 2145, NSW, Australia
| | - Umaimainthan Palendira
- Department of Immunology and Infectious Diseases, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney 2000, NSW, Australia
| |
Collapse
|
49
|
Liikanen I, Lauhan C, Quon S, Omilusik K, Phan AT, Bartrolí LB, Ferry A, Goulding J, Chen J, Scott-Browne JP, Yustein JT, Scharping NE, Witherden DA, Goldrath AW. Hypoxia-inducible factor activity promotes antitumor effector function and tissue residency by CD8+ T cells. J Clin Invest 2021; 131:143729. [PMID: 33792560 PMCID: PMC8011896 DOI: 10.1172/jci143729] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
Adoptive T cell therapies (ACTs) hold great promise in cancer treatment, but low overall response rates in patients with solid tumors underscore remaining challenges in realizing the potential of this cellular immunotherapy approach. Promoting CD8+ T cell adaptation to tissue residency represents an underutilized but promising strategy to improve tumor-infiltrating lymphocyte (TIL) function. Here, we report that deletion of the HIF negative regulator von Hippel-Lindau (VHL) in CD8+ T cells induced HIF-1α/HIF-2α-dependent differentiation of tissue-resident memory-like (Trm-like) TILs in mouse models of malignancy. VHL-deficient TILs accumulated in tumors and exhibited a core Trm signature despite an exhaustion-associated phenotype, which led to retained polyfunctionality and response to αPD-1 immunotherapy, resulting in tumor eradication and protective tissue-resident memory. VHL deficiency similarly facilitated enhanced accumulation of chimeric antigen receptor (CAR) T cells with a Trm-like phenotype in tumors. Thus, HIF activity in CD8+ TILs promotes accumulation and antitumor activity, providing a new strategy to enhance the efficacy of ACTs.
Collapse
Affiliation(s)
- Ilkka Liikanen
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Colette Lauhan
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Sara Quon
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Kyla Omilusik
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Anthony T Phan
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Laura Barceló Bartrolí
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Amir Ferry
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - John Goulding
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Joyce Chen
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, California, USA
| | - James P Scott-Browne
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Jason T Yustein
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, Texas, USA
| | - Nicole E Scharping
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Deborah A Witherden
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| | - Ananda W Goldrath
- Division of Biological Sciences, Section of Molecular Biology, University of California San Diego, San Diego, California, USA
| |
Collapse
|
50
|
Cao H, Quan S, Zhang L, Chen Y, Jiao G. BMPR2 expression level is correlated with low immune infiltration and predicts metastasis and poor survival in osteosarcoma. Oncol Lett 2021; 21:391. [PMID: 33777214 PMCID: PMC7988701 DOI: 10.3892/ol.2021.12652] [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: 02/14/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most common malignant bone tumor in adolescents and young adults, and identifying biomarkers for prognosis and therapy is necessary. Bone morphogenetic protein receptor 2 (BMPR2) is involved in various cellular functions, including cell adhesion, proliferation and invasion, inflammation, apoptosis and metastatic spread. However, the correlation between BMPR2 expression levels and prognosis and tumor-infiltrating immune cells in osteosarcoma is not well understood. In the present study, the expression level of BMPR2 was investigated using the Oncomine and R2 databases. The association between the expression level of BMPR2 and the clinical prognosis of patients with cancer was analyzed using the R2 database. The relationship between the expression level of BMPR2 and immune cell infiltration in the stroma of osteosarcoma was assessed using the Tumor Immune Estimation Resource (TIMER) and CIBERSORT. The correlations between BMPR2 expression level and infiltrated immune cell gene marker sets in osteosarcoma were validated in the TIMER and R2 databases. Analysis of a cohort of patients with osteosarcoma revealed that BMPR2 expression was significantly higher in osteosarcoma compared with in normal tissue and was correlated with poor prognosis. M0 macrophages, M2 macrophages, resting mast, γ δ T and CD8+ T cells were the top five immune cells with the highest degrees of infiltration in osteosarcoma. In addition, BMPR2 expression level showed a significant negative correlation with the gene markers of CD8+ T cells, monocytes and M2 macrophages. Low levels of infiltrating CD8+ T cells, monocytes or M2 macrophages in osteosarcoma was significantly associated with poor survival. These data suggested that CD8+ T cells, monocytes and M2 macrophages play significant roles in the establishment of the immune microenvironment of osteosarcoma. High BMPR2 expression was associated with poor prognosis and low infiltration of CD8+ T cells, monocytes and M2 macrophages in osteosarcoma. Hence, BMPR2 can be considered a biomarker of the immune infiltration, metastasis and prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Hongxin Cao
- Department of Medical Oncology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Key Laboratory of Chemical Biology, Ministry of Education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shuang Quan
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lu Zhang
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Spine and Spinal Cord Disease Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yunzhen Chen
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Spine and Spinal Cord Disease Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Guangjun Jiao
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Spine and Spinal Cord Disease Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|