201
|
Ai LJ, Li GD, Chen G, Sun ZQ, Zhang JN, Liu M. Molecular subtyping and the construction of a predictive model of colorectal cancer based on ion channel genes. Eur J Med Res 2024; 29:219. [PMID: 38576045 PMCID: PMC10993535 DOI: 10.1186/s40001-024-01819-2] [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: 11/28/2023] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
PURPOSE Colorectal cancer (CRC) is a highly heterogeneous malignancy with an unfavorable prognosis. The purpose of this study was to address the heterogeneity of CRC by categorizing it into ion channel subtypes, and to develop a predictive modeling based on ion channel genes to predict the survival and immunological states of patients with CRC. The model will provide guidance for personalized immunotherapy and drug treatment. METHODS A consistent clustering method was used to classify 619 CRC samples based on the expression of 279 ion channel genes. Such a method was allowed to investigate the relationship between molecular subtypes, prognosis, and immune infiltration. Furthermore, a predictive modeling was constructed for ion channels to evaluate the ion channel properties of individual tumors using the least absolute shrinkage and selection operator. The expression patterns of the characteristic genes were validated through molecular biology experiments. The effect of potassium channel tetramerization domain containing 9 (KCTD9) on CRC was verified by cellular functional experiments. RESULTS Four distinct ion channel subtypes were identified in CRC, each characterized by unique prognosis and immune infiltration patterns. Notably, Ion Cluster3 exhibited high levels of immune infiltration and a favorable prognosis, while Ion Cluster4 showed relatively lower levels of immune infiltration and a poorer prognosis. The ion channel score could predict overall survival, with lower scores correlated with longer survival. This score served as an independent prognostic factor and presented an excellent predictive efficacy in the nomogram. In addition, the score was closely related to immune infiltration, immunotherapy response, and chemotherapy sensitivity. Experimental evidence further confirmed that low expression of KCTD9 in tumor tissues was associated with an unfavorable prognosis in patients with CRC. The cellular functional experiments demonstrated that KCTD9 inhibited the proliferation, migration and invasion capabilities of LOVO cells. CONCLUSIONS Ion channel subtyping and scoring can effectively predict the prognosis and evaluate the immune microenvironment, immunotherapy response, and drug sensitivity in patients with CRC.
Collapse
Affiliation(s)
- Lian-Jie Ai
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Guo-Dong Li
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Gang Chen
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Zi-Quan Sun
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Jin-Ning Zhang
- Colorectal Tumor Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Ming Liu
- General Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
202
|
Qiu H, Liang J, Yang G, Xie Z, Wang Z, Wang L, Zhang J, Nanda HS, Zhou H, Huang Y, Peng X, Lu C, Chen H, Zhou Y. Application of exosomes in tumor immunity: recent progresses. Front Cell Dev Biol 2024; 12:1372847. [PMID: 38633106 PMCID: PMC11021734 DOI: 10.3389/fcell.2024.1372847] [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: 01/18/2024] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Exosomes are small extracellular vesicles secreted by cells, ranging in size from 30 to 150 nm. They contain proteins, nucleic acids, lipids, and other bioactive molecules, which play a crucial role in intercellular communication and material transfer. In tumor immunity, exosomes present various functions while the following two are of great importance: regulating the immune response and serving as delivery carriers. This review starts with the introduction of the formation, compositions, functions, isolation, characterization, and applications of exosomes, and subsequently discusses the current status of exosomes in tumor immunotherapy, and the recent applications of exosome-based tumor immunity regulation and antitumor drug delivery. Finally, current challenge and future prospects are proposed and hope to demonstrate inspiration for targeted readers in the field.
Collapse
Affiliation(s)
- Haiyan Qiu
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Junting Liang
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Guang Yang
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zhenyu Xie
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zhenpeng Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Liyan Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jingying Zhang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Himansu Sekhar Nanda
- Biomedical Engineering and Technology Lab, Discipline of Mechanical Engineering, PDPM Indian Institute of Information Technology Design and Manufacturing Jabalpur, Jabalpur, Madhya Pradesh, India
| | - Hui Zhou
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yong Huang
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xinsheng Peng
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Chengyu Lu
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Huizhi Chen
- School of Pharmacy, Guangdong Medical University, Dongguan, China
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yubin Zhou
- School of Pharmacy, Guangdong Medical University, Dongguan, China
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
203
|
Lei X, de Groot DC, Welters MJP, de Wit T, Schrama E, van Eenennaam H, Santegoets SJ, Oosenbrug T, van der Veen A, Vos JL, Zuur CL, de Miranda NFCC, Jacobs H, van der Burg SH, Borst J, Xiao Y. CD4 + T cells produce IFN-I to license cDC1s for induction of cytotoxic T-cell activity in human tumors. Cell Mol Immunol 2024; 21:374-392. [PMID: 38383773 PMCID: PMC10978876 DOI: 10.1038/s41423-024-01133-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/05/2024] [Indexed: 02/23/2024] Open
Abstract
CD4+ T cells can "help" or "license" conventional type 1 dendritic cells (cDC1s) to induce CD8+ cytotoxic T lymphocyte (CTL) anticancer responses, as proven in mouse models. We recently identified cDC1s with a transcriptomic imprint of CD4+ T-cell help, specifically in T-cell-infiltrated human cancers, and these cells were associated with a good prognosis and response to PD-1-targeting immunotherapy. Here, we delineate the mechanism of cDC1 licensing by CD4+ T cells in humans. Activated CD4+ T cells produce IFNβ via the STING pathway, which promotes MHC-I antigen (cross-)presentation by cDC1s and thereby improves their ability to induce CTL anticancer responses. In cooperation with CD40 ligand (L), IFNβ also optimizes the costimulatory and other functions of cDC1s required for CTL response induction. IFN-I-producing CD4+ T cells are present in diverse T-cell-infiltrated cancers and likely deliver "help" signals to CTLs locally, according to their transcriptomic profile and colocalization with "helped/licensed" cDCs and tumor-reactive CD8+ T cells. In agreement with this scenario, the presence of IFN-I-producing CD4+ T cells in the TME is associated with overall survival and the response to PD-1 checkpoint blockade in cancer patients.
Collapse
Affiliation(s)
- Xin Lei
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniël C de Groot
- Department of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marij J P Welters
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom de Wit
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen Schrama
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Saskia J Santegoets
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Timo Oosenbrug
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Joris L Vos
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Charlotte L Zuur
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Otorhinolaryngology Leiden University Medical Center, Leiden, The Netherlands
| | | | - Heinz Jacobs
- Department of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sjoerd H van der Burg
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Jannie Borst
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands.
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Yanling Xiao
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands.
- Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
204
|
Guo M, Liu MYR, Brooks DG. Regulation and impact of tumor-specific CD4 + T cells in cancer and immunotherapy. Trends Immunol 2024; 45:303-313. [PMID: 38508931 DOI: 10.1016/j.it.2024.02.005] [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: 02/20/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/22/2024]
Abstract
CD4+ T cells are crucial in generating and sustaining immune responses. They orchestrate and fine-tune mammalian innate and adaptive immunity through cell-based interactions and the release of cytokines. The role of these cells in contributing to the efficacy of antitumor immunity and immunotherapy has just started to be uncovered. Yet, many aspects of the CD4+ T cell response are still unclear, including the differentiation pathways controlling such cells during cancer progression, the external signals that program them, and how the combination of these factors direct ensuing immune responses or immune-restorative therapies. In this review, we focus on recent advances in understanding CD4+ T cell regulation during cancer progression and the importance of CD4+ T cells in immunotherapies.
Collapse
Affiliation(s)
- Mengdi Guo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Melissa Yi Ran Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - David G Brooks
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
205
|
Sharma N, Fan X, Atolagbe OT, Ge Z, Dao KN, Sharma P, Allison JP. ICOS costimulation in combination with CTLA-4 blockade remodels tumor-associated macrophages toward an antitumor phenotype. J Exp Med 2024; 221:e20231263. [PMID: 38517331 PMCID: PMC10959121 DOI: 10.1084/jem.20231263] [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: 07/19/2023] [Revised: 01/19/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024] Open
Abstract
We have previously demonstrated synergy between ICOS costimulation (IVAX; ICOSL-transduced B16-F10 cellular vaccine) and CTLA-4 blockade in antitumor therapy. In this study, we employed CyTOF and single-cell RNA sequencing and observed significant remodeling of the lymphoid and myeloid compartments in combination therapy. Compared with anti-CTLA-4 monotherapy, the combination therapy enriched Th1 CD4 T cells, effector CD8 T cells, and M1-like antitumor proinflammatory macrophages. These macrophages were critical to the therapeutic efficacy of anti-CTLA-4 combined with IVAX or anti-PD-1. Macrophage depletion with clodronate reduced the tumor-infiltrating effector CD4 and CD8 T cells, impairing their antitumor functions. Furthermore, the recruitment and polarization of M1-like macrophages required IFN-γ. Therefore, in this study, we show that there is a positive feedback loop between intratumoral effector T cells and tumor-associated macrophages (TAMs), in which the IFN-γ produced by the T cells polarizes the TAMs into M1-like phenotype, and the TAMs, in turn, reshape the tumor microenvironment to facilitate T cell infiltration, immune function, and tumor rejection.
Collapse
Affiliation(s)
- Naveen Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaozhou Fan
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Zhongqi Ge
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly N. Dao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Padmanee Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Immunotherapy Platform, James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James P. Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
206
|
Gupta R, Gupta J, Roy S. Exosomes: Key Players for Treatment of Cancer and Their Future Perspectives. Assay Drug Dev Technol 2024; 22:118-147. [PMID: 38407852 DOI: 10.1089/adt.2023.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Affiliation(s)
- Reena Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Suchismita Roy
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| |
Collapse
|
207
|
Zhao Q, Bai L, Zhu D, Li T, Xu J, Xu Y, Zhou X. Clinical efficacy and potential mechanism of ginseng polysaccharides in the treatment of non-small cell lung cancer based on meta-analysis associated with network pharmacology. Heliyon 2024; 10:e27152. [PMID: 38496882 PMCID: PMC10944195 DOI: 10.1016/j.heliyon.2024.e27152] [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: 10/25/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 03/19/2024] Open
Abstract
Background The ginseng polysaccharide injection is a well-known traditional Chinese medicine often employed as a supplementary treatment for cancer. This treatment can not only alleviate the adverse effects caused by tumor radiotherapy and chemotherapy but also enhance the immune system of individuals diagnosed with lung cancer. It is important to acknowledge the efficacy of ginseng polysaccharide injection in the treatment of non-small cell lung cancer (NSCLC). However, these small-sample studies may have certain biases, and the underlying mechanisms of ginseng polysaccharides therapy for NSCLC are still unclear. Methods The present study involved a systematic review of the literature on randomized controlled trials (RCTs) focusing on using ginseng polysaccharide injection as a therapeutic approach for NSCLC. Seven databases were searched for eligible studies published before April 2023. Two researchers independently managed data extraction, risk of bias assessment, and data analyses using RevMan 5.3 software. In network pharmacology, we thoroughly searched the relevant literature on ginseng polysaccharides (GPs) and the PubChem database. This search aimed to identify the main active ingredients and targets associated with ginseng polysaccharides. Subsequently, we compared these targets with those of NSCLC and utilized bioinformatics techniques to analyze and explore their potential interactions. Results A total of 11 RCTs involving 845 patients with NSCLC were included in the meta-analysis. The meta-analysis revealed that ginseng polysaccharide injection combined significantly improved the objective response rate [RR = 1.45, 95% CI (1.26, 1.67), P < 0.00001]. Furthermore, it was observed that ginseng polysaccharide injection increased the serum levels of CD4+ T-lymphocytes (CD4+ T) [MD = 8.98, 95% CI (5.18, 12.78), P < 0.00001], and decreased the serum levels of CD8+ T-lymphocytes (CD8+ T) [MD = -2.68, 95% CI (-4.66, -0.70), P = 0.008]. Through network pharmacology analysis, a total of 211 target genes of GPs and 81 common targets were identified. GAPDH, EGFR, VEGFA, JUN, SRC, CASP3, STAT3, CCND1, HSP90AA1, and MMP9 were identified as the core target proteins. Additionally, KEGG enrichment analysis revealed 122 relevant signaling pathways, including Pathways in cancer, PD-L1 expression and PD-1 checkpoint pathway in cancer, and Proteoglycans in cancer. Conclusion Ginseng polysaccharide injection can improve the ORR of patients with NSCLC, increase the serum levels of CD4+ T, and decrease the serum levels of CD8+ T. The potential mechanism may be associated with the PD-1/PD-L1 signaling pathway.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Le Bai
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Dongwei Zhu
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Tingyuan Li
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Jie Xu
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Yong Xu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianmei Zhou
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| |
Collapse
|
208
|
Safaei S, Fadaee M, Farzam OR, Yari A, Poursaei E, Aslan C, Samemaleki S, Shanehbandi D, Baradaran B, Kazemi T. Exploring the dynamic interplay between exosomes and the immune tumor microenvironment: implications for breast cancer progression and therapeutic strategies. Breast Cancer Res 2024; 26:57. [PMID: 38553754 PMCID: PMC10981336 DOI: 10.1186/s13058-024-01810-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
Breast cancer continues to pose a substantial worldwide health concern, demanding a thorough comprehension of the complex interaction between cancerous cells and the immune system. Recent studies have shown the significant function of exosomes in facilitating intercellular communication and their participation in the advancement of cancer. Tumor-derived exosomes have been identified as significant regulators in the context of breast cancer, playing a crucial role in modulating immune cell activity and contributing to the advancement of the illness. This study aims to investigate the many effects of tumor-derived exosomes on immune cells in the setting of breast cancer. Specifically, we will examine their role in influencing immune cell polarization, facilitating immunological evasion, and modifying the tumor microenvironment. Furthermore, we explore the nascent domain of exosomes produced from immune cells and their prospective involvement in the prevention of breast cancer. This paper focuses on new research that emphasizes the immunomodulatory characteristics of exosomes produced from immune cells. It also explores the possibility of these exosomes as therapeutic agents or biomarkers for the early identification and prevention of breast cancer. The exploration of the reciprocal connections between exosomes formed from tumors and immune cells, together with the rising significance of exosomes derived from immune cells, presents a potential avenue for the advancement of novel approaches in the field of breast cancer therapy and prevention.
Collapse
Affiliation(s)
- Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Manouchehr Fadaee
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Elham Poursaei
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cynthia Aslan
- Research Center for Integrative Medicine in Aging, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Samemaleki
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran.
| |
Collapse
|
209
|
Li J, Wei Y, Liu J, Cheng S, Zhang X, Qiu H, Li J, He C. Integrative analysis of metabolism subtypes and identification of prognostic metabolism-related genes for glioblastoma. Biosci Rep 2024; 44:BSR20231400. [PMID: 38419527 PMCID: PMC10965397 DOI: 10.1042/bsr20231400] [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: 08/16/2023] [Revised: 02/01/2024] [Accepted: 02/28/2024] [Indexed: 03/02/2024] Open
Abstract
Increasing evidence has demonstrated that cancer cell metabolism is a critical factor in tumor development and progression; however, its role in glioblastoma (GBM) remains limited. In the present study, we classified GBM into three metabolism subtypes (MC1, MC2, and MC3) through cluster analysis of 153 GBM samples from the RNA-sequencing data of The Cancer Genome Atlas (TCGA) based on 2752 metabolism-related genes (MRGs). We further explored the prognostic value, metabolic signatures, immune infiltration, and immunotherapy sensitivity of the three metabolism subtypes. Moreover, the metabolism scoring model was established to quantify the different metabolic characteristics of the patients. Results showed that MC3, which is associated with a favorable survival outcome, had higher proportions of isocitrate dehydrogenase (IDH) mutations and lower tumor purity and proliferation. The MC1 subtype, which is associated with the worst prognosis, shows a higher number of segments and homologous recombination defects and significantly lower mRNA expression-based stemness index (mRNAsi) and epigenetic-regulation-based mRNAsi. The MC2 subtype has the highest T-cell exclusion score, indicating a high likelihood of immune escape. The results were validated using an independent dataset. Five MRGs (ACSL1, NDUFA2, CYP1B1, SLC11A1, and COX6B1) correlated with survival outcomes were identified based on metabolism-related co-expression module analysis. Laboratory-based validation tests further showed the expression of these MRGs in GBM tissues and how their expression influences cell function. The results provide a reference for developing clinical management approaches and treatments for GBM.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Rehabilitation Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215228, China
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Yutian Wei
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jiali Liu
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Shupeng Cheng
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Xia Zhang
- Center of Rehabilitation Medicine, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi Province 710054, China
| | - Huaide Qiu
- Faculty of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing, Jiangsu Province 210038, China
| | - Jianan Li
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Chuan He
- Department of Rehabilitation Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu Province 215228, China
| |
Collapse
|
210
|
Ninmer EK, Zhu H, Chianese-Bullock KA, von Mehren M, Haas NB, Ross MI, Dengel LT, Slingluff CL. Multipeptide vaccines for melanoma in the adjuvant setting: long-term survival outcomes and post-hoc analysis of a randomized phase II trial. Nat Commun 2024; 15:2570. [PMID: 38519525 PMCID: PMC10959948 DOI: 10.1038/s41467-024-46877-6] [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: 11/20/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024] Open
Abstract
The critical roles of CD4+ T cells have been understudied for cancer vaccines. Here we report long-term clinical outcomes of a randomized multicenter phase II clinical trial (NCT00118274), where patients with high-risk melanoma received a multipeptide vaccine targeting CD8+ T cells (12MP) and were randomized to receive either of two vaccines for CD4+ (helper) T cells: 6MHP (6 melanoma-specific helper peptides), or tet (a nonspecific helper peptide from tetanus toxoid). Cyclophosphamide (Cy) pre-treatment was also assessed. Primary outcomes for T cell responses to 12MP, 6MHP, and tet were previously reported, suggesting immunogenicity of both vaccines but that CD8 T cell responses to 12MP were lower when tet was replaced with 6MHP. Here, in post-hoc analyses, we report durable prolongation of overall survival by adding 6MHP instead of tet. That benefit was experienced only by male patients. A favorable interaction of 6MHP and Cy is also suggested. Multivariable Cox regression analysis of the intent-to-treat population identify vaccine arm (12MP + 6MHP+Cy) and patient sex (male) as the two significant predictors of enhanced survival. These findings support the value of adding cognate T cell help to cancer vaccines and also suggest a need to assess the impact of patient sex on immune therapy outcomes.
Collapse
Affiliation(s)
- Emily K Ninmer
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Hong Zhu
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- University of Virginia, School of Medicine, Cancer Center, Charlottesville, VA, USA
| | - Kimberly A Chianese-Bullock
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
- University of Virginia, School of Medicine, Cancer Center, Charlottesville, VA, USA
| | | | - Naomi B Haas
- Fox Chase Cancer Center, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Merrick I Ross
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Lynn T Dengel
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA.
- University of Virginia, School of Medicine, Cancer Center, Charlottesville, VA, USA.
| |
Collapse
|
211
|
Peng S, Lin A, Jiang A, Zhang C, Zhang J, Cheng Q, Luo P, Bai Y. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer 2024; 23:58. [PMID: 38515134 PMCID: PMC10956324 DOI: 10.1186/s12943-024-01972-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) play critical antitumor roles, encompassing diverse subsets including CD4+, NK, and γδ T cells beyond conventional CD8+ CTLs. However, definitive CTLs biomarkers remain elusive, as cytotoxicity-molecule expression does not necessarily confer cytotoxic capacity. CTLs differentiation involves transcriptional regulation by factors such as T-bet and Blimp-1, although epigenetic regulation of CTLs is less clear. CTLs promote tumor killing through cytotoxic granules and death receptor pathways, but may also stimulate tumorigenesis in some contexts. Given that CTLs cytotoxicity varies across tumors, enhancing this function is critical. This review summarizes current knowledge on CTLs subsets, biomarkers, differentiation mechanisms, cancer-related functions, and strategies for improving cytotoxicity. Key outstanding questions include refining the CTLs definition, characterizing subtype diversity, elucidating differentiation and senescence pathways, delineating CTL-microbe relationships, and enabling multi-omics profiling. A more comprehensive understanding of CTLs biology will facilitate optimization of their immunotherapy applications. Overall, this review synthesizes the heterogeneity, regulation, functional roles, and enhancement strategies of CTLs in antitumor immunity, highlighting gaps in our knowledge of subtype diversity, definitive biomarkers, epigenetic control, microbial interactions, and multi-omics characterization. Addressing these questions will refine our understanding of CTLs immunology to better leverage cytotoxic functions against cancer.
Collapse
Affiliation(s)
- Shengkun Peng
- Department of Radiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and ImmunologySchool of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South University, Hunan, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
212
|
Guan XL, Guan XY, Zhang ZY. Roles and application of exosomes in the development, diagnosis and treatment of gastric cancer. World J Gastrointest Oncol 2024; 16:630-642. [PMID: 38577463 PMCID: PMC10989387 DOI: 10.4251/wjgo.v16.i3.630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/18/2023] [Accepted: 01/15/2024] [Indexed: 03/12/2024] Open
Abstract
As important messengers of intercellular communication, exosomes can regulate local and distant cellular communication by transporting specific exosomal contents and can also promote or suppress the development and progression of gastric cancer (GC) by regulating the growth and proliferation of tumor cells, the tumor-related immune response and tumor angiogenesis. Exosomes transport bioactive molecules including DNA, proteins, and RNA (coding and noncoding) from donor cells to recipient cells, causing reprogramming of the target cells. In this review, we will describe how exosomes regulate the cellular immune response, tumor angiogenesis, proliferation and metastasis of GC cells, and the role and mechanism of exosome-based therapy in human cancer. We will also discuss the potential application value of exosomes as biomarkers in the diagnosis and treatment of GC and their relationship with drug resistance.
Collapse
Affiliation(s)
- Xiao-Li Guan
- Department of General Medicine, The Second Hospital of Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Xiao-Ying Guan
- Department of Pathology, The Second Hospital of Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Zheng-Yi Zhang
- Department of General Medicine, The Second Hospital of Lanzhou University, Lanzhou 730030, Gansu Province, China
| |
Collapse
|
213
|
Gao Y, Gong Y, Lu J, Hao H, Shi X. Targeting YAP1 to improve the efficacy of immune checkpoint inhibitors in liver cancer: mechanism and strategy. Front Immunol 2024; 15:1377722. [PMID: 38550587 PMCID: PMC10972981 DOI: 10.3389/fimmu.2024.1377722] [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: 01/28/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
Liver cancer is the third leading of tumor death, including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Immune checkpoint inhibitors (ICIs) are yielding much for sufferers to hope for patients, but only some patients with advanced liver tumor respond. Recent research showed that tumor microenvironment (TME) is critical for the effectiveness of ICIs in advanced liver tumor. Meanwhile, metabolic reprogramming of liver tumor leads to immunosuppression in TME. These suggest that regulating the abnormal metabolism of liver tumor cells and firing up TME to turn "cold tumor" into "hot tumor" are potential strategies to improve the therapeutic effect of ICIs in liver tumor. Previous studies have found that YAP1 is a potential target to improve the efficacy of anti-PD-1 in HCC. Here, we review that YAP1 promotes immunosuppression of TME, mainly due to the overstimulation of cytokines in TME by YAP1. Subsequently, we studied the effects of YAP1 on metabolic reprogramming in liver tumor cells, including glycolysis, gluconeogenesis, lipid metabolism, arachidonic acid metabolism, and amino acid metabolism. Lastly, we summarized the existing drugs targeting YAP1 in the treatment of liver tumor, including some medicines from natural sources, which have the potential to improve the efficacy of ICIs in the treatment of liver tumor. This review contributed to the application of targeted YAP1 for combined therapy with ICIs in liver tumor patients.
Collapse
Affiliation(s)
- Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Junlan Lu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Huiqin Hao
- Chinese Medicine Gene Expression Regulation Laboratory, State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
- Basic Laboratory of Integrated Traditional Chinese and Western, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xinli Shi
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| |
Collapse
|
214
|
Wu R, Li D, Zhang S, Wang J, Chen K, Tuo Z, Miyamoto A, Yoo KH, Wei W, Zhang C, Feng D, Han P. A pan-cancer analysis of the oncogenic and immunological roles of transglutaminase 1 (TGM1) in human cancer. J Cancer Res Clin Oncol 2024; 150:123. [PMID: 38472489 PMCID: PMC10933153 DOI: 10.1007/s00432-024-05640-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND There is currently a limited number of studies on transglutaminase type 1 (TGM1) in tumors. The objective of this study is to perform a comprehensive analysis across various types of cancer to determine the prognostic significance of TGM1 in tumors and investigate its role in the immune environment. METHOD Pan-cancer and mutational data were retrieved from the TCGA database and analyzed using R (version 3.6.4) and its associated software package. The expression difference and prognosis of TGM1 were examined, along with its correlation with tumor heterogeneity, stemness, mutation landscape, and RNA modification. Additionally, the relationship between TGM1 expression and tumor immunity was investigated using the TIMER method. RESULTS TGM1 is expressed differently in various tumors and normal samples and is associated with the overall survival and progression-free time of KIRC, ACC, SKCM, LIHC, and STES. In LICH, we found a negative correlation between TGM1 expression and 6 indicators of tumor stemness. The mutation frequencies of BLCA, LIHC, and KIRC were 1.7%, 0.3%, and 0.3% respectively. In BLCA and BRCA, there was a significant correlation between TGM1 expression and the infiltration of CD4 + T cells, CD8 + T cells, neutrophils, and dendritic cells. CONCLUSION TGM1 has the potential to serve as both a prognostic marker and a drug target.
Collapse
Affiliation(s)
- Ruicheng Wu
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuxia Zhang
- Research Core Facilities, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Chen
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Akira Miyamoto
- Department of Rehabilitation, West Kyushu University, Fukuoka, Japan
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, South Korea
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| | - Ping Han
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
215
|
Franken A, Bila M, Mechels A, Kint S, Van Dessel J, Pomella V, Vanuytven S, Philips G, Bricard O, Xiong J, Boeckx B, Hatse S, Van Brussel T, Schepers R, Van Aerde C, Geurs S, Vandecaveye V, Hauben E, Vander Poorten V, Verbandt S, Vandereyken K, Qian J, Tejpar S, Voet T, Clement PM, Lambrechts D. CD4 + T cell activation distinguishes response to anti-PD-L1+anti-CTLA4 therapy from anti-PD-L1 monotherapy. Immunity 2024; 57:541-558.e7. [PMID: 38442708 DOI: 10.1016/j.immuni.2024.02.007] [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: 12/26/2022] [Revised: 11/30/2023] [Accepted: 02/08/2024] [Indexed: 03/07/2024]
Abstract
Cancer patients often receive a combination of antibodies targeting programmed death-ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen-4 (CTLA4). We conducted a window-of-opportunity study in head and neck squamous cell carcinoma (HNSCC) to examine the contribution of anti-CTLA4 to anti-PD-L1 therapy. Single-cell profiling of on- versus pre-treatment biopsies identified T cell expansion as an early response marker. In tumors, anti-PD-L1 triggered the expansion of mostly CD8+ T cells, whereas combination therapy expanded both CD4+ and CD8+ T cells. Such CD4+ T cells exhibited an activated T helper 1 (Th1) phenotype. CD4+ and CD8+ T cells co-localized with and were surrounded by dendritic cells expressing T cell homing factors or antibody-producing plasma cells. T cell receptor tracing suggests that anti-CTLA4, but not anti-PD-L1, triggers the trafficking of CD4+ naive/central-memory T cells from tumor-draining lymph nodes (tdLNs), via blood, to the tumor wherein T cells acquire a Th1 phenotype. Thus, CD4+ T cell activation and recruitment from tdLNs are hallmarks of early response to anti-PD-L1 plus anti-CTLA4 in HNSCC.
Collapse
Affiliation(s)
- Amelie Franken
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Michel Bila
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Department of General Medical Oncology, UZ Leuven, 3000 Leuven, Belgium; Department of Oral and Maxillofacial Surgery, UZ Leuven, Leuven 3000, Belgium
| | - Aurelie Mechels
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Sam Kint
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Jeroen Van Dessel
- Department of Oral and Maxillofacial Surgery, UZ Leuven, Leuven 3000, Belgium
| | | | - Sebastiaan Vanuytven
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Gino Philips
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Orian Bricard
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Jieyi Xiong
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Department of General Medical Oncology, UZ Leuven, 3000 Leuven, Belgium
| | - Thomas Van Brussel
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Rogier Schepers
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium
| | - Cedric Van Aerde
- Department of Imaging and Pathology, KU Leuven, UZ Leuven, Leuven 3000, Belgium
| | - Sarah Geurs
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium; Department of Biomolecular Medicine, UZ Ghent, Ghent 9052, Belgium
| | | | - Esther Hauben
- Otorhinolaryngology, Head and Neck Surgery, Leuven 3000, Belgium
| | - Vincent Vander Poorten
- Otorhinolaryngology, Head and Neck Surgery, Leuven 3000, Belgium; Department of Oncology, Section Head and Neck Oncology, Leuven 3000, Belgium
| | - Sara Verbandt
- Digestive Oncology, KU Leuven, UZ Leuven, Leuven 3000, Belgium
| | - Katy Vandereyken
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sabine Tejpar
- Digestive Oncology, KU Leuven, UZ Leuven, Leuven 3000, Belgium
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium
| | - Paul M Clement
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Department of General Medical Oncology, UZ Leuven, 3000 Leuven, Belgium.
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; VIB Center for Cancer Biology, Leuven 3000, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), Leuven 3000, Belgium.
| |
Collapse
|
216
|
Wang YX, Zhou CP, Wang DT, Ma J, Sun XH, Wang Y, Zhang YM. Unraveling the causal role of immune cells in gastrointestinal tract cancers: insights from a Mendelian randomization study. Front Immunol 2024; 15:1343512. [PMID: 38533503 PMCID: PMC10963466 DOI: 10.3389/fimmu.2024.1343512] [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: 11/23/2023] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Background Despite early attempts, the relationship between immune characteristics and gastrointestinal tract cancers remains incompletely elucidated. Hence, rigorous and further investigations in this domain hold significant clinical relevance for the development of novel potential immunotherapeutic targets. Methods We conducted a two-sample Mendelian randomization (MR) analysis using the tools available in the "TwoSampleMR" R package. The GWAS data for these 731 immune traits were sourced from the GWAS Catalog database. Concurrently, data on gastrointestinal tract cancers, encompassing malignant tumors in the esophagus, stomach, small intestine, colon, and rectum, were extracted from the FinnGen database. The immune traits subjected to MR analysis predominantly fall into four categories: median fluorescence intensities (MFI), relative cell (RC), absolute cell (AC), and morphological parameters (MP). To ensure the reliability of our findings, sensitivity analyses were implemented to address robustness, account for heterogeneity, and alleviate the impact of horizontal pleiotropy. Results A total of 78 immune traits causally linked to gastrointestinal tract cancers were identified, encompassing esophageal cancer (12 traits), gastric cancer (13 traits), small intestine cancer (22 traits), colon cancer (12 traits), and rectal cancer (19 traits). Additionally, 60 immune traits were recognized as protective factors associated with gastrointestinal tract cancers, distributed across esophageal cancer (14 traits), gastric cancer (16 traits), small intestine cancer (7 traits), colon cancer (14 traits), and rectal cancer (9 traits). Furthermore, it was observed that seven immune traits are causally related to gastrointestinal tract cancers in at least two locations. These traits include "CCR2 on CD14- CD16+ monocyte," "CD19 on IgD+ CD38-," "CD19 on IgD+ CD38- naive," "CD25hi CD45RA+ CD4 not Treg AC," "CD27 on unsw mem," "CD28 on CD39+ activated Treg," and "CD45 on CD4+." Conclusion This study elucidates a causal link between immune cells and gastrointestinal tract cancers at various sites through genetic investigation. The findings of this research open up new perspectives and resources for exploring tumor prevention strategies and immunotherapeutic targets.
Collapse
Affiliation(s)
- Yu-xiang Wang
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Chao-ping Zhou
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Da-tian Wang
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Jun Ma
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Xue-hu Sun
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yao Wang
- Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ya-ming Zhang
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| |
Collapse
|
217
|
Sun Z, Sun Z, Liu J, Gao X, Jiao L, Zhao Q, Chu Y, Wang X, Deng G, Cai L. Engineered Extracellular Vesicles Expressing Siglec-10 Camouflaged AIE Photosensitizer to Reprogram Macrophages to Active M1 Phenotype and Present Tumor-Associated Antigens for Photodynamic Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307147. [PMID: 37941517 DOI: 10.1002/smll.202307147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/15/2023] [Indexed: 11/10/2023]
Abstract
Cancer immunotherapy has attracted considerable attention due to its advantages of persistence, targeting, and ability to kill tumor cells. However, the efficacy of tumor immunotherapy in practical applications is limited by tumor heterogeneity and complex tumor immunosuppressive microenvironments in which abundant of M2 macrophages and immune checkpoints (ICs) are present. Herein, two type-I aggregation-induced emission (AIE)-active photosensitizers with various reactive oxygen species (ROS)-generating efficiencies are designed and synthesized. Engineered extracellular vesicles (EVs) that express ICs Siglec-10 are first obtained from 4T1 tumor cells. The engineered EVs are then fused with the AIE photosensitizer-loaded lipidic nanosystem to form SEx@Fc-NPs. The ROS generated by the inner type-I AIE photosensitizer of the SEx@Fc-NPs through photodynamic therapy (PDT) can convert M2 macrophages into M1 macrophages to improve tumor immunosuppressive microenvironment. The outer EV-antigens that carry 4T1 tumor-associated antigens directly stimulate dendritic cells maturation to activate different types of tumor-specific T cells in overcoming tumor heterogeneity. In addition, blocking Siglec-10 reversed macrophage exhaustion for enhanced antitumor ability. This study presents that a combination of PDT, immune checkpoints, and EV-antigens can greatly improve the efficiency of tumor immunotherapy and is expected to serve as an emerging strategy to improve tumor immunosuppressive microenvironment and overcome immune escape.
Collapse
Affiliation(s)
- Zhihong Sun
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
- Queen Mary School, Nanchang University, Nanchang, 330031, P. R. China
| | - Zhuokai Sun
- Queen Mary School, Nanchang University, Nanchang, 330031, P. R. China
| | - Jie Liu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Xiaohan Gao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Liping Jiao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Qi Zhao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Yongli Chu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Xiaozhong Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330031, P. R. China
- School of Public Health, Nanchang University, Nanchang, 330031, P. R. China
| | - Guanjun Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Sino-Euro Center of Biomedicine and Health, Shenzhen, 518024, P. R. China
| |
Collapse
|
218
|
Kagamu H. Immunotherapy for non-small cell lung cancer. Respir Investig 2024; 62:307-312. [PMID: 38310751 DOI: 10.1016/j.resinv.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/19/2023] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
Immune checkpoint inhibitors (ICI) bind to programmed cell death-1 (PD-1)/PD-1 ligand-1 (PD-L1) and Cytotoxic T-lymphocyte antigen-4 (CTLA-4), which suppress T-cell function and inhibit their inhibitory function, resulting in T-cell activation. ICI have been approved for a wide range of cancers, including malignant melanoma, renal cell carcinoma, non-small cell lung cancer, head and neck cancer, Hodgkin's disease, small-cell lung cancer, malignant pleural mesothelioma, gastric cancer, esophageal cancer, breast cancer, uterine cancer, and hepatocellular carcinoma, and the number of indications continues to grow. In addition to the treatment of advanced disease, the anti-tumor effect has been demonstrated across disease stages, from locally advanced disease to early-stage operative disease. The treatment of lung cancer is at the forefront of this trend and long-term durable responses and survival benefits in lung cancer have been exhibited that were unimaginable when cytotoxic anticancer agents were the only treatment options. However, treatment efficacy varies greatly from case to case, and no biomarkers have been developed to accurately predict efficacy. In this article, we discuss the past and future of ICI therapy for lung cancer, based on clinical and basic evidence accumulated to-date.
Collapse
Affiliation(s)
- Hiroshi Kagamu
- Department of Respiratory Medicine, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan.
| |
Collapse
|
219
|
Huang H, Gu J, Kuang X, Yu Y, Rao B, Fang S, Lu J, Qiu F. An integrative pan-cancer analysis of WWC family genes and functional validation in lung cancer. Cell Signal 2024; 115:111034. [PMID: 38190957 DOI: 10.1016/j.cellsig.2024.111034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 01/10/2024]
Abstract
The WW and C2 domain containing (WWC) protein family functions as scaffolds regulating cell proliferation and organ growth control through the Hippo signaling pathway. However, their pan-cancer dysregulation and mechanistic roles in signaling transduction have remained unclear. We performed integrated pan-cancer analyses of WWC family gene expression using data from The Cancer Genome Atlas (TCGA) across 33 different cancer types. Prognostic relevance was evaluated by survival analyses. WWC genetic alterations, DNA methylation, pathway activities, drug response, and tumor immunology were analyzed using online databases. Furthermore, we examined the functional roles of WWCs in lung cancer cells. We observed aberrant WWC expression in various cancers, which associated with patient prognosis. WWC hypermethylation occurred in many cancers and exhibited negative correlation with expression, alongside mutations linked to poor outcomes. Pathway analysis implicated WWCs as Hippo pathway scaffolds, while drug sensitivity analysis suggested associations with diverse chemotherapies. Additionally, pan-cancer analyses elucidated vital immunomodulatory roles for WWC through heterogeneous correlations with immune cell infiltrates, checkpoint molecules, tumor mutation burden, microsatellite instability, and chemokine pathways across cancers. Experimentally, WWCs suppressed lung cancer cell proliferation, migration, and invasion while enhancing apoptosis and paclitaxel chemosensitivity. Mechanistically, WWCs bound large tumor suppressor 1 and 2 (LATS1/2) kinases to stimulate phosphorylation cascades, thereby inhibiting nuclear translocation of the Yes-associated protein (YAP) oncoprotein. Taken together, our multi-omics characterization provides comprehensive evidence for WWCs as putative tumor suppressors across cancers via Hippo pathway modulation. WWCs may serve as prognostic markers and therapeutic targets in lung cancer.
Collapse
Affiliation(s)
- Hongmei Huang
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Jiaji Gu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Xinjie Kuang
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Yonghui Yu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Boqi Rao
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Shenying Fang
- The fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, PR China
| | - Jiachun Lu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Fuman Qiu
- State Key Lab of Respiratory Disease, Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China.
| |
Collapse
|
220
|
Lee S, Kang BH, Lee HB, Jang BS, Han W, Kim IA. B-Cell-Mediated Immunity Predicts Survival of Patients With Estrogen Receptor-Positive Breast Cancer. JCO Precis Oncol 2024; 8:e2300263. [PMID: 38452311 DOI: 10.1200/po.23.00263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 03/09/2024] Open
Abstract
PURPOSE The estrogen receptor-positive (ER+) breast cancer (BC), which constitutes the majority of BC cases, exhibits highly heterogeneous clinical behavior. To aid precision treatments, we aimed to find molecular subtypes of ER+ BC representing the tumor microenvironment and prognosis. METHODS We analyzed RNA-seq data of 113 patients with BC and classified them according to the PAM50 intrinsic subtypes using gene expression profiles. Among them, we further focused on 44 patients with luminal-type (ER+) BC for subclassification. The Cancer Genome Atlas (TCGA) data of patients with BC were used as a validation data set to verify the new classification. We estimated the immune cell composition using CIBERSORT and further analyzed its association with clinical or molecular parameters. RESULTS Principal component analysis clearly divided the patients into two subgroups separately from the luminal A and B classification. The top differentially expressed genes between the subgroups were distinctly characterized by immunoglobulin and B-cell-related genes. We could also cluster a separate cohort of patients with luminal-type BC from TCGA into two subgroups on the basis of the expression of a B-cell-specific gene set, and patients who were predicted to have high B-cell immune activity had better prognoses than other patients. CONCLUSION Our transcriptomic approach emphasize a molecular phenotype of B-cell immunity in ER+ BC that may help to predict disease prognosis. Although further researches are required, B-cell immunity for patients with ER+ BC may be helpful for identifying patients who are good responders to chemotherapy or immunotherapy.
Collapse
Affiliation(s)
- Seungbok Lee
- Department of Genomic Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byung-Hee Kang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Radiation Oncology, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Han-Byoel Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Bum-Sup Jang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - In Ah Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Radiation Oncology and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
221
|
Dou T, Li J, Zhang Y, Pei W, Zhang B, Wang B, Wang Y, Jia H. The cellular composition of the tumor microenvironment is an important marker for predicting therapeutic efficacy in breast cancer. Front Immunol 2024; 15:1368687. [PMID: 38487526 PMCID: PMC10937353 DOI: 10.3389/fimmu.2024.1368687] [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: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
At present, the incidence rate of breast cancer ranks first among new-onset malignant tumors in women. The tumor microenvironment is a hot topic in tumor research. There are abundant cells in the tumor microenvironment that play a protumor or antitumor role in breast cancer. During the treatment of breast cancer, different cells have different influences on the therapeutic response. And after treatment, the cellular composition in the tumor microenvironment will change too. In this review, we summarize the interactions between different cell compositions (such as immune cells, fibroblasts, endothelial cells, and adipocytes) in the tumor microenvironment and the treatment mechanism of breast cancer. We believe that detecting the cellular composition of the tumor microenvironment is able to predict the therapeutic efficacy of treatments for breast cancer and benefit to combination administration of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Binyue Zhang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| |
Collapse
|
222
|
Lin M, Chen D, Shao Z, Liu Q, Hao Z, Xin Z, Chen Y, Wu W, Chen X, He T, Wu D, Wu P. Inflammatory dendritic cells restrain CD11b +CD4 + CTLs via CD200R in human NSCLC. Cell Rep 2024; 43:113767. [PMID: 38354085 DOI: 10.1016/j.celrep.2024.113767] [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: 07/26/2023] [Revised: 12/12/2023] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
CD4+ cytotoxic T lymphocytes (CD4+ CTLs) are suggested to play a crucial role in inflammatory diseases, including cancer, but their characteristics in human non-small cell lung cancer (NSCLC) remain unknown. Here, using the cell surface marker CD11b, we identify CD11b+CD4+ CTLs as a cytotoxic subset of CD4+ T cells in multiple tissues of NSCLC patients. In addition, tumor-infiltrating CD11b+CD4+ CTLs show a dysfunctional phenotype with elevated expression of CD200 receptor (CD200R), a negatively immunomodulatory receptor. CD4+ regulatory T (Treg) cells restrain the anti-tumor role of CD11b+CD4+ CTLs via CD200. Mechanistically, inflammatory dendritic cells promote the CD200R expression of CD11b+CD4+ CTLs by secreting interleukin-1β (IL-1β). Finally, we demonstrate that CD200 blockade can revive the tumor-killing role of CD11b+CD4+ CTLs and prolong the survival of tumor-bearing mice. Taken together, our study identifies CD11b+CD4+ CTLs in NSCLC with decreased cytotoxicity that can be reinvigorated by CD200 blockade, suggesting that targeting CD200 is a promising immunotherapy strategy in NSCLC.
Collapse
Affiliation(s)
- Mingjie Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Di Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zheyu Shao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Qinyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zhixing Hao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zhongwei Xin
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yongyuan Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Wenxuan Wu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiaoke Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Teng He
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Infectious Disease, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Dang Wu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| | - Pin Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
223
|
Chen J, Yu F, He G, Hao W, Hu W. A nomogram based on peripheral lymphocyte for predicting 8-year survival in patients with prostate cancer: a single-center study using LASSO-cox regression. BMC Cancer 2024; 24:254. [PMID: 38395827 PMCID: PMC10885398 DOI: 10.1186/s12885-024-11929-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
PURPOSE The purpose of this study was to develop a functional clinical nomogram for predicting 8-year overall survival (OS) of patients with prostate cancer (PCa) primary based on peripheral lymphocyte. PATIENTS AND METHODS Using data from a single-institutional registry of 94 patients with PCa in China, this study identified and integrated significant prognostic factors for survival to build a nomogram. The discriminative ability was measured by concordance index (C-index) and ROC curves (Receiver Operating Characteristic Curves). And the predictive accuracy was measured by the calibration curves. Decision curve analyses (DCA) was used to measure the clinical usefulness. RESULTS A total of 94 patients were included for analysis. Five independent prognostic factors were identified by LASSO-Cox regression and incorporated into the nomogram: age, the T stage, the absolute counts of peripheral CD3(+)CD4(+) T lymphocytes, CD3(-)CD16(+)CD56(+) NK cells and CD4(+)/CD8(+) ratio. The area under the curve (AUC) values of the predictive model for 5-, 8-, and 10-year overall survival were 0.81, 0.76, and 0.73, respectively. The calibration curves for probability of 5-,8- and 10-year OS showed optimal agreement between nomogram prediction and actual observation. The stratification into different risk groups allowed significant distinction. DCA indicated the good clinical application value of the model. CONCLUSION We developed a novel nomogram that enables personalized prediction of OS for patients diagnosed with PCa. This finding revealed a relative in age and survival rate in PCa, and a more favorable prognosis in patients exhibiting higher levels of CD4 + T, CD4+/CD8 + ratio and CD3(-)CD16(+)CD56(+) NK cells specifically. This clinically applicable prognostic model exhibits promising predictive capabilities, offering valuable support to clinicians in informed decision-making process.
Collapse
Affiliation(s)
- Jiayi Chen
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
| | - Feng Yu
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
| | - Ganyuan He
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China
| | - Wenke Hao
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China.
| | - Wenxue Hu
- Department of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangdong Provincial Geriatrics Institute, Southern Medical University, Guangzhou, China.
| |
Collapse
|
224
|
Krishna C, Tervi A, Saffern M, Wilson EA, Yoo SK, Mars N, Roudko V, Cho BA, Jones SE, Vaninov N, Selvan ME, Gümüş ZH, Lenz TL, Merad M, Boffetta P, Martínez-Jiménez F, Ollila HM, Samstein RM, Chowell D. An immunogenetic basis for lung cancer risk. Science 2024; 383:eadi3808. [PMID: 38386728 PMCID: PMC11998992 DOI: 10.1126/science.adi3808] [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: 04/21/2023] [Accepted: 01/16/2024] [Indexed: 02/24/2024]
Abstract
Cancer risk is influenced by inherited mutations, DNA replication errors, and environmental factors. However, the influence of genetic variation in immunosurveillance on cancer risk is not well understood. Leveraging population-level data from the UK Biobank and FinnGen, we show that heterozygosity at the human leukocyte antigen (HLA)-II loci is associated with reduced lung cancer risk in smokers. Fine-mapping implicated amino acid heterozygosity in the HLA-II peptide binding groove in reduced lung cancer risk, and single-cell analyses showed that smoking drives enrichment of proinflammatory lung macrophages and HLA-II+ epithelial cells. In lung cancer, widespread loss of HLA-II heterozygosity (LOH) favored loss of alleles with larger neopeptide repertoires. Thus, our findings nominate genetic variation in immunosurveillance as a critical risk factor for lung cancer.
Collapse
Affiliation(s)
- Chirag Krishna
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
| | - Anniina Tervi
- Institute for Molecular Medicine (FIMM), HiLIFE, University of Helsinki; Helsinki, Finland
| | - Miriam Saffern
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Eric A. Wilson
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Seong-Keun Yoo
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Nina Mars
- Institute for Molecular Medicine (FIMM), HiLIFE, University of Helsinki; Helsinki, Finland
| | - Vladimir Roudko
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Byuri Angela Cho
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Samuel Edward Jones
- Institute for Molecular Medicine (FIMM), HiLIFE, University of Helsinki; Helsinki, Finland
| | - Natalie Vaninov
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | | | - Tobias L. Lenz
- Research Unit for Evolutionary Immunogenomics, Department of Biology, Universität Hamburg; 20146 Hamburg, Germany
| | - Miriam Merad
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna; 40138 Bologna, Italy
- Stony Brook Cancer Center, Stony Brook University; New York, NY 11794, USA
| | - Francisco Martínez-Jiménez
- Vall d’Hebron Institute of Oncology, Barcelona, Spain
- Hartwig Medical Foundation, Amsterdam, the Netherlands
| | - Hanna M. Ollila
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
- Institute for Molecular Medicine (FIMM), HiLIFE, University of Helsinki; Helsinki, Finland
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School; Boston, MA 02114, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Robert M. Samstein
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| | - Diego Chowell
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY 10029, USA
| |
Collapse
|
225
|
Guldvik IJ, Ramberg H, Kristensen G, Røder A, Mills IG, Lilleby W, Taskén KA. Systemic interrogation of immune-oncology-related proteins in patients with locally advanced prostate cancer undergoing androgen deprivation and intensity-modulated radiotherapy. World J Urol 2024; 42:95. [PMID: 38386171 PMCID: PMC10884049 DOI: 10.1007/s00345-024-04787-8] [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: 07/31/2023] [Accepted: 01/10/2024] [Indexed: 02/23/2024] Open
Abstract
PURPOSE The primary objective was to establish whether blood-based leucine-rich alpha-2-glycoprotein (LRG1) can predict outcomes in patients with locally advanced prostate cancer undergoing androgen-deprivation therapy (ADT) and radiotherapy (RT) and to determine how it may relate to 92 immune-oncology (I-O)-related proteins in this setting. METHODS Baseline blood level of LRG1 from patients treated with ADT and RT enrolled in the CuPCa (n = 128) and IMRT (n = 81) studies was measured using ELISA. A longitudinal cohort with matched blood samples from start of ADT, start of RT, and end of RT protocol from 47 patients from the IMRT cohort was used to establish levels of I-O proteins by high-multiplexing Proximal Extension Assay by Olink Proteomics. Statistical analyses using Kaplan-Meier, Cox regression, and LIMMA analyses were applied to predict the prognostic value of LRG1 and its correlation to I-O proteins. RESULTS High baseline levels of LRG1 predicted a low frequency of treatment failure in patients undergoing ADT + RT in both the CuPCa and the IMRT cohorts. LRG1 was moderately correlated with CD4, IL6, and CSF1. We identified I-O proteins predicting metastatic failure (MF) at different timepoints. CONCLUSION LRG1 biomarker is associated with I-O proteins and can be used to improve stratification and monitoring of prostate cancer patients undergoing ADT + RT. This work will require further in-depth analyses in independent cohorts with treatment outcome data.
Collapse
Affiliation(s)
- Ingrid Jenny Guldvik
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Håkon Ramberg
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Gitte Kristensen
- Department of Urology, Center for Cancer and Organ Diseases, Copenhagen Prostate Cancer Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Andreas Røder
- Department of Urology, Center for Cancer and Organ Diseases, Copenhagen Prostate Cancer Center, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ian G Mills
- Cancer Research UK, Li Ka Shing Centre, Cambridge Research Institute, Cambridge, UK
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Patrick G. Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast, UK
| | | | - Kristin Austlid Taskén
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
226
|
Rakké YS, Buschow SI, IJzermans JNM, Sprengers D. Engaging stimulatory immune checkpoint interactions in the tumour immune microenvironment of primary liver cancers - how to push the gas after having released the brake. Front Immunol 2024; 15:1357333. [PMID: 38440738 PMCID: PMC10910082 DOI: 10.3389/fimmu.2024.1357333] [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: 12/17/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) are the first and second most common primary liver cancer (PLC). For decades, systemic therapies consisting of tyrosine kinase inhibitors (TKIs) or chemotherapy have formed the cornerstone of treating advanced-stage HCC and CCA, respectively. More recently, immunotherapy using immune checkpoint inhibition (ICI) has shown anti-tumour reactivity in some patients. The combination regimen of anti-PD-L1 and anti-VEGF antibodies has been approved as new first-line treatment of advanced-stage HCC. Furthermore, gemcibatine plus cisplatin (GEMCIS) with an anti-PD-L1 antibody is awaiting global approval for the treatment of advanced-stage CCA. As effective anti-tumour reactivity using ICI is achieved in a minor subset of both HCC and CCA patients only, alternative immune strategies to sensitise the tumour microenvironment of PLC are waited for. Here we discuss immune checkpoint stimulation (ICS) as additional tool to enhance anti-tumour reactivity. Up-to-date information on the clinical application of ICS in onco-immunology is provided. This review provides a rationale of the application of next-generation ICS either alone or in combination regimen to potentially enhance anti-tumour reactivity in PLC patients.
Collapse
Affiliation(s)
- Yannick S. Rakké
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Sonja I. Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| | - Jan N. M. IJzermans
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
227
|
Jones J, Shi Q, Nath RR, Brito IL. Keystone pathobionts associated with colorectal cancer promote oncogenic reprograming. PLoS One 2024; 19:e0297897. [PMID: 38363784 PMCID: PMC10871517 DOI: 10.1371/journal.pone.0297897] [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: 06/07/2023] [Accepted: 01/12/2024] [Indexed: 02/18/2024] Open
Abstract
Fusobacterium nucleatum (Fn) and enterotoxigenic Bacteroides fragilis (ETBF) are two pathobionts consistently enriched in the gut microbiomes of patients with colorectal cancer (CRC) compared to healthy counterparts and frequently observed for their direct association within tumors. Although several molecular mechanisms have been identified that directly link these organisms to features of CRC in specific cell types, their specific effects on the epithelium and local immune compartment are not well-understood. To fill this gap, we leveraged single-cell RNA sequencing (scRNA-seq) on wildtype mice and mouse model of CRC. We find that Fn and ETBF exacerbate cancer-like transcriptional phenotypes in transit-amplifying and mature enterocytes in a mouse model of CRC. We also observed increased T cells in the pathobiont-exposed mice, but these pathobiont-specific differences observed in wildtype mice were abrogated in the mouse model of CRC. Although there are similarities in the responses provoked by each organism, we find pathobiont-specific effects in Myc-signaling and fatty acid metabolism. These findings support a role for Fn and ETBF in potentiating tumorigenesis via the induction of a cancer stem cell-like transit-amplifying and enterocyte population and the disruption of CTL cytotoxic function.
Collapse
Affiliation(s)
- Josh Jones
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Qiaojuan Shi
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Rahul R. Nath
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Ilana L. Brito
- Meinig School for Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
228
|
Fang Z, Xue Y, Leng Y, Zhang L, Ren X, Yang N, Chen J, Chen L, Wang H. Erzhi pills reverse PD-L1-mediated immunosuppression in melanoma microenvironment. Heliyon 2024; 10:e24988. [PMID: 38317912 PMCID: PMC10839997 DOI: 10.1016/j.heliyon.2024.e24988] [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: 11/23/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/07/2024] Open
Abstract
Background Cancer immunotherapies aimed at activating immune system, especially by blocking immune checkpoints, have become a successful modality for treating patients with advanced cancers. However, its clinical practice is frequently conceded by high outcomes, low initial response rates and severe side effects. New strategies are necessary to complement and advance this biological therapy. Erzhi Pills (EZP) have diverse pharmaceutical effects including immune regulation, anti-tumor and anti-senescence. We hypothesized that EZP could exert its antitumor effect through immunomodulation. Purpose The aim of this study was to investigate the effects of EZP on anti-tumor activities, and define its molecular mechanisms. Methods By applying melanoma model with high immune infiltrates, we determined the anti-melanoma effect of EZP. To identify whether this effect was mediated by direct targeting tumor cells, cell viability and apoptosis were examined in vitro. Network pharmacology analysis was used to predict the potential mechanisms of EZP for melanoma via immune response. Flow cytometry, immunohistochemistry (IHC), enzyme-linked immunosorbent assay (ELISA) and crystal violet (CV) experiments were performed to detect T cell infiltrations and functions mediated by EZP. The mechanism of EZP was further investigated by western blotting both in vivo and in vitro. Results The administration of EZP significantly inhibited tumor weight and volume. EZP extract could only slightly reduce cell viability and induce melanoma apoptosis. Network pharmacology analysis predicted that JAK-STAT signaling pathway and T cell receptor signaling pathway might be involved during EZP treatment. Flow cytometry and IHC analyses showed that EZP increased the number of CD4+ T cells and enhanced the function of CD8+ T cells. In co-culture experiments, EZP elevated killing ability of T cells. Western blotting showed that EZP treatment reduced PD-L1 signaling pathway. Conclusion These findings indicated that EZP exerted anti-melanoma effects by inducing apoptosis and blocking PD-L1 to activate T cells. EZP might represent a promising candidate drug for cancer immunotherapies.
Collapse
Affiliation(s)
- Zhirui Fang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| | - Yuejin Xue
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| | - Yuze Leng
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| | - Lusha Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| | - Xiuyun Ren
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| | - Ning Yang
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Department of Dermatology, 300250, Tianjin, China
| | - Jing Chen
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Department of Dermatology, 300250, Tianjin, China
| | - Lu Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
- Instrumental Analysis and Research Center, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| | - Hong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 301617, Tianjin, China
| |
Collapse
|
229
|
Ding W, Chen WW, Wang YQ, Xu XZ, Wang YB, Yan YM, Tan YL. Immune-related long noncoding RNA zinc finger protein 710-AS1-201 promotes the metastasis and invasion of gastric cancer cells. World J Gastrointest Oncol 2024; 16:458-474. [PMID: 38425400 PMCID: PMC10900153 DOI: 10.4251/wjgo.v16.i2.458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/02/2023] [Accepted: 12/20/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a prevalent malignant tumor of the gastrointestinal system. ZNF710 is a transcription factor (TF), and zinc finger protein 710 (ZNF710)-AS1-201 is an immune-related long noncoding RNA (lncRNA) that is upregulated in GC cells. AIM To assess the correlation between ZNF710-AS1-201 and immune microenvironment features and to investigate the roles of ZNF710-AS1-201 in the invasion and metastasis processes of GC cells. METHODS We obtained data from The Cancer Genome Atlas and Wujin Hospital. We assessed cell growth, migration, invasion, and programmed cell death using cell counting kit-8, EdU, scratch, Transwell, and flow cytometry assays. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to identify the potential downstream targets of ZNF710-AS1-201. RESULTS In GC tissues with low ZNF710-AS1-201 expression, immunoassays detected significant infiltration of various antitumor immune cells, such as memory CD8 T cells and activated CD4 T cells. In the low-expression group, the half-maximal inhibitory concentrations (IC50s) of 5-fluorouracil, cisplatin, gemcitabine, and trametinib were lower, whereas the IC50s of dasatinib and vorinostat were higher. The malignant degree of GC was higher and the stage was later in the high-expression group. Additionally, patients with high expression of ZNF710-AS1-201 had lower overall survival and disease-free survival rates. In vitro, the overexpression of ZNF710-AS1-201 greatly enhanced growth, metastasis, and infiltration while suppressing cell death in HGC-27 cells. In contrast, the reduced expression of ZNF710-AS1-201 greatly hindered cell growth, enhanced apoptosis, and suppressed the metastasis and invasion of MKN-45 cells. The expression changes in ZNF710 were significant, but the corresponding changes in isocitrate dehydrogenase-2, Semaphorin 4B, ARHGAP10, RGMB, hsa-miR-93-5p, and ZNF710-AS1-202 were not consistent or statistically significant after overexpression or knockdown of ZNF710-AS1-201, as determined by qRT-PCR. CONCLUSION Immune-related lncRNA ZNF710-AS1-201 facilitates the metastasis and invasion of GC cells. It appears that ZNF710-AS1-201 and ZNF710 have potential as effective targets for therapeutic intervention in GC. Nevertheless, it is still necessary to determine the specific targets of the ZNF710 TF.
Collapse
Affiliation(s)
- Wei Ding
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, Jiangsu Province, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213017, Jiangsu Province, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213003, Jiangsu Province, China
| | - Wei-Wei Chen
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, Jiangsu Province, China
| | - Yi-Qin Wang
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, Jiangsu Province, China
| | - Xue-Zhong Xu
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, Jiangsu Province, China
| | - Yi-Bo Wang
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, Jiangsu Province, China
| | - Yong-Min Yan
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213017, Jiangsu Province, China
| | - Yu-Lin Tan
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, Jiangsu Province, China
- Department of General Surgery, The Wujin Clinical College of Xuzhou Medical University, Changzhou 213003, Jiangsu Province, China
| |
Collapse
|
230
|
Yang L, Chen Y, Liu K, Chen Y, Zhang Y, Zhang Z, Li H. Investigating the immune mechanism of natural products in the treatment of lung cancer. Front Pharmacol 2024; 15:1289957. [PMID: 38420194 PMCID: PMC10899684 DOI: 10.3389/fphar.2024.1289957] [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: 09/06/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
With the deepening of people's understanding of lung cancer, the research of lung cancer immunotherapy has gradually become the focus of attention. As we all know, the treatment of many diseases relies on the rich sources, complex and varied compositions and wide range of unique biological properties of natural products. Studies have shown that natural products can exert anticancer effects by inducing tumor cell death, inhibiting tumor cell proliferation, and enhancing tumor cell autophagy. More notably, natural products can adjust and strengthen the body's immune response, which includes enhancing the function of NK cells and promoting the differentiation and proliferation of T lymphocytes. In addition, these natural products may enhance their anticancer effects by affecting inhibitory factors in the immune system, hormone levels, enzymes involved in biotransformation, and modulating other factors in the tumor microenvironment. The importance of natural products in lung cancer immunotherapy should not be underestimated. However, the specific links and correlations between natural products and lung cancer immunity are not clear enough, and further studies are urgently needed to clarify the relationship between the two. In this paper, we will focus on the correlation between natural products and lung cancer immune responses, with a view to providing new research perspectives for immunotherapy of lung cancer.
Collapse
Affiliation(s)
- Lian Yang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yukun Chen
- Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kaile Liu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hegen Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
231
|
Yi B, Wei X, Liu D, Jing L, Xu S, Zhang M, Liang Z, Liu R, Zhang Z. Comprehensive analysis of disulfidptosis-related genes: a prognosis model construction and tumor microenvironment characterization in clear cell renal cell carcinoma. Aging (Albany NY) 2024; 16:3647-3673. [PMID: 38358909 PMCID: PMC10929811 DOI: 10.18632/aging.205550] [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: 07/25/2023] [Accepted: 12/01/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Disulfidptosis, a form of cell death induced by abnormal intracellular accumulation of disulfides, is a newly recognized variety of cell death. Clear cell renal cell carcinoma (ccRCC) is a usual urological tumor that poses serious health risks. There are few studies of disulfidptosis-related genes (DRGs) in ccRCC so far. METHODS The expression, transcriptional variants, and prognostic role of DRGs were assessed. Based on DRGs, consensus unsupervised clustering analysis was performed to stratify ccRCC patients into various subtypes and constructed a DRG risk scoring model. Patients were stratified into high or low-risk groups by this model. We focused on assessing the discrepancy in prognosis, TME, chemotherapeutic susceptibility, and landscape of immune between the two risk groups. Finally, we validated the expression and explored the biological function of the risk scoring gene FLRT3 through in vitro experiments. RESULTS The different subtypes had significantly different gene expression, immune, and prognostic landscapes. In the two risk groups, the high-risk group had higher TME scores, more significant immune cell infiltration, and a higher probability of benefiting from immunotherapy, but had a worse prognosis. There were also remarkable differences in chemotherapeutic susceptibility between the two risk groups. In ccRCC cells, the expression of FLRT3 was shown to be lower and its overexpression caused a decrease in cell proliferation and metastatic capacity. CONCLUSIONS Starting from disulfidptosis, we established a new risk scoring model which can provide new ideas for doctors to forecast patient survival and determine clinical treatment plans.
Collapse
Affiliation(s)
- Bocun Yi
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xifeng Wei
- Department of Urology, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Dongze Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Liwei Jing
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shengxian Xu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Man Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Institute of Endocrinology, Chu Hsien-I Memorial Hospital of Tianjin Medical University, Tianjin, China
| | - Zhengxin Liang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ranlu Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhihong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
232
|
Babalola KT, Arora M, Ganugula R, Agarwal SK, Mohan C, Kumar MNVR. Leveraging Lymphatic System Targeting in Systemic Lupus Erythematosus for Improved Clinical Outcomes. Pharmacol Rev 2024; 76:228-250. [PMID: 38351070 PMCID: PMC10877736 DOI: 10.1124/pharmrev.123.000938] [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: 07/20/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 02/16/2024] Open
Abstract
The role of advanced drug delivery strategies in drug repositioning and minimizing drug attrition rates, when applied early in drug discovery, is poised to increase the translational impact of various therapeutic strategies in disease prevention and treatment. In this context, drug delivery to the lymphatic system is gaining prominence not only to improve the systemic bioavailability of various pharmaceutical drugs but also to target certain specific diseases associated with the lymphatic system. Although the role of the lymphatic system in lupus is known, very little is done to target drugs to yield improved clinical benefits. In this review, we discuss recent advances in drug delivery strategies to treat lupus, the various routes of drug administration leading to improved lymph node bioavailability, and the available technologies applied in other areas that can be adapted to lupus treatment. Moreover, this review also presents some recent findings that demonstrate the promise of lymphatic targeting in a preclinical setting, offering renewed hope for certain pharmaceutical drugs that are limited by efficacy in their conventional dosage forms. These findings underscore the potential and feasibility of such lymphatic drug-targeting approaches to enhance therapeutic efficacy in lupus and minimize off-target effects of the pharmaceutical drugs. SIGNIFICANCE STATEMENT: The World Health Organization estimates that there are currently 5 million humans living with some form of lupus. With limited success in lupus drug discovery, turning to effective delivery strategies with existing drug molecules, as well as those in the early stage of discovery, could lead to better clinical outcomes. After all, effective delivery strategies have been proven to improve treatment outcomes.
Collapse
Affiliation(s)
- K T Babalola
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - M Arora
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - R Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - S K Agarwal
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - C Mohan
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| | - M N V Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM) (K.T.B., M.A., R.G., M.N.V.R.K.), Division of Translational Science and Medicine, College of Community Health Sciences (K.T.B., M.A., R.G., M.N.V.R.K.), Alabama Life Research Institute (K.T.B., M.A., R.G., M.N.V.R.K.), and Department of Biological Sciences (M.A., R.G., M.N.V.R.K.), The University of Alabama, Tuscaloosa, Alabama; Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, Texas (S.K.A.); Department of Biomedical Engineering, University of Houston, Houston, Texas (C.M.); Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama (M.N.V.R.K.); and Center for Free Radical Biology (M.N.V.R.K.) and Nephrology Research and Training Center, Division of Nephrology, Department of Medicine (M.N.V.R.K.), University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
233
|
Kong X, Li Q, Wang D, Wang M, Yang F, Meng J. Mechanism of Qizhen decoction-mediated maturation of DC cells to activate the IL-12/JAK2/STAT4 pathway to sensitise PD-1 inhibitors in the treatment of colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117399. [PMID: 37956913 DOI: 10.1016/j.jep.2023.117399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine has been utilized to treat colorectal cancer (CRC). Qizhen decoction (QZD), a potential compound prescription of traditional Chinese medicine, possesses multiple biological activities. It has been used to treat CRC in clinical practice and has been proven to be effective. AIM OF THE STUDY To investigate the impact of QZD supported by intestinal flora in combination with PD-1 inhibitor on colorectal cancer, and to elucidate the mechanism by which QZD enhances the sensitivity of PD-1 inhibitor against colorectal cancer. MATERIALS AND METHODS Observation of Intestinal Flora Mediating the Effect of QZD Combined with PD-1 Inhibitor in the Treatment of Colorectal Cancer. We used Flow cytometry and qPCR to detect the effect of QZD combined with PD-1 inhibitor on the activation of effector T cells in a wild mouse model of colorectal cancer. In wild and germ-free mouse models, the differences in inflammatory factors, pathological change, body mass, colorectal length, and tumour load were observed. In the study of the mechanism of QZD combined with PD-1 inhibitor in the treatment of colorectal cancer, the study evaluated the abundance of Akkermansia, the phenotypes of effector T cells and DC cells, as well as inflammatory factors in each group of mice to determine whether Akkermansia played a role in activating DC cells. Based on the JAK2/TYK2/STAT4 pathway, the mechanism of PD-1 inhibitor sensitisation by QZD in colorectal cancer was further investigated. RESULTS We found that QZD combined with PD-1 inhibitor could improve the therapeutic effect on colorectal cancer by inducing more critical immune functions. QZD promotes increased Akkermansia abundance in the gut. Akkermansia promotes maturation of DC cells, and mature DC cells activate the IL-12/JAK2/STAT4 pathway, which significantly activates effector T cells. Akkermansia is key to QZD combined with PD-1 inhibitor-mediated immunity exerting a therapeutic effect on colorectal cancer. CONCLUSION The mechanism of action of the QZD sensitizing PD-1 inhibitor is to promote the maturation of DC cells to release IL-12 and activate the JAK2/STAT4 pathway to induce effector T cell activation by increasing the abundance of Akkermansia.
Collapse
Affiliation(s)
- Xianbin Kong
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Qingbo Li
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Dong Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Miao Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Fan Yang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Jingyan Meng
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
234
|
Tenggara JB, Rachman A, Prihartono J, Rachmadi L, Panigoro SS, Heriyanto DS, Sutandyo N, Nasution IR, Rahadiati FB, Steven R, Betsy R, Juanputra S, Sudoyo AW. The relationship between high ratios of CD4/FOXP3 and CD8/CD163 and the improved survivability of metastatic triple-negative breast cancer patients: a multicenter cohort study. BMC Res Notes 2024; 17:44. [PMID: 38308298 PMCID: PMC10835864 DOI: 10.1186/s13104-024-06704-z] [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: 07/19/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) has been documented as the most aggressive subtype of breast cancer. This study aimed to analyze antitumor and protumor immune activities, and their ratios as significant prognostic biomarkers in metastatic TNBC (mTNBC). METHODS A multicenter cohort study was conducted among 103 de novo mTNBC patients. The expression of CD8 and CD163 was evaluated using immunohistochemistry staining, CD4 and FOXP3 using double-staining immunohistochemistry, and PD-L1 using immunohistochemistry and RT-PCR. RESULTS Multivariate analysis revealed that high CD4/FOXP3 (HR 1.857; 95% CI 1.049-3.288; p = 0.034) and the CD8/CD163 ratio (HR 2.089; 95% CI 1.174-3.717; p = 0.012) yield significantly improved 1 year overall survival (OS). Kaplan-Meier analysis showed that high levels of CD4 (p = 0.023), CD8 (p = 0.043), CD4/FOXP3 (p = 0.016), CD8/FOXP3 (p = 0.005), CD8/CD163 (p = 0.005) ratios were significantly associated with higher rate of 1 year OS. Furthermore, 1 year OS was directly correlated with antitumor CD4 (R = 0.233; p = 0.018) and CD8 (R = 0.219; p = 0.026) and was indirectly correlated with protumor CD163 and FOXP3 through CD4/FOXP3 (R = 0.282; p = 0.006), CD4/CD163 (R = 0.239; p = 0.015), CD8/FOXP3 (R = 0.260; p = 0.008), and CD8/CD163 (R = 0.258; p = 0.009). CONCLUSION This is the first study to demonstrate that high levels of CD4/FOXP3 and CD8/CD163 significantly improved the 1 year OS in de novo mTNBC patients. Thus, we recommend the application of these markers as prognosis determination and individual treatment decision.
Collapse
Affiliation(s)
- Jeffry Beta Tenggara
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia.
| | - Andhika Rachman
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia
| | - Joedo Prihartono
- Department of Community Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Lisnawati Rachmadi
- Department of Anatomical Pathology, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Sonar Soni Panigoro
- Department of Surgical Oncology, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Dr. Sardjito Hospital-Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Noorwati Sutandyo
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dharmais National Cancer Hospital, Jakarta, Indonesia
| | - Intan Russianna Nasution
- Division of Hematology and Medical Oncology, Gatot Soebroto Army Hospital Jakarta, Jakarta, Indonesia
| | - Familia Bella Rahadiati
- Department of Anatomical Pathology, Gatot Soebroto Army Hospital Jakarta, Jakarta, Indonesia
| | - Ricci Steven
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia
| | - Rachelle Betsy
- Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Samuel Juanputra
- Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Aru Wisaksono Sudoyo
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Cipto Mangunkusumo General Hospital-Faculty of Medicine Universitas Indonesia, Jl. Pangeran Diponegoro No. 71, RW.5, Kec. Senen, Central Jakarta, Jakarta, 10430, Indonesia
- Division of Hematology and Medical Oncology, Department of Internal Medicine, MRCCC Siloam Hospital Jakarta, Jakarta, Indonesia
| |
Collapse
|
235
|
Jung I, Shin S, Baek MC, Yea K. Modification of immune cell-derived exosomes for enhanced cancer immunotherapy: current advances and therapeutic applications. Exp Mol Med 2024; 56:19-31. [PMID: 38172594 PMCID: PMC10834411 DOI: 10.1038/s12276-023-01132-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/05/2023] [Indexed: 01/05/2024] Open
Abstract
Cancer immunotherapy has revolutionized the approach to cancer treatment of malignant tumors by harnessing the body's immune system to selectively target cancer cells. Despite remarkable advances, there are still challenges in achieving successful clinical responses. Recent evidence suggests that immune cell-derived exosomes modulate the immune system to generate effective antitumor immune responses, making them a cutting-edge therapeutic strategy. However, natural exosomes are limited in clinical application due to their low drug delivery efficiency and insufficient antitumor capacity. Technological advancements have allowed exosome modifications to magnify their intrinsic functions, load different therapeutic cargoes, and preferentially target tumor sites. These engineered exosomes exert potent antitumor effects and have great potential for cancer immunotherapy. In this review, we describe ingenious modification strategies to attain the desired performance. Moreover, we systematically summarize the tumor-controlling properties of engineered immune cell-derived exosomes in innate and adaptive immunity. Collectively, this review provides a comprehensive and intuitive guide for harnessing the potential of modified immune cell-derived exosome-based approaches, offering valuable strategies to enhance and optimize cancer immunotherapy.
Collapse
Affiliation(s)
- Inseong Jung
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Sanghee Shin
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| | - Kyungmoo Yea
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea.
- New Biology Research Center, DGIST, Daegu, 43024, Republic of Korea.
| |
Collapse
|
236
|
Xu H, Chen C, Chen L, Pan S. Pan-cancer analysis identifies the IRF family as a biomarker for survival prognosis and immunotherapy. J Cell Mol Med 2024; 28:e18084. [PMID: 38130025 PMCID: PMC10844690 DOI: 10.1111/jcmm.18084] [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: 02/14/2023] [Revised: 11/08/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
IRF family genes have been shown to be crucial in tumorigenesis and tumour immunity. However, information about the role of IRF in the systematic assessment of pan-cancer and in predicting the efficacy of tumour therapy is still unknown. In this work, we performed a systematic analysis of IRF family genes in 33 tumour samples, including expression profiles, genomics and clinical characteristics. We then applied Single-Sample Gene-Set Enrichment Analysis (ssGSEA) to calculate IRF-scores and analysed the impact of IRF-scores on tumour progression, immune infiltration and treatment efficacy. Our results showed that genomic alterations, including SNPs, CNVs and DNA methylation, can lead to dysregulation of IRFs expression in tumours and participate in regulating multiple tumorigenesis. IRF-score expression differed significantly between 12 normal and tumour samples and the impact on tumour prognosis and immune infiltration depended on tumour type. IRF expression was correlated to drug sensitivity and to the expression of immune checkpoints and immune cell infiltration, suggesting that dysregulation of IRF family expression may be a critical factor affecting tumour drug response. Our study comprehensively characterizes the genomic and clinical profile of IRFs in pan-cancer and highlights their reliability and potential value as predictive markers of oncology drug efficacy. This may provide new ideas for future personalized oncology treatment.
Collapse
Affiliation(s)
- Hua‐Guo Xu
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Can Chen
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Lin‐Yuan Chen
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Shiyang Pan
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| |
Collapse
|
237
|
Xiong Z, Raphael I, Olin M, Okada H, Li X, Kohanbash G. Glioblastoma vaccines: past, present, and opportunities. EBioMedicine 2024; 100:104963. [PMID: 38183840 PMCID: PMC10808938 DOI: 10.1016/j.ebiom.2023.104963] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/08/2024] Open
Abstract
Glioblastoma (GBM) is one of the most lethal central nervous systems (CNS) tumours in adults. As supplements to standard of care (SOC), various immunotherapies improve the therapeutic effect in other cancers. Among them, tumour vaccines can serve as complementary monotherapy or boost the clinical efficacy with other immunotherapies, such as immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapy. Previous studies in GBM therapeutic vaccines have suggested that few neoantigens could be targeted in GBM due to low mutation burden, and single-peptide therapeutic vaccination had limited efficacy in tumour control as monotherapy. Combining diverse antigens, including neoantigens, tumour-associated antigens (TAAs), and pathogen-derived antigens, and optimizing vaccine design or vaccination strategy may help with clinical efficacy improvement. In this review, we discussed current GBM therapeutic vaccine platforms, evaluated and potential antigenic targets, current challenges, and perspective opportunities for efficacy improvement.
Collapse
Affiliation(s)
- Zujian Xiong
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA; Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Michael Olin
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008 PR China.
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
238
|
Bai Y, Li T, Wang Q, You W, Yang H, Xu X, Li Z, Zhang Y, Yan C, Yang L, Qiu J, Liu Y, Chen S, Wang D, Huang B, Liu K, Song BL, Wang Z, Li K, Liu X, Wang G, Yang W, Chen J, Hao P, Zhang Z, Wang Z, Zhu ZJ, Xu C. Shaping immune landscape of colorectal cancer by cholesterol metabolites. EMBO Mol Med 2024; 16:334-360. [PMID: 38177537 PMCID: PMC10897227 DOI: 10.1038/s44321-023-00015-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024] Open
Abstract
Cancer immunotherapies have achieved unprecedented success in clinic, but they remain largely ineffective in some major types of cancer, such as colorectal cancer with microsatellite stability (MSS CRC). It is therefore important to study tumor microenvironment of resistant cancers for developing new intervention strategies. In this study, we identify a metabolic cue that determines the unique immune landscape of MSS CRC. Through secretion of distal cholesterol precursors, which directly activate RORγt, MSS CRC cells can polarize T cells toward Th17 cells that have well-characterized pro-tumor functions in colorectal cancer. Analysis of large human cancer cohorts revealed an asynchronous pattern of the cholesterol biosynthesis in MSS CRC, which is responsible for the abnormal accumulation of distal cholesterol precursors. Inhibiting the cholesterol biosynthesis enzyme Cyp51, by pharmacological or genetic interventions, reduced the levels of intratumoral distal cholesterol precursors and suppressed tumor progression through a Th17-modulation mechanism in preclinical MSS CRC models. Our study therefore reveals a novel mechanism of cancer-immune interaction and an intervention strategy for the difficult-to-treat MSS CRC.
Collapse
Affiliation(s)
- Yibing Bai
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tongzhou Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Qinshu Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Weiqiang You
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Haochen Yang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xintian Xu
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, China
| | - Ziyi Li
- Beijing Advanced Innovation Center for Genomics, BIOPIC and School of Life Sciences, Peking University, Beijing, China
| | - Yu Zhang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chengsong Yan
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lei Yang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiaqian Qiu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yuanhua Liu
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, China
| | - Shiyang Chen
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dongfang Wang
- Beijing Advanced Innovation Center for Genomics, BIOPIC and School of Life Sciences, Peking University, Beijing, China
| | - Binlu Huang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kexin Liu
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bao- Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhuozhong Wang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kang Li
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Xin Liu
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guangchuan Wang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weiwei Yang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianfeng Chen
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pei Hao
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Shanghai, China
| | - Zemin Zhang
- Beijing Advanced Innovation Center for Genomics, BIOPIC and School of Life Sciences, Peking University, Beijing, China
| | - Zhigang Wang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Chenqi Xu
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
239
|
Hansen UK, Church CD, Carnaz Simões AM, Frej MS, Bentzen AK, Tvingsholm SA, Becker JC, Fling SP, Ramchurren N, Topalian SL, Nghiem PT, Hadrup SR. T antigen-specific CD8+ T cells associate with PD-1 blockade response in virus-positive Merkel cell carcinoma. J Clin Invest 2024; 134:e177082. [PMID: 38618958 PMCID: PMC11014655 DOI: 10.1172/jci177082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/23/2024] [Indexed: 04/16/2024] Open
Abstract
Merkel cell carcinoma (MCC) is a highly immunogenic skin cancer primarily induced by Merkel cell polyomavirus, which is driven by the expression of the oncogenic T antigens (T-Ags). Blockade of the programmed cell death protein-1 (PD-1) pathway has shown remarkable response rates, but evidence for therapy-associated T-Ag-specific immune response and therapeutic strategies for the nonresponding fraction are both limited. We tracked T-Ag-reactive CD8+ T cells in peripheral blood of 26 MCC patients under anti-PD1 therapy, using DNA-barcoded pMHC multimers, displaying all peptides from the predicted HLA ligandome of the oncoproteins, covering 33 class I haplotypes. We observed a broad T cell recognition of T-Ags, including identification of 20 T-Ag-derived epitopes we believe to be novel. Broadening of the T-Ag recognition profile and increased T cell frequencies during therapy were strongly associated with clinical response and prolonged progression-free survival. T-Ag-specific T cells could be further boosted and expanded directly from peripheral blood using artificial antigen-presenting scaffolds, even in patients with no detectable T-Ag-specific T cells. These T cells provided strong tumor-rejection capacity while retaining a favorable phenotype for adoptive cell transfer. These findings demonstrate that T-Ag-specific T cells are associated with the clinical outcome to PD-1 blockade and that Ag-presenting scaffolds can be used to boost such responses.
Collapse
Affiliation(s)
- Ulla Kring Hansen
- Section of Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- PokeAcell Aps, BioInnovation Institute, Copenhagen, Denmark
| | - Candice D. Church
- Department of Dermatology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Marcus Svensson Frej
- Section of Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- PokeAcell Aps, BioInnovation Institute, Copenhagen, Denmark
| | - Amalie Kai Bentzen
- Section of Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Siri A. Tvingsholm
- Section of Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jürgen C. Becker
- Department of Translational Skin Cancer Research, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | | | | | - Suzanne L. Topalian
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Paul T. Nghiem
- Department of Dermatology, Department of Medicine, University of Washington, Seattle, Washington, USA
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sine Reker Hadrup
- Section of Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
240
|
Li D, Cao D, Sun Y, Cui Y, Zhang Y, Jiang J, Cao X. The roles of epigallocatechin gallate in the tumor microenvironment, metabolic reprogramming, and immunotherapy. Front Immunol 2024; 15:1331641. [PMID: 38348027 PMCID: PMC10859531 DOI: 10.3389/fimmu.2024.1331641] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Cancer, a disease that modern medicine has not fully understood and conquered, with its high incidence and mortality, deprives countless patients of health and even life. According to global cancer statistics, there were an estimated 19.3 million new cancer cases and nearly 10 million cancer deaths in 2020, with the age-standardized incidence and mortality rates of 201.0 and 100.7 per 100,000, respectively. Although remarkable advancements have been made in therapeutic strategies recently, the overall prognosis of cancer patients remains not optimistic. Consequently, there are still many severe challenges to be faced and difficult problems to be solved in cancer therapy today. Epigallocatechin gallate (EGCG), a natural polyphenol extracted from tea leaves, has received much attention for its antitumor effects. Accumulating investigations have confirmed that EGCG can inhibit tumorigenesis and progression by triggering apoptosis, suppressing proliferation, invasion, and migration, altering tumor epigenetic modification, and overcoming chemotherapy resistance. Nevertheless, its regulatory roles and biomolecular mechanisms in the immune microenvironment, metabolic microenvironment, and immunotherapy remain obscure. In this article, we summarized the most recent updates about the effects of EGCG on tumor microenvironment (TME), metabolic reprogramming, and anti-cancer immunotherapy. The results demonstrated EGCG can promote the anti-cancer immune response of cytotoxic lymphocytes and dendritic cells (DCs), attenuate the immunosuppression of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), and inhibit the tumor-promoting functions of tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), and various stromal cells including cancer-associated fibroblasts (CAFs), endothelial cells (ECs), stellate cells, and mesenchymal stem/stromal cells (MSCs). Additionally, EGCG can suppress multiple metabolic reprogramming pathways, including glucose uptake, aerobic glycolysis, glutamine metabolism, fatty acid anabolism, and nucleotide synthesis. Finally, EGCG, as an immunomodulator and immune checkpoint blockade, can enhance immunotherapeutic efficacy and may be a promising candidate for antitumor immunotherapy. In conclusion, EGCG plays versatile regulatory roles in TME and metabolic reprogramming, which provides novel insights and combined therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Dongming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yuanlin Sun
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yingnan Cui
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yangyu Zhang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
241
|
van der Heide V, Davenport B, Cubitt B, Roudko V, Choo D, Humblin E, Jhun K, Angeliadis K, Dawson T, Furtado G, Kamphorst A, Ahmed R, de la Torre JC, Homann D. Functional impairment of "helpless" CD8 + memory T cells is transient and driven by prolonged but finite cognate antigen presentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576725. [PMID: 38328184 PMCID: PMC10849538 DOI: 10.1101/2024.01.22.576725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Generation of functional CD8 + T cell memory typically requires engagement of CD4 + T cells. However, in certain scenarios, such as acutely-resolving viral infections, effector (T E ) and subsequent memory (T M ) CD8 + T cell formation appear impervious to a lack of CD4 + T cell help during priming. Nonetheless, such "helpless" CD8 + T M respond poorly to pathogen rechallenge. At present, the origin and long-term evolution of helpless CD8 + T cell memory remain incompletely understood. Here, we demonstrate that helpless CD8 + T E differentiation is largely normal but a multiplicity of helpless CD8 T M defects, consistent with impaired memory maturation, emerge as a consequence of prolonged yet finite exposure to cognate antigen. Importantly, these defects resolve over time leading to full restoration of CD8 + T M potential and recall capacity. Our findings provide a unified explanation for helpless CD8 + T cell memory and emphasize an unexpected CD8 + T M plasticity with implications for vaccination strategies and beyond.
Collapse
|
242
|
Gong C, Tu Z, Long X, Liu X, Liu F, Liu J, Zhu X, Li J, Huang K. Predictive role of E2F6 in cancer prognosis and responses of immunotherapy. Int Immunopharmacol 2024; 127:111302. [PMID: 38071912 DOI: 10.1016/j.intimp.2023.111302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/16/2023] [Accepted: 11/24/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND E2F6 is a member of the E2F transcription factor family. Numerous studies have demonstrated that E2F6 is critical to cancer development and progression, but its role in cancer immunotherapy remains unclear. METHODS Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases were used to obtain RNA-seq data for cancer and normal tissues, and we utilized the cBioPortal to analyze E2F6 genomic alterations in pan-cancer. The protein localization of E2F6 was obtained using the Human Protein Atlas (HPA), and the upregulation of E2F6 expression in clinical glioblastoma multiforme (GBM) tissues was detected by Western blot analysis. The ComPPI website was used to analyze the protein interaction information of E2F6. To evaluate the role of E2F6 in pan-cancer prognosis, we used univariate Cox regression and Kaplan-Meier methods, and gene set enrichment analysis (GSEA) was utilized to identify markers associated with E2F6 expression in tumors. TIMER 2.0 was used to study E2F6-related immune cell infiltration in tumor tissues, and the correlation of E2F6 with immunotherapy biomarkers was investigated using Spearman correlation analysis. The role of E2F6 in the cell cycle was analyzed by flow cytometry, and the Cell Counting Kit-8 (CCK-8) and colony formation assays were utilized to determine the proliferative ability of cells. RESULTS In most tumor types, E2F6 was highly expressed and was a good predictor of prognosis. E2F6 was significantly related to markers of immune activation, tumor immune cell infiltration, and immune regulators. Furthermore, E2F6 knockdown significantly attenuated the proliferative ability of glioma cells. Finally, E2F6 effectively predicted anti-programmed cell death 1 (PD1) treatment response. CONCLUSION E2F6 is an effective biomarker that predicts the prognosis of cancer patients treated with anti-immune checkpoint therapy.
Collapse
Affiliation(s)
- Chuandong Gong
- Department of Neurosurgery, the 2(nd) affiliated hospital, Jiangxi Medical College, Nanchang University, Jiangxi 330006, PR China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, PR China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, PR China; JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, PR China
| | - Zewei Tu
- Department of Neurosurgery, the 2(nd) affiliated hospital, Jiangxi Medical College, Nanchang University, Jiangxi 330006, PR China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, PR China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, PR China; JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, PR China
| | - Xiaoyan Long
- East China Institute of Digital Medical Engineering, Shangrao, Jiangxi 330006, PR China
| | - Xinjun Liu
- People's Hospital of Yingtan City, Yingtan, Jiangxi 330006, PR China
| | - Feng Liu
- Department of Neurosurgery, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi 330006, PR China
| | - Jia Liu
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06511, USA
| | - Xingen Zhu
- Department of Neurosurgery, the 2(nd) affiliated hospital, Jiangxi Medical College, Nanchang University, Jiangxi 330006, PR China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, PR China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, PR China; JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, PR China.
| | - Jingying Li
- Department of Comprehensive Intensive Care Unit, the 2(nd) affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| | - Kai Huang
- Department of Neurosurgery, the 2(nd) affiliated hospital, Jiangxi Medical College, Nanchang University, Jiangxi 330006, PR China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, PR China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, PR China; JXHC Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
243
|
Jenika D, Pounraj S, Wibowo D, Flaxl LM, Rehm BHA, Mintern JD. In vivo assembly of epitope-coated biopolymer particles that induce anti-tumor responses. NPJ Vaccines 2024; 9:18. [PMID: 38263169 PMCID: PMC10805745 DOI: 10.1038/s41541-023-00787-8] [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: 10/31/2022] [Accepted: 10/02/2023] [Indexed: 01/25/2024] Open
Abstract
There is an unmet need for antigen delivery systems that elicit efficient T cell priming to prevent infectious diseases or for treatment of cancers. Here, we explored the immunogenic potential of biologically assembled biopolymer particles (BPs) that have been bioengineered to display the antigenic MHC I and MHC II epitopes of model antigen ovalbumin (OVA). Purified dendritic cells (DCs) captured BP-OVA and presented the associated antigenic epitopes to CD4+ T cells and CD8+ T cells. Vaccination with BP-OVA in the absence of adjuvant elicited antigen presentation to OVA-specific CD8+ and CD4+ T cells and cross-primed effective cytotoxic T lymphocyte (CTL) killers. BP-OVA induction of CTL killing did not require CD4+ T cell help, with active CTLs generated in BP-OVA vaccinated I-Ab-/- and CD40-/- mice. In contrast, IL-15 and type I IFN were required, with abrogated CTL activity in vaccinated IL-15-/- and IFNAR1-/- mice. cDC1 and/or CD103+ DCs were not essential for BP-OVA specific CTL with immunization eliciting responses in Batf3-/- mice. Poly I:C, but not LPS or CpG, co-administered as an adjuvant with BP-OVA boosted CTL responses. Finally, vaccination with BP-OVA protected against B16-OVA melanoma and Eμ-myc-GFP-OVA lymphoma inoculation. In summary, we have demonstrated that epitope-displaying BPs represent an antigen delivery platform exhibiting a unique mechanism to effectively engage T cell immune responses.
Collapse
Affiliation(s)
- Devi Jenika
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, 3010, Australia
| | - Saranya Pounraj
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, 4111, Australia
| | - David Wibowo
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, 4111, Australia
| | - Leonhard M Flaxl
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, 3010, Australia
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, 4111, Australia.
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, 4215, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, 3010, Australia.
| |
Collapse
|
244
|
Morath K, Sadhu L, Dyckhoff G, Gapp M, Keppler OT, Fackler OT. Activation-neutral gene editing of tonsillar CD4 T cells for functional studies in human ex vivo tonsil cultures. CELL REPORTS METHODS 2024; 4:100685. [PMID: 38211593 PMCID: PMC10831948 DOI: 10.1016/j.crmeth.2023.100685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/13/2023] [Accepted: 12/13/2023] [Indexed: 01/13/2024]
Abstract
The molecular and immunological properties of tissue-resident resting CD4 T cells are understudied due to the lack of suitable gene editing methods. Here, we describe the ex vivo culture and gene editing methodology ediTONSIL for CD4 T cells from human tonsils. Optimized CRISPR-Cas9 RNP nucleofection results in knockout efficacies of over 90% without requiring exogenous activation. Editing can be performed on multiple cell types in bulk cultures or on isolated CD4 T cells that can be labeled and reintroduced into their tissue environment. Importantly, CD4 T cells maintain their tissue-specific properties such as viability, activation state, or immunocompetence following reassembly into lymphoid aggregates. This highly efficient and versatile gene editing workflow for tonsillar CD4 T cells enables the dissection of molecular mechanisms in ex vivo cultures of human lymphoid tissue and can be adapted to other tonsil-resident cell types.
Collapse
Affiliation(s)
- Katharina Morath
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany
| | - Lopamudra Sadhu
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany
| | - Gerhard Dyckhoff
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Madeleine Gapp
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig-Maximilians-Universität München, Pettenkoferstraße 9a, 80336 Munich, Germany
| | - Oliver T Keppler
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig-Maximilians-Universität München, Pettenkoferstraße 9a, 80336 Munich, Germany; German Centre for Infection Research (DZIF), Partner Site München, Munich, Germany
| | - Oliver T Fackler
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany; German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
245
|
Fu S, Tan Z, Shi H, Chen J, Zhang Y, Guo C, Feng W, Xu H, Wang J, Wang H. Development of a stemness-related prognostic index to provide therapeutic strategies for bladder cancer. NPJ Precis Oncol 2024; 8:14. [PMID: 38245587 PMCID: PMC10799910 DOI: 10.1038/s41698-024-00510-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024] Open
Abstract
Bladder cancer (BC) is a heterogeneous disease with varying clinical outcomes. Recent evidence suggests that cancer progression involves the acquisition of stem-like signatures, and assessing stemness indices help uncover patterns of intra-tumor molecular heterogeneity. We used the one-class logistic regression algorithm to compute the mRNAsi for each sample in BLCA cohort. We subsequently classified BC patients into two subtypes based on 189 mRNAsi-related genes, using the unsupervised consensus clustering. Then, we identified nine hub genes to construct a stemness-related prognostic index (SRPI) using Cox regression, LASSO regression and Random Forest methods. We further validated SRPI using two independent datasets. Afterwards, we examined the molecular and immune characterized of SRPI. Finally, we conducted multiply drug screening and experimental approaches to identify and confirm the most proper agents for patients with high SRPI. Based on the mRNAsi-related genes, BC patients were classified into two stemness subtypes with distinct prognosis, functional annotations, genomic variations and immune profiles. Using the SRPI, we identified a specific subgroup of BC patients with high SRPI, who had a poor response to immunotherapy, and were less sensitive to commonly used chemotherapeutic agents, FGFR inhibitors, and EGFR inhibitors. We further identified that dasatinib was the most promising therapeutic agent for this subgroup of patients. This study provides further insights into the stemness classification of BC, and demonstrates that SRPI is a promising tool for predicting prognosis and therapeutic opportunities for BC patients.
Collapse
Affiliation(s)
- Shi Fu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | - Zhiyong Tan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | - Hongjin Shi
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | - Junhao Chen
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | | | - Chunming Guo
- School for Life Science, Yunnan University, Kunming, China
| | - Wei Feng
- Kunming Medical University, Kunming, China
| | - Haole Xu
- Kunming Medical University, Kunming, China
| | - Jiansong Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China.
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China.
| |
Collapse
|
246
|
Bawden EG, Wagner T, Schröder J, Effern M, Hinze D, Newland L, Attrill GH, Lee AR, Engel S, Freestone D, de Lima Moreira M, Gressier E, McBain N, Bachem A, Haque A, Dong R, Ferguson AL, Edwards JJ, Ferguson PM, Scolyer RA, Wilmott JS, Jewell CM, Brooks AG, Gyorki DE, Palendira U, Bedoui S, Waithman J, Hochheiser K, Hölzel M, Gebhardt T. CD4 + T cell immunity against cutaneous melanoma encompasses multifaceted MHC II-dependent responses. Sci Immunol 2024; 9:eadi9517. [PMID: 38241401 DOI: 10.1126/sciimmunol.adi9517] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024]
Abstract
Whereas CD4+ T cells conventionally mediate antitumor immunity by providing help to CD8+ T cells, recent clinical studies have implied an important role for cytotoxic CD4+ T cells in cancer immunity. Using an orthotopic melanoma model, we provide a detailed account of antitumoral CD4+ T cell responses and their regulation by major histocompatibility complex class II (MHC II) in the skin. Intravital imaging revealed prominent interactions of CD4+ T cells with tumor debris-laden MHC II+ host antigen-presenting cells that accumulated around tumor cell nests, although direct recognition of MHC II+ melanoma cells alone could also promote CD4+ T cell control. CD4+ T cells stably suppressed or eradicated tumors even in the absence of other lymphocytes by using tumor necrosis factor-α and Fas ligand (FasL) but not perforin-mediated cytotoxicity. Interferon-γ was critical for protection, acting both directly on melanoma cells and via induction of nitric oxide synthase in myeloid cells. Our results illustrate multifaceted and context-specific aspects of MHC II-dependent CD4+ T cell immunity against cutaneous melanoma, emphasizing modulation of this axis as a potential avenue for immunotherapies.
Collapse
Affiliation(s)
- Emma G Bawden
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Teagan Wagner
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jan Schröder
- Computational Sciences Initiative, Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Maike Effern
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Daniel Hinze
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Lewis Newland
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Grace H Attrill
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Ariane R Lee
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sven Engel
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David Freestone
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Marcela de Lima Moreira
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Elise Gressier
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Nathan McBain
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ashraful Haque
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ruining Dong
- Computational Sciences Initiative, Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Pathology and Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Angela L Ferguson
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Centenary Institute, University of Sydney, Sydney, NSW, Australia
- Infection, Immunity and Inflammation theme, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Jarem J Edwards
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Peter M Ferguson
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- NSW Health Pathology, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Department of Tissue Oncology and Diagnostic Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- NSW Health Pathology, Sydney, NSW, Australia
| | - James S Wilmott
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| | - Andrew G Brooks
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David E Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre Melbourne, Melbourne, VIC, Australia
| | - Umaimainthan Palendira
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jason Waithman
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Katharina Hochheiser
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre Melbourne, Melbourne, VIC, Australia
| | - Michael Hölzel
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, Bonn 53105, Germany
| | - Thomas Gebhardt
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
247
|
Lensch V, Gabba A, Hincapie R, Bhagchandani SH, Basak A, Alam MM, Irvine DJ, Shalek AK, Johnson JA, Finn MG, Kiessling LL. Glycan-costumed virus-like particles promote type 1 anti-tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.575711. [PMID: 38293025 PMCID: PMC10827186 DOI: 10.1101/2024.01.18.575711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Cancer vaccine development is inhibited by a lack of strategies for directing dendritic cell (DC) induction of effective tumor-specific cellular immunity. Pathogen engagement of DC lectins and toll-like receptors (TLRs) shapes immunity by directing T cell function. Strategies to activate specific DC signaling pathways via targeted receptor engagement are crucial to unlocking type 1 cellular immunity. Here, we engineered a glycan-costumed virus-like particle (VLP) vaccine that delivers programmable peptide antigens to induce tumor-specific cellular immunity in vivo. VLPs encapsulating TLR7 agonists and decorated with a selective mannose-derived ligand for the lectin DC-SIGN induced robust DC activation and type 1 cellular immunity, whereas VLPs lacking this key DC-SIGN ligand failed to promote DC-mediated immunity. Vaccination with glycan-costumed VLPs generated tumor antigen-specific Th1 CD4+ and CD8+ T cells that infiltrated solid tumors, inhibiting tumor growth in a murine melanoma model. Thus, VLPs employing lectin-driven immune reprogramming provide a framework for advancing cancer immunotherapies.
Collapse
Affiliation(s)
- Valerie Lensch
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adele Gabba
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Hincapie
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sachin H. Bhagchandani
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ankit Basak
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | | | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Alex K. Shalek
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jeremiah A. Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - M. G. Finn
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Laura L. Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
248
|
Sobral MC, Mooney DJ. Materials-Based Approaches for Cancer Vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:179-187. [PMID: 38166245 DOI: 10.4049/jimmunol.2300482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/27/2023] [Indexed: 01/04/2024]
Abstract
Therapeutic cancer vaccines offer the promise of stimulating the immune system to specifically eradicate tumor cells and establish long-term memory to prevent tumor recurrence. However, despite showing benign safety profiles and the ability to generate Ag-specific cellular responses, cancer vaccines have been hampered by modest clinical efficacy. Lessons learned from these studies have led to the emergence of innovative materials-based strategies that aim to boost the clinical activity of cancer vaccines. In this Brief Review, we provide an overview of the key elements needed for an effective vaccine-induced antitumor response, categorize current approaches to therapeutic cancer vaccination, and explore recent advances in materials-based strategies to potentiate cancer vaccines.
Collapse
Affiliation(s)
- Miguel C Sobral
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA; and Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA; and Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA
| |
Collapse
|
249
|
Hua Y, Yang S, Zhang Y, Li J, Wang M, Yeerkenbieke P, Liao Q, Liu Q. Modulating ferroptosis sensitivity: environmental and cellular targets within the tumor microenvironment. J Exp Clin Cancer Res 2024; 43:19. [PMID: 38217037 PMCID: PMC10787430 DOI: 10.1186/s13046-023-02925-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/06/2023] [Indexed: 01/14/2024] Open
Abstract
Ferroptosis, a novel form of cell death triggered by iron-dependent phospholipid peroxidation, presents significant therapeutic potential across diverse cancer types. Central to cellular metabolism, the metabolic pathways associated with ferroptosis are discernible in both cancerous and immune cells. This review begins by delving into the intricate reciprocal regulation of ferroptosis between cancer and immune cells. It subsequently details how factors within the tumor microenvironment (TME) such as nutrient scarcity, hypoxia, and cellular density modulate ferroptosis sensitivity. We conclude by offering a comprehensive examination of distinct immunophenotypes and environmental and metabolic targets geared towards enhancing ferroptosis responsiveness within the TME. In sum, tailoring precise ferroptosis interventions and combination strategies to suit the unique TME of specific cancers may herald improved patient outcomes.
Collapse
Affiliation(s)
- Yuze Hua
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yalu Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of General Surgery, Anhui Provincial Hospital, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230027, China
| | - Jiayi Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Mengyi Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Palashate Yeerkenbieke
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China
- Department of General Surgery, Xinjiang Yili Kazak Autonomous Prefecture Friendship Hospital, Xinjiang, 835099, China
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 1# Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
250
|
Wang L, Peng B, Yan Y, Liu G, Yang D, Wang Q, Li Y, Mao Q, Chen Q. The tRF-3024b hijacks miR-192-5p to increase BCL-2-mediated resistance to cytotoxic T lymphocytes in Esophageal Squamous Cell Carcinoma. Int Immunopharmacol 2024; 126:111135. [PMID: 37977065 DOI: 10.1016/j.intimp.2023.111135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
The limited efficacy of immune checkpoint inhibitors (ICIs) in the treatment of advanced Esophageal Squamous Cell Carcinoma (ESCC) poses a challenge. Recent evidence suggests that tumor cells' insensitivity to cytotoxic T lymphocytes (CTLs) contributes to drug resistance against ICIs. Here, a particular tRNA-derived fragment called tRF-3024b has been identified as playing a significant role in tumor cell resistance to CTLs. Through tRF sequencing (tRF-seq), we observed a high expression of tRF-3024b in ESCC cells that survived co-culture with CTLs. Further in vitro studies demonstrated that tRF-3024b reduced the apoptosis of tumor cells when co-cultured with CTLs. The mechanism behind this resistance involves tRF-3024b promoting the expression of B-cell lymphoma-2 (BCL-2) by sequestering miR-192-5p, a microRNA that would normally inhibit BCL-2 expression. This means that tRF-3024b indirectly enhances the protective effects of BCL-2, reducing apoptosis in tumor cells. Rescue assays confirmed that the suppressive function of tRF-3024b relies on BCL-2. In summary, the tRF-3024b/miR-192-5p/BCL-2 axis sheds light on the crucial role of tRF-3024b in regulating BCL-2 expression. These findings offer valuable insights into strategies to enhance the response of ESCC to CTLs and improve the effectiveness of immunotherapy approaches in treating ESCC.
Collapse
Affiliation(s)
- Lin Wang
- Department of Oncology, Department of Geriatric Lung Cancer Laboratory, The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Bo Peng
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan Yan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guangjun Liu
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dunpeng Yang
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qibin Wang
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongcheng Li
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Qixing Mao
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Qiang Chen
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|