1
|
Zhang P, Liu Y, Zhou X, Chen Y, Pan L, Xu F, Zhang X, Shen L, Chen H. Evaluation of a multiplexed assay for six urinary proteins based on liquid microarray analysis technology. Anal Biochem 2025; 701:115820. [PMID: 39986412 DOI: 10.1016/j.ab.2025.115820] [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: 12/22/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Proteinuria serves as a critical indicator in the progression of chronic kidney disease. The quantification of urinary protein is instrumental in diagnosing kidney disease and monitoring therapeutic efficacy. Nephelometry and turbidimetric immunoassay are predominantly employed for the quantitative detection of various urinary proteins. We developed a quantitative detection method for six proteins in urine utilizing the flow fluorescence luminescence method on the Luminex®200™ detection platform. This study aims to evaluate the clinical performance and accuracy of the method. The sensitivity, precision, recovery, linearity, and interference metrics for the method meet the needs of clinical applications. The correlation coefficient (r) between the results of the method and nephelometry immunoassay on SIEMENS BNII platform ranged from 0.9375 to 0.9847, indicating a strong linear relationship. The results of the Bland-Altman analysis indicated that the systematic bias between the two methods was minimal. Additionally, Passing-Bablok regression analysis, both including and excluding outliers, was conducted on the clinical trial data. The findings demonstrated consistency between the two methods, with the expected confidence interval of bias remaining within the allowable error limits, indicating no significant discrepancy. This urine six protein quantification method demonstrates satisfactory performance, meeting the standards necessary for medical diagnosis and clinical application.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China
| | - Yi Liu
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China
| | - Xinyun Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinyu Chen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liudi Pan
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feiyi Xu
- Shanghai CP Adaltis Diagnostics Co., Ltd., Shanghai, China
| | - Xiaolong Zhang
- Shanghai CP Adaltis Diagnostics Co., Ltd., Shanghai, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China.
| | - Hui Chen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, China.
| |
Collapse
|
2
|
Zhu YJ, Li SY, Yang SS, Du Y, Zhang ZY, Liu JY. CD44 on cancer stem cell is a potential immunological and prognostic pan-cancer biomarker. Cancer Cell Int 2025; 25:134. [PMID: 40200220 PMCID: PMC11978154 DOI: 10.1186/s12935-025-03748-4] [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: 08/15/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND CD44, a widely recognized cancer stem cell marker, displayed a vital participation in the cancer immune invasion and may related with the response to the immunotherapy. However, the role of CD44 in cancer immunology is not well defined. Therefore, we intended to explore its prognostic value and potential immunological functions across 33 human cancer types. METHODS Based on the data of patients from The Cancer Genome Atlas (TCGA), Sangerbox was used to analyze the correlations between CD44 expression and tumor-infiltrated immune cells, immune checkpoints, neoantigens, microsatellite instability (MSI), and tumor mutational burden (TMB) in human cancers. A mouse model xenografted with shRNA-CD44 MC38 was established. RESULTS The elevated CD44 was associated with tumor stage and prognosis in several different cancers. GSEA results showed that upregulated CD44 involved in cancer stem cell associated process, antigen processing and presentation, and immune cells proliferation and activation. CD44 plays an essential role in the tumor immune regulation and immune checkpoints inhibitor response. The correlation of CD44 gene expression and infiltration levels of immune cells varied across different cancer types. Notably, the upregulation of CD44 expression is positively correlated with regulatory CD4 T cells, macrophages M1 and M2 in several analyzed cancers. Furthermore, we verified the effect of CD44 on tumor growth and immune microenvironment in mouse xenografted with shRNA-CD44 MC38. Moreover, DNA methylation existed in CD44 expression and associated with dysfunctional T-cell phenotypes via different mechanisms, thus resulting in tissue-dependent prognoses. CONCLUSION CD44 is both a cancer stem cell marker and a potential prognostic and immunological biomarker in various malignant tumors. Moreover, CD44 could be a novel target for immune-based therapy.
Collapse
Affiliation(s)
- Ya-Juan Zhu
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Si-Ying Li
- Department of Liver Surgery, Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shan-Shan Yang
- Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Du
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuo-Yuan Zhang
- Department of Head and Neck Cancer Surgery, West China School of Stomatology, Sichuan University, Chengdu, China.
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China.
| | - Ji-Yan Liu
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Lin Y, Song Y, Zhang Y, Li X, Kan L, Han S. New insights on anti-tumor immunity of CD8 + T cells: cancer stem cells, tumor immune microenvironment and immunotherapy. J Transl Med 2025; 23:341. [PMID: 40097979 PMCID: PMC11912710 DOI: 10.1186/s12967-025-06291-y] [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: 10/21/2024] [Accepted: 02/23/2025] [Indexed: 03/19/2025] Open
Abstract
Recent breakthroughs in tumor immunotherapy have confirmed the capacity of the immune system to fight several cancers. The effective means of treating cancer involves accelerating the death of tumor cells and improving patient immunity. Dynamic changes in the tumor immune microenvironment alter the actual effects of anti-tumor drug production and may trigger favorable or unfavorable immune responses by modulating tumor-infiltrating lymphocytes. Notably, CD8+ T cells are one of the primary tumor-infiltrating immune cells that provide anti-tumor response. Tumor cells and tumor stem cells will resist or evade destruction through various mechanisms as CD8+ T cells exert their anti-tumor function. This paper reviews the research on the regulation of tumor development and prognosis by cancer stem cells that directly or indirectly alter the role of tumor-infiltrating CD8+ T cells. We also discuss related immunotherapy strategies.
Collapse
Affiliation(s)
- Yibin Lin
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaodong Li
- Department of Neurosurgery, Siping Central People's Hospital, Siping, Jilin, 136000, China
| | - Liang Kan
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
4
|
Zheng F, Zhang S, Chang AE, Moon JJ, Wicha MS, Wang SX, Chen J, Liu J, Cheng F, Li Q. Breaking Immunosuppression to Enhance Cancer Stem Cell-Targeted Immunotherapy. Int J Biol Sci 2025; 21:1819-1836. [PMID: 39990669 PMCID: PMC11844285 DOI: 10.7150/ijbs.101025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/03/2024] [Indexed: 02/25/2025] Open
Abstract
Cancer stem cell (CSC)-targeted immunotherapy has emerged as a novel strategy in cancer treatment in the past decade. However, its efficacy is significantly limited due to the existence of host immune suppressive activity. Specifically, programmed cell death ligand-1 (PD-L1) is overexpressed in CSCs, and PD-L1 overexpressed CSCs create immunosuppressive milieu via interacting with various immune cells in tumor microenvironments (TME). Hence, novel immunotherapeutic strategies targeting CSCs with concurrent immunosuppression interruption will be promising in enhancing anti-CSC effects. These include dendritic cell (DC) and nanodisc (ND)-based vaccines to present CSC antigens in the forms of CSC lysate, CSC-marker proteins, and CSC-derived peptides to induce anti-CSC immunity. In addition, CSC-directed bispecific antibodies (BiAbs) and antibody drug conjugates (ADCs) have been developed to target CSCs effectively. Furthermore, chimeric antigen receptor (CAR)-T cell therapy and natural killer (NK) cell-based therapy targeting CSCs have achieved progress in both solid and hematologic tumors, and inhibition of CSC associated signaling pathways has proven successful. In this review, we aimed to outline the roles and regulatory mechanisms of PD-L1 in the properties of CSCs; the crosstalk between CSCs and immunosuppressive cells in TME, and recent progress and future promises of immunosuppression blockage to enhance CSC-targeted immunotherapy.
Collapse
Affiliation(s)
- Fang Zheng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Shan Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Alfred E. Chang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Max S. Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | - Junhui Chen
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Jixian Liu
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Fanjun Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
5
|
Brosseau S, Abreu P, Bouchez C, Charon L, Kieffer Y, Gentric G, Picant V, Veith I, Camonis J, Descroix S, Mechta-Grigoriou F, Parrini MC, Zalcman G. YAP/TEAD involvement in resistance to paclitaxel chemotherapy in lung cancer. Mol Cell Biochem 2025; 480:231-248. [PMID: 38427166 DOI: 10.1007/s11010-024-04949-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
The Yes-associated protein (YAP) oncoprotein has been linked to both metastases and resistance to targeted therapy of lung cancer cells. We aimed to investigate the effect of YAP pharmacological inhibition, using YAP/TEA domain (TEAD) transcription factor interaction inhibitors in chemo-resistant lung cancer cells. YAP subcellular localization, as a readout for YAP activation, cell migration, and TEAD transcription factor functional transcriptional activity were investigated in cancer cell lines with up-regulated YAP, with and without YAP/TEAD interaction inhibitors. Parental (A549) and paclitaxel-resistant (A549R) cell transcriptomes were analyzed. The half-maximal inhibitory concentration (IC50) of paclitaxel or trametinib, which are Mitogen-Activated protein kinase and Erk Kinase (MEK) inhibitors, combined with a YAP/TEAD inhibitor (IV#6), was determined. A three-dimensional (3D) microfluidic culture device enabled us to study the effect of IV#6/paclitaxel combination on cancer cells isolated from fresh resected lung cancer samples. YAP activity was significantly higher in paclitaxel-resistant cell lines. The YAP/TEAD inhibitor induced a decreased YAP activity in A549, PC9, and H2052 cells, with reduced YAP nuclear staining. Wound healing assays upon YAP inhibition revealed impaired cell motility of lung cancer A549 and mesothelioma H2052 cells. Combining YAP pharmacological inhibition with trametinib in K-Ras mutated A549 cells recapitulated synthetic lethality, thereby sensitizing these cells to MEK inhibition. The YAP/TEAD inhibitor lowered the IC50 of paclitaxel in A549R cells. Differential transcriptomic analysis of parental and A549R cells revealed an increased YAP/TEAD transcriptomic signature in resistant cells, downregulated upon YAP inhibition. The YAP/TEAD inhibitor restored paclitaxel sensitivity of A549R cells cultured in a 3D microfluidic system, with lung cancer cells from a fresh tumor efficiently killed by YAP/TEAD inhibitor/paclitaxel doublet. Evidence of the YAP/TEAD transcriptional program's role in chemotherapy resistance paves the way for YAP therapeutic targeting.
Collapse
Affiliation(s)
- S Brosseau
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- Medicine Faculty, Université Paris Cité, 26 rue Henri Henri Huchard, 75018, Paris, France
- Thoracic Oncology Department, Clinical Investigation Centre (CIC) 1425 INSERM, Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - P Abreu
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
| | - C Bouchez
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
| | - L Charon
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
| | - Y Kieffer
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- PSL Research University, Paris, France
| | - G Gentric
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- PSL Research University, Paris, France
| | - V Picant
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- PSL Research University, Paris, France
| | - I Veith
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- PSL Research University, Paris, France
| | - J Camonis
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- PSL Research University, Paris, France
| | - S Descroix
- PSL Research University, Paris, France
- UMR 168 CNRS "Physics and Chemistry Curie" Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
| | - F Mechta-Grigoriou
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- PSL Research University, Paris, France
| | - M C Parrini
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France
- PSL Research University, Paris, France
| | - G Zalcman
- U830 INSERM "Cancer, Heterogenity, Instability, Plasticity", Team "Stress and Cancer", Institut Curie Research Centre, 26 rue d'Ulm, 75248 Cedex 05, Paris, France.
- Medicine Faculty, Université Paris Cité, 26 rue Henri Henri Huchard, 75018, Paris, France.
- Thoracic Oncology Department, Clinical Investigation Centre (CIC) 1425 INSERM, Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
6
|
Caruso JA, Chen-Tanyolac C, Tlsty TD. A hybrid epithelial-mesenchymal transition program enables basal epithelial cells to bypass stress-induced stasis and contributes to a metaplastic breast cancer progenitor state. Breast Cancer Res 2024; 26:184. [PMID: 39696672 DOI: 10.1186/s13058-024-01920-8] [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: 08/26/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Human mammary epithelial cell (HMEC) cultures encounter a stress-associated barrier termed stasis, during which most cells adopt a senescence-like phenotype. From these cultures, rare variants emerge from the basal epithelial population, re-initiating growth. Variants exhibit pre-malignant properties, including an aberrant epigenetic program that enables continued proliferation and acquisition of genetic changes. Following oncogenic transformation, variants produce tumors that recapitulate the histopathological characteristics of metaplastic breast cancer (MBC), a rare and aggressive subtype marked by the differentiation of neoplastic epithelium into squamous and mesenchymal elements. METHODS Using a serum-free HMEC culture system, we probed the capacity for phenotypic plasticity inherent to basal epithelial cell populations from human breast tissue as they navigated stasis and emerged as variant populations. RESULTS We observed robust activation of a TGF-β-dependent epithelial-mesenchymal transition (EMT) program in basal epithelial cells during stasis, followed by subsequent attenuation of this program in emerging variants. Inhibition of the TGF-β pathway or depleting the EMT regulators Snail or Slug allowed basal epithelial cells to collectively bypass stasis, demonstrating that cellular dysfunction and arrest resulting from TGF-β and EMT activation are central to this in vitro barrier. The spontaneous emergence of variants from stasis cultures was associated with a restricted EMT trajectory, characterized by the stabilization of hybrid EMT states associated with greater proliferative capacity, rather than progressing to a complete mesenchymal state characterized by irreversible growth arrest. Epigenetic mechanisms, which contributed to the dysregulated growth control characteristic of the variant phenotype, also contributed to the stability of the hybrid EMT program in variants. By overcoming the cellular dysfunction and growth arrest resulting from TGF-β and complete EMT, variants exhibited a higher oncogenic transformation efficiency compared to pre-stasis basal epithelial cells. Inhibiting the TGF-β pathway prior to stasis significantly reduced EMT in the basal epithelial population, alleviated selective pressure driving variant emergence, and also enhanced oncogenic transformation efficiency, resulting in tumors with markedly diminished metaplastic differentiation. CONCLUSIONS This study reveals how an epigenetic program governs basal epithelial cell fate decisions and contributes to the development of MBC progenitors by restricting access to terminal mesenchymal states that induce growth arrest and, instead, favoring hybrid EMT states with enhanced tumorigenic potential.
Collapse
Affiliation(s)
- Joseph A Caruso
- Department of Pathology, University of California, San Francisco, San Francisco, CA, 94143, USA.
| | - Chira Chen-Tanyolac
- Department of Pathology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Thea D Tlsty
- Department of Pathology, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
7
|
Galassi C, Esteller M, Vitale I, Galluzzi L. Epigenetic control of immunoevasion in cancer stem cells. Trends Cancer 2024; 10:1052-1071. [PMID: 39244477 DOI: 10.1016/j.trecan.2024.08.004] [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: 03/30/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
Cancer stem cells (CSCs) are a poorly differentiated population of malignant cells that (at least in some neoplasms) is responsible for tumor progression, resistance to therapy, and disease relapse. According to a widely accepted model, all stages of cancer progression involve the ability of neoplastic cells to evade recognition or elimination by the host immune system. In line with this notion, CSCs are not only able to cope with environmental and therapy-elicited stress better than their more differentiated counterparts but also appear to better evade tumor-targeting immune responses. We summarize epigenetic modifications of DNA and histones through which CSCs evade immune recognition or elimination, and propose that such alterations constitute promising therapeutic targets to increase the sensitivity of some malignancies to immunotherapy.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Qian WJ, Yan JS, Gang XY, Xu L, Shi S, Li X, Na FJ, Cai LT, Li HM, Zhao MF. Intercellular adhesion molecule-1 (ICAM-1): From molecular functions to clinical applications in cancer investigation. Biochim Biophys Acta Rev Cancer 2024; 1879:189187. [PMID: 39317271 DOI: 10.1016/j.bbcan.2024.189187] [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: 03/18/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is a versatile molecule that plays a critical role in various physiological and pathological processes, particularly in tumor development where its impact is bidirectional. On the one hand, it augments the immune response by promoting immune cell migration, infiltration, and the formation of immunological synapses, thus facilitating potent antitumor effects. Simultaneously, it contributes to tumor immune evasion and influences metastasis by mediating transendothelial migration (TEM), epithelial-to-mesenchymal transition (EMT), and epigenetic modification of tumor cells. Despite its significant potential, the full clinical utility of ICAM-1 has yet to be fully realized. In this review, we thoroughly examine recent advancements in understanding the role of ICAM-1 in tumor development, its relevance in predicting therapeutic efficacy and prognosis, as well as the progress in clinical translational research on anti-ICAM-1-based therapies, encompassing including monoclonal antibodies, immunotherapy, antibody-drug conjugate (ADC), and conventional treatments. By shedding light on these innovative strategies, we aim to underscore ICAM-1's significance as a valuable and multifaceted target for cancer treatment, igniting enthusiasm for further research and facilitating translation into clinical applications.
Collapse
Affiliation(s)
- Wen-Jing Qian
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jin-Shan Yan
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xiao-Yu Gang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Lu Xu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Sha Shi
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xin Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Fang-Jian Na
- Network Information Center, China Medical University, Shenyang, China
| | - Lu-Tong Cai
- Psychological Medicine, Shenyang Medical College, Shenyang, China
| | - He-Ming Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China; Guangdong Association of Clinical Trials (GACT)/Chinese Thoracic Oncology Group (CTONG) and Guangdong Provincial Key Lab of Translational Medicine in Lung Cancer, Guangzhou, China.
| | - Ming-Fang Zhao
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
9
|
Caruso JA, Tlsty TD. An adaptive Epithelial-Mesenchymal Transition Program Enables Basal Epithelial Cells to Bypass Stress-Induced Stasis and Contributes to Metaplastic Breast Cancer Progenitor State. RESEARCH SQUARE 2024:rs.3.rs-4980285. [PMID: 39399685 PMCID: PMC11469408 DOI: 10.21203/rs.3.rs-4980285/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Background Human mammary epithelial cell (HMEC) cultures encounter a stress-associated barrier termed stasis, during which most cells adopt a senescence-like phenotype. From these cultures, rare variants emerge from the basal epithelial population, re-initiating growth. Variants exhibit pre-malignant properties, including an aberrant epigenetic program that enables continued proliferation and acquisition of genetic changes. Following oncogenic transformation, variants produce tumors that recapitulate the histopathological characteristics of metaplastic breast cancer (MBC), a rare subtype characterized by squamous and mesenchymal differentiation. Methods Using the conventional serum-free HMEC culture system, we probed the capacity for phenotypic plasticity inherent to basal epithelial cell populations from human breast tissue as they navigated stasis and emerged as variant populations. Results We observed robust activation of a TGF-β-dependent epithelial-mesenchymal transition (EMT) program in basal epithelial cells during stasis, followed by subsequent attenuation of this program in emerging variants. Inhibiting the TGF-β pathway or depleting the EMT regulators Snail or Slug allowed basal epithelial cells to collectively bypass stasis, demonstrating that cellular dysfunction and arrest resulting from TGF-β and EMT activation are central to this in vitro barrier. The spontaneous emergence of variants from stasis cultures was associated with a restricted EMT trajectory, which diverted cells away from a complete mesenchymal state characterized by irreversible growth arrest, and instead limited variants to epithelial and intermediate EMT states associated with greater proliferative capacity and stemness. Epigenetic mechanisms, which contributed to the dysregulated growth control characteristic of the variant phenotype, also contributed to the constrained EMT program in variants. By overcoming the cellular dysfunction and growth arrest resulting from TGF-β and EMT activation, variants exhibited increased oncogenic transformation efficiency compared to pre-stasis basal epithelial cells. Inhibiting the TGF-β pathway prior to stasis significantly reduced EMT in the basal epithelial population, alleviated selective pressure driving variant emergence, and enhanced oncogenic transformation efficiency, resulting in tumors with markedly diminished metaplastic differentiation. Conclusions This study reveals how adaptive EMT reprogramming governs basal epithelial cell fate decisions and contributes to the development of MBC progenitors by restricting access to terminal mesenchymal states that induce growth arrest and, instead, favoring intermediate states with enhanced tumorigenic potential.
Collapse
|
10
|
Sun Y, Yinwang E, Wang S, Wang Z, Wang F, Xue Y, Zhang W, Zhao S, Mou H, Chen S, Jin L, Li B, Ye Z. Phenotypic and spatial heterogeneity of CD8 + tumour infiltrating lymphocytes. Mol Cancer 2024; 23:193. [PMID: 39251981 PMCID: PMC11382426 DOI: 10.1186/s12943-024-02104-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
CD8+ T cells are the workhorses executing adaptive anti-tumour response, and targets of various cancer immunotherapies. Latest advances have unearthed the sheer heterogeneity of CD8+ tumour infiltrating lymphocytes, and made it increasingly clear that the bulk of the endogenous and therapeutically induced tumour-suppressive momentum hinges on a particular selection of CD8+ T cells with advantageous attributes, namely the memory and stem-like exhausted subsets. A scrutiny of the contemporary perception of CD8+ T cells in cancer and the subgroups of interest along with the factors arbitrating their infiltration contextures, presented herein, may serve as the groundwork for future endeavours to probe further into the regulatory networks underlying their differentiation and migration, and optimise T cell-based immunotherapies accordingly.
Collapse
Affiliation(s)
- Yikan Sun
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Eloy Yinwang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shengdong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Zenan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Fangqian Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Yucheng Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Wenkan Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shenzhi Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Haochen Mou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shixin Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Lingxiao Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Binghao Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China.
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
11
|
Zhang X, Tan Y, Li T, Tan D, Fu B, Yang M, Chen Y, Cao M, Xuan C, Du Q, Hu R, Wang Q. Intercellular adhesion molecule-1 suppresses TMZ chemosensitivity in acquired TMZ-resistant gliomas by increasing assembly of ABCB1 on the membrane. Drug Resist Updat 2024; 76:101112. [PMID: 38924997 DOI: 10.1016/j.drup.2024.101112] [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: 08/28/2023] [Revised: 05/31/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
AIMS Despite aggressive treatment, the recurrence of glioma is an inevitable occurrence, leading to unsatisfactory clinical outcomes. A plausible explanation for this phenomenon is the phenotypic alterations that glioma cells undergo aggressive therapies, such as TMZ-therapy. However, the underlying mechanisms behind these changes are not well understood. METHODS The TMZ chemotherapy resistance model was employed to assess the expression of intercellular adhesion molecule-1 (ICAM1) in both in vitro and in vivo settings. The potential role of ICAM1 in regulating TMZ chemotherapy resistance was investigated through knockout and overexpression techniques. Furthermore, the mechanism underlying ICAM1-mediated TMZ chemotherapy resistance was examined using diverse molecular biological methods, and the lipid raft protein was subsequently isolated to investigate the cellular subcomponents where ICAM1 operates. RESULTS Acquired TMZ resistant (TMZ-R) glioma models heightened production of intercellular adhesion molecule-1 (ICAM1) in TMZ-R glioma cells. Additionally, we observed a significant suppression of TMZ-R glioma proliferation upon inhibition of ICAM1, which was attributed to the enhanced intracellular accumulation of TMZ. Our findings provide evidence supporting the role of ICAM1, a proinflammatory marker, in promoting the expression of ABCB1 on the cell membrane of TMZ-resistant cells. We have elucidated the mechanistic pathway by which ICAM1 modulates phosphorylated moesin, leading to an increase in ABCB1 expression on the membrane. Furthermore, our research has revealed that the regulation of moesin by ICAM1 was instrumental in facilitating the assembly of ABCB1 exclusively on the lipid raft of the membrane. CONCLUSIONS Our findings suggest that ICAM1 is an important mediator in TMZ-resistant gliomas and targeting ICAM1 may provide a new strategy for enhancing the efficacy of TMZ therapy against glioma.
Collapse
Affiliation(s)
- Xin Zhang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, PR China; State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Yingying Tan
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Tao Li
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China; General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Dashan Tan
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Bin Fu
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Mengdi Yang
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Yaxin Chen
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Mengran Cao
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Chenyuan Xuan
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| | - Rong Hu
- State Key Laboratory of Natural Medicines, School of Basic Medical and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, PR China.
| | - Qing Wang
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, Jiangsu, PR China.
| |
Collapse
|
12
|
Zhao L, Jin S, Wang S, Zhang Z, Wang X, Chen Z, Wang X, Huang S, Zhang D, Wu H. Tertiary lymphoid structures in diseases: immune mechanisms and therapeutic advances. Signal Transduct Target Ther 2024; 9:225. [PMID: 39198425 PMCID: PMC11358547 DOI: 10.1038/s41392-024-01947-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are defined as lymphoid aggregates formed in non-hematopoietic organs under pathological conditions. Similar to secondary lymphoid organs (SLOs), the formation of TLSs relies on the interaction between lymphoid tissue inducer (LTi) cells and lymphoid tissue organizer (LTo) cells, involving multiple cytokines. Heterogeneity is a distinguishing feature of TLSs, which may lead to differences in their functions. Growing evidence suggests that TLSs are associated with various diseases, such as cancers, autoimmune diseases, transplant rejection, chronic inflammation, infection, and even ageing. However, the detailed mechanisms behind these clinical associations are not yet fully understood. The mechanisms by which TLS maturation and localization affect immune function are also unclear. Therefore, it is necessary to enhance the understanding of TLS development and function at the cellular and molecular level, which may allow us to utilize them to improve the immune microenvironment. In this review, we delve into the composition, formation mechanism, associations with diseases, and potential therapeutic applications of TLSs. Furthermore, we discuss the therapeutic implications of TLSs, such as their role as markers of therapeutic response and prognosis. Finally, we summarize various methods for detecting and targeting TLSs. Overall, we provide a comprehensive understanding of TLSs and aim to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Lianyu Zhao
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Song Jin
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyao Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zhe Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Xuan Wang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Zhanwei Chen
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Xiaohui Wang
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| |
Collapse
|
13
|
He W, Li Y, Liu SB, Chang Y, Han S, Han X, Ma Z, Amin HM, Song YH, Zhou J. From mitochondria to tumor suppression: ACAT1's crucial role in gastric cancer. Front Immunol 2024; 15:1449525. [PMID: 39247186 PMCID: PMC11377227 DOI: 10.3389/fimmu.2024.1449525] [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: 06/15/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Acetyl CoA acetyltransferase 1 (ACAT1), a mitochondrial enzyme, is mainly involved in the formation and decomposition of ketones, isoleucine, and fatty acids. Previous clinical studies showed that mutations in the ACAT1 gene lead to ketoacidosis, Notably the role of ACAT1 in human cancer' pathogenesis varies depending on cancer type, and its specific role in gastric cancer remains largely unknown. In the current study, we found that the expression of ACAT1 in primary late-stage gastric cancer tumor tissues was significantly lower than in early-stage tumors. This observation was further confirmed in high-grade gastric cancer cell line MKN45. The expression of CD44 and OCT4 was decreased, while CD24 expression was increased by overexpressing ACAT1 in MKN45 gastric cancer cells. Moreover, the ability of gastric cancer cells to form colonies on soft agar was also reduced by ACAT1 overexpression. Likewise, overexpression of ACAT1 inhibited epithelial mesenchymal transition (EMT) in gastric cancer cells evidenced by increased expression of the epithelial marker E-Cadherin, decreased expression of mesenchymal marker vimentin, and decreased expression levels of SNAI 1/3. In addition, ACAT1 overexpression inhibited cell migration and invasion, improved the response to 5-Fluorouracil (5-FU) and etoposide. In contrast, inhibition of ACAT1 activity promoted the proliferation of gastric cancer cells. The xenotransplantation results in nude mice showed that overexpression of ACAT1 in gastric cancer cells inhibited tumor growth in vivo. In addition, the low expression of ACAT1 in gastric cancer was further validated by searching public databases and conducting bioinformatic analyses. Mechanistically, bioinformatic analysis found that the inhibitory effect of ACAT1 in gastric cancer may be related to the Adipocytokine Signaling Pathway, Ppar Signaling Pathway, Propanoate Metabolism and P53 Signaling Pathway. Correlation analysis indicated ACAT1 mRNA expression was correlated with immune infiltrates. Collectively, our data show that ACAT1 induces pronounced inhibitory effects on gastric cancer initiation and development, which may impact future strategies to treat this aggressive cancer.
Collapse
Affiliation(s)
- Wei He
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Yanfang Li
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Song-Bai Liu
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, China
| | - Ying Chang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Shiyuan Han
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Xingyu Han
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Zixin Ma
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Hesham M Amin
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yao-Hua Song
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Jin Zhou
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Alqarni A, Jasim SA, Altalbawy FMA, Kaur H, Kaur I, Rodriguez-Benites C, Deorari M, Alwaily ER, Al-Ani AM, Redhee AH. Challenges and opportunities for cancer stem cell-targeted immunotherapies include immune checkpoint inhibitor, cancer stem cell-dendritic cell vaccine, chimeric antigen receptor immune cells, and modified exosomes. J Biochem Mol Toxicol 2024; 38:e23719. [PMID: 38764138 DOI: 10.1002/jbt.23719] [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: 02/10/2024] [Revised: 04/15/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024]
Abstract
Cancer stem cells (CSCs) are associated with the tumor microenvironment (TME). CSCs induce tumorigenesis, tumor recurrence and progression, and resistance to standard therapies. Indeed, CSCs pose an increasing challenge to current cancer therapy due to their stemness or self-renewal properties. The molecular and cellular interactions between heterogeneous CSCs and surrounding TME components and tumor-supporting immune cells show synergistic effects toward treatment failure. In the immunosuppressive TME, CSCs express various immunoregulatory proteins, growth factors, metabolites and cytokines, and also produce exosomes, a type of extracellular vesicles, to protect themselves from host immune surveillance. Among these, the identification and application of CSC-derived exosomes could be considered for the development of therapeutic approaches to eliminate CSCs or cancer, in addition to targeting the modulators that remodel the composition of the TME, as reviewed in this study. Here, we introduce the role of CSCs and how their interaction with TME complicates immunotherapies, and then present the CSC-based immunotherapy and the limitation of these therapies. We describe the biology and role of tumor/CSC-derived exosomes that induce immune suppression in the TME, and finally, introduce their potentials for the development of CSC-based targeted immunotherapy in the future.
Collapse
Affiliation(s)
- Abdullah Alqarni
- Department of Diagnostics Dental Sciences and Oral Biology, College of Dentistry, King Khalid University, Abha, Saudi Arabia
| | | | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Harpreet Kaur
- School of Basic and Applied Sciences, Shobhit University, Gangoh, India
- Department of Health and Allied Sciences, Arka Jain University, Jamshedpur, India
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, India
| | - Carlos Rodriguez-Benites
- Departamento Académico de Física, Facultad de Ciencias Físicas y Matemáticas, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi Qar, Iraq
| | - Ahmed M Al-Ani
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Ahmed H Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
15
|
Imodoye SO, Adedokun KA. EMT-induced immune evasion: connecting the dots from mechanisms to therapy. Clin Exp Med 2023; 23:4265-4287. [PMID: 37966552 DOI: 10.1007/s10238-023-01229-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a dynamic program crucial for organismal development and tissue regeneration. Unfortunately, this program is often hijacked by epithelial tumors to facilitate metastasis. Beyond its role in cancer spread, EMT increases cancer cell survival by activating stem cell programs and bypassing apoptotic programs. Importantly, the capacity of EMT to enforce tumor progression by altering the tumor cell phenotype without triggering immune responses opens the intriguing possibility of a mechanistic link between EMT-driven cancers and immune evasion. Indeed, EMT has been acknowledged as a of driver immune evasion, but the mechanisms are still evolving. Here, we review recent insights into the influence of EMT on tumor immune evasion. Specifically, we focus on the mechanistic roles of EMT in immune escape as the basis that may provide a platform for innovative therapeutic approaches in advanced tumors. We summarize promising therapeutic approaches currently in clinical trials and trending preclinical studies aimed at reinvigorating the tumor microenvironment to create immune-permissive conditions that facilitates immune-mediated tumor clearance. We anticipate that this will assist researchers and pharmaceutical companies in understanding how EMT compromises the immune response, potentially paving the way for effective cancer therapies.
Collapse
Affiliation(s)
- Sikiru O Imodoye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, USA.
| | - Kamoru A Adedokun
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| |
Collapse
|
16
|
Damei I, Trickovic T, Mami-Chouaib F, Corgnac S. Tumor-resident memory T cells as a biomarker of the response to cancer immunotherapy. Front Immunol 2023; 14:1205984. [PMID: 37545498 PMCID: PMC10399960 DOI: 10.3389/fimmu.2023.1205984] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TIL) often include a substantial subset of CD8+ tissue-resident memory T (TRM) cells enriched in tumor-specific T cells. These TRM cells play a major role in antitumor immune response. They are identified on the basis of their expression of the CD103 (αE(CD103)β7) and/or CD49a (α1(CD49a)β1) integrins, and the C-type lectin CD69, which are involved in tissue residency. TRM cells express several T-cell inhibitory receptors on their surface but they nevertheless react strongly to malignant cells, exerting a strong cytotoxic function, particularly in the context of blocking interactions of PD-1 with PD-L1 on target cells. These TRM cells form stable conjugates with autologous tumor cells and interact with dendritic cells and other T cells within the tumor microenvironment to orchestrate an optimal in situ T-cell response. There is growing evidence to indicate that TGF-β is essential for the formation and maintenance of TRM cells in the tumor, through the induction of CD103 expression on activated CD8+ T cells, and for the regulation of TRM effector functions through bidirectional integrin signaling. CD8+ TRM cells were initially described as a prognostic marker for survival in patients with various types of cancer, including ovarian, lung and breast cancers and melanoma. More recently, these tumor-resident CD8+ T cells have been shown to be a potent predictive biomarker of the response of cancer patients to immunotherapies, including therapeutic cancer vaccines and immune checkpoint blockade. In this review, we will highlight the major characteristics of tumor TRM cell populations and the possibilities for their exploitation in the design of more effective immunotherapy strategies for cancer.
Collapse
|
17
|
Wang R, Liu Z, Fan Z, Zhan H. Lipid metabolism reprogramming of CD8 + T cell and therapeutic implications in cancer. Cancer Lett 2023:216267. [PMID: 37315709 DOI: 10.1016/j.canlet.2023.216267] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Effector, memory and exhaustion are three phenotypes of CD8+ T cell. In tumor microenvironment (TME), metabolism dysfunction of the three should take the blame for immune escape. Against background of CD8+ T cell in normal development, multiple determinants in TME, including nutrition competition, PD-1 signals and other cancer- CD8+ T cell interaction, cause metabolism reprograming, including failure in energy metabolism and other abnormal lipid metabolism. Further, incompatibility of different CD8+ T cell metabolism pattern results in unresponsiveness of immune checkpoint blockade (ICB). Therefore, combination of ICB and drugs aiming at abnormal lipid metabolism provides promising direction to improve cancer therapy.
Collapse
Affiliation(s)
- Runxian Wang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Zhenya Liu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
18
|
Gavil NV, Scott MC, Weyu E, Smith OC, O’Flanagan SD, Wijeyesinghe S, Lotfi-Emran S, Shiao SL, Vezys V, Masopust D. Chronic antigen in solid tumors drives a distinct program of T cell residence. Sci Immunol 2023; 8:eadd5976. [PMID: 37267383 PMCID: PMC10569081 DOI: 10.1126/sciimmunol.add5976] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 05/10/2023] [Indexed: 06/04/2023]
Abstract
Analyses of healthy tissue reveal signatures that identify resident memory CD8+ T cells (TRM), which survey tissues without recirculating. The density of TRM phenotype cells within solid tumors correlates favorably with prognosis, suggesting that intratumoral residents control cancer. However, residence has not been directly tested, and intratumoral TRM phenotype cells could instead reflect aspects of the microenvironment that correlate with prognosis. Using a breast cancer model in mice, we found that conventional TRM markers do not inform the tumor residence of either bystander or tumor-specific cells, which exhibit further distinct phenotypes in the tumor microenvironment and healthy mammary tissue. Rather, tumor-specific, stem progenitor CD8+ T cells migrate to tumors and become resident while acquiring select markers of exhaustion. These data indicate that tonic antigen stimulation and the tumor environment drive distinct programs of residence compared with healthy tissues and that tumor immunity is sustained by continued migration of tumor-specific stem cells.
Collapse
Affiliation(s)
- Noah V. Gavil
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - Milcah C. Scott
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - Eyob Weyu
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - Olivia C. Smith
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - Stephen D. O’Flanagan
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - Sahar Lotfi-Emran
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - Stephen L. Shiao
- Department of Radiation Oncology, Cedars-Sinai Medical Center; Los Angeles, CA 90048, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota Medical School; Minneapolis, MN 55455, USA
| |
Collapse
|
19
|
Harnessing epithelial-mesenchymal plasticity to boost cancer immunotherapy. Cell Mol Immunol 2023; 20:318-340. [PMID: 36823234 PMCID: PMC10066239 DOI: 10.1038/s41423-023-00980-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/17/2023] [Indexed: 02/25/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy is a powerful option for cancer treatment. Despite demonstrable progress, most patients fail to respond or achieve durable responses due to primary or acquired ICB resistance. Recently, tumor epithelial-to-mesenchymal plasticity (EMP) was identified as a critical determinant in regulating immune escape and immunotherapy resistance in cancer. In this review, we summarize the emerging role of tumor EMP in ICB resistance and the tumor-intrinsic or extrinsic mechanisms by which tumors exploit EMP to achieve immunosuppression and immune escape. We discuss strategies to modulate tumor EMP to alleviate immune resistance and to enhance the efficiency of ICB therapy. Our discussion provides new prospects to enhance the ICB response for therapeutic gain in cancer patients.
Collapse
|
20
|
Isomoto K, Haratani K, Tsujikawa T, Makutani Y, Kawakami H, Takeda M, Yonesaka K, Tanaka K, Iwasa T, Hayashi H, Ito A, Nishio K, Nakagawa K. Mechanisms of primary and acquired resistance to immune checkpoint inhibitors in advanced non-small cell lung cancer: A multiplex immunohistochemistry-based single-cell analysis. Lung Cancer 2022; 174:71-82. [PMID: 36347190 DOI: 10.1016/j.lungcan.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/07/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Immune checkpoint inhibitors (ICIs) have become a key therapeutic modality for advanced non-small cell lung cancer (NSCLC), but most patients experience primary or acquired resistance to these drugs. We here explored the mechanisms underlying both types of ICI resistance by analysis of the tumor immune microenvironment (TME). MATERIALS AND METHODS Four patients who experienced a long-term response to ICI treatment (progression-free survival [PFS] of ≥12 months) followed by disease progression, after which a rebiopsy was immediately performed (cohort-A), as well as four patients who experienced early tumor progression during ICI treatment (PFS of <9 weeks, cohort-B) were enrolled in this retrospective study. The pretreatment TME was evaluated by 16- or 17-color multiplex immunohistochemistry (mIHC)-based spatial profiling at the single-cell level for both cohorts. In cohort-A, changes in the TME after disease progression during ICI treatment were also investigated by mIHC analysis and transcriptomic analysis. RESULTS Pretreatment tumor tissue from cohort-B manifested poor infiltration of tumor-reactive CD8+ T cells characterized by CD39 and CD103 expression or by programmed cell death-1 expression, implicating insufficient recognition of tumor cells by CD8+ T cells as a mechanism of primary ICI resistance. Analysis of the paired tumor specimens from cohort-A revealed various changes in the TME associated with acquired ICI resistance, including substantial infiltration of myeloid-derived suppressor cells and M2-type tumor-associated macrophages without a marked decline in the number of tumor-reactive CD8+ T cells; a decrease in the number of tumor-reactive CD8+ T cells; and an apparent decrease in neoantigen presentation by tumor cells. CONCLUSION The presence of intratumoral tumor-reactive CD8+ T cells may be a prerequisite for a long-term response to ICI treatment in advanced NSCLC, but it is not sufficient for cancer cell eradication. Various TME profiles are associated with acquired ICI resistance, suggesting that patient-specific strategies to overcome such resistance may be necessary.
Collapse
Affiliation(s)
- Kohsuke Isomoto
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Koji Haratani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan.
| | - Takahiro Tsujikawa
- Department of Otolaryngology-Head & Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Makutani
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kimio Yonesaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kaoru Tanaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tsutomu Iwasa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
21
|
NDR1 activates CD47 transcription by increasing protein stability and nuclear location of ASCL1 to enhance cancer stem cell properties and evasion of phagocytosis in small cell lung cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:254. [PMID: 36224405 DOI: 10.1007/s12032-022-01859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/21/2022] [Indexed: 10/17/2022]
Abstract
Small cell lung cancer (SCLC) is one of the most malignant types of lung cancer. Cancer stem cell (CSC) and tumor immune evasion are critical for the development of SCLC. We previously reported that NDR1 enhances breast CSC properties. NDR1 might also have a role in the regulation of immune responses. In the current study, we explore the function of NDR1 in the control of CSC properties and evasion of phagocytosis in SCLC. We find that NDR1 enhances the enrichment of the ALDEFLUORhigh and CD133high population, and promotes sphere formation in SCLC cells. Additionally, NDR1 upregulates CD47 expression to enhance evasion of phagocytosis in SCLC. Furthermore, the effects of NDR1 enhanced CD47 expression and evasion of phagocytosis are more prominent in CSC than in non-CSC. Importantly, NDR1 promotes ASCL1 expression to enhance NDR1-promoted CSC properties and evasion of phagocytosis in SCLC cells. Mechanically, NDR1 enhances protein stability and the nuclear location of ASCL1 to activate the transcription of CD47 in SCLC. Finally, CD47-blocking antibody can be used to target NDR1 enhanced CSC properties and evasion of phagocytosis by suppressing EGFR activation in SCLC. In summary, our data indicate that NDR1 could be a critical factor for modulating CSC properties and phagocytosis in SCLC.
Collapse
|
22
|
Sharma A, Blériot C, Currenti J, Ginhoux F. Oncofetal reprogramming in tumour development and progression. Nat Rev Cancer 2022; 22:593-602. [PMID: 35999292 DOI: 10.1038/s41568-022-00497-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 12/12/2022]
Abstract
Embryonic development is characterized by rapidly dividing cells, cellular plasticity and a highly vascular microenvironment. These features are similar to those of tumour tissue, in that malignant cells are characterized by their ability to proliferate and exhibit cellular plasticity. The tumour microenvironment also often includes immunosuppressive features. Reciprocal communication between various cellular subpopulations enables fetal and tumour tissues to proliferate, migrate and escape immune responses. Fetal-like reprogramming has been demonstrated in the tumour microenvironment, indicating extraordinary cellular plasticity and bringing an additional layer of cellular heterogeneity. More importantly, some of these features are also present during inflammation. This Perspective discusses the similarity between embryogenesis, inflammation and tumorigenesis, and describes the mechanisms of oncofetal reprogramming that enable tumour cells to escape from immune responses, promoting tumour growth and metastasis.
Collapse
Affiliation(s)
- Ankur Sharma
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia.
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia.
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia.
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | | | - Jennifer Currenti
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Florent Ginhoux
- INSERM U1015, Institut Gustave Roussy, Villejuif, France.
- Singapore Immunology Network, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
23
|
Liao W, Li Y, Wang J, Zhao M, Chen N, Zheng Q, Wan L, Mou Y, Tang J, Wang Z. Natural Products-Based Nanoformulations: A New Approach Targeting CSCs to Cancer Therapy. Int J Nanomedicine 2022; 17:4163-4193. [PMID: 36134202 PMCID: PMC9482958 DOI: 10.2147/ijn.s380697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer stem cells (CSCs) lead to the occurrence and progression of cancer due to their strong tumorigenic, self-renewal, and multidirectional differentiation abilities. Existing cancer treatment methods cannot effectively kill or inhibit CSCs but instead enrich them and produce stronger proliferation, invasion, and metastasis capabilities, resulting in cancer recurrence and treatment resistance, which has become a difficult problem in clinical treatment. Therefore, targeting CSCs may be the most promising approach for comprehensive cancer therapy in the future. A variety of natural products (NP) have significant antitumor effects and have been identified to target and inhibit CSCs. However, pharmacokinetic defects and off-target effects have greatly hindered their clinical translation. NP-based nanoformulations (NPNs) have tremendous potential to overcome the disadvantages of NP against CSCs through site-specific delivery and by improving their pharmacokinetic parameters. In this review, we summarize the recent progress of NPNs targeting CSCs in cancer therapy, looking forward to transforming preclinical research results into clinical applications and bringing new prospects for cancer treatment.
Collapse
Affiliation(s)
- Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Yuchen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Bishan Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Maoyuan Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Nianzhi Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Qiao Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Lina Wan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Yu Mou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.,TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Zhilei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.,TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| |
Collapse
|