1
|
Gao S, Liu M, Zhang Y, He Z, Li Y, Ji J, Ye L, Yang X, Zhai G. A precision intelligent nanomissile for inhibiting tumor metastasis, boosting energy deprivation and immunotherapy. Biomaterials 2025; 315:122953. [PMID: 39531747 DOI: 10.1016/j.biomaterials.2024.122953] [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: 06/19/2024] [Revised: 10/19/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
The epithelial-mesenchymal transition (EMT), tumor stroma and local metabolic alterations cooperate to establish a unique tumor microenvironment (TME) that fosters tumor progression and metastasis. To tackle this challenge, a precision intelligent nanomissile named HA@AT-Pd has been designed for dual-pronged cancer-associated fibroblast (CAF) transformation and tumor cell elimination. It is observed that HA@AT-Pd inhibits the production of cancer stem cells (CSCs) by blocking the TGF-β/Smad signaling pathway-mediated EMT and reversing activated CAFs to quiescence. Notably, HA@AT-Pd induces energy depletion in breast cancer cells through simultaneously suppressing cellular oxidative phosphorylation and glycolysis. The inhibition of glycolysis results in reduced lactic acid production, thereby converting an immunosuppressive TME into an immune-activating environment. Furthermore, the photothermal effect generated by HA@AT-Pd evokes immunogenic cell death, which can further enhance the anti-tumor immune response. Overall, this multifunctional combination strategy unveils potential therapeutic avenues to inhibit tumor progression and metastasis.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Department of Clinical Research, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Meng Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yu Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhijing He
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yingying Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
2
|
Wu NC, Quevedo R, Nurse M, Hezaveh K, Liu H, Sun F, Muffat J, Sun Y, Simmons CA, McGaha TL, Prinos P, Arrowsmith CH, Ailles L, D'Arcangelo E, McGuigan AP. The use of a multi-metric readout screen to identify EHMT2/G9a-inhibition as a modulator of cancer-associated fibroblast activation state. Biomaterials 2025; 314:122879. [PMID: 39395244 DOI: 10.1016/j.biomaterials.2024.122879] [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: 01/11/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a pivotal role in cancer progression, including mediating tumour cell invasion via their pro-invasive secretory profile and ability to remodel the extracellular matrix (ECM). Given that reduced CAF abundance in tumours correlates with improved outcomes in various cancers, we set out to identify epigenetic targets involved in CAF activation in regions of tumour-stromal mixing with the goal of reducing tumour aggressiveness. Using the GLAnCE (Gels for Live Analysis of Compartmentalized Environments) platform, we performed an image-based, phenotypic screen that enabled us to identify modulators of CAF abundance and the capacity of CAFs to induce tumour cell invasion. We identified EHMT2 (also known as G9a), an enzyme that targets the methylation of histone 3 lysine 9 (H3K9), as a potent modulator of CAF abundance and CAF-mediated tumour cell invasion. Transcriptomic and functional analysis of EHMT2-inhibited CAFs revealed EHMT2 participated in driving CAFs towards a pro-invasive phenotype and mediated CAF hyperproliferation, a feature typically associated with activated fibroblasts in tumours. Our study suggests that EHMT2 regulates CAF state within the tumour microenvironment by impacting CAF activation, as well as by magnifying the pro-invasive effects of these activated CAFs on tumour cell invasion through promoting CAF hyperproliferation.
Collapse
Affiliation(s)
- Nila C Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rene Quevedo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Michelle Nurse
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Kebria Hezaveh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Haijiao Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Fumao Sun
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Hospital for Sick Children, Toronto, ON, Canada
| | - Julien Muffat
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Hospital for Sick Children, Toronto, ON, Canada
| | - Yu Sun
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Tracy L McGaha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Panagiotis Prinos
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Cheryl H Arrowsmith
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Elisa D'Arcangelo
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
| | - Alison P McGuigan
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Wan W, Zhu K, Ran Z, Zhu X, Wang D. Development of a Nomogram-Integrated Model Incorporating Intra-tumoral and Peri-tumoral Ultrasound Radiomics Alongside Clinical Parameters for the Prediction of Histological Grading in Invasive Breast Cancer. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:262-272. [PMID: 39477745 DOI: 10.1016/j.ultrasmedbio.2024.09.025] [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: 07/11/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE To develop a comprehensive nomogram to predict the histological grading of breast cancer and further examine its clinical significance by integrating both intra-tumoral and peri-tumoral ultrasound radiomics features. METHODS In a retrospective study 468 female breast cancer patients were analyzed from 2017 to 2020 at the Second Affiliated Hospital of Harbin Medical University. Patients were grouped into high-grade (n = 215) and low-grade (n = 253) categories based on pathological evaluation. Tumor regions of interest were defined and expanded automatically to peri-tumor regions of interest. Ultrasound radiomics features were extracted independently. To ensure rigor, cases were randomly divided into 80% training and 20% test sets. Optimal features were selected using statistical and machine learning methods. Intra-tumor, peri-tumor, and combined radiomics models were constructed. To determine the best predictors of breast cancer histological grading, we screened the features using single- and multi-factor logistic regression analyses. Finally, a nomogram was developed and evaluated for its predictive value in this context. RESULTS By applying logistic regression, we integrated ultrasound, clinicopathologic, and radiomics features to generate a nomogram. The combined model outperformed others, achieving areas under the curve of 0.934 and 0.812 in training and test sets. Calibration curves also showed high accuracy and reliability. CONCLUSION A nomogram constructed through the integration of combined intra-tumor-peri-tumor ultrasound radiomics features along with clinicopathologic characteristics exhibited remarkable performance in distinguishing the histologic grades of invasive breast cancer.
Collapse
Affiliation(s)
- Wenjia Wan
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Kai Zhu
- Radiology Department, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhicheng Ran
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xinyu Zhu
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Dongmo Wang
- Ultrasound Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
4
|
He S, Hao L, Chen Y, Gong B, Xu X. Chinese herbal Jianpi Jiedu formula suppressed colorectal cancer growth in vitro and in vivo via modulating hypoxia-inducible factor 1 alpha-mediated fibroblasts activation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118753. [PMID: 39209001 DOI: 10.1016/j.jep.2024.118753] [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: 07/04/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jianpi Jiedu Formula (JPJDF) is a traditional Chinese medicinal decoction clinically used for its anti-cancer properties, particularly in colorectal cancer (CRC). AIM OF THE STUDY This study aims to investigate the therapeutic effects of JPJDF on CRC and elucidate its potential molecular mechanisms, with a focus on its impact on hypoxia-inducible factor 1 alpha (HIF1α) and cancer-associated fibroblasts (CAFs) both in vitro and in vivo. MATERIALS AND METHODS UPLC-Q-TOF-MS was used to identify the constituents of JPJDF. A chemical-induced colorectal cancer model was established and treated with JPJDF to evaluate its effects. Tumor size was measured, and histopathological analyses were performed to examine JPJDF's regulatory potential on CRC. The functional mechanism of JPJDF was predicted through network pharmacology, molecular docking, and transcriptomics. Co-culture techniques involving CRC cells and CCD-18Co fibroblasts were used to assess JPJDF's impact on fibroblast activation. The effects of HIF1α on CAFs were evaluated using CCK-8 proliferation, clonal formation, and apoptotic assays, with differential marker expression quantified via qPCR and Western blotting. RESULTS Pharmacodynamic assessment demonstrated that JPJDF reduced tumor size without affecting body weight, indicating its safety in the chemical-induced murine CRC model. Network pharmacology analysis, combined with molecular docking and transcriptomics, revealed that JPJDF regulates HIF-1 signaling pathways and identified HIF1α as a potential target for JPJDF's anti-CRC effect. JPJDF effectively suppressed CRC growth in vivo by attenuating fibroblast activation, reducing α-SMA expression and POSTN secretion through HIF1α inhibition. HIF1α knockdown in CRC cells inhibited fibroblast proliferation and clonal formation, while overexpression promoted these processes. Additionally, downregulating HIF1α suppressed α-SMA and POSTN expression in fibroblasts, whereas overexpression enhanced fibroblast activation. CONCLUSION JPJDF emerges as a promising therapeutic candidate for inhibiting CAFs activation by targeting HIF1α, offering potential avenues for modulating fibroblast activation towards CAFs in CRC therapy.
Collapse
Affiliation(s)
- Shenglan He
- Department of Digestive Endoscopy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lixiao Hao
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Youlan Chen
- Institute of Integrated Traditional Chinese and Western Medicine Digestive Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Biao Gong
- Department of Digestive Endoscopy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xiaowen Xu
- Department of Digestive Endoscopy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
5
|
Mai RY, Ye JZ, Gao X, Wen T, Li SZ, Zeng C, Cen WJ, Wu GB, Lin Y, Liang R, Luo XL. Up-regulated ITGB4 promotes hepatocellular carcinoma metastasis by activating hypoxia-mediated glycolysis and cancer-associated fibroblasts. Eur J Pharmacol 2025; 986:177102. [PMID: 39603378 DOI: 10.1016/j.ejphar.2024.177102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
The pre-metastatic niche constructed by cancer-associated fibroblasts (CAFs) plays a key role in the hypoxic tumor microenvironment (TME), promoting hepatocellular carcinoma (HCC) metastasis. Integrin, which is involved in cell-to-cell or cell-to-matrix interactions and TME regulation, affects tumor metastasis. However, the complex interactions between integrin-mediated HCC cells and CAFs remain unclear. Co-culture experiments were used to assess the behaviors of HCC cells and CAFs, demonstrating HCC metastatic traits and CAFs activation in vitro. Transcriptome sequencing analysis and molecular detection identified key genes, with overexpression and knockdown experiments further confirming their roles in HCC progression. Xenograft models validated these findings in vivo. We showed that HCC cells induced the conversion of normal hepatic stellate cells (HSCs) into CAFs and recruit additional CAFs, driven by lactate produced by HCC. Integrin beta 4 (ITGB4) was identified as a key gene in the process. Inhibiting ITGB4 reduced lactate secretion, reversed CAFs activation and recruitment, and decreased HCC metastasis, while overexpressing ITGB4 significantly enhanced these malignant phenotypes. ITGB4 influences glycolysis and HCC metastasis through the AKT/HK2 signaling pathway, and CAFs activation and recruitment through the TGF-β/Smads signaling pathway. Compared to tumors derived from control cells, ITGB4-knockdown tumors showed fewer and smaller intrahepatic metastatic nodules, reduced lactate production and CAFs formation, along with inhibition of AKT/HK2 and TGF-β/Smads signaling pathways. Our findings highlighted the impact of hypoxia on HCC progression, revealing the roles of ITGB4-mediated glycolysis and lactate-induced CAFs in the pre-metastatic niche on HCC metastasis.
Collapse
Affiliation(s)
- Rong-Yun Mai
- Department of Hepatobilliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China; Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Jia-Zhou Ye
- Department of Hepatobilliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Xing Gao
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China; Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Tong Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Shi-Zhou Li
- Department of Hepatobilliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China; Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Can Zeng
- Department of Hepatobilliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China; Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Wei-Jie Cen
- Department of Hepatobilliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Guo-Bin Wu
- Department of Hepatobilliary & Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Yan Lin
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| | - Rong Liang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| | - Xiao-Ling Luo
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| |
Collapse
|
6
|
Sun D, Lu J, Tian H, Li H, Chen X, Hua F, Yang W, Yu J, Chen D. The impact of POSTN on tumor cell behavior and the tumor microenvironment in lung adenocarcinoma. Int Immunopharmacol 2025; 145:113713. [PMID: 39672019 DOI: 10.1016/j.intimp.2024.113713] [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: 03/20/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND The role of cancer-associated fibroblasts (CAFs) in modulating the anti-tumor immune response in lung adenocarcinoma (LUAD) remains elusive, primarily due to the heterogeneous nature of these cells. This heterogeneity muddles the understanding of their impact on immunotherapy effectiveness. METHODS We utilized the LUAD single-cell dataset to precisely classify tumor cells and CAFs. By employing CSOmap, we predicted cell interactions and reconstructed the three-dimensional spatial organization, highlighting the close association of myofibroblasts with specific tumor cell subsets. A prognostic signature based on myofibroblast-specific genes was developed and validated to predict LUAD patient survival. In vivo, we conducted subcutaneous tumorigenesis assays in mice, treating with PD-L1 and the POSTN inhibitor RGD to assess the combined effects of POSTN pathway blockade and immunotherapy on tumor growth and immune cell dynamics. For analyzing the tumor microenvironment, we used flow cytometry and multiplex immunofluorescence staining. In vitro, with cell lines like A549, H1299, and RAW264.7, we investigated POSTN's role in macrophage recruitment and polarization. Through ELISA, Western blot, and immunofluorescence staining, we explored how POSTN acts via ITGB3, providing a more comprehensive understanding of its mechanism in LUAD. RESULTS Our analysis discerned six distinct tumor cell subsets, with cluster 1 displaying pronounced cellular communication with myofibroblasts, evidenced by spatial accessibility in three dimensions. The myofibroblast-specific genomic signature was established and confirmed as a robust, independent prognostic indicator. Among the signature genes, CTHRC1, POSTN, and MMP11 emerged as high-variant genes in myofibroblasts, identified via the FindAllMarkers function in Seurat. Of these, only POSTN's differential expression correlated with LUAD prognosis, with high POSTN expression being indicative of poor patient outcomes. In vitro, recombinant POSTN was observed to enhance tumor invasiveness, motility, and proliferation, while attenuating apoptosis and fostering an EMT phenotype. Additionally, Transwell assays showed that rPOSTN could induce macrophage infiltration via ITGB3 and drive M2 polarization via the PI3K-Akt-JNK pathway. Importantly, blocking the POSTN pathway augmented the efficacy of PD-L1 inhibitors. In vivo, in a mouse subcutaneous tumorigenesis model, the combination of POSTN pathway blockade with PD-L1 inhibitor treatment notably inhibited tumor growth and changed the tumor microenvironment's immune cell composition, with an increase in CD8+ T cells and a favorable shift in the M1/M2 macrophage ratio. CONCLUSION This study sheds light on the intricate interplay between tumor cells and myofibroblasts in LUAD, pinpointing the pivotal role of the highly mutated gene POSTN. It underscores POSTN's instrumental role in manipulating the tumor microenvironment, primarily by promoting EMT and inhibiting apoptosis in lung cancer cells, alongside enhancing macrophage recruitment and fostering M2 polarization. These insights provide a foundation for enriching immunotherapy strategies, particularly through the inhibition of the POSTN pathway in LUAD.
Collapse
Affiliation(s)
- Dongfeng Sun
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Jie Lu
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Hui Tian
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hao Li
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China; Interventional Department, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xiaozheng Chen
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Feng Hua
- Department of Thoracic Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, China
| | - Wenfeng Yang
- Department of Thoracic Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, China.
| | - Jinming Yu
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China; Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong, China.
| | - Dawei Chen
- Department of Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China; Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong, China.
| |
Collapse
|
7
|
Hussen BM, Abdullah SR, Jaafar RM, Rasul MF, Aroutiounian R, Harutyunyan T, Liehr T, Samsami M, Taheri M. Circular RNAs as Key Regulators in Cancer Hallmarks: New Progress and Therapeutic Opportunities. Crit Rev Oncol Hematol 2025:104612. [PMID: 39755160 DOI: 10.1016/j.critrevonc.2024.104612] [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: 09/28/2024] [Revised: 12/22/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
Circular RNAs (circRNAs) have emerged as critical regulators in cancer biology, contributing to various cancer hallmarks, including cell proliferation, apoptosis, metastasis, and drug resistance. Defined by their covalently closed loop structure, circRNAs possess unique characteristics like high stability, abundance, and tissue-specific expression. These non-coding RNAs function through mechanisms such as miRNA sponging, interactions with RNA-binding proteins (RBPs), and modulating transcription and splicing. Advances in RNA sequencing and bioinformatics tools have enabled the identification and functional annotation of circRNAs across different cancer types. Clinically, circRNAs demonstrate high specificity and sensitivity in samples, offering potential as diagnostic and prognostic biomarkers. Additionally, therapeutic strategies involving circRNA mimics, inhibitors, and delivery systems are under investigation. However, their precise mechanisms remain unclear, and more clinical evidence is needed regarding their roles in cancer hallmarks. Understanding circRNAs will pave the way for novel diagnostic and therapeutic approaches, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, Iraq; Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Snur Rasool Abdullah
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Rayan Mazin Jaafar
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Rouben Aroutiounian
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, 0025 Yerevan, Armenia; Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, 0025 Yerevan, Armenia
| | - Tigran Harutyunyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, 0025 Yerevan, Armenia; Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, 0025 Yerevan, Armenia
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| | - Majid Samsami
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Research Institute for Urology and Nephrology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Gao B, Gou X, Feng C, Zhang Y, Gu H, Chai F, Wang Y, Ye Y, Hong N, Hu G, Sun B, Cheng J, Yang H. Identification of cancer-associated fibrolast subtypes and distinctive role of MFAP5 in CT-detected extramural venous invasion in gastric cancer. Transl Oncol 2025; 51:102188. [PMID: 39531783 PMCID: PMC11600027 DOI: 10.1016/j.tranon.2024.102188] [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: 07/19/2024] [Revised: 09/26/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Extramural venous invasion (EMVI) detected by computed tomography has been identified as an independent risk factor for distant metastasis in patients with advanced gastric cancer (GC). Cancer-associated fibroblasts (CAFs) are critical for remodeling the tumor microenvironment in GCs. Here, we report that MFAP5+ CAFs promote the formation of EMVI imaging in GC. We detected gene expression in pathological samples from 13 advanced GC patients with EMVI. Radiogenomics results showed the degree of CAFs infiltration was directly proportional to the EMVI score and EMT pathway in GC patients. Single-cell sequencing data analysis results showed that MFAP5+CAFs subtypes in GC were negatively correlated with patient prognosis and were enriched in tumor lactylation modification and EMT pathways. Immunohistochemistry results showed that the expression of MFAP5, L-lactyl and EMT markers in GC tissues was proportional to the EMVI score. CAF from gastric cancer tissue was extracted using collagenase method and co-cultured with GC cell line in vitro. After lentivirus knockdown of MFAP5 in CAFs, the levels of L-lactoyl and histone lactylation modifications were significantly reduced, and the sphere-forming and vascularization abilities of CAFs were significantly inhibited. Cell function experiments showed that MFAP5+ CAFs can affect the EMT, metastasis and invasion capabilities of GC cells. In vivo experimental results of the nude mouse in situ EMVI model suggest that MFAP5+ CAF may promote the formation of EMVI imaging features in GC by regulating lactylation modification. This innovative work may provide important new references for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Bo Gao
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Xinyi Gou
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Caizhen Feng
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yinli Zhang
- Department of Pathology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Huining Gu
- Department of Immunology, School of Basic Medical Sciences, Peking University and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Guohua Hu
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Peking University Health Science Center, Beijing, China
| | - Boshi Sun
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Peking University Health Science Center, Beijing, China.
| | - Hao Yang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
9
|
Zhang X, Zhang M, Sun H, Wang X, Wang X, Sheng W, Xu M. The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells. J Adv Res 2025; 67:121-132. [PMID: 38309692 DOI: 10.1016/j.jare.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Transcription factors (TFs) fulfill a critical role in the formation and maintenance of different cell types during the developmental process as well as disease. It is believed that cancer-associated fibroblasts (CAFs) are activation status of tissue-resident fibroblasts or derived from form other cell types via transdifferentiation or dedifferentiation. Despite a subgroup of CAFs exhibit anti-cancer effects, most of them are reported to exert effects on tumor progression, further indicating their heterogeneous origin. AIM OF REVIEW This review aimed to summarize and review the roles of TFs in the reciprocal crosstalk between CAFs and tumor cells, discuss the emerging mechanisms, and their roles in cell-fate decision, cellular reprogramming and advancing our understanding of the gene regulatory networks over the period of cancer initiation and progression. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript delves into the key contributory factors of TFs that are involved in activating CAFs and maintaining their unique states. Additionally, it explores how TFs play a pivotal and multifaceted role in the reciprocal crosstalk between CAFs and tumor cells. This includes their involvement in processes such as epithelial-mesenchymal transition (EMT), proliferation, invasion, and metastasis, as well as metabolic reprogramming. TFs also have a role in constructing an immunosuppressive microenvironment, inducing resistance to radiation and chemotherapy, facilitating angiogenesis, and even 'educating' CAFs to support the malignancies of tumor cells. Furthermore, this manuscript delves into the current status of TF-targeted therapy and considers the future directions of TFs in conjunction with anti-CAFs therapies to address the challenges in clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
10
|
Wu S, Fang R, Rietveld MH, Torremans JRG, Liu Y, Gu Z, Bouwes Bavinck JN, Vermeer MH, El Ghalbzouri A. Identification of Small-Molecule Inhibitors Targeting Different Signaling Pathways in Cancer-Associated Fibroblast Reprogramming under Tumor-Stroma Interaction. J Invest Dermatol 2025; 145:65-76.e13. [PMID: 38848988 DOI: 10.1016/j.jid.2024.04.026] [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: 08/29/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 06/09/2024]
Abstract
Cancer-associated fibroblasts (CAFs) interact reciprocally with tumor cells through various signaling pathways in many cancer types, including cutaneous squamous cell carcinoma. Among normal fibroblast subtypes, papillary fibroblasts (PFs) and reticular fibroblasts (RFs) respond distinctly to tumor cell signaling, eventuating the differentiation of RFs rather than PFs into CAFs. The regulation of subtype differentiation in fibroblasts remains poorly explored. In this study, we assessed the differences between PFs, RFs, and CAFs and examined the effects of small-molecule inhibitors targeting the TGFβ, phosphoinositide 3-kinase/protein kinase B/mTOR, and NOTCH pathways on the tumor-promoting property of CAFs and CAF reprogramming in 2-dimensional and 3-dimensional cultures. Blocking TGFβ and phosphoinositide 3-kinase strongly deactivated and concurrently induced a PF phenotype in RFs and CAFs. Three-dimensional coculturing of a cutaneous squamous cell carcinoma cell line MET2 with RFs or CAFs led to enhanced tumor invasion, RF-CAF transition, and cytokine production, which were further repressed by blocking TGFβ and phosphoinositide 3-kinase/mTOR pathways but not NOTCH pathway. In conclusion, the study identified biomarkers for PFs, RFs, and CAFs and displayed different effects of blocking key signaling pathways in CAFs and tumor cell-CAF interplay. These findings prompted a CAF-to-PF therapeutic strategy and provided perspectives of using included inhibitors in CAF-based cancer therapy.
Collapse
Affiliation(s)
- Shidi Wu
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rui Fang
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK Partner Site Essen) and German Cancer Research Center, Heidelberg, Germany
| | - Marion H Rietveld
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen R G Torremans
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yang Liu
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Zili Gu
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan N Bouwes Bavinck
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarten H Vermeer
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
11
|
Zeng Q, Zhang S, Leng N, Xing Y. Advancing tumor vaccines: Overcoming TME challenges, delivery strategies, and biomaterial-based vaccine for enhanced immunotherapy. Crit Rev Oncol Hematol 2025; 205:104576. [PMID: 39581246 DOI: 10.1016/j.critrevonc.2024.104576] [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: 10/08/2024] [Revised: 11/03/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024] Open
Abstract
Tumor vaccines, as an immunotherapeutic approach, harness the body's immune cells to provoke antitumor responses, which have shown promising efficacy in clinical settings. However, the immunosuppressive tumor microenvironment (TME) and the ineffective vaccine delivery systems hinder the progression of many vaccines beyond phase II trials. This article begins with a comprehensive review of the complex interactions between tumor vaccines and TME, summarizing the current state of vaccine clinical research. Subsequently, we review recent advancements in targeted vaccine delivery systems and explore biomaterial-based tumor vaccines as a strategy to improve the efficacy of both delivery systems and treatment. Finally, we have presented our perspectives on tumor vaccine development, aiming to advance the field towards the creation of more effective tumor vaccines.
Collapse
Affiliation(s)
- Qingsong Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Shibo Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Ning Leng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
12
|
Acimovic I, Gabrielová V, Martínková S, Eid M, Vlažný J, Moravčík P, Hlavsa J, Moráň L, Cakmakci RC, Staňo P, Procházka V, Kala Z, Trnka J, Vaňhara P. Ex-Vivo 3D Cellular Models of Pancreatic Ductal Adenocarcinoma: From Embryonic Development to Precision Oncology. Pancreas 2025; 54:e57-e71. [PMID: 39074056 DOI: 10.1097/mpa.0000000000002393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
ABSTRACT Pancreas is a vital gland of gastrointestinal system with exocrine and endocrine secretory functions, interweaved into essential metabolic circuitries of the human body. Pancreatic ductal adenocarcinoma (PDAC) represents one of the most lethal malignancies, with a 5-year survival rate of 11%. This poor prognosis is primarily attributed to the absence of early symptoms, rapid metastatic dissemination, and the limited efficacy of current therapeutic interventions. Despite recent advancements in understanding the etiopathogenesis and treatment of PDAC, there remains a pressing need for improved individualized models, identification of novel molecular targets, and development of unbiased predictors of disease progression. Here we aim to explore the concept of precision medicine utilizing 3-dimensional, patient-specific cellular models of pancreatic tumors and discuss their potential applications in uncovering novel druggable molecular targets and predicting clinical parameters for individual patients.
Collapse
Affiliation(s)
- Ivana Acimovic
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Viktorie Gabrielová
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Stanislava Martínková
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | - Michal Eid
- Departments of Internal Medicine, Hematology and Oncology
| | | | - Petr Moravčík
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Hlavsa
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | | | - Riza Can Cakmakci
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Peter Staňo
- Departments of Internal Medicine, Hematology and Oncology
| | - Vladimír Procházka
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Zdeněk Kala
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Trnka
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | | |
Collapse
|
13
|
Jain SM, Nagainallur Ravichandran S, Murali Kumar M, Banerjee A, Sun-Zhang A, Zhang H, Pathak R, Sun XF, Pathak S. Understanding the molecular mechanism responsible for developing therapeutic radiation-induced radioresistance of rectal cancer and improving the clinical outcomes of radiotherapy - A review. Cancer Biol Ther 2024; 25:2317999. [PMID: 38445632 PMCID: PMC10936619 DOI: 10.1080/15384047.2024.2317999] [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: 08/16/2023] [Accepted: 02/08/2024] [Indexed: 03/07/2024] Open
Abstract
Rectal cancer accounts for the second highest cancer-related mortality, which is predominant in Western civilizations. The treatment for rectal cancers includes surgery, radiotherapy, chemotherapy, and immunotherapy. Radiotherapy, specifically external beam radiation therapy, is the most common way to treat rectal cancer because radiation not only limits cancer progression but also significantly reduces the risk of local recurrence. However, therapeutic radiation-induced radioresistance to rectal cancer cells and toxicity to normal tissues are major drawbacks. Therefore, understanding the mechanistic basis of developing radioresistance during and after radiation therapy would provide crucial insight to improve clinical outcomes of radiation therapy for rectal cancer patients. Studies by various groups have shown that radiotherapy-mediated changes in the tumor microenvironment play a crucial role in developing radioresistance. Therapeutic radiation-induced hypoxia and functional alterations in the stromal cells, specifically tumor-associated macrophage (TAM) and cancer-associated fibroblasts (CAF), play a crucial role in developing radioresistance. In addition, signaling pathways, such as - the PI3K/AKT pathway, Wnt/β-catenin signaling, and the hippo pathway, modulate the radiation responsiveness of cancer cells. Different radiosensitizers, such as small molecules, microRNA, nanomaterials, and natural and chemical sensitizers, are being used to increase the effectiveness of radiotherapy. This review highlights the mechanism responsible for developing radioresistance of rectal cancer following radiotherapy and potential strategies to enhance the effectiveness of radiotherapy for better management of rectal cancer.
Collapse
Affiliation(s)
- Samatha M Jain
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Shruthi Nagainallur Ravichandran
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Makalakshmi Murali Kumar
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Alexander Sun-Zhang
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Hong Zhang
- School of Medicine, Department of Medical Sciences, Orebro University, Örebro, Sweden
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| |
Collapse
|
14
|
Yu Y, Tao Y, Ma J, Li J, Song Z. Targeting the tumor microenvironment with mesenchymal stem cells based delivery approach for efficient delivery of anticancer agents: An updated review. Biochem Pharmacol 2024; 232:116725. [PMID: 39746456 DOI: 10.1016/j.bcp.2024.116725] [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: 09/02/2024] [Revised: 11/14/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Drug delivery to cancer cells continues to present a major therapeutic challenge. Mesenchymal stem cells (MSCs) possess an intrinsic ability to migrate specifically to tumor tissues, making them promising candidates for targeted drug delivery. Evidence from preclinical studies indicates that MSCs loaded with therapeutic anti-cancer agents exhibit considerable anti-tumor activity. Moreover, several clinical trials are currently evaluating their effectiveness in cancer patients. The integration of MSCs with synthetic nanoparticles (NPs) enhances their therapeutic potential, particularly through the use of cell membrane-coated NPs, which represent a significant advancement in the field. This review systematically investigates the tumor microenvironment, the sources of MSCs, the tumor homing mechanisms, and the methods of loading and releasing anticancer drugs from MSCs. Furthermore, cutting-edge strategies to improve the efficacy of MSCs based drug delivery systems (DDS) including the innovative use of MSC membrane coated nanoparticles have been discussed. The study concludes with an overview of the therapeutic use of MSCs as drug carriers, including a detailed analysis of the mechanisms by which MSCs deliver therapeutics to cancer cells, enabling targeted drug delivery. It aims to elucidate the current state of this approach, identify key areas for development, and outline potential future directions for advancing MSCs based cancer therapies.
Collapse
Affiliation(s)
- Yang Yu
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Jian Li
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Zhidu Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
15
|
Zhang X, Lee KC, Choi JY, Lee KH, Choe YS. Comparative Study of Dimeric Fibroblast Activation Protein-Targeting Radioligands Labeled with Fluorine-18, Copper-64, and Gallium-68. Mol Pharm 2024. [PMID: 39736080 DOI: 10.1021/acs.molpharmaceut.4c01080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025]
Abstract
Fibroblast activation protein inhibitors (FAPIs) labeled with gallium-68 and lutetium-177 show potential for use in the diagnosis and treatment of various cancers expressing FAP. However, 177Lu-labeled FAPIs often exhibit short tumor retention time, limiting their therapeutic applications. To improve tumor retention, we synthesized three radiolabeled dimeric FAPIs, [18F]1, [64Cu]2, and [68Ga]3. These were prepared by chelating Al[18F]F to 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA)-l-glutamic acid (E)-(FAPI)2 and copper-64 or gallium-68 to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-E-(FAPI)2. NOTA-E-(FAPI)2 and DOTA-E-(FAPI)2 showed higher binding affinities for FAP compared with that of FAPI-04 (IC50 = 0.47 and 0.16 nM vs 0.89 nM, respectively). All radioligands were synthesized in high decay-corrected radiochemical yields (59-96%) and were stable in fetal bovine serum and phosphate-buffered saline. The more hydrophilic radioligand, [68Ga]3, was selected for cellular uptake studies, which confirmed FAP-specific uptake. Positron emission tomography imaging and ex vivo biodistribution studies in U87MG tumor-bearing mice revealed high tumor uptake of all three radioligands, with significant blocking observed after preinjection of FAPI-04. [64Cu]2 and [68Ga]3 exhibited favorable in vivo pharmacokinetics compared to those of [18F]1. Notably, [68Ga]3 showed lower normal organ uptake than did the other two radioligands, and moreover, it exhibited higher, more prolonged tumor uptake than its monomeric counterpart [68Ga]Ga-FAPI-04 over a 3 h period, suggesting its potential as a promising FAP-specific theranostic radioligand.
Collapse
Affiliation(s)
- Xuran Zhang
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Yearn Seong Choe
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| |
Collapse
|
16
|
Feng C, Wang Y, Song W, Liu T, Mo H, Liu H, Wu S, Qin Z, Wang Z, Tao Y, He L, Tang S, Xie Y, Wang Q, Li T. Spatially-resolved analyses of muscle invasive bladder cancer microenvironment unveil a distinct fibroblast cluster associated with prognosis. Front Immunol 2024; 15:1522582. [PMID: 39759522 PMCID: PMC11695344 DOI: 10.3389/fimmu.2024.1522582] [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: 11/04/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
Background Muscle-invasive bladder cancer (MIBC) is a prevalent cancer characterized by molecular and clinical heterogeneity. Assessing the spatial heterogeneity of the MIBC microenvironment is crucial to understand its clinical significance. Methods In this study, we used imaging mass cytometry (IMC) to assess the spatial heterogeneity of MIBC microenvironment across 185 regions of interest in 40 tissue samples. We focused on three primary parameters: tumor (T), leading-edge (L), and nontumor (N). Cell gating was performed using the Cytobank platform. We calculated the Euclidean distances between cells to determine cellular interactions and performed single-cell RNA sequencing (scRNA-seq) to explore the molecular characteristics and mechanisms underlying specific fibroblast (FB) clusters. scRNA-seq combined with spatial transcriptomics (ST) facilitated the identification of ligand-receptor (L-R) pairs that mediate interactions between specific FB clusters and endothelial cells. Machine learning algorithms were used to construct a prognostic gene signature. Results The microenvironments in the N, L, and T regions of MIBC exhibited spatial heterogeneity and regional diversity in their components. A distinct FB cluster located in the L region-identified as S3-is strongly associated with poor prognosis. IMC analyses demonstrated a close spatial association between S3 and endothelial cells, with S3-positive tumors exhibiting increased blood vessel density and altered vascular morphology. The expression of vascular endothelial growth factor receptor and active vascular sprouting were significant in S3-positive tumors. scRNA-seq and ST analyses indicated that the genes upregulated in S3 were associated with angiogenesis. NOTCH1-JAG2 signaling pathway was identified as a significant L-R pair specific to S3 and endothelial cell interactions. Further analysis indicated that YAP1 was a potential regulator of S3. Machine learning algorithms and Gene Set Variation Analysis were used to establish an S3-related gene signature that was associated with the poor prognosis of tumors including MIBC, mesothelioma, glioblastoma multiforme, lower-grade glioma, stomach adenocarcinoma, uveal melanoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, and lung squamous cell carcinoma. Conclusions We assessed the spatial landscape of the MIBC microenvironment and revealed a specific FB cluster with prognostic potential. These findings offer novel insights into the spatial heterogeneity of the MIBC microenvironment and highlight its clinical significance.
Collapse
Affiliation(s)
- Chao Feng
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yaobang Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wuyue Song
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tao Liu
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Han Mo
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hui Liu
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shulin Wu
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zezu Qin
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenxing Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuting Tao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Liangyu He
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shaomei Tang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Yuanliang Xie
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
| | - Tianyu Li
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Raaijmakers KTPM, Adema GJ, Bussink J, Ansems M. Cancer-associated fibroblasts, tumor and radiotherapy: interactions in the tumor micro-environment. J Exp Clin Cancer Res 2024; 43:323. [PMID: 39696386 DOI: 10.1186/s13046-024-03251-0] [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: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) represent a group of genotypically non-malignant stromal cells in the tumor micro-environment (TME) of solid tumors that encompasses up to 80% of the tumor volume. Even though the phenotypic diversity and plasticity of CAFs complicates research, it is well-established that CAFs can affect many aspects of tumor progression, including growth, invasion and therapy resistance. Although anti-tumorigenic properties of CAFs have been reported, the majority of research demonstrates a pro-tumorigenic role for CAFs via (in)direct signaling to cancer cells, immunomodulation and extracellular matrix (ECM) remodeling. Following harsh therapeutic approaches such as radio- and/or chemotherapy, CAFs do not die but rather become senescent. Upon conversion towards senescence, many pro-tumorigenic characteristics of CAFs are preserved or even amplified. Senescent CAFs continue to promote tumor cell therapy resistance, modulate the ECM, stimulate epithelial-to-mesenchymal transition (EMT) and induce immunosuppression. Consequently, CAFs play a significant role in tumor cell survival, relapse and potentially malignant transformation of surviving cancer cells following therapy. Modulating CAF functioning in the TME therefore is a critical area of research. Proposed strategies to enhance therapeutic efficacy include reverting senescent CAFs towards a quiescent phenotype or selectively targeting (non-)senescent CAFs. In this review, we discuss CAF functioning in the TME before and during therapy, with a strong focus on radiotherapy. In the future, CAF functioning in the therapeutic TME should be taken into account when designing treatment plans and new therapeutic approaches.
Collapse
Affiliation(s)
- Kris T P M Raaijmakers
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
18
|
Sun Y, Li Z, Liu J, Xiao Y, Pan Y, Lv B, Wang X, Lin Z. Pan-cancer analysis shows that BCAP31 is a potential prognostic and immunotherapeutic biomarker for multiple cancer types. Front Immunol 2024; 15:1507375. [PMID: 39737177 PMCID: PMC11683684 DOI: 10.3389/fimmu.2024.1507375] [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: 10/07/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Background B-cell receptor-associated protein 31 (BCAP31) is a widely expressed transmembrane protein primarily located in the endoplasmic reticulum (ER), including the ER-mitochondria associated membranes. Emerging evidence suggests that BCAP31 may play a role in cancer development and progression, although its specific effects across different cancer types remain incompletely understood. Methods The raw data on BCAP31 expression in tumor and adjacent non-tumor (paracancerous) samples were obtained from the Broad Institute Cancer Cell Line Encyclopedia (CCLE) and UCSC databases. We also examined the association between BCAP31 expression and clinicopathological factors. Using the Cox proportional hazards model, we found that high BCAP31 levels were linked to poor prognosis. To further explore BCAP31's role, we analyzed the relationship between copy number variations (CNV) and BCAP31 mRNA expression using data from The Cancer Genome Atlas (TCGA). Additionally, the association between BCAP31 expression and signature pathway scores from the MsigDB database provided insights into the tumor biology and immunological characteristics of BCAP31.We assessed the relationship between tumor immune infiltration and BCAP31 expression using the TIMER2 and ImmuCellAI databases. The ESTIMATE computational method was employed to estimate the proportion of immune cells infiltrating the tumors, as well as the stromal and immune components, based on TCGA data. To investigate drug sensitivity in relation to BCAP31 expression, we utilized GDSC2 data, which included responses to 198 medications. We explored the relationship between BCAP31 gene expression and response to immunotherapy. Additionally, the study involved culturing KYSE-150 cells under standard conditions and using siRNA-mediated knockdown of BCAP31 to assess its function. Key experiments included Western blotting (WB) to confirm BCAP31 knockdown, MTT assays for cell proliferation, colony formation assays for growth potential, Transwell assays for migration and invasion, and wound healing assays for motility. Additionally, immunohistochemistry (IHC) was performed on tumor and adjacent normal tissue samples to evaluate BCAP31 expression levels. Results BCAP31 was found to be significantly overexpressed in several prevalent malignancies and was associated with poor prognosis. Cox regression analysis across all cancer types revealed that higher BCAP31 levels were predominantly linked to worse overall survival (OS), disease-free interval (DFI), disease-specific survival (DSS), and progression-free interval (PFI). In most malignancies, increased BCAP31 expression was positively correlated with higher CNV. Additionally, BCAP31 expression was strongly associated with the tumor microenvironment (TME), influencing the levels of infiltrating immune cells, immune-related genes, and immune-related pathways. Drug sensitivity analysis identified six medications that showed a significant positive correlation with BCAP31 expression. Furthermore, BCAP31 expression impacted the outcomes and prognosis of cancer patients undergoing immune therapy. The functional assays demonstrated that BCAP31 knockdown in KYSE-150 cells significantly inhibited cell migration, invasion, and proliferation while enhancing colony formation ability. WB and immunohistochemistry analyses confirmed elevated BCAP31 expression in tumor tissues compared to adjacent normal tissues in esophageal cancer, lung adenocarcinoma, and gastric adenocarcinoma. Conclusion BCAP31 has the potential to serve as a biomarker for cancer immunology, particularly in relation to immune cell infiltration, and as an indicator of poor prognosis. These findings provide a new perspective that could inform the development of more targeted cancer therapy strategies.
Collapse
Affiliation(s)
- Yangyong Sun
- Department of Cardiothoracic Surgery, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhi Li
- Department of Emergency, Nanjing Jiangning Hospital, Nanjing, Jiangsu, China
| | - Jianchao Liu
- Department of Cardiothoracic Surgery, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Xiao
- Department of Emergency, Nanjing Jiangning Hospital, Nanjing, Jiangsu, China
| | - Yaqiang Pan
- Department of Cardiothoracic Surgery, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Benbo Lv
- Department of Cardiothoracic Surgery, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xufeng Wang
- Department of Cardiothoracic Surgery, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhiqiang Lin
- Department of Otolaryngology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
19
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
20
|
Gao H, Chen J, Yang Z, Zhu Z, He L, Zhang W, Chen X, Zhang J. Comparative Study of [ 18F]AlF-LNC1007, [ 18F]FDG, and [ 18F]AlF-NOTA-FAPI-04 PET/CT in Breast Cancer Diagnosis: A Methodological Exploration and Analytical Insight. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67523-67531. [PMID: 39607746 DOI: 10.1021/acsami.4c17912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Objective: To compare the diagnostic value of [18F]AlF-LNC1007, [18F]FDG, and [18F]AlF-NOTA-FAPI-04 PET/CT in breast cancer. Methods: 33 patients with highly suspected or already diagnosed but untreated breast cancer were enrolled in the study and underwent [18F]AlF-LNC1007 (30 patients), [18F]FDG (22 patients), and [18F]AlF-NOTA-FAPI-04 (8 patients) PET/CT. Quantitative measurements included the SUVmax and tumor-to-background ratio (TBR) for all lesions and background tissues. The Chi-square test was used for intergroup diagnostic efficacy, and the Wilcoxon test was used for intergroup SUVmax or TBR. Diagnostic efficacy for lymph node metastasis was evaluated using receiver operating characteristic (ROC) analysis. Results: Compared to [18F]FDG, [18F]AlF-LNC1007 had a higher positive predictive value (100% vs 91%, P = 0.0004) in lymph node metastases (42 vs 46) and higher sensitivity (100 vs 76%, P = 0.0003) in bone metastases (33 vs 25) but lower sensitivity (93 vs 100%, P = 0.001) in liver metastases. Apart from liver metastases, [18F]AlF-LNC1007 PET/CT had higher SUVmax in primary tumor and other metastases, with no statistical difference in TBR. Compared to [18F]AlF-NOTA-FAPI-04 PET/CT, [18F]AlF-LNC1007 had less false-positive and a higher positive predictive value in bone metastases (99 vs 95%, P = 0.0003) but had lower SUVmax(P < 0.01) in all primary and metastases lesions. The TBR difference between [18F]AlF-LNC1007 and [18F]AlF-NOTA-FAPI-04 was statistically significant only in bone metastases (5.97 vs 5.02, P = 0.001). The comparison of lymph node detection efficacy between [18F]AlF-LNC1007 and [18F]FDG PET/CT showed significant differences in SUVmax cutoff values for diagnosing lymph node metastases (2.62 vs 3.90), sensitivity (95.2% vs 66.67), and specificity (100% vs 85.00) (all P < 0.001). Conclusion: [18F]AlF-LNC1007 demonstrated superior efficacy compared to [18F]FDG and [18F]AlF-NOTA-FAPI-04 and higher uptake than [18F]FDG in primary tumor, lymph node and bone metastases, and higher TBR than [18F]AlF-NOTA-FAPI-04, especially in bone metastases. [18F]AlF-LNC1007 also showed high specificity in differentiating inflammatory and metastatic lymph nodes.
Collapse
Affiliation(s)
- Haiyan Gao
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jie Chen
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhichuan Yang
- Department of Emergency Surgery, The Affilliated Chengdu 363 Hospital of Southwest Medical University, Chengdu 610041, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Limeng He
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Zhang
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Departments of Chemical and Biomolecular Engineering, and Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Lower Kent Ridge Road, 4 Science Drive 2, 117544 Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellenece, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
21
|
Zhang J, Huang J, Yang Q, Zeng L, Deng K. Regulatory mechanisms of macrophage-myofibroblast transdifferentiation: A potential therapeutic strategy for fibrosis. Biochem Biophys Res Commun 2024; 737:150915. [PMID: 39486135 DOI: 10.1016/j.bbrc.2024.150915] [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: 07/09/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Macrophage-myofibroblast transdifferentiation (MMT), a fibrotic process impacting diverse tissue types, has garnered recent scholarly interest. Within damaged tissues, the role of myofibroblasts is pivotal in the accumulation of excessive fibrous connective tissue, leading to persistent scarring or organ dysfunction. Consequently, the examination of MMT-related fibrosis is imperative. This review underscores MMT as a fundamental mechanism in myofibroblast generation during tissue fibrosis, and its exploration is crucial for elucidating the regulatory mechanisms underlying this process. Gaining insight into these mechanisms promises to facilitate the development of therapeutic approaches aimed at inhibiting and reversing fibrosis, thereby offering potential avenues for the treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Junchao Zhang
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Jinfa Huang
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Qian Yang
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Lingling Zeng
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Kaixian Deng
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China.
| |
Collapse
|
22
|
Yang C, Shu J, Li Y, Zhao N, Liu X, Tian X, Sun Z, Tabish MS, Hong Y, Chen K, Sun M. Long non-coding RNAs are involved in the crosstalk between cancer-associated fibroblasts and tumor cells. Front Immunol 2024; 15:1469918. [PMID: 39717771 PMCID: PMC11663902 DOI: 10.3389/fimmu.2024.1469918] [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: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
The proliferation of tumors is not merely self-regulated by the cancer cells but is also intrinsically connected to the tumor microenvironment (TME). Within this complex TME, cancer-associated fibroblasts (CAFs) are pivotal in the modulation of tumor onset and progression. Rich signaling interactions exist between CAFs and tumor cells, which are crucial for tumor regulation. Long non-coding RNAs (LncRNAs) emerge from cellular transcription as a class of functionally diverse RNA molecules. Recent studies have revealed that LncRNAs are integral to the crosstalk between CAFs and tumor cells, with the capacity to modify cellular transcriptional activity and secretion profiles, thus facilitating CAFs activation, tumor proliferation, metastasis, drug resistance, and other related functionalities. This comprehensive review revisits the latest research on LncRNA-mediated interactions between CAFs and tumor cells, encapsulates the biological roles of LncRNAs, and delves into the molecular pathways from a broader perspective, aspiring to offer novel perspectives for a deeper comprehension of the etiology of tumors and the enhancement of therapeutic approaches.
Collapse
Affiliation(s)
- Chenbo Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Jiao Shu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yiwei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Zhao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiaonan Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiangyu Tian
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Zexin Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Muhammad Saud Tabish
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yichen Hong
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
23
|
Lei Y, Shu D, Xia J, Zhang T, Wei H. Extracellular nicotinamide phosphoribosyltransferase visfatin activates JAK2-STAT3 pathway in cancer-associated fibroblasts to promote colorectal cancer metastasis. Genes Genomics 2024:10.1007/s13258-024-01596-6. [PMID: 39643827 DOI: 10.1007/s13258-024-01596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/29/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Metastasis is one of the major challenges in the treatment of colorectal cancer (CRC), during which cancer-associated fibroblasts (CAFs) in the tumor microenvironment are critically involved. OBJECTIVE In this study, we aim to explore the regulatory role of extracellular nicotinamide phosphoribosyltransferase Visfatin and its impact on CRC metastasis. METHODS To examine the effect of visfatin on CAFs, human CRC tissue-derived CAFs were exposed to visfatin, and the expression of inflammatory factors, activation of JAK-STAT pathway and production of ROS in CAFs were assessed. To examine the effect of visfatin-treated CAFs on CRC metastasis, human CRC cell line SW480 or SW620 were cultured with the conditioned medium derived from visfatin-treated CAFs, and the invasion and migration ability of SW480 or SW620 cells were evaluated by transwell migration and matrigel invasion assays. RESULTS Our previous study found that visfatin, a secreted form of nicotinamide phosphoribosyltransferase that governs the rate-limiting step of NAD synthesis, promoted CRC metastasis. However, little is known about the effect of visfatin on CAFs. The conditioned medium derived from visfatin- treated CAFs promotes the migratory and invasive capability of CRC cells, and enhance lung metastasis in mouse model. Visfatin treatment stimulated the expression of a couple of inflammatory factors in CAFs, which was mediated by visfatin-induced activation of JAK- STAT pathway and accumulation of ROS. Inhibition of JAK-STAT pathway or neutralization of cellular ROS attenuated visfatin-mediated migration and invasion of CRC cells. CONCLUSIONS The present work highlights a critical role of visfatin in the crosstalk between CRC cells and CAFs, which moonlight as a non-metabolic extracellular signal molecule to hijacks JAK-STAT pathway in CAFs to promote CRC metastasis.
Collapse
Affiliation(s)
- Yun Lei
- Pathological Diagnosis Center, Zhoushan Hospital of Zhejiang Province, Zhejiang, China
| | - Dan Shu
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Jianyu Xia
- School of Basic Medical Science, Chengdu Medical College, Chengdu, China
| | - Tao Zhang
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China.
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China.
| | - He Wei
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China.
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China.
| |
Collapse
|
24
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
25
|
Xiong Z, Zhuang RL, Yu SL, Xie ZX, Peng SR, Li ZA, Li BH, Xie JJ, Li YN, Li KW, Huang H. Cancer-associated fibroblasts regulate mitochondrial metabolism and inhibit chemosensitivity via ANGPTL4-IQGAP1 axis in prostate cancer. J Adv Res 2024:S2090-1232(24)00559-9. [PMID: 39647634 DOI: 10.1016/j.jare.2024.12.003] [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: 09/23/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
INTRODUCTION Cancer-associated fibroblasts (CAFs) are a critical component of the tumor microenvironment, being implicated in enhancing tumor growth and fostering drug resistance. Nonetheless, the mechanisms underlying their function in prostate cancer (PCa) remain incompletely understood, which is essential for devising effective therapeutic strategies. OBJECTIVES The main objective of this study was to explore the mechanisms by which CAFs mediate PCa growth and chemoresistance. METHODS We validated through data analysis and experimentation that CAFs significantly impact PCa cell proliferation and chemoresistance. Subsequently, we conducted a comprehensive proteomic analysis of the conditioned media from CAFs and PCa cells and identified angiopoietin-like protein 4 (ANGPTL4) as a key factor. We employed ELISA and multiplex immunofluorescence assays, all of which indicated that ANGPTL4 was primarily secreted by CAFs.Next, we conducted metabolomics analysis, GST pull-down assays, Co-IP, and other experiments to explore the specific molecular mechanisms of ANGPTL4 and its precise effects on PCa cells. Through drug screening, we identified Quercetin 3-O-(6'-galactopyranosyl)-β-D-galactopyranoside (QGGP) as an effective inhibitor of CAFs function. Finally, we thoroughly assessed the therapeutic potential of QGGP both as a monotherapy and in combination with docetaxel in PCa cells. RESULTS We discovered that the extracrine factor ANGPTL4 is primarily expressed in CAFs in PCa. When ANGPTL4 binds to IQ motif-containing GTPase-activating protein 1 (IQGAP1) on the PCa cell membrane, it activates the Raf-MEK-ERK-PGC1α axis, promoting mitochondrial biogenesis and OXPHOS metabolism, and thereby facilitating PCa growth and chemoresistance. Furthermore, virtual and functional screening strategies identified QGGP as a specific inhibitor of IQGAP1 that promotes its degradation. Combined with docetaxel treatment, QGGP can reverse the effects of CAFs and improve the responsiveness of PCa to chemotherapy. CONCLUSIONS This study uncovers a paracrine mechanism of chemoresistance in PCa and proposes that targeting the stroma could be a therapeutic choice.
Collapse
Affiliation(s)
- Zhi Xiong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Rui-Lin Zhuang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shun-Li Yu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Zhao-Xiang Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Shi-Rong Peng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ze-An Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Bing-Heng Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jun-Jia Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yi-Ning Li
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China.
| | - Kai-Wen Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China.
| |
Collapse
|
26
|
Yin QZ, Liu YJ, Zhang Q, Xi SY, Yang TB, Li JP, Gao J. Overexpression of Basonuclin Zinc Finger Protein 2 in stromal cell is related to mesenchymal phenotype and immunosuppression of mucinous colorectal adenocarcinoma. Int Immunopharmacol 2024; 142:113184. [PMID: 39306894 DOI: 10.1016/j.intimp.2024.113184] [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: 07/02/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Mucinous carcinoma (MC) is a distinct histologic subtype of colorectal cancer (CRC) that is less studied and associated with poor prognosis. This study aimed to identify MC-specific therapeutic targets and biomarkers to improve the prognosis of this aggressive disease. METHODS CRC samples from The Cancer Genome Atlas (TCGA) were categorized into MC and non-MC (NMC) groups based on histologic type. A multi-scale embedded gene co-expression network analysis (MEGENA) was constructed to identify gene modules associated with the MC group. The potential functions of Basonuclin Zinc Finger Protein 2 (BNC2) were further analyzed using the Biomarker Exploration for Solid Tumors (BEST) database. In vivo and in vitro experiments were conducted to validate the predicted results. RESULTS We identified the stromal component-related gene, BNC2, in the MC population. This gene is associated with a shorter progression-free interval (PFI) in CRC patients. BNC2 promotes FAP (encoding Fibroblast Activation Protein Alpha) transcription in cancer-associated fibroblasts (CAFs) and is involved in angiogenesis through two pathways. Additionally, BNC2 enhances tumor cell invasiveness in a CAF-dependent manner. Patients with high BNC2 expression benefited less from immunotherapy compared to those with low BNC2 expression. CONCLUSIONS Our study highlights the clinical importance of BNC2 in MC, and targeting BNC2 on stromal cells (fibroblasts and endothelial cells) may be an effective strategy for treating MC.
Collapse
Affiliation(s)
- Qing-Zhong Yin
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuan-Jie Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Qian Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Song-Yang Xi
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, Jiangsu 212000, China
| | - Tian-Bao Yang
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jie-Pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Ju Gao
- The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, Jiangsu 225009, China; Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
27
|
Zhao J, Li Y, Huang Y, Su P, Nie F, Yang P, Yang C. Tumor-Derived GDF15 Induces Tumor Associated Fibroblast Transformation From BMSCs and Fibroblasts in Oral Squamous Cell Carcinoma. J Cell Physiol 2024:e31498. [PMID: 39639678 DOI: 10.1002/jcp.31498] [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: 05/23/2024] [Revised: 11/02/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Cancer associated fibroblasts (CAFs) are the predominant stromal cell-type in the solid tumor microenvironment, originating from various cell types and playing a crucial role in promoting tumor progression and metastasis The generation of CAFs is influenced by complex factors secreted by tumor cells, with particular emphasis on transforming growth factor-β (TGF-β). However, it remains largely unknown whether growth/differentiation factor-15 (GDF15), as a member of the TGF-β superfamily, exerts similar effects to TGF-β in oral squamous cell carcinoma (OSCC). In this study, we investigated the impact of GDF15 derived from tumor cells on CAF transformation and elucidated the underlying mechanisms. Exogenous GDF15 and OSCC cells induced the transformation of bone marrow mesenchymal stem cells (BMSCs) and human gingival fibroblasts (HGFs) into CAFs, as evidenced by α-smooth muscle actin (α-SMA) as a phenotypic marker and TGF-β, interleukin 6 (IL-6), and vascular endothelial-derived growth factor (VEGF) as functional markers. Conversely, knockdown of GDF15 in OSCC cells reversed CAF transformation. Mechanistically, extracellular signal-regulated kinases 1/2(ERK1/2) pathway was associated with GDF15-mediated promotion of CAF transformation. Furthermore, OSCC-induced CAFs enhanced migration and invasion abilities of OSCC cells; but this pro-cancer effect was abolished upon knockdown of GDF15 in OSCC cells. Subcutaneous coinjection of OSCC cells with BMSCs or HGFs into mice revealed the promoted tumor growth along with increased expression levels of α-SMA and Ki67 compared with alone OSCC cells injection; these effects were attenuated when GDF15 was knocked down in OSCC cells. Collectively, our findings suggest that tumor-derived GDF15 contributes to the progression of OSCC by promoting CAF transformation through activation of the ERK1/2 pathway.
Collapse
Affiliation(s)
- Jingjing Zhao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Yahui Li
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, China
- 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 Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shanghai, China
| | - Yingying Huang
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Peng Su
- Department of Pathology, Jinan, China
| | - Fujiao Nie
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Pishan Yang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Chengzhe Yang
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
28
|
Tang R, Liu XQ. Modeling development of breast cancer: from tumor microenvironment to preclinical applications. Front Pharmacol 2024; 15:1466017. [PMID: 39697553 PMCID: PMC11652193 DOI: 10.3389/fphar.2024.1466017] [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: 07/17/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Breast cancer is a complex disease and its progression is related not only to tumor cells but also to its microenvironment, which can not be sufficiently reflected by the traditional monolayer cell culture manner. The novel human cancer models comprising tumor microenvironment (TME), such as tumor organoids and organs-on-a-chip, has been established in recent years to help elucidate the underlying mechanisms of tumorigenesis and promote the development of cancer therapies. In this review, we first discuss the current state of breast cancer and their treatment strategies, and elucidates the complex properties of TME of breast cancer in vivo. The culture models used in breast cancer research are then summarized with insights into recent development. Finally, we also conclude by discussing the current limitations and future directions of culture models in breast cancer research for providing a preclinical reference for the precise treatment of cancer patients.
Collapse
Affiliation(s)
- Ruizhi Tang
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi-Qiu Liu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Chen L, Zhang H, Gao K, Meng F, Yang F, Li J, Wang L, Tai J. Investigation of the correlation between AGRN expression and perineural invasion in colon cancer. Front Mol Biosci 2024; 11:1510478. [PMID: 39691475 PMCID: PMC11649504 DOI: 10.3389/fmolb.2024.1510478] [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: 10/13/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Background and Purpose Colon cancer is one of the most common gastrointestinal malignancies. According to the traditional view, the primary modes of transmission include direct dissemination, hematogenous metastasis, and lymph node metastasis. In recent years, the role of perineural invasion (PNI) in the spread and metastasis of tumors has received immense attention. However, there are still relatively few reports on the potential mechanisms and biomarkers of PNI occurrence and development in colon cancer. Method We identified genes linked to the onset and progression of PNI in colon cancer using bioinformatics tools and extensive databases. Gene function enrichment analysis was used to explore the potential roles of these genes in tumor proliferation, invasion, and PNI. A collection of postoperative pathological specimens from colon cancer patients who underwent surgery, related clinicopathological data, and immunohistochemistry were used to validate AGRN expression in PNI tissues. Results Bioinformatics analysis revealed that AGRN is overexpressed in colon cancer tissues and correlates with poor patient prognosis. The findings from gene association and enrichment studies indicate that AGRN and its associated genes may play a role in PNI development and progression in colon cancer by simultaneously enhancing tumor cell invasion and neural cell growth. Immunohistochemical analysis of clinical samples confirmed that AGRN expression is elevated in colon cancer tissues with PNI. Conclusion We found that AGRN is significantly overexpressed in colon cancer tissues exhibiting PNI and is linked to poor patient survival. AGRN and its related genes may contribute to PNI by promoting tumor cell invasion and neural cell growth. Hence, AGRN may play a crucial role in the initiation and progression of PNI in colon cancer.
Collapse
Affiliation(s)
- Lei Chen
- Department of Colorectal and Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Haijia Zhang
- Department of Colorectal and Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Kaiyue Gao
- Department of Colorectal and Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Fanqi Meng
- Department of Colorectal and Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Funing Yang
- Pediatric Outpatient Clinic, The First Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Lijie Wang
- Department of Colorectal and Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Jiandong Tai
- Department of Colorectal and Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Qin L, Zeng X, Qiu X, Chen X, Liu S. The role of N6-methyladenosine modification in tumor angiogenesis. Front Oncol 2024; 14:1467850. [PMID: 39691597 PMCID: PMC11649548 DOI: 10.3389/fonc.2024.1467850] [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: 07/21/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024] Open
Abstract
Tumor angiogenesis is a characteristics of malignant cancer progression that facilitates cancer cell growth, diffusion and metastasis, and has an indispensable role in cancer development. N6-methyladenosine (m6A) is among the most prevalent internal modifications in eukaryotic RNAs, and has considerable influence on RNA metabolism, including its transcription, splicing, localization, translation, recognition, and degradation. The m6A modification is generated by m6A methyltransferases ("writers"), removed by m6A demethylases ("erasers"), and recognized by m6A-binding proteins ("readers"). There is accumulating evidence that abnormal m6A modification is involved in the pathogenesis of multiple diseases, including cancers, and promotes cancer occurrence, development, and progression through its considerable impact on oncoprotein expression. Furthermore, increasing studies have demonstrated that m6A modification can influence angiogenesis in cancers through multiple pathways to regulate malignant processes. In this review, we elaborate the role of m6A modification in tumor angiogenesis-related molecules and pathways in detail, providing insights into the interactions between m6A and tumor angiogenesis. Moreover, we describe how targeting m6A modification in combination with anti-angiogenesis drugs is expected to be a promising anti-tumor treatment strategy, with potential value for addressing the challenge of drug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Shiquan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical
University, Nanning, Guangxi, China
| |
Collapse
|
31
|
Zeng C, Zhang Y, Lin C, Liang W, Chen J, Chen Y, Xiao H, Li Y, Guan H. TFCP2L1, a potential differentiation regulator, predicts favorable prognosis and dampens thyroid cancer progression. J Endocrinol Invest 2024; 47:2953-2968. [PMID: 38753296 DOI: 10.1007/s40618-024-02392-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/11/2024] [Indexed: 11/09/2024]
Abstract
PURPOSE Thyroid cancer has an overwhelming incidence in the population. Thus, there is an urgent need to understand the underlying mechanism of its occurrence and development, which may provide new insights into therapeutic strategies. The role and mechanism of TFCP2L1 in regulating the progression of thyroid cancer remains unclear. METHODS Public databases and clinical samples were used to detect the expression of TFCP2L1 in cancer and non-cancer tissues. Kaplan-Meier and Cox regression analyses were used to compare the differences in survival probability of the TFCP2L1 highly expressing group and the TFCP2L1 lowly expressing group. Functional assays were used to evaluate the biological effect of TFCP2L1 on thyroid cancer cells. RNA sequencing and enrichment analyses were used to find out pathways that were activated or inactivated by TFCP2L1. RESULTS We demonstrated that TFCP2L1 was significantly downregulated in thyroid cancer. Decreased expression of TFCP2L1 was associated with malignant clinicopathological characteristics. Kaplan-Meier and Cox regression analyses indicated that thyroid tumor patients with low TFCP2L1 expression presented shorter disease-free interval and progression-free interval. Additionally, TFCP2L1 expression was positively correlated with thyroid differentiation degree. Overexpression of TFCP2L1 in thyroid cancer cells inhibited cell growth and motility in vitro, and tumorigenicity and metastasis in vivo. Mechanistically, the NF-κB signaling pathway was found inactivated by overexpressing TFCP2L1. CONCLUSION Our results suggest that TFCP2L1 is a tumor suppressor and potential differentiation regulator, and might be a potential therapeutic target in thyroid cancer.
Collapse
Affiliation(s)
- C Zeng
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Y Zhang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - C Lin
- Department of Geriatrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200000, China
| | - W Liang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - J Chen
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Y Chen
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - H Xiao
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Y Li
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China.
| | - H Guan
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China.
| |
Collapse
|
32
|
Napoli M, Bauer J, Bonod C, Vadon-Le Goff S, Moali C. PCPE-2 (procollagen C-proteinase enhancer-2): The non-identical twin of PCPE-1. Matrix Biol 2024; 134:59-78. [PMID: 39251075 DOI: 10.1016/j.matbio.2024.09.001] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
PCPE-2 was discovered at the beginning of this century, and was soon identified as a close homolog of PCPE-1 (procollagen C-proteinase enhancer 1). After the demonstration that it could also stimulate the proteolytic maturation of fibrillar procollagens by BMP-1/tolloid-like proteinases (BTPs), PCPE-2 did not attract much attention as it was thought to fulfill the same functions as PCPE-1 which was already well-described. However, the tissue distribution of PCPE-2 shows both common points and significant differences with PCPE-1, suggesting that their activities are not fully overlapping. Also, the recently established connections between PCPE-2 (gene name PCOLCE2) and several important diseases such as atherosclerosis, inflammatory diseases and cancer have highlighted the need for a thorough reappraisal of the in vivo roles of this regulatory protein. In this context, the recent finding that, while retaining the ability to bind fibrillar procollagens and to activate their C-terminal maturation, PCPE-2 can also bind BTPs and inhibit their activity has substantially extended its potential functions. In this review, we describe the current knowledge about PCPE-2 with a focus on collagen fibrillogenesis, lipid metabolism and inflammation, and discuss how we could further advance our understanding of PCPE-2-dependent biological processes.
Collapse
Affiliation(s)
- Manon Napoli
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Julien Bauer
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Christelle Bonod
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Catherine Moali
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France.
| |
Collapse
|
33
|
Yumoto S, Horiguchi H, Kadomatsu T, Horino T, Sato M, Terada K, Miyata K, Moroishi T, Baba H, Oike Y. Host ANGPTL2 establishes an immunosuppressive tumor microenvironment and resistance to immune checkpoint therapy. Cancer Sci 2024; 115:3846-3858. [PMID: 39321028 PMCID: PMC11611770 DOI: 10.1111/cas.16348] [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: 03/26/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
Use of immune checkpoint inhibitors (ICIs) as cancer immunotherapy has advanced rapidly in the clinic; however, mechanisms underlying resistance to ICI therapy, including impaired T cell infiltration, low immunogenicity, and tumor "immunophenotypes" governed by the host, remain unclear. We previously reported that in some cancer contexts, tumor cell-derived angiopoietin-like protein 2 (ANGPTL2) has tumor-promoting functions. Here, we asked whether ANGPTL2 deficiency could enhance antitumor ICI activity in two inflammatory contexts: a murine syngeneic model of colorectal cancer and a mouse model of high-fat diet (HFD)-induced obesity. Systemic ANGPTL2 deficiency potentiated ICI efficacy in the syngeneic model, supporting an immunosuppressive role for host ANGPTL2. Relevant to the mechanism, we found that ANGPTL2 induces pro-inflammatory cytokine production in adipose tissues, driving generation of myeloid-derived suppressor cells (MDSCs) in bone marrow and contributing to an immunosuppressive tumor microenvironment and resistance to ICI therapy. Moreover, HFD-induced obese mice showed impaired responsiveness to ICI treatment, suggesting that obesity-induced chronic inflammation facilitated by high ANGPTL2 expression blocks ICI antitumor effects. Our findings overall provide novel insight into protumor ANGPTL2 functions and illustrate the essential role of the host system in ICI responsiveness.
Collapse
Affiliation(s)
- Shinsei Yumoto
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Haruki Horiguchi
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Aging and Geriatric Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Taichi Horino
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Michio Sato
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kazutoyo Terada
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Keishi Miyata
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Toshiro Moroishi
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Molecular and Medical Pharmacology, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Aging and Geriatric Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
34
|
Wang Y, Ju X, Hua R, Chen J, Dai X, Liu L, Wang G, Bai Y, Hu H, Li X. Deep learning analysis of histopathological images predicts immunotherapy prognosis and reveals tumour microenvironment features in non-small cell lung cancer. Br J Cancer 2024; 131:1833-1845. [PMID: 39455880 PMCID: PMC11589918 DOI: 10.1038/s41416-024-02856-8] [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: 02/04/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer mortality worldwide. Immune checkpoint inhibitors (ICIs) have emerged as a crucial treatment option for patients with advanced NSCLC. However, only a subset of patients experience clinical benefit from ICIs. Therefore, identifying biomarkers that can predict response to ICIs is imperative for optimising patient selection. METHODS Hematoxylin and eosin (H&E) images of NSCLC patients were obtained from the local cohort (n = 106) and The Cancer Genome Atlas (TCGA) (n = 899). We developed an ICI-related pathological prognostic signature (ir-PPS) based on H&E stained histopathology images to predict prognosis in NSCLC patients treated with ICIs using deep learning. To accomplish this, we employed a modified ResNet model (ResNet18-PG), a widely-used deep learning architecture well-known for its effectiveness in handling complex image recognition tasks. Our modifications include a progressive growing strategy to improve the stability of model training and the use of the AdamW optimiser, which enhances the optimisation process by adjusting the learning rate based on training dynamics. RESULTS The deep learning model, ResNet18-PG, achieved an area under the receiver operating characteristic curve (AUC) of 0.918 and a recall of 0.995 on the local cohort. The ir-PPS effectively risk-stratified NSCLC patients. Patients in the low-risk group (n = 40) had significantly improved progression-free survival (PFS) after ICI treatment compared to those in the high-risk group (n = 66, log-rank P = 0.004, hazard ratio (HR) = 3.65, 95%CI: 1.75-7.60). The ir-PPS demonstrated good discriminatory power for predicting 6-month PFS (AUC = 0.750), 12-month PFS (AUC = 0.677), and 18-month PFS (AUC = 0.662). The low-risk group exhibited increased expression of immune checkpoint molecules, cytotoxicity-related genes, an elevated abundance of tumour-infiltrating lymphocytes, and enhanced activity in immune stimulatory pathways. CONCLUSIONS The ir-PPS signature derived from H&E images using deep learning could predict ICIs prognosis in NSCLC patients. The ir-PPS provides a novel imaging biomarker that may help select optimal candidates for ICIs therapy in NSCLC.
Collapse
Affiliation(s)
- Youyu Wang
- Department of Thoracic Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xueming Ju
- Department of Ultrasound, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Rong Hua
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ji Chen
- Department of Medical Oncology, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lunxu Liu
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Guifang Wang
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Honglin Hu
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Xiaohua Li
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China.
| |
Collapse
|
35
|
Rastegar-Pouyani N, Abdolvahab MH, Farzin MA, Zare H, Kesharwani P, Sahebkar A. Targeting cancer-associated fibroblasts with pirfenidone: A novel approach for cancer therapy. Tissue Cell 2024; 91:102624. [PMID: 39581071 DOI: 10.1016/j.tice.2024.102624] [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: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population within the tumor that have recently come into the spotlight. By extracellular matrix (ECM) remodeling and robust cross-talk with cancer cells via different secretions such as cytokines, chemokines, and growth factors, CAFs contribute to cancer progression and poorer prognoses in patients. Novel candidates have been developed to inhibit CAFs; however, due to safety and efficacy issues, none have successfully passed clinical trials. Despite these shortcomings, one concept embraced by many researchers is to repurpose non-oncology drugs with potential anti-cancer properties for cancer treatment. One such example is pirfenidone (PFD), an oral anti-fibrotic medication, primarily administered for idiopathic pulmonary fibrosis. Emerging evidence suggests that PFD has promising anti-cancer effects, mainly manifesting through targeting CAFs. With inhibitory effects on CAFs, PFD restricts cancer proliferation, metastasis, immunosuppression, drug resistance, and tumor stiffness. To improve efficacy and minimize adverse effects, several innovative approaches have been proposed for targeting CAFs via PFD. Interestingly, combination therapy comprising PFD and chemotherapeutics e.g. doxorubicin has shown synergistic anti-cancer effects while protecting normal tissue. Furthermore, novel drug delivery systems, e.g. biomimetic liposomes and multilayer core-shell nanoparticles, have enhanced the pharmacokinetic properties of PFD and further increased its intratumoral delivery. Single-cell RNA sequencing (scRNA-seq) has also been suggested to characterize different subpopulations of CAFs and design precise PFD-based therapeutic strategies. Herein, we discuss the promising anti-cancer effects of PFD via inhibition of CAFs. We then provide findings on novel PFD-based approaches to target CAFs using combination therapy, nanocarrier-based drug delivery, and scRNA-seq.
Collapse
Affiliation(s)
- Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Tehran University of Medical Sciences, Tehran, Iran; Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohadeseh Haji Abdolvahab
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Amin Farzin
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hamed Zare
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Wang Z, Chen H, Sun L, Wang X, Xu Y, Tian S, Liu X. Uncovering the potential of APOD as a biomarker in gastric cancer: A retrospective and multi-center study. Comput Struct Biotechnol J 2024; 23:1051-1064. [PMID: 38455068 PMCID: PMC10918487 DOI: 10.1016/j.csbj.2024.02.015] [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: 10/21/2023] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
Gastric cancer (GC) poses a significant health challenge worldwide, necessitating the identification of predictive biomarkers to improve prognosis. Dysregulated lipid metabolism is a well-recognized hallmark of tumorigenesis, prompting investigation into apolipoproteins (APOs). In this study, we focused on apolipoprotein D (APOD) following comprehensive analyses of APOs in pan-cancer. Utilizing data from the TCGA-STAD and GSE62254 cohorts, we elucidated associations between APOD expression and multiple facets of GC, including prognosis, tumor microenvironment (TME), cancer biomarkers, mutations, and immunotherapy response, and identified potential anti-GC drugs. Single-cell analyses and immunohistochemical staining confirmed APOD expression in fibroblasts within the GC microenvironment. Additionally, we independently validated the prognostic significance of APOD in the ZN-GC cohort. Our comprehensive analyses revealed that high APOD expression in GC patients was notably associated with unfavorable clinical outcomes, reduced microsatellite instability and tumor mutation burden, alterations in the TME, and diminished response to immunotherapy. These findings provide valuable insights into the potential prognostic and therapeutic implications of APOD in GC.
Collapse
Affiliation(s)
- Zisong Wang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Hongshan Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Le Sun
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xuanyu Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yihang Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Sufang Tian
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xiaoping Liu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
37
|
Mi X, Duan Y, Sun J, Tai Q, Yao H, Meng L, Yang X, Shi X, Shi B, Chen J, Sun L, Zhou D, Xiao S, Yao Y, He S. The ketogenic diet modulates tumor-associated neutrophil polarization via the AMOT-YAP/TAZ axis to inhibit colorectal cancer progression. Pharmacol Res 2024; 210:107494. [PMID: 39510146 DOI: 10.1016/j.phrs.2024.107494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Despite significant advances in the diagnosis and treatment of colorectal cancer (CRC), the prognosis for late-stage patients remains poor, highlighting the urgent need for new preventive and therapeutic strategies. Recent studies have focused on the ketogenic diet (KD) and its metabolite, β-hydroxybutyrate (BHB), for their tumor-suppressive effects and modulation of inflammatory responses. Using the azoxymethane (AOM) / dextran sulfate sodium (DSS)-induced mouse CRC model, we found that the ketogenic diet and BHB inhibit pro-tumor N2-type tumor-associated neutrophils (TANs) while promoting the polarization of TANs towards the anti-tumor N1 type. This shift in TANs polarization affects tumor growth and metastasis. The underlying mechanism involves BHB acting on the intracellular receptor histone deacetylases 3 (HDAC3), which modulates the activation of the AMOT-YAP/TAZ axis, leading to the inhibition of pro-carcinogenic factor transcription and release. Moreover, clinical cohort data corroborate these findings, showing that CRC patients with elevated BHB levels have significantly lower rates of lymph node involvement, which is associated with a higher infiltration ratio of anti-carcinogenic N1-type TANs in the tumor microenvironment (TME). These results suggest that BHB levels could serve as a prognostic biomarker for CRC. In conclusion, our findings indicate that BHB derived from KD regulates TANs polarization in CRC via the HDAC3-AMOT-YAP/TAZ axis, effectively inhibiting tumor growth and metastasis. These insights establish a novel theoretical basis for employing the KD in the treatment of CRC and for developing cancer adjuvant immunotherapy strategy based on the polarization of neutrophils.
Collapse
Affiliation(s)
- Xiuwei Mi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Yudong Duan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiying Sun
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China; Department of Respiratory Diseases, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, China
| | - Qingliang Tai
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Huihui Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Lijun Meng
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Xiaoshan Yang
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Xinyu Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Bo Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Junjie Chen
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215299, China
| | - Liang Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Diyuan Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
38
|
Xiao M, Guo Z, Yang Y, Hu C, Cheng Q, Zhang C, Wu Y, Cheng Y, Benson WLM, Shamay SMN, Leung GPH, Li J, Gao H, Zhang J. Glycyrrhizic acid-based multifunctional nanoplatform for tumor microenvironment regulation. Chin J Nat Med 2024; 22:1089-1099. [PMID: 39725510 DOI: 10.1016/s1875-5364(24)60685-0] [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: 07/19/2024] [Indexed: 12/28/2024]
Abstract
Natural compounds demonstrate unique therapeutic advantages for cancer treatment, primarily through direct tumor suppression or interference with the tumor microenvironment (TME). Glycyrrhizic acid (GL), a bioactive ingredient derived from the medicinal herb Glycyrrhiza uralensis Fisch., and its sapogenin glycyrrhetinic acid (GA), have been recognized for their ability to inhibit angiogenesis and remodel the TME. Consequently, the combination of GL with other therapeutic agents offers superior therapeutic benefits. Given GL's amphiphilic structure, self-assembly capability, and liver cancer targeting capacity, various GL-based nanoscale drug delivery systems have been developed. These GL-based nanosystems exhibit angiogenesis suppression and TME regulation properties, synergistically enhancing anti-cancer effects. This review summarizes recent advances in GL-based nanosystems, including polymer-drug micelles, drug-drug assembly nanoparticles (NPs), liposomes, and nanogels, for cancer treatment and tumor postoperative care, providing new insights into the anti-cancer potential of natural compounds. Additionally, the review discusses existing challenges and future perspectives for translating GL-based nanosystems from bench to bedside.
Collapse
Affiliation(s)
- Meng Xiao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhiqing Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yating Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chuan Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanfen Cheng
- School of Medicine, Chengdu University, Chengdu 610106, China
| | - Wui Lau Man Benson
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Sheung Mei Ng Shamay
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong 999077, China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong 999077, China; The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hong Kong 999077, China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
39
|
Verocq C, Noël JC, Charry M, Zindy E, Rorive S, Salmon I, Decaestecker C, Catteau X. Inverse correlation between the amounts of lymphocytic infiltrate and stroma in breast carcinoma. Heliyon 2024; 10:e40295. [PMID: 39641033 PMCID: PMC11617241 DOI: 10.1016/j.heliyon.2024.e40295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Background Previous breast carcinoma studies focused on the evaluation of tumour-infiltrating lymphocytes (TILs) or of tumoural stroma via the tumour stroma ratio (TSR). Few studies assessed peritumoural lymphocytes and almost no studies investigated a possible relationship between lymphocytes and stroma. This prompted us to evaluate the amount of tumour cells, intra- and peritumoural lymphocytes, and stroma in breast cancer to support the hypothesis that the stroma may block the infiltration of lymphocytes inside the tumour. Methods We collected a retrospective series of 158 breast cancers (<25 mm). In addition to standard TILs and TSR evaluations, we assessed the percentages of tumour cells, stromal myofibroblasts, intra- and peritumoural lymphocytes on full-section tumours with haematoxylin and eosin and immunohistochemical staining. Results We showed significant negative correlations between the amounts of stroma and both intra- and peritumoural lymphocyte percentages. Considering the estrogen receptor positive invasive breast cancer of no special type cases, we showed that TSR had a positive prognostic value with an optimal threshold of 10 %. Conclusions This study is one of the first to show inverse correlations between tumoural stroma amount and intra- and peritumoural lymphocyte percentages, which supports the hypothesis that tumoural stroma can prevent the recruitment of lymphocytes around and within the tumour.
Collapse
Affiliation(s)
- Camille Verocq
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| | - Jean-Christophe Noël
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Manon Charry
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| | - Egor Zindy
- DIAPath - Center for Microscopy and Molecular Imaging, ULB, Rue Adrienne Bolland 8, 6041, Gosselies, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
- DIAPath - Center for Microscopy and Molecular Imaging, ULB, Rue Adrienne Bolland 8, 6041, Gosselies, Belgium
| | - Christine Decaestecker
- DIAPath - Center for Microscopy and Molecular Imaging, ULB, Rue Adrienne Bolland 8, 6041, Gosselies, Belgium
- Laboratory of Image Synthesis and Analysis, Ecole Polytechnique de Bruxelles, ULB, 50, Avenue F. Roosevelt, 1050, Brussels, Belgium
| | - Xavier Catteau
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| |
Collapse
|
40
|
Yoo W, Choi H, Lee J, Lee Y, Park KC, Noh K. CRHBP, a novel multiple cancer biomarker connected with better prognosis and anti-tumorigenicity. Cancer Cell Int 2024; 24:391. [PMID: 39614295 DOI: 10.1186/s12935-024-03562-4] [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: 07/18/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND The corticotropin-releasing hormone-binding protein (CRHBP) plays a crucial role in regulating corticotropin release. Little is known about the role of CRHBP, a major regulator of neuroendocrine, autonomic, and stress adaptation, in tumors. In this study, we aimed to investigate the clinical and molecular landscapes of CRHBP in various types of tumors. METHODS We investigated the role of CRHBP in different types of tumors using publicly available databases and performed a comparative expression analysis of CRHBP-related genes in pan-cancer prognosis using methylation profiling, tumor-infiltrating immune cell expression analysis, gene enrichment analysis, and protein-protein interaction analysis, identified common pathways, and in vitro evaluation. RESULTS We evaluated CRHBP expression across tumor and corresponding normal tissues using the data from The Cancer Genome Atlas and the Genotype-Tissue Expression database. CRHBP was downregulated in most tumors and was identified as an important factor for predicting the prognosis of patients with cancer. Intracellular metabolic pathways and hormone-related processes were involved in the functional mechanisms of CRHBP. Mechanistically, the downregulation of CRHBP was attributed to the upregulation of four miRNAs in most tumors, and CRHBP expression was related to tumor-infiltrating immune cells in tumors. Overexpression of CRHBP significantly inhibited cell proliferation of LUAD, LIHC, and KIRC cell lines, while inhibition of cell mobility was found only in KIRC and HCC cells. CONCLUSIONS This study provides a comprehensive summary of the systemic role of CRHBP expression in various types of tumors, highlighting the prognostic importance and clinical significance of tumors. Furthermore, CRHBP decreases cell proliferation and mobility in cancer cell lines associated with OS and DFS, further research is needed to understand the underlying mechanisms and explore clinical applications.
Collapse
Affiliation(s)
- Wonbeak Yoo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hyunji Choi
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jieun Lee
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Yeeun Lee
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Kyung Chan Park
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| | - Kyunghee Noh
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
- Department of Nanobiotechnology, University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
41
|
Mussa A, Ismail NH, Hamid M, Al-Hatamleh MAI, Bragoli A, Hajissa K, Mokhtar NF, Mohamud R, Uskoković V, Hassan R. Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer. J Exp Clin Cancer Res 2024; 43:312. [PMID: 39609700 PMCID: PMC11603874 DOI: 10.1186/s13046-024-03218-1] [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: 08/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.
Collapse
Affiliation(s)
- Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Mahasin Hamid
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Hunan Province, Changsha, 410013, China
- Department of Zoology, Faculty of Sciences and Information Technology, University of Nyala, Nyala, 63311, Sudan
| | - Mohammad A I Al-Hatamleh
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anthony Bragoli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Khalid Hajissa
- Department of Zoology, Faculty of Science and Technology, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (iNFORMM), Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, 92604, USA
- Division of Natural Sciences, Fullerton College, Fullerton, CA, 92832, USA
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| |
Collapse
|
42
|
Zhang S, Wang J, Chen Y, Liang W, Liu H, Du R, Sun Y, Hu C, Shang Z. CAFs-derived lactate enhances the cancer stemness through inhibiting the MST1 ubiquitination degradation in OSCC. Cell Biosci 2024; 14:144. [PMID: 39605072 PMCID: PMC11603751 DOI: 10.1186/s13578-024-01329-y] [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: 08/03/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs), a predominant stromal cell type in the tumor microenvironment, significantly affect the progression of oral squamous cell carcinoma (OSCC). RESULTS The specific mechanisms through which CAFs influence the cancer stem cell phenotype in OSCC are not fully understood. This study explored the effects of lactic acid produced by CAFs on the cancer stem cells (CSCs) phenotype of OSCC cells. Our results demonstrated that CAFs exhibit increased glycolysis and lactic acid production. Lactic acid treatment enhances CSCs-related markers expression, sphere formation, and clonogenic ability of OSCC cells. RNA sequencing revealed that lactic acid treatment elevates Discs Large Homolog 5 (DLG5) expression and markedly affects the Hippo pathway. Further investigation revealed that DLG5 mediates the effects of lactic acid on the CSCs phenotype. DLG5 knockdown results in elevated expression of E3 ubiquitin ligase Cullin 3, which can promote the ubiquitination and degradation of MST1, but the expression of phosphorylated MST1 remains unchanged. This leads to enhanced binding of phosphorylated MST1 to YAP1, increasing YAP1 phosphorylation and activating the Hippo pathway. CONCLUSION Collectively, our findings suggest that lactic acid from CAFs promotes the CSCs phenotype in OSCC through the DLG5/CUL3/MST1 axis. Therefore, targeting lactic acid exchange between CAFs and tumor cells may provide a novel therapeutic approach to suppress the CSCs phenotype in OSCC.
Collapse
Affiliation(s)
- Shuzhen Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Central Department School & Hospital of Stomatology, Wuhan University, Wuhan, 430022, China
| | - Jingjing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Weilian Liang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Day Surgery Center, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hanzhe Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ruixue Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yunqing Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China.
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
43
|
Lu Y, Quan J, Liu F, Huang B. Systematic pan-cancer analysis of the prognostic value of MECOM in human cancer. Discov Oncol 2024; 15:694. [PMID: 39576394 PMCID: PMC11584820 DOI: 10.1007/s12672-024-01599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024] Open
Abstract
Recently, emerging evidence suggests an association between MECOM (MDS1 and EVI1 complex locus) and cancers. However, a comprehensive pan-cancer analysis to fully investigate this relationship is lacking. Herein, public platforms with large-scale genomics, including The Cancer Genome Atlas, Gene Expression Omnibus dataset, and the Human Protein Atlas were explored to investigate the prognostic and immunological roles of MECOM across certain cancer types. Our findings revealed differential expression of MECOM in various cancer types, indicating its potential to predict diverse clinical outcomes, such as overall survival time and disease-free survival time in patients with various malignancies. Additionally, we observed an association between the mutation burden in MECOM in various cancers and patient survival. Furthermore, the mechanism of MECOM-mediated oncogenesis was tentatively explored by immune infiltration analysis. This study provided a relatively comprehensive overview of the prognostic and immunological roles of MECOM in multiple cancers.
Collapse
Affiliation(s)
- Yingqiang Lu
- Department of Urology, Urology and Nephrology Center, Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Jing Quan
- Department of Urology, Urology and Nephrology Center, Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Feng Liu
- Department of Urology, Urology and Nephrology Center, Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Banggao Huang
- Department of Urology, Urology and Nephrology Center, Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
44
|
Roszkowska M. Multilevel Mechanisms of Cancer Drug Resistance. Int J Mol Sci 2024; 25:12402. [PMID: 39596466 PMCID: PMC11594576 DOI: 10.3390/ijms252212402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer drug resistance represents one of the most significant challenges in oncology and manifests through multiple interconnected molecular and cellular mechanisms. Objective: To provide a comprehensive analysis of multilevel processes driving treatment resistance by integrating recent advances in understanding genetic, epigenetic, and microenvironmental factors. This is a systematic review of the recent literature focusing on the mechanisms of cancer drug resistance, including genomic studies, clinical trials, and experimental research. Key findings include the following: (1) Up to 63% of somatic mutations can be heterogeneous within individual tumors, contributing to resistance development; (2) cancer stem cells demonstrate enhanced DNA repair capacity and altered metabolic profiles; (3) the tumor microenvironment, including cancer-associated fibroblasts and immune cell populations, plays a crucial role in promoting resistance; and (4) selective pressure from radiotherapy drives the emergence of radioresistant phenotypes through multiple adaptive mechanisms. Understanding the complex interplay between various resistance mechanisms is essential for developing effective treatment strategies. Future therapeutic approaches should focus on combination strategies that target multiple resistance pathways simultaneously, guided by specific biomarkers.
Collapse
Affiliation(s)
- Malgorzata Roszkowska
- Department of Clinical Neuropsychology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
45
|
Pang N, Yang Z, Zhang W, Du Y, Zhang L, Li X, Peng Y, Qi X. Cancer-associated fibroblasts barrier breaking via TGF-β blockade paved way for docetaxel micelles delivery to treat pancreatic cancer. Int J Pharm 2024; 665:124706. [PMID: 39277152 DOI: 10.1016/j.ijpharm.2024.124706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
TGF-β is a crucial regulator in tumor microenvironment (TME), especially for myofibroblastic cancer-associated fibroblasts (myCAFs). The myCAFs can be motivated by TGF-β signaling to erect pro-tumor TME, meanwhile, myCAFs overexpress TGF-β to mediate the crosstalk between tumor and stromal cells. The blockade of TGF-β can break cancer-associated fibroblasts barrier, consequently opening the access for drugs into tumor. The TGF-β is a promising target in anti-tumor therapy. Herein, we introduced a two-stage combination therapy (TC-Therapy), including TGF-β receptor I inhibitor SB525334 (SB) and cytotoxicity agent docetaxel micelle (DTX-M). We found that SB and DTX-M synergistically inhibited myCAFs proliferation and elevated p53 protein expression in BxPC-3/3T3 mixed cells. Gene and protein tests demonstrated that SB cut off TGF-β signaling via receptor blockade and it did not arouse TGF-β legend compensated internal autocrine. On the contrary, two agents combined decreased TGF-β secretion and inhibited myCAFs viability marked by α-SMA and FAPα. TC-Therapy was applied in BxPc-3/3T3 mixed tumor-bearing mice model. After TC-Therapy, the α-SMA+/ FAPα+ myCAFs faded increasingly and collagenous fibers mainly secreted by myCAFs decreased dramatically as well. More than that, the myCAFs barrier breaking helped to normalize micro-vessels and paved way for micelle penetration. The TGF-β protein level of TC-Therapy in TME was much lower than that of simplex DTX-M, which might account for TME restoration. In conclusion, TGF-β inhibitor acted as the pioneer before nano chemotherapeutic agents. The TC-Therapy of TGF-β signaling inhibition and anti-tumor agent DTX-M is a promising regimen without arising metastasis risk to treat pancreatic cancer. The therapeutic regimen focused on TGF-β related myCAFs reminds clinicians to have a comprehensive understanding of pancreatic cancer.
Collapse
Affiliation(s)
- Ning Pang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Zhenzhen Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenjie Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yitian Du
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lu Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xin Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiwei Peng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xianrong Qi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
46
|
Huang J, Zhang H, Ma L, Ma N, Luo N, Jin W, Shi J, Xu S, Xiong Y. Rhein and hesperidin nanoparticles remodel tumor immune microenvironment by reducing CAFs and CCL2 secreted by CAAs for efficient triple-negative breast cancer therapy. Int Immunopharmacol 2024; 141:113001. [PMID: 39186835 DOI: 10.1016/j.intimp.2024.113001] [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: 07/13/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
In triple-negative breast cancer (TNBC), the tumor immune microenvironment (TIME) is a highly heterogeneous ecosystem that exerts indispensable roles in tumorigenesis and tumor progression. Cancer-associated fibroblasts (CAFs) and cancer-associated adipocytes (CAAs) are the main matrix components in the TIME of TNBC. CAFs mediate the edesmoplastic response, which is a major driver of the immunosuppressive microenvironment to promote tumor growth. In addition, CAAs, a type of tumor-educated adipocyte, participate in crosstalk with breast cancer and are capable of secreting various cytokines, adipokines and chemokines, especially C-C Motif Chemokine Ligand 2 (CCL2), resulting in changes of cancer cell phenotype and function. Therefore, how to treat tumors by regulating the CAFs and the secretion of CCL2 by CAAs in TIME is investigated here. Our research group previously found that rhein (Rhe) has been identified as effective against CAFs, while hesperidin (Hes) could effectively diminish CCL2 secretion by CAAs. Inspired by the above, we developed unique PLGA-based nanoparticles loaded with Rhe and Hes (RH-NP) using the emulsion solvent diffusion method. The RH-NP particles have an average size of 114.1 ± 0.98 nm. RH-NP effectively reduces CAFs and inhibits CCL2 secretion by CAAs, promoting increased infiltration of cytotoxic T cells and reducing immunosuppressive cell presence within tumors. This innovative, safe, low-toxic, and highly effective anti-tumor strategy could be prospective in TNBC treatment.
Collapse
Affiliation(s)
- Jingyi Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Lisha Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ninghui Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ningchao Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wanyu Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jingbin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shujun Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
47
|
Osuala KO, Heyza J, Zhao Z, Xu Y, Moin K, Ji K, Mattingly RR. Carcinoma-Associated Fibroblasts Accelerate Growth and Invasiveness of Breast Cancer Cells in 3D Long-Term Breast Cancer Models. Cancers (Basel) 2024; 16:3840. [PMID: 39594795 PMCID: PMC11593312 DOI: 10.3390/cancers16223840] [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: 10/14/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Carcinoma-associated fibroblasts (CAFs), a prominent cell type in the tumor microenvironment (TME), significantly contributes to cancer progression through interactions with cancer cells and other TME components. Consequently, targeting signaling pathways driven by CAFs has potential to yield new therapeutic approaches to inhibit cancer progression. However, the mechanisms underlying their long-term interactions with cancer cells in vivo remains poorly understood. Methods: To address this, we developed a three-dimensional (3D) parallel coculture model of human triple-negative breast cancer (TNBC) cells and CAFs using our innovative TAME devices. This model allowed for the analysis of TNBC paracrine interactions via their secretome over extended culture periods (at least 70 days). Results: Using TNBC cell lines (MDA-MB-231, MCF10.DCIS, and HCC70), we found that TNBC spheroids in 3D parallel cocultures with CAFs exhibited more pronounced invasive finger-like outgrowths than those in cocultures of TNBC cells and normal fibroblasts (NFs) over a period of 50-70 days. We also established that the CAF-derived secretome affects TNBC migration towards the CAF secretome region. Additionally, we observed a preferential migration of CAFs, but not NFs, toward TNBC spheroids. Conclusions: Overall, our results suggest that paracrine interactions between TNBC cells and CAFs enhance TNBC invasive phenotypes and promote reciprocal migration.
Collapse
Affiliation(s)
- Kingsley O. Osuala
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA; (K.O.O.); (J.H.); (K.M.)
| | - Joshua Heyza
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA; (K.O.O.); (J.H.); (K.M.)
| | - Zhiguo Zhao
- Department of Electrical and Computer Engineering, Wayne State University, Detroit, MI 48201, USA; (Z.Z.); (Y.X.)
| | - Yong Xu
- Department of Electrical and Computer Engineering, Wayne State University, Detroit, MI 48201, USA; (Z.Z.); (Y.X.)
| | - Kamiar Moin
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA; (K.O.O.); (J.H.); (K.M.)
| | - Kyungmin Ji
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA; (K.O.O.); (J.H.); (K.M.)
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
| | - Raymond R. Mattingly
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA; (K.O.O.); (J.H.); (K.M.)
- Department of Pharmacology and Toxicology, Brody Medical School, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
48
|
Peng Z, Zhao T, Gao P, Zhang G, Wu X, Tian H, Qu M, Tan X, Zhang Y, Zhao X, Qi X. Tumor-Derived Extracellular Vesicles Enable Tumor Tropism Chemo-Genetherapy for Local Immune Activation in Triple-Negative Breast Cancer. ACS NANO 2024; 18:30943-30956. [PMID: 39474658 PMCID: PMC11562804 DOI: 10.1021/acsnano.3c12967] [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: 12/22/2023] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/13/2024]
Abstract
Triple-negative breast cancer (TNBC) is highly heterogeneous, lacks accessible therapeutic targets, and features an immunosuppressive tumor microenvironment (TME). Anthracycline-based chemotherapy remains the primary treatment method for TNBC, while the current popular immune checkpoint inhibitors persistently encounter therapeutic resistance. Therefore, there is an urgent need to explore combined therapeutic strategies to remodel the TME and improve the treatment response. Considering the highly specific homing ability of tumor cell-derived vesicles and the key role of the signal transduction and activation of the transcription factor 3 (STAT3) pathway in TNBC, we propose a synergistic therapeutic strategy that integrates gene therapy, chemotherapy, and immunotherapy based on STAT3 short interfering RNA (siSTAT3) and doxorubicin (DOX)-functionalized tumor-derived extracellular vesicles (TEVs) (siSTAT3-DOX@TEV). The in vitro and in vivo results demonstrate that siSTAT3-DOX@TEV target tumor tissues precisely, downregulate STAT3 expression, and synergistically and efficiently induce immunogenic death, thereby reversing the immunosuppressive TME. Moreover, mass cytometry and immunohistochemistry reveal the local immune activation effect of siSTAT3-DOX@TEV, with a significant increase in M1 macrophages, CD4+ T cells, and CD8+ T cells in tumor tissues. These results provide strong hints for the development of TEV-based chemo-gene therapeutic agents for TNBC treatment at the clinical level.
Collapse
Affiliation(s)
- Zaihui Peng
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tingting Zhao
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Pingping Gao
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Guozhi Zhang
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiujuan Wu
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hao Tian
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Man Qu
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xuanni Tan
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi Zhang
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiang Zhao
- Department
of Oncology, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaowei Qi
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
49
|
Qiu X, Li S, Fan T, Zhang Y, Wang B, Zhang B, Zhang M, Zhang L. Advances and prospects in tumor infiltrating lymphocyte therapy. Discov Oncol 2024; 15:630. [PMID: 39514075 PMCID: PMC11549075 DOI: 10.1007/s12672-024-01410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy in adoptive T-cell therapy (ACT) has already caused durable regression in a variety of cancer types due to T-cell persistence, clinical activity, and duration of objective response and safety. TILs are composed of polyclonal effector T-cells specific to heterogenetic tumor antigens, reasonably providing a promising means for tumor therapy. In addition, their expansion in vitro can release them from the suppressive tumor microenvironment. Even though significant advances have been made in the procedure of TIL therapy, from TIL isolation, modification, expansion, and infusion back to the patient to target the tumor, strategy optimization is always ongoing to overcome drawbacks such as a complex process, options for the lineage differentiation status of TILs, and sufficient trafficking of TILs to the tumor. In this review, we summarize the current advances of TIL therapy, raise problem-based optimization strategies, and provide future perspectives on next-generation TIL therapy as a potential avenue for enhancing cell-based immunotherapy.
Collapse
Affiliation(s)
- Xu Qiu
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Shengjun Li
- Clinical Laboratory, Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Tianyu Fan
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Taian City Central Hospital, Taian, Shandong, China
| | - Yue Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Bin Wang
- The Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Bei Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Mingzhe Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
50
|
Lopes CDH, Braganca Xavier C, Torrado C, Veneziani AC, Megid TBC. A Comprehensive Exploration of Agents Targeting Tumor Microenvironment: Challenges and Future Perspectives. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:283-299. [PMID: 39524466 PMCID: PMC11541921 DOI: 10.36401/jipo-24-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 11/16/2024]
Abstract
The tumor microenvironment (TME) encompasses the complex and diverse surroundings in which tumors arise. Emerging insights highlight the TME's critical role in tumor development, progression, metastasis, and treatment response. Consequently, the TME has attracted significant research and clinical interest, leading to the identification of numerous novel therapeutic targets. Advances in molecular technologies now enable detailed genomic and transcriptional analysis of cancer cells and the TME and the integration of microenvironmental data to the tumor genomic landscape. This comprehensive review discusses current progress in targeting the TME for drug development, addressing associated challenges, strategies for modulating the pro-tumor microenvironment, and the discovery of new targets.
Collapse
Affiliation(s)
| | | | - Carlos Torrado
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|