1
|
Lee C, Lee J, Jeong M, Nam D, Rhee I. Emerging strategies for targeting tumor-associated macrophages in glioblastoma: A focus on chemotaxis blockade. Life Sci 2025; 376:123762. [PMID: 40419107 DOI: 10.1016/j.lfs.2025.123762] [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: 03/04/2025] [Revised: 05/14/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain tumors, with poor prognosis for affected patients. A key player in the GBM tumor microenvironment is the tumor-associated macrophage (TAM), which promotes tumor progression, immune evasion, and therapeutic resistance. The recruitment of TAMs to the tumor site is driven by specific chemotactic signals, including CSF-1/CSF-1R, CXCR4/CXCL12, and HGF/MET pathways. This review explores the current understanding of these chemotaxis mechanisms and their role in GBM progression. It highlights the potential therapeutic benefits of targeting TAM chemotaxis pathways to disrupt TAM infiltration, reduce immunosuppression, and enhance the efficacy of conventional treatments. Additionally, we discuss the preclinical and clinical evidence surrounding key inhibitors, such as PLX3397, AMD3100, and Crizotinib, which have shown promise in reprogramming TAMs and improving treatment outcomes in GBM. While these strategies offer hope for overcoming some of the challenges of GBM therapy, the review also addresses the limitations and obstacles in clinical translation, emphasizing the need for further research and the development of combination therapies to achieve sustained therapeutic benefit.
Collapse
Affiliation(s)
- Chaelin Lee
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Jaehyun Lee
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Moongyu Jeong
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Dayoung Nam
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea
| | - Inmoo Rhee
- Department of Biotechnology and Bioscience, Sejong University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Zhu X, Zhang L, Yu X, Yan P, Zhang X, Zhao Y, Wang D, Yang XA. Elucidating the tumor microenvironment interactions in breast, cervical, and ovarian cancer through single-cell RNA sequencing. Sci Rep 2025; 15:17846. [PMID: 40404741 PMCID: PMC12098903 DOI: 10.1038/s41598-025-03017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
This study aimed to identify the key cell types and their interactions in gynecological oncology of breast cancer, cervical cancer, and ovarian cancer. Single-cell RNA sequencing was performed on tumor samples of gynecological oncology from the GEO database. Cell types were identified using SingleR and cell composition was analyzed to understand the tumor microenvironment (TME). CellChat was used to analyze cell interactions, and pseudotemporal analysis was conducted on cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) to understand their differentiation status. Four CAF subtypes were identified: iCAF, myCAF, proCAF, and matCAF. The iCAF subpopulation secreted COL1A1 and promoted tumor cell migration, while myCAF was involved in angiogenesis. The matCAF subpopulation was present throughout tumor development. TAMs were found to promote angiogenesis through the VEGFA_VEGFR2 signaling pathway. CAFs and TAMs play pivotal roles in tumor progression through their interactions and signaling pathways.
Collapse
Affiliation(s)
- Xiaoyue Zhu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Liang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaomin Yu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Pengxian Yan
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Xiaoyu Zhang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Yunlong Zhao
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Dongze Wang
- Clinical and Basic Medical College, Shandong First Medical University, Jinan, 250000, China
| | - Xiu-An Yang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China.
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, 067000, China.
| |
Collapse
|
3
|
Cao Y, Qian R, Yao R, Zheng Q, Yang C, Yang X, Ji S, Zhang L, Zhan S, Wang Y, Wang T, Wang H, Wong CM, Yuan S, Heeschen C, Gao Q, Bernards R, Qin W, Wang C. DYRK1A-TGF-β signaling axis determines sensitivity to OXPHOS inhibition in hepatocellular carcinoma. Dev Cell 2025; 60:1483-1497.e7. [PMID: 39798576 DOI: 10.1016/j.devcel.2024.12.035] [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: 04/05/2024] [Revised: 09/14/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025]
Abstract
Intervening in mitochondrial oxidative phosphorylation (OXPHOS) has emerged as a potential therapeutic strategy for certain types of cancers. Employing kinome-based CRISPR screen, we find that knockout of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) synergizes with OXPHOS inhibitor IACS-010759 in liver cancer cells. Targeting DYRK1A combined with OXPHOS inhibitors activates TGF-β signaling, which is crucial for OXPHOS-inhibition-triggered cell death. Mechanistically, upregulation of glutamine transporter solute carrier family 1 member 5 (SLC1A5) transcription compensates for the increased glutamine requirement upon OXPHOS inhibition. DYRK1A directly phosphorylates SMAD3 Thr132, thereby suppressing the negative impact of TGF-β signaling on transcription of SLC1A5, leading to intrinsic resistance of liver cancer cells to OXPHOS inhibition. Moreover, we demonstrate the therapeutic efficacy of IACS-010759 in combination with DYRK1A inhibition in multiple liver cancer models, including xenografts, patient-derived xenografts, and spontaneous tumor model. Our study elucidates how the DYRK1A-TGF-β signaling axis controls the response of tumor cells to OXPHOS inhibition and provides valuable insights into targeting OXPHOS for liver cancer therapy.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruolan Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruilian Yao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Quan Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xupeng Yang
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuyi Ji
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linmen Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Zhan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiping Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianshi Wang
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Ming Wong
- State Key Laboratory for Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Shengxian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Christopher Heeschen
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Guo W, Mu K, Geng JC, Xing HY, Dong Y, Liu WD, Wang SC, Shi JX, Xing BR, Zhao JY, Li XM. ATF1 and miR-27b-3p drive intervertebral disc degeneration through the PPARG/NF-κB signaling axis. Commun Biol 2025; 8:751. [PMID: 40369110 PMCID: PMC12078598 DOI: 10.1038/s42003-025-08186-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 05/07/2025] [Indexed: 05/16/2025] Open
Abstract
Intervertebral disc degeneration (IDD) is a primary cause of degenerative disc disease; however, the mechanisms underlying it remain unknown. Although great efforts have been made to develop new regenerative therapies, their clinical success is limited. Recent research has indicated that microRNAs (miRNAs) are significantly involved in the progression of IDD. Investigating the role of miRNA intervention in IDD could facilitate the development of therapeutic strategies based on miRNAs. However, circulating miRNAs have not yet been recognized as standard biomarkers for IDD. In this study, we observed that the expression of miR-27b-3p was elevated in the blood and nucleus pulposus (NP) tissue of patients with IDD. Furthermore, reducing the expression of miR-27b-3p was shown to impede the progression of IDD. MiR-27b-3p could reduce the expression of collagen II and ACAN and promote the expression of MMP13 and ADAMT-5 in vitro and in vivo. miR-27b-3p aggravated IDD progression by directly targeting peroxisome proliferator-activated receptor gamma (PPARG), a negative regulator of the NF-κB signal pathway. This study also established that PPARG serves a protective role in IDD. The overexpression of PPARG was able to mitigate the detrimental effects caused by miR-27b-3p in NP cells and animal models of IDD, indicating that miR-27b-3p facilitates the progression of IDD through its interaction with PPARG. Additionally, the transcription factor ATF1 was found to enhance the expression of miR-27b-3p by targeting its promoter region, thereby promoting the degenerative impact of miR-27b-3p on NP cells. Given that miR-27b-3p can promote IDD both in vitro and in vivo, it holds potential as a biomarker, and the inhibition of miR-27b-3p expression may represent a novel therapeutic target for IDD.
Collapse
Affiliation(s)
- Wei Guo
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China.
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, PR China.
- Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou, PR China.
| | - Kun Mu
- Department of Breast Surgery, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
| | - Jing-Chao Geng
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
| | - Hai-Yang Xing
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
| | - Yu Dong
- Department of Anaesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
| | - Wen-Dong Liu
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
- Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou, PR China
| | - Shuan-Chi Wang
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, PR China
| | - Jia-Xiao Shi
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, PR China
| | - Bao-Rui Xing
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, PR China
- Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou, PR China
| | - Jian-Yong Zhao
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, PR China
- Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou, PR China
| | - Xiao-Ming Li
- Department of Orthopaedics, Hebei Province Cangzhou Hospital of Integrated Traditional Chinese Medicine-Western Medicine, Cangzhou, PR China.
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, Cangzhou, PR China.
- Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou, PR China.
| |
Collapse
|
5
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [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: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
6
|
Babiker NA, Nadeem S, Abu Kariem H, Abdul Hameed A, Negmeldin AT, El-labbad EM. Medicinal Chemistry Strategies in Targeting TGF-βR1 Kinase Domain: Unveiling Insights into Inhibitor Structure-Activity Relationship (SAR). Pharmaceuticals (Basel) 2025; 18:716. [PMID: 40430535 PMCID: PMC12114876 DOI: 10.3390/ph18050716] [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: 04/15/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The transforming growth factor-β (TGF-β) signaling pathway is involved in various cellular functions, including immunological response, extracellular matrix formation, differentiation, growth and development, and cell cycle regulation. The TGF β receptor type 1 (TGF-βR1) has emerged as a key component of this pathway, exhibiting significant overexpression in diverse malignancies, including hepatocellular carcinoma, gastric cancer, breast cancer, and colon cancer. Multiple therapeutic targets have been identified for the TGF-β signaling pathway, encompassing antibodies, ligand traps, vaccines, antisense oligonucleotides, and small-molecule TGF-βR1 kinase inhibitors. This review delineates the structural and functional characteristics of the small-molecule TGF-βR1 kinase inhibitors. The inhibitors discussed herein are categorized based on shared pharmacophoric features, notably a five-membered heterocyclic ring linked to three distinct features (R1, R2, and R3). These features interact with amino acids within the selectivity pocket, hinge region, or solvent-exposed area, respectively. These insights contribute to a clearer understanding of the structural requirements for selective TGF-βR1 inhibition. The presented findings in this review article offer a valuable foundation for future drug discovery efforts targeting the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Nusaiba A. Babiker
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Soam Nadeem
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Hasan Abu Kariem
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Afra Abdul Hameed
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Ahmed T. Negmeldin
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Eman M. El-labbad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| |
Collapse
|
7
|
Huang K, Yang L, Ma Y, Cao L, Li S, Zhao Z, Zhou J, Wang S. Integrated multi-omics characterization of neuroblastoma with bone or bone marrow metastasis. Genes Dis 2025; 12:101511. [PMID: 40070366 PMCID: PMC11894310 DOI: 10.1016/j.gendis.2024.101511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/17/2024] [Accepted: 12/18/2024] [Indexed: 03/14/2025] Open
Abstract
The pathogenesis of neuroblastoma with bone or bone marrow metastasis (NB-BBM) and its complex immune microenvironment remain poorly elucidated, hampering the advancement of effective risk prediction for BBM and limiting therapeutic strategies. Feature recognition of 142 paraffin-embedded hematoxylin-eosin-stained tumor section images was conducted using a Swin-Transformer for pathological histology to predict NB-BBM occurrence. Single-cell transcriptomics identified a tumor cell subpopulation (NB3) and two tumor-associated macrophage (TAM) subpopulations (SPP1+ TAMs and IGHM+ TAMs) closely associated with BBM and highlighted transketolase (TKT) as a key molecular marker for metastatic progression in NB. This extensive multi-omics investigation into NB-BBM enhances our understanding of single-cell transcriptional dynamics in NB beyond existing research, outlining the evolution from in situ carcinoma through tumorigenesis to bone marrow metastases. Furthermore, exploration of the immune microenvironment identified specific subpopulations of TAMs crucial in promoting NB-BBM, presenting new avenues for immunotherapy. These insights enhance our understanding of the metastatic process from NB to BBM and facilitate the development of more effective diagnostic and therapeutic strategies for this aggressive pediatric cancer.
Collapse
Affiliation(s)
| | | | - Yue Ma
- Department of Pediatric Surgical Oncology, The Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Lijian Cao
- Department of Pediatric Surgical Oncology, The Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Suwen Li
- Department of Pediatric Surgical Oncology, The Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Zhenzhen Zhao
- Department of Pediatric Surgical Oncology, The Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Jianwu Zhou
- Department of Pediatric Surgical Oncology, The Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| | - Shan Wang
- Department of Pediatric Surgical Oncology, The Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing 400014, China
| |
Collapse
|
8
|
Abooali M, Schlichtner S, Lei X, Aliu N, Ruggiero S, Loges S, Ziegler M, Hertel F, Volckmar AL, Stenzinger A, Christopoulos P, Thomas M, Klenova E, Meyer NH, Boussios S, Heaton N, Zen Y, Zamalloa A, Chokshi S, Urbani L, Richard S, Kirubendran K, Hussain R, Siligardi G, Cholewa D, Berger SM, Yasinska IM, Fasler-Kan E, Sumbayev VV. Intracellular and extracellular activities of V-domain Ig-containing suppressor of T cell activation (VISTA) modulated by immunosuppressive factors of tumour microenvironment. Cancer Lett 2025; 616:217581. [PMID: 39983894 DOI: 10.1016/j.canlet.2025.217581] [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: 12/26/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/23/2025]
Abstract
V-domain Ig-containing suppressor of T cell activation (VISTA) is a unique immune checkpoint protein, which was reported to display both receptor and ligand activities. However, the mechanisms of regulation of VISTA activity and functions by factors of tumour microenvironment (TME) remain unclear and understanding these processes is required in order to develop successful personalised cancer immunotherapeutic strategies and approaches. Here we report for the very first time that VISTA interacts with another immune checkpoint protein galectin-9 inside the cell most likely facilitating its interaction with TGF-β-activated kinase 1 (TAK1). This process is required for protection of lysosomes, which is crucial for many cell types and tissues. We found that VISTA expression can be differentially controlled by crucial factors present in TME, such as transforming growth factor beta type 1 (TGF-β) and hypoxia as well as other factors activating hypoxic signalling. We confirmed that involvement of these important pathways modulated by TME differentially influences VISTA expression in different cell types. These networks include: TGF-β-Smad3 pathway, TAK1 (TGF-β-activated kinase 1) or apoptosis signal-regulating kinase 1 (ASK1)-induced activation of activating transcription factor 2 (ATF-2) and hypoxic signalling pathway. Based on this work we determined the five critical functions of VISTA and the role of TME factors in controlling (modulating or downregulating) VISTA expression.
Collapse
Affiliation(s)
- Maryam Abooali
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Stephanie Schlichtner
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Xi Lei
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Nijas Aliu
- Department of Human Genetics, Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Sonia Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Ziegler
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Franziska Hertel
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Elena Klenova
- School of Life Sciences, University of Essex, Colchester, UK
| | - N Helge Meyer
- Division of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, CT1 1QU, Kent, UK; Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King's College London, Strand, London, WC2R 2LS, UK; Kent Medway Medical School, University of Kent, Canterbury, CT2 7LX, Kent, UK; Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, ME7 5NY, Kent, UK; AELIA Organization, 9th Km Thessaloniki-Thermi, 57001, Thessaloniki, Greece
| | - Nigel Heaton
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Yoh Zen
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Ane Zamalloa
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, UK
| | - Shilpa Chokshi
- Peninsula Medical School, Faculty of Health, University of Plymouth, UK; Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Luca Urbani
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Sophie Richard
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | - Kavitha Kirubendran
- Roger Williams Institute of Liver Studies, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, Foundation for Liver Research and King's College Hospital, London, UK
| | | | | | - Dietmar Cholewa
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Steffen M Berger
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Inna M Yasinska
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children's Hospital, Inselspital Bern, University of Bern and Department of Biomedical Research, University of Bern, Bern, Switzerland.
| | - Vadim V Sumbayev
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, UK.
| |
Collapse
|
9
|
Zhang H, Zhang X, Zhang Y, Han D, Ha H, Zhang B, Shang P. Pan-Cancer Analysis Shows that KIFC2 is a Potential Prognostic and Immunotherapeutic Biomarker for Multiple Cancer Types Including Bladder Cancer. Mol Biotechnol 2025; 67:1641-1658. [PMID: 38658471 DOI: 10.1007/s12033-024-01149-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/29/2024] [Indexed: 04/26/2024]
Abstract
KIFC2 plays an important role in prostate cancer progression and chemotherapy resistance, but the mechanism of its involvement in other malignancies remains unclear. Therefore, this study aimed to analyze and validate the mechanism of effect of KIFC2 in multiple tumors. Bioinformatic analysis was performed in conjunction with multiple databases (The Cancer Genome Atlas, Genotype-Tissue Expression Project, Human Protein Atlas, etc.) to fully explore the potential role of KIFC2 within individual tumors and to analyze the correlation with major research components such as prognosis, mutations, and the tumor microenvironment. The expression of KIFC2 demonstrates a significant correlation with the prognosis, clinical phenotype, tumor mutational burden, microsatellite instability, and tumor microenvironment across various malignancies and is associated with the modulation of diverse functional and signaling pathways. The differences in the expression of KIFC2 in the bladder cancer tissues (14 pairs) were statistically significant. The pan-cancer analysis in this study revealed the multifunctionality of KIFC2 in a variety of tumors, indicating a possible prognostic predictor and potential therapeutic target for tumors.
Collapse
Affiliation(s)
- Helin Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Key Laboratory of Urological Diseases in Gansu Province, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, 730030, Gansu, China
| | - Xingxing Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Key Laboratory of Urological Diseases in Gansu Province, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, 730030, Gansu, China
| | - Yuelin Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Key Laboratory of Urological Diseases in Gansu Province, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, 730030, Gansu, China
| | - Dali Han
- The Second Hospital & Clinical Medical School, Lanzhou University, Key Laboratory of Urological Diseases in Gansu Province, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, 730030, Gansu, China
| | - Hualan Ha
- The Second Hospital & Clinical Medical School, Lanzhou University, Key Laboratory of Urological Diseases in Gansu Province, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, 730030, Gansu, China
| | - Biao Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Key Laboratory of Urological Diseases in Gansu Province, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, 730030, Gansu, China
| | - Panfeng Shang
- The Second Hospital & Clinical Medical School, Lanzhou University, Key Laboratory of Urological Diseases in Gansu Province, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
10
|
Aftabi S, Barzegar Behrooz A, Cordani M, Rahiman N, Sadeghdoust M, Aligolighasemabadi F, Pistorius S, Alavizadeh SH, Taefehshokr N, Ghavami S. Therapeutic targeting of TGF-β in lung cancer. FEBS J 2025; 292:1520-1557. [PMID: 39083441 PMCID: PMC11970718 DOI: 10.1111/febs.17234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/22/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Transforming growth factor-β (TGF-β) plays a complex role in lung cancer pathophysiology, initially acting as a tumor suppressor by inhibiting early-stage tumor growth. However, its role evolves in the advanced stages of the disease, where it contributes to tumor progression not by directly promoting cell proliferation but by enhancing epithelial-mesenchymal transition (EMT) and creating a conducive tumor microenvironment. While EMT is typically associated with enhanced migratory and invasive capabilities rather than proliferation per se, TGF-β's influence on this process facilitates the complex dynamics of tumor metastasis. Additionally, TGF-β impacts the tumor microenvironment by interacting with immune cells, a process influenced by genetic and epigenetic changes within tumor cells. This interaction highlights its role in immune evasion and chemoresistance, further complicating lung cancer therapy. This review provides a critical overview of recent findings on TGF-β's involvement in lung cancer, its contribution to chemoresistance, and its modulation of the immune response. Despite the considerable challenges encountered in clinical trials and the development of new treatments targeting the TGF-β pathway, this review highlights the necessity for continued, in-depth investigation into the roles of TGF-β. A deeper comprehension of these roles may lead to novel, targeted therapies for lung cancer. Despite the intricate behavior of TGF-β signaling in tumors and previous challenges, further research could yield innovative treatment strategies.
Collapse
Affiliation(s)
- Sajjad Aftabi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of BiologyComplutense UniversityMadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Mohammadamin Sadeghdoust
- Division of BioMedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sCanada
| | - Farnaz Aligolighasemabadi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
| | - Stephen Pistorius
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Nima Taefehshokr
- Apoptosis Research CentreChildren's Hospital of Eastern Ontario Research InstituteOttawaCanada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Faculty Academy of Silesia, Faculty of MedicineKatowicePoland
- Children Hospital Research Institute of ManitobaUniversity of ManitobaWinnipegCanada
| |
Collapse
|
11
|
Sarangi R, Mishra S, Mahapatra S. Cancer Vaccines: A Novel Revolutionized Approach to Cancer Therapy. Indian J Clin Biochem 2025; 40:191-200. [PMID: 40123637 PMCID: PMC11928706 DOI: 10.1007/s12291-024-01201-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/19/2024] [Indexed: 03/25/2025]
Abstract
Over the past few decades, there has been significant advancement in the field of tumor immunotherapy. For many years vaccination against infectious diseases have been available. On the other hand very few cancer vaccines have been approved for human use. Ideal Cancer vaccines are biological response modifier work by stimulating both humoral and cellular immunity while overcoming the immunological suppression found in tumor. Two types of cancer vaccine: Prophylactic and therapeutic cancer vaccines are recommended for clinical use of individuals. HPV and HBV vaccines are the two widely used preventive vaccine used for treatment of cervical and hepatocellular carcinoma respectively and are approved by Food and Drug Administration (FDA). In therapeutic vaccine only three are approved: Sipuleucel T-cell vaccine for treatment refractory prostatic cancer, BCG vaccine for early bladder cancer and T-VEC for inoperable melanoma. Active ingredient in all cancer vaccines is an antigen. Antigens used for formulating cancer vaccines along with adjuvants optimizes immunogenicity in it. Heterogeneity within and between cancer types, screening and identifying suitable antigen specific to tumors and selection of vaccine delivery platforms are challenges in the development of vaccines. Adoptive cell therapy, Chimeric antigen receptor T cell therapy are recent breakthrough for cancer treatment.
Collapse
Affiliation(s)
- RajLaxmi Sarangi
- Departments of Biochemistry, Kalinga Institute of Medical Sciences (KIMS), Bhubaneswar, Odisha 751024 India
| | - Sanjukta Mishra
- Departments of Biochemistry, Kalinga Institute of Medical Sciences (KIMS), Bhubaneswar, Odisha 751024 India
| | - Srikrushna Mahapatra
- Departments of Biochemistry, Kalinga Institute of Medical Sciences (KIMS), Bhubaneswar, Odisha 751024 India
| |
Collapse
|
12
|
Zhang Y, Guo Z, Sun J, Bi Y, Cai R, Zhang R, Ren H, Qian J, Meng F. Tumor immune microenvironment in adenoid cystic carcinoma of the lacrimal gland: relationship with histopathology and prognosis. BMC Cancer 2025; 25:581. [PMID: 40165156 PMCID: PMC11959856 DOI: 10.1186/s12885-025-13438-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 04/02/2025] Open
Abstract
PURPOSE To quantitatively investigate the pathological subtypes of lacrimal gland adenoid cystic carcinoma (LGACCs), the tumor immune microenvironment in each pathological subtype, and the relation to survival. METHODS In this retrospective study, the tumor subtype was determined by H&E staining. Multiplex immunochemistry was performed to define specific immune cells. The tumor immune microenvironment (TIME) was sketched by sequential image scanning and reconstructed by a cytometry platform. RESULTS Eighteen patients with adequate paraffin blocks diagnosed with LGACC from 2012 to 2021 were included in this study. Thirteen patients out of the eighteen patients (72.2%)showed a mixture of different pathological subtypes. Each pathological subtype took different percentages on different tumors. The cribriform was the most common subtype, taking an overall percentage of 39%. The rest of the pathological subtypes were tubular (19%), basaloid (17%), cribriform and tubular mixture (C + T) 14%. The sclerosing and comedocarcinomic subtypes were the least seen in LGACC, taking a percentage of 11% altogether. Patients with cribriform dominant component had better overall survival than the non-cribriform dominant patients. Patients with basaloid dominant component had worse clinical outcomes than the non-basaloid dominant ones. The TIME showed high immunogenicity in the tumor margin but declined in the tumor areas. Pathological subtypes rather than individual differences determined the TIME phenotype. The cribriform subtype possessed more immune cell infiltration than other pathological subtypes. CONCLUSIONS LGACC is composed of multiple pathological subtypes. Each pathological subtype takes different percentages on different tumors, which is related to the prognosis. TIME pattern in LGACC varies among different pathological subtypes, which could indicate novel strategies in immunotherapy.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Zhipeng Guo
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jie Sun
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Yingwen Bi
- Department of Pathology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Rongrong Cai
- Department of Pathology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Rui Zhang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Hui Ren
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
| | - Jiang Qian
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China.
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.
| | - Fengxi Meng
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China.
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China.
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Wu S, Wu Z, Lu Z, Qi F, Cheng J, Chu T, Li B, Zhao Y, Nie G, Li S. Selective apoptosis of tumor-associated platelets boosts the anti-metastatic potency of PD-1 blockade therapy. Cell Rep Med 2025; 6:101984. [PMID: 40020674 PMCID: PMC11970387 DOI: 10.1016/j.xcrm.2025.101984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/24/2024] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Despite the transformative impact of programmed cell death protein-1 (PD-1) blockade therapy on metastatic/advanced solid tumor treatment, its efficacy is hindered by a limited response rate. Platelets play a pivotal role in tumor metastasis by shielding circulating tumor cells and secreting immunosuppressive factors. We here demonstrate that selectively inducing apoptosis in tumor-associated platelets (TAPs) using ABT-737-loaded nanoparticles (cyclic arginine-glycine-aspartate containing peptide-modified ABT-737-loaded nanoparticles [cRGD-NP@A]) enhances the anti-metastatic efficacy of the anti-PD-1 antibody (aPD-1). cRGD-NP@A specifically binds to TAPs, disrupting platelet-tumor cell interactions and exposing tumor cells to immune surveillance in vivo. Combined with aPD-1, cRGD-NP@A substantially augments immune activation and reduces TAP-derived immunosuppressive factors, notably transforming growth factor β1 (TGF-β1), consequently improving anti-metastatic outcomes across multiple metastasis-bearing animal models without observable adverse effects. Our study underscores the importance of depleting TAPs to enhance PD-1 blockade therapy, presenting a promising strategy to improve response rates and clinical outcomes for patients with metastatic cancer.
Collapse
Affiliation(s)
- Suying Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Zhouliang Wu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin 300211, P.R. China
| | - Zefang Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Feilong Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Jin Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Tianjiao Chu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; School of Astronautics, Harbin Institute of Technology, Harbin 150001, P.R. China
| | - Bozhao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China; School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P.R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| |
Collapse
|
14
|
Pei Y, Liang H, Guo Y, Wang B, Wu H, Jin Z, Lin S, Zeng F, Wu Y, Shi Q, Xu J, Huang Y, Ren T, Liu J, Guo W. Liquid-liquid phase separation drives immune signaling transduction in cancer: a bibliometric and visualized study from 1992 to 2024. Front Oncol 2025; 15:1509457. [PMID: 40104511 PMCID: PMC11913689 DOI: 10.3389/fonc.2025.1509457] [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: 10/11/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025] Open
Abstract
Background Liquid-liquid phase separation (LLPS) is a novel concept that could explain how living cells precisely modulate internal spatial and temporal functions. However, a comprehensive bibliometric analysis on LLPS and immune signaling processes in cancer is still scarce. This study aims to perform a bibliometric assessment of research to explore the landscape of LLPS research in immune signaling pathways for cancer. Methods Utilizing the Web of Science Core Collection database and multiple analysis software, we performed quantitative and qualitative analyses of the study situation between LLPS and immune signaling in cancer from 1992 to 2024. Results The corresponding authors were primarily from China and the USA. The most relevant references were the "International Journal of Molecular Sciences", "Proteomics". The annual number of publications exhibited a fast upward tendency from 2020 to 2024. The most frequent key terms included expression, separation, activation, immunotherapy, and mechanisms. Qualitative evaluation emphasized the TCR, BCR, cGAS-STING, RIG-1, NF-κB signaling pathways associated with LLPS processes. Conclusion This research is the first to integratively map out the knowledge structure and forward direction in the area of immune transduction linked with LLPS over the past 30 years. In summary, although this research area is still in its infancy, illustrating the coordinated structures and communications between cancer and immune signaling with LLPS within a spatial framework will offer deeper insights into the molecular mechanisms of cancer development and further enhance the effectiveness of existing immunotherapies.
Collapse
Affiliation(s)
- Yanhong Pei
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Haijie Liang
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Yu Guo
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Boyang Wang
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Han Wu
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Zhijian Jin
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Shanyi Lin
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Fanwei Zeng
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Yifan Wu
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Qianyu Shi
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Jiuhui Xu
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Yi Huang
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Tingting Ren
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Wei Guo
- Department of Bone Tumor, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
15
|
Sparano C, Solís-Sayago D, Zangger NS, Rindlisbacher L, Van Hove H, Vermeer M, Westermann F, Mussak C, Rallo E, Dergun S, Litscher G, Xu Y, Bijnen M, Friedrich C, Greter M, Juranić Lisnić V, Becher B, Gasteiger G, Oxenius A, Tugues S. Autocrine TGF-β1 drives tissue-specific differentiation and function of resident NK cells. J Exp Med 2025; 222:e20240930. [PMID: 39692745 DOI: 10.1084/jem.20240930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 12/19/2024] Open
Abstract
Group 1 innate lymphoid cells (ILCs) encompass NK cells and ILC1s, which have non-redundant roles in host protection against pathogens and cancer. Despite their circulating nature, NK cells can establish residency in selected tissues during ontogeny, forming a distinct functional subset. The mechanisms that initiate, maintain, and regulate the conversion of NK cells into tissue-resident NK (trNK) cells are currently not well understood. Here, we identify autocrine transforming growth factor-β (TGF-β) as a cell-autonomous driver for NK cell tissue residency across multiple glandular tissues during development. Cell-intrinsic production of TGF-β was continuously required for the maintenance of trNK cells and synergized with Hobit to enhance cytotoxic function. Whereas autocrine TGF-β was redundant in tumors, our study revealed that NK cell-derived TGF-β allowed the expansion of cytotoxic trNK cells during local infection with murine cytomegalovirus (MCMV) and contributed to viral control in the salivary gland. Collectively, our findings reveal tissue-specific regulation of trNK cell differentiation and function by autocrine TGF-β1, which is relevant for antiviral immunity.
Collapse
Affiliation(s)
- Colin Sparano
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Darío Solís-Sayago
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | | | - Lukas Rindlisbacher
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Hannah Van Hove
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Marijne Vermeer
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Frederike Westermann
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Caroline Mussak
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Elisa Rallo
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Stanislav Dergun
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Gioana Litscher
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Yishu Xu
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Mitchell Bijnen
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Christin Friedrich
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg , Würzburg, Germany
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka , Rijeka, Croatia
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg , Würzburg, Germany
| | | | - Sonia Tugues
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
- Department of Immunology, Medical Faculty Mannheim, Mannheim Institute for Innate Immunosciences (MI3), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
16
|
Cui X, Song Y, Han J, Yuan Z. The multifaceted role of SMAD4 in immune cell function. Biochem Biophys Rep 2025; 41:101902. [PMID: 39802394 PMCID: PMC11721226 DOI: 10.1016/j.bbrep.2024.101902] [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: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) signaling pathway, with SMAD4 as its central mediator, plays a pivotal role in regulating cellular functions, including growth, differentiation, apoptosis, and immune responses. While extensive research has elucidated SMAD4's role in tumorigenesis, its functions within immune cells remain underexplored. This review synthesizes current knowledge on SMAD4's diverse roles in various immune cells such as T cells, B cells, dendritic cells, and macrophages, highlighting its impact on immune homeostasis and pathogen response. Understanding SMAD4's role in immune cells is crucial, as its dysregulation can lead to autoimmune disorders, chronic inflammation, and immune deficiencies. The review emphasizes the significance of SMAD4 in immune regulation, proposing that deeper investigation could reveal novel therapeutic targets for immune-mediated conditions. Insights into SMAD4's involvement in processes like T cell differentiation, B cell class switch recombination, and macrophage polarization underscore its potential as a therapeutic target for a range of diseases, including autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Xinmu Cui
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Yu Song
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Jianfeng Han
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
- Cellular Biomedicine Group Inc, Shanghai, 201203, China
| | - Zhaoxin Yuan
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| |
Collapse
|
17
|
Kwon JY, Vera RE, Fernandez-Zapico ME. The multi-faceted roles of cancer-associated fibroblasts in pancreatic cancer. Cell Signal 2025; 127:111584. [PMID: 39756502 PMCID: PMC11807759 DOI: 10.1016/j.cellsig.2024.111584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/13/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
The tumor microenvironment (TME) has been linked with the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype of pancreatic cancer. A central component of the TME are cancer-associated fibroblasts (CAFs), which can either suppress or promote tumor growth in a context-dependent manner. In this review, we will discuss the multi-faceted roles of CAFs in tumor-stroma interactions influencing cancer initiation, progression and therapeutic response.
Collapse
Affiliation(s)
- John Y Kwon
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | - Renzo E Vera
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | | |
Collapse
|
18
|
Emir SM, Karaoğlan BS, Kaşmer R, Şirin HB, Sarıyıldız B, Karakaş N. Hunting glioblastoma recurrence: glioma stem cells as retrospective targets. Am J Physiol Cell Physiol 2025; 328:C1045-C1061. [PMID: 39818986 DOI: 10.1152/ajpcell.00344.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/11/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain malignancies in adults. Standard approaches, including surgical resection followed by adjuvant radio- and chemotherapy with temozolomide (TMZ), provide only transient control, as GBM frequently recurs due to its infiltrative nature and the presence of therapy-resistant subpopulations such as glioma stem cells (GSCs). GSCs, with their quiescent state and robust resistance mechanisms, evade conventional therapies, contributing significantly to relapse. Consequently, current treatment methods for GBM face significant limitations in effectively targeting GSCs. In this review, we emphasize the relationship between GBM recurrence and GSCs, discuss the current limitations, and provide future perspectives to overwhelm the challenges associated with targeting GSCs. Eliminating GSCs may suppress recurrence, achieve durable responses, and improve therapeutic outcomes for patients with GBM.
Collapse
Affiliation(s)
- Sümeyra Mengüç Emir
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Birnur Sinem Karaoğlan
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Ramazan Kaşmer
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Hilal Buse Şirin
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Batuhan Sarıyıldız
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
| | - Nihal Karakaş
- Cancer Research Center, Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, Istanbul, Türkiye
- Department of Medical Biology, International School of Medicine, İstanbul Medipol University, Istanbul, Türkiye
| |
Collapse
|
19
|
Maggi E, Landolina N, Munari E, Mariotti FR, Tumino N, Vacca P, Azzarone B, Moretta L. T cells in the microenvironment of solid pediatric tumors: the case of neuroblastoma. Front Immunol 2025; 16:1544137. [PMID: 40092980 PMCID: PMC11906424 DOI: 10.3389/fimmu.2025.1544137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Neuroblastoma (NB) is an immunologically "cold" tumor with poor or no inflamed substrates as most of solid pediatric tumors (SPT). Consistent data indicate that NB tumor microenvironment (TME) is dominated by myeloid cells, with little (but variable) T cell infiltration. The obstacles to lymphocyte infiltration and to their anti-tumor activity are due to different tumor immune evasion strategies, including loss of HLA Class I molecules, high expression of immune checkpoint molecular ligands leading to exhaustion of T effector (and NK) cells, induction of T regulatory, myeloid and stromal cells and secretion of immunosuppressive mediators. In odds with adult solid tumors, NB displays weak immunogenicity caused by intrinsic low mutational burden and scant expression of neoepitopes in the context of MHC-class I antigens which, in turn, are particularly poorly expressed on NB cells, thus inducing low anti-tumor T cell responses. In addition, NB is generated from embryonal cells and is the result of transcriptional abnormalities and not of the accumulation of genetic mutations over time, thus further explaining the low immunogenicity. The poor expression of immunogenic molecules on tumor cells is associated with the high production of immunosuppressive factors which further downregulate lymphocyte infiltration and activity, thus explaining the limited efficacy of new drugs in NB, as immune checkpoint inhibitors. This review is focused on examining the role of T effector and regulatory cells infiltrating TME of NB, taking into account their repertoire, phenotype, function, plasticity and, importantly, predictive value for defining novel targets for therapy.
Collapse
Affiliation(s)
- Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Enrico Munari
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | | | - Nicola Tumino
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Vacca
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
20
|
Tasis A, Spyropoulos T, Mitroulis I. The Emerging Role of CD8 + T Cells in Shaping Treatment Outcomes of Patients with MDS and AML. Cancers (Basel) 2025; 17:749. [PMID: 40075597 PMCID: PMC11898900 DOI: 10.3390/cancers17050749] [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: 01/13/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). In this review, we focus on the implication of CD8+ T cells in the treatment response of patients with MDS and AML. First, we review reported studies of aberrant functionality and clonality of CD8+ T cells in MDS and AML, often driven by the immunosuppressive bone marrow microenvironment, which can hinder effective antitumor immunity. Additionally, we discuss the potential use of CD8+ T cell subpopulations, including memory and senescent-like subsets, as predictive biomarkers for treatment response to a variety of treatment regimens, such as hypomethylating agents, which is the standard of care for patients with higher-risk MDS, and chemotherapy which is the main treatment of patients with AML. Understanding the multifaceted role of CD8+ T cells and their interaction with malignant cells in MDS and AML will provide useful insights into their potential as prognostic/predictive biomarkers, but also uncover alternative approaches to novel treatment strategies that could reshape the therapeutic landscape, thus improving treatment efficacy, aiding in overcoming treatment resistance and improving patient survival in these challenging myeloid neoplasms.
Collapse
Affiliation(s)
- Athanasios Tasis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Theodoros Spyropoulos
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Ioannis Mitroulis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
21
|
Shi Y, Zhang J, Li Y, Feng C, Shao C, Shi Y, Fang J. Engineered mesenchymal stem/stromal cells against cancer. Cell Death Dis 2025; 16:113. [PMID: 39971901 PMCID: PMC11839947 DOI: 10.1038/s41419-025-07443-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/03/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Mesenchymal stem/stromal cells (MSCs) have garnered attention for their potential in cancer therapy due to their ability to home to tumor sites. Engineered MSCs have been developed to deliver therapeutic proteins, microRNAs, prodrugs, chemotherapy drugs, and oncolytic viruses directly to the tumor microenvironment, with the goal of enhancing therapeutic efficacy while minimizing off-target effects. Despite promising results in preclinical studies and clinical trials, challenges such as variability in delivery efficiency and safety concerns persist. Ongoing research aims to optimize MSC-based cancer eradication and immunotherapy, enhancing their specificity and efficacy in cancer treatment. This review focuses on advancements in engineering MSCs for tumor-targeted therapy.
Collapse
Affiliation(s)
- Yuzhu Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jia Zhang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Basic Medical Sciences, Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yanan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chao Feng
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200025, China.
| | - Jiankai Fang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
22
|
Felix Oghenemaro E, Uthirapathy S, Nathiya D, Kaur P, Ravi Kumar M, Verma A. Role of glutaminyl-peptide cyclo-transferase-like protein (QPCTL) in cancer: From molecular mechanisms to immunotherapy. Gene 2025; 937:149153. [PMID: 39653089 DOI: 10.1016/j.gene.2024.149153] [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: 09/10/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Glutaminyl-peptide cyclotransferase-like protein (QPCTL) is a newly discovered enzyme that has sparked interest owing to its possible role in cancer genesis and progression. Initially discovered as a post-translational modification regulator of protein maturation, QPCTL has emerged as a key participant in cancer biology. Recent research has linked QPCTL to numerous essential cancer-related processes, including cell proliferation, migration, invasion, and apoptosis. Furthermore, QPCTL expression changes have been seen in a variety of cancer types, underlining its potential as a diagnostic and prognostic marker. The molecular mechanisms behind QPCTL's participation in cancer will be examined in this review. We investigate its involvement in the control of signaling pathways and the modification of cellular activities that are important in cancer. We also examine the clinical importance of QPCTL, including as its relationship with tumor development, metastasis, and response to treatment. We also discuss the possible therapeutic implications of targeting QPCTL in cancer therapy. QPCTL is a prospective target for the development of innovative anticancer treatments due to its participation in several cancer-associated pathways.
Collapse
Affiliation(s)
- Enwa Felix Oghenemaro
- Delta State University, Department of Pharmaceutical Microbiology, Faculty of Pharmacy, PMB 1, Abraka, Delta State, Nigeria
| | - Subasini Uthirapathy
- Faculty of Pharmacy, Pharmacology Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India.
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Ashish Verma
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| |
Collapse
|
23
|
Garcia-Fabiani MB, Haase S, Banerjee K, Zhu Z, McClellan BL, Mujeeb AA, Li Y, Tronrud CE, Varela ML, West ME, Yu J, Kadiyala P, Taher AW, Núñez FJ, Alghamri MS, Comba A, Mendez FM, Nicola Candia AJ, Salazar B, Nunez FM, Edwards MB, Qin T, Cartaxo RT, Niculcea M, Koschmann C, Venneti S, Vallcorba MP, Nasajpour E, Pericoli G, Vinci M, Kleinman CL, Jabado N, Chandler JP, Sonabend AM, DeCuypere M, Hambardzumyan D, Prolo LM, Mahaney KB, Grant GA, Petritsch CK, Welch JD, Sartor MA, Lowenstein PR, Castro MG. H3.3-G34R Mutation-Mediated Epigenetic Reprogramming Leads to Enhanced Efficacy of Immune Stimulatory Gene Therapy in Diffuse Hemispheric Gliomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.06.13.544658. [PMID: 37398299 PMCID: PMC10312611 DOI: 10.1101/2023.06.13.544658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Diffuse hemispheric glioma (DHG), H3 G34-mutant, representing 9-15% of cases, are aggressive Central Nervous System (CNS) tumors with poor prognosis. This study examines the role of epigenetic reprogramming of the immune microenvironment and the response to immune-mediated therapies in G34-mutant DHG. To this end, we utilized human G34-mutant DHG biopsies, primary G34-mutant DHG cultures, and genetically engineered G34-mutant mouse models (GEMMs). Our findings show that the G34 mutation alters histone marks' deposition at promoter and enhancer regions, leading to the activation of the JAK/STAT pathway, which in turn results in an immune-permissive tumor microenvironment. The implementation of Ad-TK/Ad-Flt3L immunostimulatory gene therapy significantly improved median survival, and lead to over 50% long term survivors. Upon tumor rechallenge in the contralateral hemisphere without any additional treatment, the long-term survivors exhibited robust anti-tumor immunity and immunological memory. These results indicate that immune-mediated therapies hold significant potential for clinical translation in treating patients harboring H3.3-G34 mutant DHGs, offering a promising strategy for improving outcomes in this challenging cancer subtype affecting adolescents and young adults (AYA). STATEMENT OF SIGNIFICANCE This study uncovers the role of the H3.3-G34 mutation in reprogramming the tumor immune microenvironment in diffuse hemispheric gliomas. Our findings support the implementation of precision medicine informed immunotherapies, aiming at improving enhanced therapeutic outcomes in adolescents and young adults harboring H3.3-G34 mutant DHGs.
Collapse
Affiliation(s)
- Maria B. Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Present address: Leloir Institute Foundation, Buenos Aires, Argentina
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ziwen Zhu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brandon L. McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anzar A. Mujeeb
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yingxiang Li
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Claire E. Tronrud
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria L. Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Molly E.J. West
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jin Yu
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
- Present address: Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ayman W. Taher
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Felipe J. Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Flor M. Mendez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alejandro J. Nicola Candia
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brittany Salazar
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fernando M. Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Marta B. Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rodrigo T. Cartaxo
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Michael Niculcea
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Carl Koschmann
- Department of Pediatrics, Chad Carr Pediatric Brain Tumor Center, University of Michigan Medical School, MI 48109, USA
| | - Sriram Venneti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Emon Nasajpour
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Giulia Pericoli
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Maria Vinci
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Claudia L. Kleinman
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - James P. Chandler
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael DeCuypere
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Lou & Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Division of Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Laura M. Prolo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Gerald A. Grant
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
- Present address: Department of Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Claudia K Petritsch
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
24
|
Deng L, Wu Z, Sun C, Liu Z. Transcriptome analysis revealed SMURF2 as a prognostic biomarker for oral cancer. J Appl Genet 2025; 66:155-170. [PMID: 38698292 PMCID: PMC11762210 DOI: 10.1007/s13353-024-00869-w] [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: 02/05/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND The activation of TGF-β pathway can facilitate tumorigenesis. Understanding the TGF-related genes (TRGs) in oral cancer and determining their prognostic value is of utmost importance. METHODS The TRGs were selected to develop a prognostic model based on lasso regression. Oral cancer patients were classified into high-risk and low-risk groups based on the risk model. Subsequently, multivariate COX regression was employed to identify the prognostic marker. Additionally, the expression of SMURF2 was validated using quantitative real-time polymerase chain reaction (qRT-PCR) and the Human Protein Atlas (HPA) database. To investigate the relationship between SMURF2 expression and immune cell infiltrations, we conducted single-sample Gene Set Enrichment Analysis (ssGSEA) analyses. RESULTS We identified 16 differentially expressed TRGs in oral cancer, all of which showed upregulation. From these, we selected eight TRGs as prognostic signatures. Furthermore, the high-risk group demonstrated lower infiltration levels of immune cells, immune score, and higher tumor purity. Interestingly, we also found that SMURF2 serves as an independent prognostic biomarker. SMURF2 was upregulated in oral cancer, as confirmed by public databases and qRT-PCR analysis. Importantly, our results indicate a close association between SMURF2 expression and the immune microenvironment. CONCLUSION The 8-TRG signature prognosis model that we constructed has the ability to predict the survival rate and immune activity of oral cancer patients. SMURF2 could be effective in recognizing prognosis and evaluating immune efficacy for oral cancer.
Collapse
Affiliation(s)
- Lu Deng
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, 332000, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, Jiangxi Province, 332000, China
| | - Zhihong Wu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Jiujiang University, Jiujiang, Jiangxi Province, 332000, China
| | - Chuanxi Sun
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, 332000, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, Jiangxi Province, 332000, China
| | - Zhe Liu
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, 332000, China.
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, Jiangxi Province, 332000, China.
| |
Collapse
|
25
|
Guo C, Sun H, Du Y, Dai X, Pang Y, Han Z, Xiong X, Li S, Zhang J, Zheng Q, Gui X. Specifically blocking αvβ8-mediated TGF-β signaling to reverse immunosuppression by modulating macrophage polarization. J Exp Clin Cancer Res 2025; 44:1. [PMID: 39743547 PMCID: PMC11697059 DOI: 10.1186/s13046-024-03250-1] [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: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Targeting the TGF-β pathway in tumor therapy has proven challenging due to the highly context-dependent functions of TGF-β. Integrin αvβ8, a pivotal activator of TGF-β, has been implicated in TGF-β signaling within tumors, as demonstrated by the significant anti-tumor effects of anti-αvβ8 antibodies. Nevertheless, the expression profile of αvβ8 remains a subject of debate, and the precise mechanisms underlying the anti-tumor effects of anti-αvβ8 antibodies are not yet fully elucidated. METHODS We utilized single-cell RNA sequencing to assess αvβ8 expression across various human tumors. An anti-αvβ8 antibody was developed and characterized for its binding and blocking properties in vitro. Cryo-EM single-particle analysis was employed to study the detailed interaction between αvβ8 and the antibody Fab fragment. The anti-tumor efficacy of the antibody was evaluated in syngeneic mouse models with varying levels of αvβ8 expression, both as a monotherapy and in combination with PD-1 antibodies. Human PBMCs were isolated to investigate αvβ8 expression in myeloid cells, and macrophages were exposed to the antibody to study its impact on macrophage polarization. Pharmacokinetic studies of the αvβ8 antibody were conducted in cynomolgus monkeys. RESULTS Integrin αvβ8 is notably expressed in certain tumor types and tumor-infiltrating macrophages. The specific αvβ8 antibody 130H2 demonstrated high affinity, specificity, and blocking potency in vitro. Cryo-EM analysis further revealed that 130H2 interacts exclusively with the β8 subunit, without binding to the αv subunit. In vivo studies showed that this antibody significantly inhibited tumor growth and alleviated immunosuppression by promoting immune cell infiltration. Furthermore, combining the antibody with PD-1 inhibition produced a synergistic anti-tumor effect. In human PBMCs, monocytes exhibited high αvβ8 expression, and the antibody directly modulated macrophage polarization. Tumors with elevated αvβ8 expression were particularly responsive to 130H2 treatment. Additionally, favorable pharmacokinetic properties were observed in cynomolgus monkeys. CONCLUSIONS In summary, integrin αvβ8 is highly expressed in certain tumors and tumor-infiltrating macrophages. Targeting αvβ8 with a blocking antibody significantly inhibits tumor growth by modulating macrophage polarization and enhancing immune cell infiltration. Combining αvβ8 targeting with PD-1 treatment markedly increases the sensitivity of immune-excluded tumors. These results support further clinical evaluation of αvβ8 antibodies.
Collapse
Affiliation(s)
- Cuicui Guo
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Hui Sun
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Yulei Du
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Xiaodong Dai
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Yu Pang
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Zhen Han
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Xinhui Xiong
- Nanjing Novoacine Biotechnology Co., Ltd, Nanjing, 210032, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Junhua Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China.
| | - Xun Gui
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China.
| |
Collapse
|
26
|
Lu Z, Dong H, Tu Z, Liu H. Expression, molecular mechanisms and therapeutic potentials of ATF1 in cancers. Life Sci 2025; 360:123256. [PMID: 39580140 DOI: 10.1016/j.lfs.2024.123256] [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: 09/17/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
Activating transcription factor 1 (ATF1) is a crucial cellular regulator, with its misregulation implicated in numerous cancers. As a key player in the ATF/CREB family, ATF1 modulates gene expression in response to extracellular signals, significantly impacting cancer progression. This review examines ATF1's structural features, its role in tumorigenesis, and its potential therapeutic applications. Data from various databases consistently show ATF1 overexpression in diverse cancers, associated with poor prognosis and aggressive phenotypes. The review explores ATF1's complex regulatory mechanisms, influencing cell proliferation, apoptosis, migration, invasion, and therapeutic resistance, and its interactions with regulatory networks. Emerging strategies targeting ATF1, such as engineered antibodies, natural compounds, and small molecule inhibitors, show efficacy in preclinical models. ATF1 may also act as a biomarker for personalized therapeutic response and resistance. Future research should focus on ATF1's role in the tumor microenvironment and its interaction with the immune system, potentially leading to new immunotherapeutic strategies. A deeper understanding of ATF1 could enhance cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Ziwen Lu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hangyu Dong
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhigang Tu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
27
|
Hanahan D, Michielin O, Pittet MJ. Convergent inducers and effectors of T cell paralysis in the tumour microenvironment. Nat Rev Cancer 2025; 25:41-58. [PMID: 39448877 DOI: 10.1038/s41568-024-00761-z] [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] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
Tumorigenesis embodies the formation of a heterotypic tumour microenvironment (TME) that, among its many functions, enables the evasion of T cell-mediated immune responses. Remarkably, most TME cell types, including cancer cells, fibroblasts, myeloid cells, vascular endothelial cells and pericytes, can be stimulated to deploy immunoregulatory programmes. These programmes involve regulatory inducers (signals-in) and functional effectors (signals-out) that impair CD8+ and CD4+ T cell activity through cytokines, growth factors, immune checkpoints and metabolites. Some signals target specific cell types, whereas others, such as transforming growth factor-β (TGFβ) and prostaglandin E2 (PGE2), exert broad, pleiotropic effects; as signals-in, they trigger immunosuppressive programmes in most TME cell types, and as signals-out, they directly inhibit T cells and also modulate other cells to reinforce immunosuppression. This functional diversity and redundancy pose a challenge for therapeutic targeting of the immune-evasive TME. Fundamentally, the commonality of regulatory programmes aimed at abrogating T cell activity, along with paracrine signalling between cells of the TME, suggests that many normal cell types are hard-wired with latent functions that can be triggered to prevent inappropriate immune attack. This intrinsic capability is evidently co-opted throughout the TME, enabling tumours to evade immune destruction.
Collapse
Affiliation(s)
- Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| | - Olivier Michielin
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mikael J Pittet
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
| |
Collapse
|
28
|
Biernacki MA, Bleakley M. Clinical trials, challenges, and changes in TCR-based therapeutics for hematologic malignancies. Expert Rev Hematol 2025; 18:21-31. [PMID: 39667756 DOI: 10.1080/17474086.2024.2441962] [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: 03/15/2024] [Revised: 08/14/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION T cells engineered to express antigen-specific T cell receptors (TCR; TCR-T) are a promising class of immunotherapeutic for patients with hematologic malignancies. Like chimeric antigen receptor-engineered T cells (CAR-T), TCR-T are cell products with defined specificity and composition. Unlike CAR-T, TCR-T can recognize targets arising both from intracellular and cell surface proteins and leverage the sensitivity of natural TCR signaling machinery. A growing number of TCR-T targeting various antigens in different hematologic malignancies are in early-phase clinical trials, and more are in preclinical development. AREAS COVERED This review covers results from early-phase TCR-T clinical trials for hematologic malignancies. Challenges in the field are reviewed, including identifying optimal targets, engaging CD4+ help for CD8+ T cells, and overcoming tumor-induced suppression; recent innovations to overcome these challenges are also highlighted. EXPERT OPINION In the future, TCR-T's promise for hematologic malignancies will be borne out in later-phase clinical trials and approvals for clinical use. Improved antigen discovery methods will help build the toolbox of targets needed for broadly applicable TCR-T. Rationally designed TCR-T modifications including incorporation of accessory receptors and gene editing will enhance TCR-T function. New hybrid receptors combining features of TCR and CAR will enter the clinic.
Collapse
MESH Headings
- Humans
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- Hematologic Neoplasms/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Clinical Trials as Topic
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Antigens, Neoplasm/immunology
- Animals
Collapse
Affiliation(s)
| | - Marie Bleakley
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
Santiago-Sánchez GS, Fabian KP, Hodge JW. A landscape of checkpoint blockade resistance in cancer: underlying mechanisms and current strategies to overcome resistance. Cancer Biol Ther 2024; 25:2308097. [PMID: 38306161 PMCID: PMC10841019 DOI: 10.1080/15384047.2024.2308097] [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: 08/31/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
The discovery of immune checkpoints and the development of immune checkpoint inhibitors (ICI) have achieved a durable response in advanced-stage cancer patients. However, there is still a high proportion of patients who do not benefit from ICI therapy due to a lack of response when first treated (primary resistance) or detection of disease progression months after objective response is observed (acquired resistance). Here, we review the current FDA-approved ICI for the treatment of certain solid malignancies, evaluate the contrasting responses to checkpoint blockade in different cancer types, explore the known mechanisms associated with checkpoint blockade resistance (CBR), and assess current strategies in the field that seek to overcome these mechanisms. In order to improve current therapies and develop new ones, the immunotherapy field still has an unmet need in identifying other molecules that act as immune checkpoints, and uncovering other mechanisms that promote CBR.
Collapse
Affiliation(s)
- Ginette S. Santiago-Sánchez
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kellsye P. Fabian
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W. Hodge
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Du W, Tang Z, Du A, Yang Q, Xu R. Bidirectional crosstalk between the epithelial-mesenchymal transition and immunotherapy: A bibliometric study. Hum Vaccin Immunother 2024; 20:2328403. [PMID: 38502119 PMCID: PMC10956627 DOI: 10.1080/21645515.2024.2328403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Immunotherapy has recently attracted considerable attention. However, currently, a thorough analysis of the trends associated with the epithelial-mesenchymal transition (EMT) and immunotherapy is lacking. In this study, we used bibliometric tools to provide a comprehensive overview of the progress in EMT-immunotherapy research. A total of 1,302 articles related to EMT and immunotherapy were retrieved from the Web of Science Core Collection (WOSCC). The analysis indicated that in terms of the volume of research, China was the most productive country (49.07%, 639), followed by the United States (16.89%, 220) and Italy (3.6%, 47). The United States was the most influential country according to the frequency of citations and citation burstiness. The results also suggested that Frontiers in Immunotherapy can be considered as the most influential journal with respect to the number of articles and impact factors. "Immune infiltration," "bioinformatics analysis," "traditional Chinese medicine," "gene signature," and "ferroptosis" were found to be emerging keywords in EMT-immunotherapy research. These findings point to potential new directions that can deepen our understanding of the mechanisms underlying the combined effects of immunotherapy and EMT and help develop strategies for improving immunotherapy.
Collapse
Affiliation(s)
- Wei Du
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| | - Zemin Tang
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| | - Ashuai Du
- Department of Infectious Diseases, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Qinglong Yang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
- Department of General Surgery, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Rong Xu
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan, China
| |
Collapse
|
31
|
Tian H, Wang W, Liang S, Ding J, Hua D. From darkness to light: Targeting CAFs as a new potential strategy for cancer treatment. Int Immunopharmacol 2024; 143:113482. [PMID: 39476569 DOI: 10.1016/j.intimp.2024.113482] [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: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024]
Abstract
Cancer-associated fibroblasts (CAFs), which are the most frequent stromal cells in the tumor microenvironment (TME), play a key role in the metastasis of tumor cells. Generally speaking, CAFs in cooperation with tumor cells can secrete various cytokines, proteins, growth factors, and metabolites to promote angiogenesis, mediate immune escape of tumor cells, enhance endothelial-to-mesenchymal transition, stimulate extracellular matrix remodeling, and preserve tumor cell stemness. These activities of CAFs provide a favorable exogenous pathway for tumor progression and metastasis, and a microenvironment that allows rapid growth of tumor cells, which always lead to poor prognosis for patients. More importantly, it seems that targeting CAFs is also a potential precision therapeutic strategy in clinical practice. Hence, this review outlines the origin of CAFs, the relationship between CAFs and cancer metastasis, and targeting CAFs as a potential strategy for cancer patients, which could give some inspirations for cancer treatment in clinic.
Collapse
Affiliation(s)
- Haixia Tian
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Weijing Wang
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Shuai Liang
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Junli Ding
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| | - Dong Hua
- Department of Oncology, The Affliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
32
|
Chen S, Cheng S, Cai J, Liu Z, Li H, Wang P, Li Y, Yang F, Chen K, Qiu M. The current therapeutic cancer vaccines landscape in non-small cell lung cancer. Int J Cancer 2024; 155:1909-1927. [PMID: 39109825 DOI: 10.1002/ijc.35088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 10/04/2024]
Abstract
Currently, conventional immunotherapies for the treatment of non-small cell lung cancer (NSCLC) have low response rates and benefit only a minority of patients, particularly those with advanced disease, so novel therapeutic strategies are urgent deeded. Therapeutic cancer vaccines, a form of active immunotherapy, harness potential to activate the adaptive immune system against tumor cells via antigen cross-presentation. Cancer vaccines can establish enduring immune memory and guard against recurrences. Vaccine-induced tumor cell death prompts antigen epitope spreading, activating functional T cells and thereby sustaining a cancer-immunity cycle. The success of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rendered cancer vaccines a promising avenue, especially when combined with immunotherapy or chemoradiotherapy for NSCLC. This review delves into the intricate antitumor immune mechanisms underlying therapeutic cancer vaccines, enumerates the tumor antigen spectrum of NSCLC, discusses different cancer vaccines progress and summarizes relevant clinical trials. Additionally, we analyze the combination strategies, current limitations, and future prospects of cancer vaccines in NSCLC treatment, aiming to offer fresh insights for their clinical application in managing NSCLC. Overall, cancer vaccines offer promising potential for NSCLC treatment, particularly combining with chemoradiotherapy or immunotherapy could further improve survival in advanced patients. Exploring inhaled vaccines or prophylactic vaccines represents a crucial research avenue.
Collapse
Affiliation(s)
- Shaoyi Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Sida Cheng
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Jingsheng Cai
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Zheng Liu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Haoran Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Kezhong Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Mantang Qiu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
33
|
Mahmud S, Ajadee A, Hossen MB, Islam MS, Ahmmed R, Ali MA, Mollah MMH, Reza MS, Mollah MNH. Gene-expression profile analysis to disclose diagnostics and therapeutics biomarkers for thyroid carcinoma. Comput Biol Chem 2024; 113:108245. [PMID: 39454454 DOI: 10.1016/j.compbiolchem.2024.108245] [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: 03/28/2024] [Revised: 09/15/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
The most frequent endocrine cancer of the head and neck is thyroid carcinoma (THCA). Although there is increasing evidence linking THCA to genetic alterations, the exact molecular mechanism behind this relationship is not yet completely known to the researchers. There is still much to learn about THCA's molecular roots and genetic biomarkers. Though drug therapies are the best choice after metastasis, unfortunately, the majority of the patients progressively develop resistance against the therapeutic drugs after receiving them for a few years. Therefore, multi-targeted different variants of therapeutic drugs may be essential for effective treatment against THCA. To understand molecular mechanisms of THCA development and progression and explore multi-targeted different variants of therapeutic drugs, we detected 80 common differentially expressed genes (cDEGs) between THCA and non-THCA samples from six microarray gene expression datasets using the statistical LIMMA approach. Through protein-protein interaction (PPI) network analysis, we identified the top-ranked eight differentially expressed genes (TIMP1, FN1, THBS1, RUNX2, SHANK2, TOP2A, LRP2, and ACTN1) as the THCA-causing key genes (KGs), where 6 KGs (TIMP1, TOP2A, FN1, ACTN1, RUNX2, THBS1) are upregulated and 2 KGs (LRP2, SHANK2) are downregulated. The expression pattern analysis of KGs with the independent TCGA database by Box plots also confirmed their upregulated and downregulated patterns. The expression analysis of KGs in different stages of THCA development indicated that these KGs might be utilized as early diagnostic and prognostic biomarkers. The pan-cancer analysis of KGs indicated a substantial correlation of KGs with multiple cancers, including THCA. Some transcription factors (TFs) and microRNAs were detected as the key transcriptional and post-transcriptional regulators of KGs using gene regulatory network (GRN) analysis. The enrichment analysis of the cDEGs revealed several key molecular functions, biological processes, cellular components, and pathways significantly associated with THCA. These findings highlight critical mechanisms influenced by the identified key genes (KGs), providing deeper insight into their roles in THCA development. Then we detected 6 repurposable drug molecules (Entrectinib, Imatinib, Ponatinib, Sorafenib, Retevmo, and Pazopanib) by molecular docking with KGs-mediated receptor proteins, ADME/T analysis, and cross-validation with the independent receptors. Therefore, these findings might be useful resources for wet lab researchers and clinicians to consider an effective treatment strategy against THCA.
Collapse
Affiliation(s)
- Sabkat Mahmud
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Alvira Ajadee
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Bayazid Hossen
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; Department of Agricultural and Applied Statistics, Bangladesh Agricultural University (BAU), Bangladesh
| | - Md Saiful Islam
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Reaz Ahmmed
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; Department of Biochemistry and Molecular Biology, University of Rajshahi, Bangladesh
| | - Md Ahad Ali
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; Department of Chemistry, University of Rajshahi, Rajshahi 6205, Bangladesh
| | | | - Md Selim Reza
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; Department of Biomedical Informatics and Genomics, Tulane University, USA
| | - Md Nurul Haque Mollah
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh.
| |
Collapse
|
34
|
Pan J, Zhang C, Huang H, Zhu Y, Zhang Y, Wu S, Zhao YC, Chen F. Deciphering the Prognostic and Therapeutic Value of a Gene Model Associated with Two Aggressive Hepatocellular Carcinoma Phenotypes Using Machine Learning. J Hepatocell Carcinoma 2024; 11:2373-2390. [PMID: 39634327 PMCID: PMC11614714 DOI: 10.2147/jhc.s480358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Background Macrotrabecular-massive (MTM) and vessels encapsulating tumor clusters (VETC)-hepatocellular carcinoma (HCC) are aggressive histopathological phenotypes with significant prognostic implications. However, the molecular markers associated with MTM-HCC and VETC-HCC and their implications for clinical outcomes and therapeutic strategies remain unclear. Methods Utilizing the TCGA-LIHC cohort, we employed machine learning techniques to develop a prognostic risk score based on MTM and VETC-related genes. The performance of the risk score was assessed by investigating various aspects including clinical outcomes, biological pathways, treatment responses, drug sensitivities, tumor microenvironment, and molecular subclasses. To validate the risk score, additional data from the ICGC-JP, GSE14520, GSE104580, GSE109211, and an in-house cohort were collected and analyzed. Results The machine learning algorithm established a 4-gene-based risk score. High-risk patients had significantly worse prognosis compared to low-risk patients, with the risk score being associated with malignant progression of HCC. Functionally, the high-risk group exhibited enrichment in tumor proliferation pathways. Additionally, patients in the low-risk group exhibited improved response to TACE and sorafenib treatments compared to the high-risk group. In contrast, the high-risk group exhibited reduced sensitivity to immunotherapy and increased sensitivity to paclitaxel. In the in-house cohort, high-risk patients displayed higher rates of early recurrence, along with an increased frequency of elevated alpha-fetoprotein, microvascular invasion, and aggressive MRI features associated with HCC. Conclusion This study has successfully developed a risk score based on MTM and VETC-related genes, providing a promising tool for prognosis prediction and personalized treatment strategies in HCC patients.
Collapse
Affiliation(s)
- Junhan Pan
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Cong Zhang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Hepatobiliary and Pancreatic Interventional Treatment Center, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Huizhen Huang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yanyan Zhu
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yuhao Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Shuzhen Wu
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yan-Ci Zhao
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Feng Chen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
35
|
Berry LK, Pullikuth AK, Stearns KL, Wang Y, Wagner CJ, Chou JW, Darby JP, Kelly MG, Mall R, Leung M, Chifman J, Miller LD. A patient stratification signature mirrors the immunogenic potential of high grade serous ovarian cancers. J Transl Med 2024; 22:1048. [PMID: 39568014 PMCID: PMC11577735 DOI: 10.1186/s12967-024-05846-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND While high-grade serous ovarian cancer (HGSC) has proven largely resistant to immunotherapy, sporadic incidents of partial and complete response have been observed in clinical trials and case reports. These observations suggest that a molecular basis for effective immunity may exist within a subpopulation of HGSC. Herein, we developed an algorithm, CONSTRU (Computing Prognostic Marker Dependencies by Successive Testing of Gene-Stratified Subgroups), to facilitate the discovery and characterization of molecular backgrounds of HGSC that confer resistance or susceptibility to protective anti-tumor immunity. METHODS We used CONSTRU to identify genes from tumor expression profiles that influence the prognostic power of an established immune cytolytic activity signature (CYTscore). From the identified genes, we developed a stratification signature (STRATsig) that partitioned patient populations into tertiles that varied markedly by CYTscore prognostic power. The tertile groups were then analyzed for distinguishing biological, clinical and immunological properties using integrative bioinformatics approaches. RESULTS Patient survival and molecular measures of immune suppression, evasion and dysfunction varied significantly across STRATsig tertiles in validation cohorts. Tumors comprising STRATsig tertile 1 (S-T1) showed no immune-survival benefit and displayed a hyper-immune suppressed state marked by activation of TGF-β, Wnt/β-catenin and adenosine-mediated immunosuppressive pathways, with concurrent T cell dysfunction, reduced potential for antigen presentation, and enrichment of cancer-associated fibroblasts. By contrast, S-T3 tumors exhibited diminished immunosuppressive signaling, heightened antigen presentation machinery, lowered T cell dysfunction, and a significant CYTscore-survival benefit that correlated with mutational burden in a manner consistent with anti-tumor immunoediting. These tumors also showed elevated activity of DNA damage/repair, cell cycle/proliferation and oxidative phosphorylation, and displayed greater proportions of Th1 CD4 + T cells. In these patients, but not those of S-T1 or S-T2, validated predictors of immunotherapy response were prognostic of longer patient survival. Further analyses showed that STRATsig tertile properties were not explained by known HGSC molecular or clinical subtypes or singular immune mechanisms. CONCLUSIONS STRATsig is a composite of parallel immunoregulatory pathways that mirrors tumor immunogenic potential. Approximately one-third of HGSC cases classify as S-T3 and display a hypo-immunosuppressed and antigenic molecular composition that favors immunologic tumor control. These patients may show heightened responsiveness to current immunotherapies.
Collapse
Affiliation(s)
- Laurel K Berry
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Ashok K Pullikuth
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Kristen L Stearns
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yuezhu Wang
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Calvin J Wagner
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jeff W Chou
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA
| | - Janelle P Darby
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Michael G Kelly
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Biotechnology Research Center, Technology Innovation Institute, P.O. Box 9639, Abu Dhabi, United Arab Emirates
| | - Ming Leung
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA
| | - Julia Chifman
- Department of Mathematics and Statistics, American University, Washington, DC, 20016, USA
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
36
|
Wang T, Liu Y, Kong J, Liu J. Identification of a novel molecular classification for hepatocellular carcinoma based on disulfideptosis-related genes and its potential prognostic significance. J Cancer Res Clin Oncol 2024; 150:506. [PMID: 39551857 PMCID: PMC11570565 DOI: 10.1007/s00432-024-06031-7] [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: 09/17/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Globally, hepatocellular carcinoma (HCC) is one of the most prevalent and deadly malignant tumors. A recent study proposed disulfidptosis, a novel form of regulated cell death (RCD), offering a new avenue for identifying tumor prognosis biomarkers and developing novel therapeutic targets. METHODS Based on the expression data of 14 disulfideptosis-related genes extracted from public databases, a new molecular classification of HCC called the "disulfidptosis score" was constructed and its relationship to tumor immunity and prognosis was evaluated. RESULTS Based on the expression of disulfideptosis-related genes, we performed cluster analysis on HCC samples from the TCGA cohort, which classified these patients into three clusters: A, B, and C, and the differentially expressed genes of different clusters were analyzed. A disulfidptosis score model was constructed by differentially expressed genes associated with prognosis. Univariate and multivariate COX regression analysis showed that disulfidptosis score was an independent prognostic factor for HCC. In addition, in various disulfidptosis score groups, notable disparities were observed concerning the tumor immune microenvironment as well as the expression of immune checkpoint. CONCLUSION Disulfidptosis score have an important role in predicting HCC prognosis and help guide us in providing better immunotherapy options for patients.
Collapse
Affiliation(s)
- Tao Wang
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China
| | - Yong Liu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China
| | - Junjie Kong
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China
| | - Jun Liu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Weiqi Road, Jinan, 250000, China.
| |
Collapse
|
37
|
Fesneau O, Samson KA, Rosales W, Jones B, Moudgil T, Fox BA, Rajamanickam V, Duhen T. IL-12 drives the expression of the inhibitory receptor NKG2A on human tumor-reactive CD8 T cells. Nat Commun 2024; 15:9988. [PMID: 39557863 PMCID: PMC11574270 DOI: 10.1038/s41467-024-54420-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
Blockade of NKG2A/HLA-E interaction is a promising strategy to unleash the anti-tumor response. Yet the role of NKG2A+ CD8 T cells in the anti-tumor response and the regulation of NKG2A expression on human tumor-infiltrating T cells are still poorly understood. Here, by performing CITE-seq on T cells derived from head and neck squamous cell carcinoma and colorectal cancer, we show that NKG2A expression is induced on CD8 T cells differentiating into cytotoxic, CD39+CD103+ double positive (DP) cells, a phenotype associated with tumor-reactive T cells. This developmental trajectory leads to TCR repertoire overlap between the NKG2A- and NKG2A+ DP CD8 T cells, suggesting shared antigen specificities. Mechanistically, IL-12 is essential for the expression of NKG2A on CD8 T cells in a CD40/CD40L- dependent manner, in conjunction with TCR stimulation. Our study thus reveals that NKG2A is induced by IL-12 on human tumor-reactive CD8 T cells exposed to a TGF-β-rich environment, highlighting an underappreciated immuno-regulatory feedback loop dependent on IL-12 stimulation.
Collapse
Affiliation(s)
- Olivier Fesneau
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Kimberly A Samson
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Wesley Rosales
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Bretton Jones
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Tarsem Moudgil
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | - Thomas Duhen
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| |
Collapse
|
38
|
Brenes AJ, Griesser E, Sinclair LV, Davidson L, Prescott AR, Singh F, Hogg EKJ, Espejo-Serrano C, Jiang H, Yoshikawa H, Platani M, Swedlow JR, Findlay GM, Cantrell DA, Lamond AI. Proteomic and functional comparison between human induced and embryonic stem cells. eLife 2024; 13:RP92025. [PMID: 39540879 PMCID: PMC11563575 DOI: 10.7554/elife.92025] [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] [Indexed: 11/16/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have great potential to be used as alternatives to embryonic stem cells (hESCs) in regenerative medicine and disease modelling. In this study, we characterise the proteomes of multiple hiPSC and hESC lines derived from independent donors and find that while they express a near-identical set of proteins, they show consistent quantitative differences in the abundance of a subset of proteins. hiPSCs have increased total protein content, while maintaining a comparable cell cycle profile to hESCs, with increased abundance of cytoplasmic and mitochondrial proteins required to sustain high growth rates, including nutrient transporters and metabolic proteins. Prominent changes detected in proteins involved in mitochondrial metabolism correlated with enhanced mitochondrial potential, shown using high-resolution respirometry. hiPSCs also produced higher levels of secreted proteins, including growth factors and proteins involved in the inhibition of the immune system. The data indicate that reprogramming of fibroblasts to hiPSCs produces important differences in cytoplasmic and mitochondrial proteins compared to hESCs, with consequences affecting growth and metabolism. This study improves our understanding of the molecular differences between hiPSCs and hESCs, with implications for potential risks and benefits for their use in future disease modelling and therapeutic applications.
Collapse
Affiliation(s)
- Alejandro J Brenes
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dow St, Dundee DD1 5EHDundeeUnited Kingdom
| | - Eva Griesser
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Linda V Sinclair
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dow St, Dundee DD1 5EHDundeeUnited Kingdom
| | - Alan R Prescott
- Dundee Imaging Facility, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Francois Singh
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Elizabeth KJ Hogg
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Carmen Espejo-Serrano
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Hao Jiang
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Harunori Yoshikawa
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Melpomeni Platani
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Jason R Swedlow
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Greg M Findlay
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Doreen A Cantrell
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Angus I Lamond
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| |
Collapse
|
39
|
Thomas CJ, Delgado K, Sawant K, Roy J, Gupta U, Song CS, Poojary R, de Figueiredo P, Song J. Harnessing Bacterial Agents to Modulate the Tumor Microenvironment and Enhance Cancer Immunotherapy. Cancers (Basel) 2024; 16:3810. [PMID: 39594765 PMCID: PMC11593222 DOI: 10.3390/cancers16223810] [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: 09/25/2024] [Revised: 10/22/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer immunotherapy has revolutionized cancer treatment by leveraging the immune system to attack tumors. However, its effectiveness is often hindered by the immunosuppressive tumor microenvironment (TME), where a complex interplay of tumor, stromal, and immune cells undermines antitumor responses and allows tumors to evade immune detection. This review explores innovative strategies to modify the TME and enhance immunotherapy outcomes, focusing on the therapeutic potential of engineered bacteria. These bacteria exploit the unique characteristics of the TME, such as abnormal vasculature and immune suppression, to selectively accumulate in tumors. Genetically modified bacteria can deliver therapeutic agents, including immune checkpoint inhibitors and cytokines, directly to tumor sites. This review highlights how bacterial therapeutics can target critical immune cells within the TME, such as myeloid-derived suppressor cells and tumor-associated macrophages, thereby promoting antitumor immunity. The combination of bacterial therapies with immune checkpoint inhibitors or adoptive cell transfer presents a promising strategy to counteract immune suppression. Continued research in this area could position bacterial agents as a powerful new modality to reshape the TME and enhance the efficacy of cancer immunotherapy, particularly for tumors resistant to conventional treatments.
Collapse
Affiliation(s)
- Christina James Thomas
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Kaylee Delgado
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Kamlesh Sawant
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Jacob Roy
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Udit Gupta
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Carly Shaw Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Rayansh Poojary
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Paul de Figueiredo
- Department of Molecular Microbiology and Immunology, The University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| |
Collapse
|
40
|
Wen J, Zhang X, Wong CC, Zhang Y, Pan Y, Zhou Y, Cheung AHK, Liu Y, Ji F, Kang X, Liu D, Yu J. Targeting squalene epoxidase restores anti-PD-1 efficacy in metabolic dysfunction-associated steatohepatitis-induced hepatocellular carcinoma. Gut 2024; 73:2023-2036. [PMID: 38744443 PMCID: PMC11671884 DOI: 10.1136/gutjnl-2023-331117] [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: 09/11/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
OBJECTIVE Squalene epoxidase (SQLE) promotes metabolic dysfunction-associated steatohepatitis-associated hepatocellular carcinoma (MASH-HCC), but its role in modulating the tumour immune microenvironment in MASH-HCC remains unclear. DESIGN We established hepatocyte-specific Sqle transgenic (tg) and knockout mice, which were subjected to a choline-deficient high-fat diet plus diethylnitrosamine to induce MASH-HCC. SQLE function was also determined in orthotopic and humanised mice. Immune landscape alterations of MASH-HCC mediated by SQLE were profiled by single-cell RNA sequencing and flow cytometry. RESULTS Hepatocyte-specific Sqle tg mice exhibited a marked increase in MASH-HCC burden compared with wild-type littermates, together with decreased tumour-infiltrating functional IFN-γ+ and Granzyme B+ CD8+ T cells while enriching Arg-1+ myeloid-derived suppressor cells (MDSCs). Conversely, hepatocyte-specific Sqle knockout suppressed tumour growth with increased cytotoxic CD8+ T cells and reduced Arg-1+ MDSCs, inferring that SQLE promotes immunosuppression in MASH-HCC. Mechanistically, SQLE-driven cholesterol accumulation in tumour microenvironment underlies its effect on CD8+ T cells and MDSCs. SQLE and its metabolite, cholesterol, impaired CD8+ T cell activity by inducing mitochondrial dysfunction. Cholesterol depletion in vitro abolished the effect of SQLE-overexpressing MASH-HCC cell supernatant on CD8+ T cell suppression and MDSC activation, whereas cholesterol supplementation had contrasting functions on CD8+ T cells and MDSCs treated with SQLE-knockout supernatant. Targeting SQLE with genetic ablation or pharmacological inhibitor, terbinafine, rescued the efficacy of anti-PD-1 treatment in MASH-HCC models. CONCLUSION SQLE induces an impaired antitumour response in MASH-HCC via attenuating CD8+ T cell function and augmenting immunosuppressive MDSCs. SQLE is a promising target in boosting anti-PD-1 immunotherapy for MASH-HCC.
Collapse
Affiliation(s)
- Jun Wen
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yating Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yasi Pan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yunfei Zhou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yali Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fenfen Ji
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xing Kang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dabin Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
41
|
Chen SY, Kung HC, Espinoza B, Washington I, Chen K, Wang J, Zlomke H, Loycano M, Wang R, Pickup M, Burns WR, Fu J, Hwang WL, Zheng L. Targeting heterogeneous tumor microenvironments in pancreatic cancer mouse models of metastasis by TGF-β depletion. JCI Insight 2024; 9:e182766. [PMID: 39298276 DOI: 10.1172/jci.insight.182766] [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: 05/08/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024] Open
Abstract
The dual tumor-suppressive and -promoting functions of TGF-β signaling has made its targeting challenging. We examined the effects of TGF-β depletion by AVID200/BMS-986416 (TGF-β-TRAP), a TGF-β ligand trap, on the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC) murine models with different organ-specific metastasis. Our study demonstrated that TGF-β-TRAP potentiates the efficacy of anti-programmed cell death 1 (anti-PD-1) in a PDAC orthotopic murine model with liver metastasis tropism, significantly reducing liver metastases. We further demonstrated the heterogeneous response of cytotoxic effector T cells to combination TGF-β-TRAP and anti-PD-1 treatment across several tumor models. Single-nuclear RNA sequencing suggested that TGF-β-TRAP modulates cancer-associated fibroblast (CAF) heterogeneity and suppresses neutrophil degranulation and CD4+ T cell response to neutrophil degranulation. Ligand-receptor analysis indicated that TGF-β-TRAP may modulate the CCL5/CCR5 axis as well as costimulatory and checkpoint signaling from CAFs and myeloid cells. Notably, the most highly expressed ligands of CCR5 shifted from the immunosuppressive CCL5 to CCL7 and CCL8, which may mediate the immune agonist activity of CCR5 following TGF-β-TRAP and anti-PD-1 combination treatment. This study suggested that TGF-β depletion modulates CAF heterogeneity and potentially reprograms CAFs and myeloid cells into antitumor immune agonists in PDAC, supporting the validation of such effects in human specimens.
Collapse
Affiliation(s)
- Sophia Y Chen
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heng-Chung Kung
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - Birginia Espinoza
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - India Washington
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - Kai Chen
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - Jianxin Wang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - Haley Zlomke
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - Michael Loycano
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - Rulin Wang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
| | | | - William R Burns
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Juan Fu
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
| | - William L Hwang
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center
- Pancreatic Cancer Precision Medicine Center of Excellence Program, and
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Xue HY, Wei F. TGF-β: an active participant in the immune and metabolic microenvironment of multiple myeloma : TGF-β in the microenvironment of multiple myeloma. Ann Hematol 2024; 103:4351-4362. [PMID: 38900304 PMCID: PMC11534828 DOI: 10.1007/s00277-024-05843-4] [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: 03/04/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Although substantial quantities of potent therapies for multiple myeloma (MM) have been established, MM remains an incurable disease. In recent years, our understanding of the initiation, development, and metastasis of cancers has made a qualitative leap. Cancers attain the abilities to maintain proliferation signals, escape growth inhibitors, resist cell death, induce angiogenesis, and more importantly, escape anti-tumor immunity and reprogram metabolism, which are the hallmarks of cancers. Besides, different cancers have different tumor microenvironments (TME), thus, we pay more attention to the TME in the pathogenesis of MM. Many researchers have identified that myeloma cells interact with the components of TME, which is beneficial for their survival, ultimately causing the formation of immunosuppressive and high-metabolism TME. In the process, transforming growth factor-β (TGF-β), as a pivotal cytokine in the TME, controls various cells' fates and influences numerous metabolic pathways, including inhibiting immune cells to infiltrate the tumors, suppressing the activation of anti-tumor immune cells, facilitating more immunosuppressive cells, enhancing glucose and glutamine metabolism, dysregulating bone metabolism and so on. Thus, we consider TGF-β as the tumor promoter. However, in healthy cells and the early stage of tumors, it functions as a tumor suppressor. Due to the effect of context dependence, TGF-β has dual roles in TME, which attracts us to further explore whether targeting it can overcome obstacles in the treatment of MM by regulating the progression of myeloma, molecular mechanisms of drug resistance, and various signaling pathways in the immune and metabolic microenvironment. In this review, we predominantly discuss that TGF-β promotes the development of MM by influencing immunity and metabolism.
Collapse
Affiliation(s)
- Han-Yue Xue
- The First Clinical Medical College of Shanxi Medical University, 56 Xinjian South Road, Yingze District, Taiyuan, Shanxi, People's Republic of China
| | - Fang Wei
- Department of Hematology, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Yingze District, Taiyuan, Shanxi, People's Republic of China.
| |
Collapse
|
43
|
Rebaudi F, De Franco F, Goda R, Obino V, Vita G, Baronti C, Iannone E, Pitto F, Massa B, Fenoglio D, Jandus C, Poggio F, Fregatti P, Melaiu O, Bozzo M, Candiani S, Papaccio F, Greppi M, Pesce S, Marcenaro E. The landscape of combining immune checkpoint inhibitors with novel Therapies: Secret alliances against breast cancer. Cancer Treat Rev 2024; 130:102831. [PMID: 39342797 DOI: 10.1016/j.ctrv.2024.102831] [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: 08/13/2024] [Revised: 09/04/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
This review focuses on the immune checkpoint inhibitors (ICIs) in the context of breast cancer (BC) management. These innovative treatments, by targeting proteins expressed on both tumor and immune cells, aim to overcome tumor-induced immune suppression and reactivate the immune system. The potential of this approach is the subject of numerous clinical studies. Here, we explore the key studies and emerging therapies related to ICIs providing a detailed analysis of their specific and combined use in BC treatment.
Collapse
Affiliation(s)
- Federico Rebaudi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Fabiana De Franco
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Rayan Goda
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Valentina Obino
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Giorgio Vita
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Camilla Baronti
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Eleonora Iannone
- Breast Surgery Clinic, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca Pitto
- Department of Pathology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Barbara Massa
- Department of Pathology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Fenoglio
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy; Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Geneva Center for Inflammation Research, Geneva, Switzerland
| | - Francesca Poggio
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Piero Fregatti
- Breast Surgery Clinic, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Integrated Surgical and Diagnostic Sciences (DISC), University of Genoa, Genoa, Italy
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Matteo Bozzo
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy.
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
44
|
Patel B, Silwal A, Eltokhy MA, Gaikwad S, Curcic M, Patel J, Prasad S. Deciphering CD59: Unveiling Its Role in Immune Microenvironment and Prognostic Significance. Cancers (Basel) 2024; 16:3699. [PMID: 39518137 PMCID: PMC11545456 DOI: 10.3390/cancers16213699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND CD59, a GPI-anchored membrane protein, protects cancer cells from complement-dependent cytotoxicity (CDC) by inhibiting the formation of the membrane attack complex (MAC). It has been demonstrated to be overexpressed in most solid tumors, where it facilitates tumor cell escape from complement surveillance. The role of CD59 in cancer growth and interactions between CD59 and immune cells that modulate immune evasion has not been well explored. METHODS Using cancer patient database from The Cancer Genome Atlas (TCGA) and other public databases, we analyzed CD59 expression, its prognostic significance, and its association with immune cell infiltration in the tumor microenvironment, identifying associated genomic and functional networks and validating findings with invitro cell-line experimental data. RESULTS This article describes the abundant expression of CD59 in multiple tumors such as cervical squamous cell carcinoma (CESC), kidney renal cell carcinoma (KIRC), glioblastoma multiforme (GBM), head and neck squamous cell carcinoma (HNSC), and stomach adenocarcinoma (STAD), as well as in pan-cancer, using The Cancer Genome Atlas (TCGA) database and confirmed using multiple cancer cell lines. The expression of CD59 significantly alters the overall survival (OS) of patients with multiple malignancies such as CESC, GBM, HNSC, and STAD. Further, the correlation between CD59 and Treg and/or MDSC in the tumor microenvironment (TME) has shown to be strongly associated with poor outcomes in CESC, GBM, HNSC, and STAD as these tumors express high FOXP3 compared to KIRC. Moreover, unfavorable outcomes were strongly associated with the expression of CD59 and M2 tumor-associated macrophage infiltration in the TME via the IL10/pSTAT3 pathway in CESC and GBM but not in KIRC. In addition, TGFβ1-dominant cancers such as CESC, GBM, and HNSC showed a high correlation between CD59 and TGFβ1, leading to suppression of cytotoxic T cell activity. CONCLUSION Overall, the correlation between CD59 and immune cells predicts its prognosis as unfavorable in CESC, GBM, HNSC, and STAD while being favorable in KIRC.
Collapse
Affiliation(s)
- Bhaumik Patel
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA; (B.P.); (A.S.); (M.A.E.); (S.G.); (M.C.); (J.P.)
| | - Ashok Silwal
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA; (B.P.); (A.S.); (M.A.E.); (S.G.); (M.C.); (J.P.)
| | - Mohamed Ashraf Eltokhy
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA; (B.P.); (A.S.); (M.A.E.); (S.G.); (M.C.); (J.P.)
| | - Shreyas Gaikwad
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA; (B.P.); (A.S.); (M.A.E.); (S.G.); (M.C.); (J.P.)
| | - Marina Curcic
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA; (B.P.); (A.S.); (M.A.E.); (S.G.); (M.C.); (J.P.)
| | - Jalpa Patel
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA; (B.P.); (A.S.); (M.A.E.); (S.G.); (M.C.); (J.P.)
| | - Sahdeo Prasad
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Sharma M, Shetty SS, Soi S, Radhakrishnan R. Myofibroblasts persist through immune privilege mechanisms to mediate oral submucous fibrosis: Uncovering the pathogenesis. J Oral Biol Craniofac Res 2024; 14:773-781. [PMID: 39502133 PMCID: PMC11535754 DOI: 10.1016/j.jobcr.2024.10.008] [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: 07/23/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Immune privilege is the ability to tolerate foreign antigens without eliciting an inflammatory immune response. Several mechanisms explain a structure's immune privilege status, which is regulated by innate and adaptive immune responses. The role of myofibroblasts in perpetuating fibrosis by acquiring an immune privileged phenotype against the backdrop of oral submucous fibrosis (OSF) is evolving. Myofibroblasts persist through the Fas/FasL autocrine pathway and induce apoptosis in epithelial cells, explaining the juxtaposition of apoptotic cells in areas of fibrosis. However, increased matrix stiffness, in addition to activating TGF-β, reduces Fas surface expression in myofibroblasts, increasing their resistance to apoptosis. The reciprocal amplification loop between the immune checkpoint proteins programmed death-ligand 1 (PD-L1) and TGF-β involves the YAP-TAZ and SMAD2,3 pathways and dramatically enhances profibrotic signalling. Increased matrix stiffness also enhances cMYC expression, which subsequently amplifies PD-L1 levels on myofibroblasts. The increase in PD-L1 on the myofibroblast microengineers the phenotype of CD4+ T cells homing to fibrotic areas by acting on the programmed cell death protein 1 (PD-1) receptor on the T-cell surface, converting these cells from antifibrotic cells to profibrotic cells that produce IL-17A and TGF-β. This manuscript provides mechanistic insight into how myofibroblasts avoid apoptosis in OSFs by evading the immune system. Targeting an immune-privileged phenotype in myofibroblasts with FAS-FASL pathway-dependent characteristics is an ideal strategy for reversing OSF.
Collapse
Affiliation(s)
- Mohit Sharma
- Department of Oral Pathology, SGT Dental College Hospital & Research Institute, Gurugram, Haryana, 122505, India
| | - Smitha Sammith Shetty
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Sonal Soi
- Department of Conservative Dentistry and Endodontics, Manav Rachna Dental College, Faridabad, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India
- Academic Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S10 2TA, UK
| |
Collapse
|
46
|
Lai YW, Liu ZW, Lin MH, Yang CC, Chu CY, Chung CH, Lin CW. Melatonin increases Olaparib sensitivity and suppresses cancer-associated fibroblast infiltration via suppressing the LAMB3-CXCL2 axis in TNBC. Pharmacol Res 2024; 209:107429. [PMID: 39306019 DOI: 10.1016/j.phrs.2024.107429] [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: 07/26/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most malignant breast cancer subtype, characterized with high aggressiveness and a high recurrence rate. Olaparib is the first US Food and Drug Administration-approved poly(ADP ribose) polymerase (PARP) inhibitor (PARPi) to treat breast cancer patients with a germline BRCA1 or BRCA2 mutation. However, resistance to Olaparib treatment restricts the therapeutic effects, and thus novel therapeutics are urgently required. In the present study, we identified that the combination of melatonin and Olaparib synergistically enhanced the sensitivity of TNBC cells. Moreover, melatonin exerted promising antitumor activities in Olaparib-resistant cells, implying the potential for its clinical application. An RNA-sequencing analysis revealed that melatonin treatment downregulated laminin subunit beta 3 (LAMB3) expression. Genetic ablation of LAMB3 significantly increased Olaparib sensitivity, and subsequently suppressed proliferation, epithelial-to-mesenchymal transition (EMT)-related gene expressions, and aggressiveness of breast cancer cells. Accordingly, LAMB3 expression was positively correlated with C-X-C motif chemokine ligand 2 (CXCL2), and they collaboratively promoted cancer-associated fibroblast (CAF) infiltration. An in vivo study demonstrated that combined treatment with melatonin and Olaparib showed enhanced inhibitory efficacy against tumor growth, LAMB3 expression, CXCL2 levels, and CAF infiltration compared to single treatment groups, and combined treatment with melatonin and Olaparib significantly ameliorated the immunosuppressive tumor microenvironment. These findings illustrate a promising therapeutic strategy using melatonin to overcome Olaparib resistance and activate antitumor immunity via attenuating the LAMB3-CXCL2 axis in breast cancer patients.
Collapse
Affiliation(s)
- Yi-Wen Lai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Zei-Wei Liu
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hsiang Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan; Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Cheng-Ying Chu
- CRISPR Gene Targeting Core Lab, Taipei Medical University, Taipei, Taiwan
| | - Chu-Hung Chung
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Wei Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
47
|
Gnanagurusamy J, Krishnamoorthy S, Muthusami S. Transforming growth factor-β micro-environment mediated immune cell functions in cervical cancer. Int Immunopharmacol 2024; 140:112837. [PMID: 39111147 DOI: 10.1016/j.intimp.2024.112837] [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: 02/19/2024] [Revised: 07/02/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024]
Abstract
Propensity to develop cervical cancer (CC) in human papilloma virus (HPV) infected individual could potentially involve the impaired immune functioning. Several stages of HPV surveillance by immune cells in tumor micro-environment (TME) is regulated mainly by transforming growth factor-beta (TGF-β) and is crucial for the establishment of CC. The role of TGF-β in the initiation and progression of CC is very complex and involve different suppressor of mothers against decapentaplegic homolog (SMAD) dependent and SMAD independent signaling mechanism(s). This review summarizes the handling of HPV by immune cells such as T lymphocytes, B lymphocytes, natural killer cells (NK), dendritic cells (DC), monocytes, macrophages, myeloid derived suppressor cells (MDSC) and their regulation by TGF-β. The hijack mechanisms adapted by HPV to evade this surveillance process is discussed. Biomarkers indicating the stages of CC and immune checkpoints that can be targeted for improved outcome are included for immune-based theragnostics. This review also addresses the direct actions of TGF-β on CC cells and tumor/immune cell interactions. Therapies focused on targeting TGF-β using small molecule inhibitors, monoclonal antibodies and TGF-β chimeric antigen receptor (CAR)T cells are collated to understand the current strategies related to TGF-β in the management of CC.
Collapse
Affiliation(s)
- Jayapradha Gnanagurusamy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Sneha Krishnamoorthy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India; Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India.
| |
Collapse
|
48
|
Thinyakul C, Sakamoto Y, Shimoda M, Liu Y, Thongchot S, Reda O, Nita A, Sakamula R, Sampattavanich S, Maeda A, Chunthaboon P, Nduru D, Niimura M, Kanamori Y, Thuwajit P, Nakayama KI, Guan KL, Satou Y, Thuwajit C, Moroishi T. Hippo pathway in cancer cells induces NCAM1 +αSMA + fibroblasts to modulate tumor microenvironment. Commun Biol 2024; 7:1343. [PMID: 39420139 PMCID: PMC11487161 DOI: 10.1038/s42003-024-07041-4] [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: 01/30/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
Cancer cells adeptly manipulate the tumor microenvironment (TME) to evade host antitumor immunity. However, the role of cancer cell-intrinsic signaling in shaping the immunosuppressive TME remains unclear. Here, we found that the Hippo pathway in cancer cells orchestrates the TME by influencing the composition of cancer-associated fibroblasts (CAFs). In a 4T1 mouse breast cancer model, Hippo pathway kinases, large tumor suppressor 1 and 2 (LATS1/2), promoted the formation of neural cell adhesion molecule 1 (NCAM1)+alpha-smooth muscle actin (αSMA)+ CAFs expressing the transforming growth factor-β, which is associated with T cell inactivation and dysfunction. Depletion of LATS1/2 in cancer cells resulted in a less immunosuppressive TME, indicated by the reduced proportions of NCAM1+αSMA+ CAFs and dysfunctional T cells. Notably, similar Hippo pathway-induced NCAM1+αSMA+ CAFs were observed in human breast cancer, highlighting the potential of TME-manipulating strategies to reduce immunosuppression in cancer immunotherapy.
Collapse
Affiliation(s)
- Chanida Thinyakul
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Yasuhisa Sakamoto
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Mayuko Shimoda
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yanliang Liu
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Omnia Reda
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Akihiro Nita
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Division of Cellular Dynamics, Medical Research Laboratory, Institute of Science Tokyo, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Romgase Sakamula
- SirirajCenter of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Somponnat Sampattavanich
- SirirajCenter of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Ayato Maeda
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Paweenapon Chunthaboon
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - David Nduru
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Mayumi Niimura
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yohei Kanamori
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, 812-8582, Japan
- Anticancer Strategies Laboratory, Advanced Research Initiative, Institute of Science Tokyo, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou, 310030, China
| | - Yorifumi Satou
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
- Division of Cellular Dynamics, Medical Research Laboratory, Institute of Science Tokyo, Bunkyo-ku, Tokyo, 113-8510, Japan.
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Kumamoto, 860-8556, Japan.
| |
Collapse
|
49
|
Orcutt-Jahns B, Lima Junior JR, Lin E, Rockne RC, Matache A, Branciamore S, Hung E, Rodin AS, Lee PP, Meyer AS. Systems profiling reveals recurrently dysregulated cytokine signaling responses in ER+ breast cancer patients' blood. NPJ Syst Biol Appl 2024; 10:118. [PMID: 39389979 PMCID: PMC11467214 DOI: 10.1038/s41540-024-00447-0] [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: 04/17/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Cytokines operate in concert to maintain immune homeostasis and coordinate immune responses. In cases of ER+ breast cancer, peripheral immune cells exhibit altered responses to several cytokines, and these alterations are correlated strongly with patient outcomes. To develop a systems-level understanding of this dysregulation, we measured a panel of cytokine responses and receptor abundances in the peripheral blood of healthy controls and ER+ breast cancer patients across immune cell types. Using tensor factorization to model this multidimensional data, we found that breast cancer patients exhibited widespread alterations in response, including drastically reduced response to IL-10 and heightened basal levels of pSmad2/3 and pSTAT4. ER+ patients also featured upregulation of PD-L1, IL6Rα, and IL2Rα, among other receptors. Despite this, alterations in response to cytokines were not explained by changes in receptor abundances. Thus, tensor factorization helped to reveal a coordinated reprogramming of the immune system that was consistent across our cohort.
Collapse
Affiliation(s)
- Brian Orcutt-Jahns
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA
| | | | - Emily Lin
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Adina Matache
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Sergio Branciamore
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Ethan Hung
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA
| | - Andrei S Rodin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles (UCLA), CA, USA.
- Jonsson Comprehensive Cancer Center, Los Angeles (UCLA), CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Los Angeles (UCLA), CA, USA.
| |
Collapse
|
50
|
Zhang Z, Zhang F, Xie W, Niu Y, Wang H, Li G, Zhao L, Wang X, Xie W. Induced Necroptosis and Its Role in Cancer Immunotherapy. Int J Mol Sci 2024; 25:10760. [PMID: 39409087 PMCID: PMC11477008 DOI: 10.3390/ijms251910760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/30/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
Necroptosis is a type of regulated cell death (RCD) that is triggered by changes in the extracellular or intracellular milieu that are picked up by certain death receptors. Thanks to its potent capacity to induce immunological responses and overcome apoptotic resistance, it has garnered significant attention as a potential cancer treatment. Basic information for the creation of nano-biomedical treatments is provided by studies on the mechanisms underlying tumor necroptosis. Receptor-interacting protein kinase 1 (RIPK1)-RIPK3-mediated necroptosis, Toll-like receptor domain-containing adapter-inducing interferon (IFN)-β (TRIF)-RIPK3-mediated necroptosis, Z-DNA-binding protein 1 (ZBP1)-RIPK3-mediated necroptosis, and IFNR-mediated necroptosis are the four signaling pathways that collectively account for triggered necroptosis in this review. Necroptosis has garnered significant interest as a possible cancer treatment strategy because, in contrast to apoptosis, it elicits immunological responses that are relevant to therapy. Thus, a thorough discussion is held on the connections between tumor cell necroptosis and the immune environment, cancer immunosurveillance, and cells such as dendritic cells (DCs), cytotoxic T cells, natural killer (NK) cells, natural killer T (NKT) cells, and their respective cytokines. Lastly, a summary of the most recent nanomedicines that cause necroptosis in order to cause immunogenic cell death is provided in order to emphasize their promise for cancer immunotherapy.
Collapse
Affiliation(s)
- Ziyao Zhang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Fangming Zhang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Wenjing Xie
- School of Chemistry and Chemical Engineering, Guizhou University, Guiyang 550025, China;
| | - Yubo Niu
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Haonan Wang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Guofeng Li
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China;
| | - Xing Wang
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| | - Wensheng Xie
- The Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (Z.Z.); (F.Z.); (Y.N.); (H.W.); (G.L.)
| |
Collapse
|