1
|
Li M, Li D, Wang HY, Zhang W, Zhuo Z, Guo H, Liu J, Zhuo Y, Tang J, He J, Miao L. Leptin decreases Th17/Treg ratio to facilitate neuroblastoma via inhibiting long-chain fatty acid catabolism in tumor cells. Oncoimmunology 2025; 14:2460281. [PMID: 39902867 PMCID: PMC11796542 DOI: 10.1080/2162402x.2025.2460281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/05/2025] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
The exploration of therapeutic targets in neuroblastoma (NB), which needs more attempts, can benefit patients with high-risk NB. Based on metabolomic and transcriptomic data in mediastinal NB tissues, we found that the content of long-chain acylcarnitine (LCAC) was increased and positively associated with leptin expression in advanced NB. Leptin over-expression forced naïve CD4+ T cells to differentiate into Treg cells instead of Th17 cells, which benefited from NB cell proliferation, migration, and drug resistance. Mechanically, leptin in NB cells blunted the activity of carnitine palmitoyltransferase 2 (CPT2), the key enzyme for LCAC catabolism, by inhibiting sirtuin 3-mediated CPT2 deacetylation, which depresses oxidative phosphorylation (OXPHOS) for energy supply and increases lactic acid (LA) production from glycolysis to modulate CD4+ T cell differentiation. These findings highlight that excess leptin contributes to lipid metabolism dysfunction in NB cells and subsequently misdirects CD4+ T cell differentiation in tumor micro-environment (TME), indicating that targeting leptin could be a therapeutic strategy for retarding NB progression.
Collapse
Affiliation(s)
- Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Di Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Hai-Yun Wang
- Department of Pathology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, Guangdong, China
| | - Weixin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Zhenjian Zhuo
- Laboratory Animal Center, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Huiqin Guo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Jiabin Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yue Zhuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jue Tang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Zhou QM, Lu YF, Yang XY, Zhang JG, Wang YN, Luo WP, Mao J, Hou J, Wu F, Wang WL, Tang GP, Bai HZ, Yu RS. Redox-driven hybrid nanoenzyme dynamically activating ferroptosis and disulfidptosis for hepatocellular carcinoma theranostics. J Colloid Interface Sci 2025; 693:137611. [PMID: 40253866 DOI: 10.1016/j.jcis.2025.137611] [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: 01/26/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Hepatocellular carcinoma (HCC) presents formidable therapeutic challenges due to its pronounced metabolic heterogeneity, particularly arising from spatially uneven glucose availability within the tumor microenvironment (TME). To address this, we developed a glutathione (GSH)-responsive, biomimetic hybrid nanoenzyme system (M@GOx/Fe-HMON) composed of hollow mesoporous organosilica nanoparticles co-loaded with glucose oxidase (GOx) and Fe2+/Fe3+ redox pairs, and cloaked in homologous tumor cell membranes for enhanced targeting. In glucose-rich regions, the nanoenzyme orchestrates a GOx-peroxidase (POD) cascade that produces reactive oxygen species (ROS) via the Fenton reaction, leading to ferroptosis through intensified oxidative stress and GSH depletion. Conversely, under glucose-deficient conditions, the nanoenzyme promotes disulfidptosis by aggravating glucose deprivation, depleting nicotinamide adenine dinucleotide phosphate (NADPH), and impairing cystine metabolism, ultimately resulting in actin cytoskeletal collapse. This dual-action platform dynamically adapts to the tumor's metabolic landscape, selectively inducing ferroptosis or disulfidptosis according to glucose levels, disrupting redox homeostasis and amplifying antitumor efficacy. Notably, this study is the first to integrate ferroptosis and disulfidptosis activation into a single, metabolism-sensitive nanoenzyme system, providing a novel paradigm for exploiting tumor metabolic heterogeneity. Furthermore, the combination of endogenous metabolic regulation with magnetic resonance imaging (MRI)-guided diagnosis introduces an innovative and noninvasive strategy for precision cancer theranostics.
Collapse
Affiliation(s)
- Qiao-Mei Zhou
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Yuan-Fei Lu
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Xiao-Yan Yang
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Jin-Guo Zhang
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Yi-Ning Wang
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Wang-Ping Luo
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Jin Mao
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Jue Hou
- Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China
| | - Fan Wu
- Department of Neurosurgery, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Wei-Lin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Gu-Ping Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China.
| | - Hong-Zhen Bai
- Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China.
| | - Ri-Sheng Yu
- Department of Radiology, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, PR China.
| |
Collapse
|
3
|
Zhou S, Jiang D, Liu Y, Wang Q, Hu M, Dai K, Chen L, Zhang T, Cai C, Wang J. The role of Sine Oculis Homeobox Homolog 2 in colon Cancer: Insights into prognosis, immune regulation, and therapeutic implications. Biochem Biophys Res Commun 2025; 772:152038. [PMID: 40414014 DOI: 10.1016/j.bbrc.2025.152038] [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: 02/22/2025] [Revised: 04/19/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Colon cancer (CC) remains a significant global health burden, and the search for novel prognostic biomarkers and therapeutic targets is crucial. This study comprehensively analyzed the role of SIX2 (Sine Oculis Homeobox Homolog 2) in CC. Utilizing data from TCGA, GTEx, and CCLE databases, differential expression of SIX2 was observed in multiple cancers, with significant upregulation in many tumors compared to normal tissues. In CC, SIX2's differential expression was notable. Cox regression analysis revealed its prognostic significance, with overexpression associated with poor survival outcomes. SIX2 was strongly associated with gene alterations and correlated with key signaling pathways like WNT and TGF-β. In the tumor microenvironment, SIX2 was related to immune cell infiltration and immune-related molecules. Notably, in CC, it was associated with immunosuppressive cells and checkpoint molecules. Additionally, ABT737 was found to sensitize tumor immunotherapy in the context of SIX2. Animal experiments demonstrated that ABT737 effectively restricted the growth of CC in mice, and its combination with antiPD-1 immunotherapy was more effective. It could reduce the infiltration of CD163+ tumor-associated macrophages but without significantly increasing the infiltration of CD8+ T cells. Our findings suggest that SIX2 is a potential key player in CC, offering insights into future research and the development of targeted therapies.
Collapse
Affiliation(s)
- Shicheng Zhou
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Dan Jiang
- Department of Surgery, Wucheng District People's Hospital, Jinhua, Zhejiang, 321000, China
| | - Yu Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qin Wang
- Department of Endocrinology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Manyi Hu
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Kangfu Dai
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Lin Chen
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China
| | - Tianming Zhang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Cheng Cai
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China.
| | - Jianping Wang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China.
| |
Collapse
|
4
|
Wang H, Xu F, Wang C. Metabolic reprogramming of tumor microenviroment by engineered bacteria. Semin Cancer Biol 2025; 112:58-70. [PMID: 40157514 DOI: 10.1016/j.semcancer.2025.03.003] [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: 01/31/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The tumor microenvironment (TME) is a complex ecosystem that plays a crucial role in tumor progression and response to therapy. The metabolic characteristics of the TME are fundamental to its function, influencing not only cancer cell proliferation and survival but also the behavior of immune cells within the tumor. Metabolic reprogramming-where cancer cells adapt their metabolic pathways to support rapid growth and immune evasion-has emerged as a key factor in cancer immunotherapy. Recently, the potential of engineered bacteria in cancer immunotherapy has gained increasing recognition, offering a novel strategy to modulate TME metabolism and enhance antitumor immunity. This review summarizes the metabolic properties and adaptations of tumor and immune cells within the TME and summarizes the strategies by which engineered bacteria regulate tumor metabolism. We discuss how engineered bacteria can overcome the immunosuppressive TME by reprogramming its metabolism to improve antitumor therapy. Furthermore, we examine the advantages, potential challenges, and future clinical translation of engineered bacteria in reshaping TME metabolism.
Collapse
Affiliation(s)
- Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
5
|
Nair RS, Patel MN, Kannan T, Gour S, Hariharan MM, Prasanna V, Thirumalai A, Chockalingam R, Vasantharekha R, ThyagaRajan S, Priyanka HP. Effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. Toxicol In Vitro 2025; 106:106057. [PMID: 40112934 DOI: 10.1016/j.tiv.2025.106057] [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: 12/16/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Estrogen signaling plays a crucial role in immune regulation and cancer metabolism, yet its impact on T-cell leukemia remains unclear. In hematological malignancies, estrogen receptor (ER) activation may influence metabolic shifts that affect cell survival and proliferation. This study investigates the in vitro effects of 17β-estradiol and estrogen receptor subtype-specific agonists on Jurkat E6.1 T-cell leukemia cells. PURPOSE To assess how estrogen signaling influences metabolic reprogramming, inflammatory response, and survival pathways in Jurkat E6.1 cells through receptor-dependent and independent mechanisms. METHODS Jurkat E6.1 cells incubated with different concentrations of 17β-estradiol (10-12 M, 10-10 M, 10-8 M) or ER-α agonist 4,4',4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (10-10 M, 10-8 M, 10-6 M) or ER-β agonist diarylproprionitrile (10-10 M, 10-8 M, 10-6 M) with and without non-specific antagonist ICI 182,780 (10-6 M). The metabolic enzyme activities of hexokinase, pyruvate kinase, and citrate synthase were measured in cell pellets, while supernatants were analyzed for IL-6 and nitric oxide (NO) production. Additionally, PI3K/Akt pathway activation was assessed by measuring p-Akt/Total Akt expression. RESULTS A shift from glycolysis to oxidative phosphorylation was observed on treatment with 17β-estradiol with significant decline in hexokinase activity and a concomitant increase in activities of pyruvate kinase and citrate synthase. CONCLUSION 17β-estradiol mediates its effects on Jurkat E6.1 cells in vitro through receptor-subtype dependent and independent mechanisms involving metabolic enzymes (hexokinase, pyruvate kinase, citrate synthase), cytokines (IL-6), nitric oxide, and signaling molecules (p-Akt).
Collapse
Affiliation(s)
- Rahul S Nair
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Mantavya N Patel
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Thangamani Kannan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Shaili Gour
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Murali M Hariharan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Vijayarengamani Prasanna
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Anupriya Thirumalai
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramanathan Chockalingam
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramasamy Vasantharekha
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Hannah P Priyanka
- Inspire Laboratory, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Chennai, Tamil Nadu, India; Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
6
|
Qu B, Wang Q, Zhou Y, Ning X, Wang Q, Zhou Z, Bai P, Zhang R. pH-driven butterfly effect for cascade-amplified tumor therapy based on thalidomide coordinated Fe-HMME nanoplatform. Mater Today Bio 2025; 32:101691. [PMID: 40225132 PMCID: PMC11986554 DOI: 10.1016/j.mtbio.2025.101691] [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: 01/13/2025] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
A promising approach for treating intractable cancers has been presented by photodynamic therapy (PDT). However, the limited penetration depth of PDT and suboptimal monotherapy efficacy of PDT significantly restrict its clinical applications. In this study, we constructed an acidic tumor microenvironment (TME)-activated carrier-free nanoplatform (HMME-Fe-Thal, abbreviated as HFT) through self-assembly of iron ions, photosensitizer hematoporphyrinmonomethyl ether (HMME) and anti-angiogenesis drug thalidomide (Thal). Near infrared (NIR) triggers PDT behavior before the degradation of the HFT nanoplatform. Subsequently, the HFT nanoplatform degrades, releasing Thal for chemotherapy, iron ions for chemodynamic therapy (CDT), which reinforce the therapeutic benefits of PDT synergistically. Moreover, the iron ions released by HFT degradation turn on the MRI signal, which can suggest the most appropriate time for PDT, divide the treatment into two stages (First-stage: PDT, Second-stage: CDT/chemotherapy), and gradually achieve cascade-amplified tumor therapy. In this sense, HFT modulates TME and leads to a "butterfly effect" of CDT/chemotherapy/glutathione (GSH) depletion for enhanced PDT efficacy. This strategy compensates the deficient shadow penetration and poor treatment efficacy from PDT monotherapy. This work presents the selection and rational design of HFT constructed by endogenous components for tumor regression, and greatly push nanomaterials towards the development of PDT application.
Collapse
Affiliation(s)
- Botao Qu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Qian Wang
- School of Forensic Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Yuxin Zhou
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaogang Ning
- School of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Qian Wang
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, 030001, China
| | - Ziyi Zhou
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Peirong Bai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
7
|
Zhang J, Kong X, Zhou B, Li R, Yu Z, Zhu J, Xi Q, Li Y, Zhao Z, Zhang R. Metabolic reprogramming of drug resistance in pancreatic cancer: mechanisms and effects. Mol Aspects Med 2025; 103:101368. [PMID: 40398192 DOI: 10.1016/j.mam.2025.101368] [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: 01/19/2025] [Revised: 05/10/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
Pancreatic cancer is a highly aggressive gastrointestinal malignancy, often termed the "king of cancers" due to its notoriously high mortality rate. Its clinical characteristics, including late diagnosis, low surgical resectability, high recurrence rates, significant chemoresistance, and poor prognosis have collectively driven the persistent rise in incidence and mortality. Despite ongoing advancements in therapeutic strategies, the management of pancreatic cancer, particularly at advanced stages, remains challenging. Chemotherapy remains the mainstay of current treatment. However, the prevalent problem of chemotherapy resistance poses a significant obstacle to effective treatment. Metabolic reprogramming, characterized by alterations in glucose metabolism, lipid biosynthesis, and amino acid utilization, supports the high energy demands and rapid proliferation of cancer cells. Emerging evidence suggests that these metabolic changes, possibly mediated by epigenetic mechanisms, also contribute to tumorigenesis and metastasis. These findings highlight the critical role of metabolic alterations in pancreatic cancer pathogenesis. This review explores the relationship between metabolic reprogramming and chemotherapy resistance, discussing underlying mechanisms and summarizing preclinical studies and drug development targeting metabolism. The aim is to provide a comprehensive perspective on potential therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Jinyi Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xueqing Kong
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Boyan Zhou
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhaoan Yu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jinrong Zhu
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Xi
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan Li
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zichao Zhao
- Department of Emergency Medicine, Shaodong People's Hospital, Shaodong City, Hunan Province, China.
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory for Biotechnology Drug Candidates, Department of Biotechnology, Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou,The Second Clinical Medical School of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
8
|
Miao J, Wang D, Pang R, Zhang H, Wu Y, Sun X, Jin Y. Role of fructose in renal cell carcinoma progression. Discov Oncol 2025; 16:897. [PMID: 40410626 PMCID: PMC12102030 DOI: 10.1007/s12672-025-02688-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
Renal cell carcinoma (RCC) is a highly malignant tumor with a poor prognosis, underscoring the urgent need for novel therapeutic strategies. RCC cells exhibit rapid proliferation and high metabolic demands, leading to hypoglycemic and hypoxic conditions within the tumor microenvironment (TME). Our study reveals that the fructose transporter Glut5 is prominently expressed in RCC, facilitating increased fructose uptake. This compensatory mechanism supports RCC survival under glucose deprivation and hypoxia. Fructose utilization sustains RCC proliferation, migration, and colony formation in vitro, significantly reduces apoptosis, and accelerates renal cancer growth in vivo. Mechanistically, fructose activates the cAMP/PKA signaling pathway, driving metabolic reprogramming and promoting tumor progression. Furthermore, 2,5-dehydro-D-mannitol (2,5-AM), a competitive inhibitor of fructose transport, significantly inhibits RCC growth both in vivo and in vitro. These findings provide new insights into the role of fructose metabolism in RCC progression and suggest potential therapeutic targets.
Collapse
Grants
- SKY2023051 Research project of China Baoyuan Investment Co., Ltd., Suzhou Science and Technology Plan Project
- SKY2023051 Research project of China Baoyuan Investment Co., Ltd., Suzhou Science and Technology Plan Project
- SKY2023051 Research project of China Baoyuan Investment Co., Ltd., Suzhou Science and Technology Plan Project
- SKY2023051 Research project of China Baoyuan Investment Co., Ltd., Suzhou Science and Technology Plan Project
- SKY2023051 Research project of China Baoyuan Investment Co., Ltd., Suzhou Science and Technology Plan Project
- SKY2023051 Research project of China Baoyuan Investment Co., Ltd., Suzhou Science and Technology Plan Project
- SKY2023051 Research project of China Baoyuan Investment Co., Ltd., Suzhou Science and Technology Plan Project
Collapse
Affiliation(s)
- Jixuan Miao
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
- Department of Ultrasonography, Affiliated Jiangyin Hospital of Nantong University, Jiangyin People's Hospital, Wuxi, 214400, Jiangsu, China
| | - Di Wang
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Ruochong Pang
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Hao Zhang
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yuyun Wu
- Department of Ultrasonography, Affiliated Jiangyin Hospital of Nantong University, Jiangyin People's Hospital, Wuxi, 214400, Jiangsu, China
| | - Xingwei Sun
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yong Jin
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
9
|
Yang J, Chen R. Radiosensitization Strategies for Hepatocellular Carcinoma: Mechanisms, Therapeutic Advances, and Clinical Perspectives. Crit Rev Oncol Hematol 2025:104773. [PMID: 40412577 DOI: 10.1016/j.critrevonc.2025.104773] [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: 04/19/2025] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with treatment efficacy limited by late-stage diagnosis, frequent recurrence, and therapeutic resistance. Radiotherapy is a key local treatment for HCC; however, its efficacy is frequently limited by intrinsic tumor radioresistance. This review discusses strategies to improve the therapeutic response of HCC to radiotherapy. Targeting DNA repair mechanisms can block tumor cells from recovering after radiation-induced damage, whereas modulating cell cycle arrest and programmed cell death pathways (e.g., apoptosis, autophagy) diminishes their survival capacity. Furthermore, remodeling the tumor microenvironment-through hypoxia alleviation, metabolic reprogramming, oxidative stress regulation, and immune activation-may potentiate radiotherapy efficacy. Technological advances, such as stereotactic body radiotherapy and nanomaterial-based approaches, have also improved the precision and effectiveness of radiotherapy. Clinically, combining radiotherapy with systemic therapies (e.g., immune checkpoint inhibitors and antiangiogenic agents) has demonstrated preliminary promise in enhancing treatment outcomes. However, translating preclinical findings into clinical practice remains challenging due to tumor heterogeneity, normal tissue toxicity, and the lack of predictive biomarkers for treatment selection. Future research should focus on integrating molecular profiling with multimodal therapies to enable personalized radiosensitization and bridge the gap between mechanistic insights and clinical outcomes.
Collapse
Affiliation(s)
- Jiahui Yang
- Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Rong Chen
- Department of Radiation Oncology, Affiliated ZhongDa Hospital, Southeast University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
10
|
Chen J, Huang J, Shen L. Construction of lung adenocarcinoma subtype and prognosis model based on fatty acid metabolism-related genes. Discov Oncol 2025; 16:866. [PMID: 40405049 PMCID: PMC12098254 DOI: 10.1007/s12672-025-02613-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/07/2025] [Indexed: 05/24/2025] Open
Abstract
OBJECTIVE To explore the role of genes related to fatty acid metabolism in lung adenocarcinoma classification and prognosis. METHODS Transcriptome and clinical data from the TCGA database and GEO database were collected, the expression of prognostic fatty acid metabolism-related genes in LUAD patients was analyzed, and key genes related to both fatty acid metabolism and subtype were identified. These key genes were further filtered via the LASSO regression method, and the retained genes were used to construct a risk-scoring model. The biological function of RPS4Y1 was verified by cell viability, colony formation, migration, and flow cytometry assays. Finally, immune infiltration and drug sensitivity were analyzed in the high- and low-risk groups. RESULTS 31 key FAMGs associated with prognosis were identified in LUAD patients. LUAD cases were divided into 3 subtypes on the basis of the expression of these genes. The DEGs between the different subtypes were associated mainly with amino acid metabolic pathways. In addition, among the 46 DEGs between subtypes, 5 key FAMGs (SCGB3 A2, PGC, ADH7, RPS4Y1, and KRT6 A) were identified as the best prognostic markers via LASSO regression to establish a risk scoring model. Patients with low risk scores had a better prognosis and a greater degree of immune cell infiltration than those with high risk scores. RPS4Y1 is highly expressed in LUAD, and its knockdown significantly inhibits the growth of tumor cells. Moreover, we also analyzed drugs likely to be effective for the high- and low-risk groups. CONCLUSION FAMGs play important roles in LUAD, and the key genes identified may be new targets for LUAD treatment.
Collapse
Affiliation(s)
- Jing Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jinyu Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
11
|
Zhang M, Yang T, Qian Y. APOL4-mediated intracellular cholesterol trafficking is essential for glioblastoma cell growth. BMC Cancer 2025; 25:906. [PMID: 40399861 PMCID: PMC12093831 DOI: 10.1186/s12885-025-14316-4] [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: 11/19/2024] [Accepted: 05/12/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Dysregulated fatty acid metabolism is a key contributor to poor prognosis in glioma, and targeting cholesterol metabolism represents a promising therapeutic strategy. Apolipoprotein L4 (APOL4), a member of the Apolipoprotein L family, has been implicated in lipid metabolism, but its role in glioma remains unclear. METHODS RNA-sequencing were performed to analysis gene expression under exogenous cholesterol treatment. Comprehensive bioinformatics analyses were performed using CGGA datasets to evaluate APOL4 expression, clinical correlations, and prognostic significance in GBM. In vitro experiments, including CRISPR-Cas9 mediated APOL4 knockdown, MTT assays, wound healing assays and immunofluorescence were performed to validate the oncogenic role of APOL4. Xenograft mouse models were employed to validate tumor growth in vivo. RESULTS RNA-sequencing showed that exogenous cholesterol upregulated the expression of APOL4 in U-87 cells. Clinical database analysis revealed that APOL4 was significantly upregulated in human glioblastoma. Genetic depletion of APOL4 markedly suppressed glioblastoma cell proliferation in vitro and impaired xenograft tumor growth in vivo. Furthermore, APOL4 localized to late endosomes and lysosomes, where it likely facilitated the cytoplasmic transport of exogenous cholesterol, supporting tumor cell growth. CONCLUSIONS Our study identifies APOL4 as a novel regulator of cholesterol trafficking in glioma cells, promoting glioblastoma progression. These findings highlight APOL4 as a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Mingxiang Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China
| | - Tao Yang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Youcun Qian
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China.
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
12
|
Jiang H, Zeng Y, Ning W, Hong J, Zhu M, Li P, Ye F, Chen Z, Chen H, Chen W, Li G, Huang H. Ketogenesis instigates immune suppression in enzalutamide resistant prostate cancer via OTUD7B β-hydroxybutyrylation. Cancer Lett 2025; 625:217808. [PMID: 40393641 DOI: 10.1016/j.canlet.2025.217808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 05/08/2025] [Accepted: 05/17/2025] [Indexed: 05/22/2025]
Abstract
Next-generation androgen receptor inhibitors are the primary treatment for metastatic prostate cancer. Unfortunately, the majority of patients rapidly develop resistance. Resistance to enzalutamide has been linked to the emergence of an immunosuppressive tumor, although the underlying mechanisms remain poorly understood. In this study, we observed a marked overexpression of enzymes involved in the ketogenic pathway in enzalutamide-induced castration-resistant prostate cancer, which contributed to immune desertification and resistance to immunotherapy. Mechanistically, upregulation of the ketogenic pathway led to the accumulation of β-hydroxybutyrate, which promoted β-hydroxybutyrylation of the cell cycle-regulated deubiquitinase OTUD7B at lysine 511. This modification impaired the degradation of APC/C substrates, resulting in a subsequent reduction in cytoplasmic double-stranded DNA accumulation, thereby attenuating cGAS-STING activation and interferon expression. These findings shed light on the metabolic adaptations and immune escape driven by androgen receptor signaling inhibitors, potentially informing the development of more effective and durable therapeutic approaches in the near future.
Collapse
Affiliation(s)
- Haoran Jiang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Zhejiang Key Laboratory of Critical Care Medicine,Wenzhou, 325000,Zhejiang China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Key Laboratory of Novel Nuclide Technologies on Precision Diagnosis and Treatment & Clinical Transformation of Wenzhou City, Wenzhou, 325035, Zhejiang, China; Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yuan Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Weiqiang Ning
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Junkai Hong
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Moyang Zhu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ping Li
- Center for Wound Repair and Regeneration, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fangdie Ye
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, 200000, China
| | - Zhifa Chen
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Haoran Chen
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Gang Li
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Hang Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Institute of Urology, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China; Zhejiang Key Laboratory of Critical Care Medicine,Wenzhou, 325000,Zhejiang China.
| |
Collapse
|
13
|
Zhou Y, Kou J, Li W, Wang Y, Su X, Zhang H. BCAA metabolism in cancer progression and therapy resistance: The balance between fuel and cell signaling. Front Pharmacol 2025; 16:1595176. [PMID: 40438606 PMCID: PMC12116492 DOI: 10.3389/fphar.2025.1595176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/01/2025] [Indexed: 06/01/2025] Open
Abstract
Branched-chain amino acids (BCAAs), including leucine, isoleucine, and valine, play a crucial role in cellular metabolism and signaling. Recent studies have demonstrated that BCAA metabolic reprogramming is a key driver of tumor progression and treatment resistance in various cancers. BCAA metabolism supports cancer cell growth, survival, and proliferation by modulating pathways such as mTOR signaling and oxidative stress responses. By promoting immunosuppressive conditions and increasing the survival rate of cancer stem cells (CSCs), BCAAs contribute to immune evasion and resistance to therapies such as chemotherapy and immune checkpoint inhibitors. This article explores the different metabolic reprogramming patterns of BCAAs in various tumors and introduces BCAA-related metabolic targets for overcoming tumor resistance, offering new directions for precision cancer treatment, reducing resistance, and improving patient outcomes.
Collapse
Affiliation(s)
- Yi Zhou
- Departments of Thoracic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiahui Kou
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Wenjin Li
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Yuyao Wang
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xingxing Su
- Shunyi Maternal and Children’s Hospital of Beijing Children’s Hospital, Beijing, China
| | - Hongguang Zhang
- Departments of Thoracic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
Liu Z, Yin J, Qiu T, Liu A, Yu Y, Yang S, Liu Z, Li Q. Reversing the immunosuppressive tumor microenvironment via "Kynurenine starvation therapy" for postsurgical triple-negative breast cancer treatment. J Control Release 2025; 383:113832. [PMID: 40349785 DOI: 10.1016/j.jconrel.2025.113832] [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: 01/25/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Immunotherapy is a potential strategy to suppress the postoperative recurrence and metastasis of triple-negative breast cancer (TNBC). However, the excessive accumulation of kynurenine (Kyn) leads to immunosuppressive tumor microenvironment (TME) and impedes immunotherapeutic efficacy. Herein, a two-pronged approach through "Kynurenine Starvation Therapy" is proposed based on the in-situ hydrogel implantation for postsurgical treatment of TNBC. The hydrogel is constructed via Schiff base reaction between oxidized dextran (ODEX) and 8-arm poly(ethylene glycol) amine (8-arm PEG-NH2), which exhibits excellent biocompatibility and gradual biodegradability. The indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor NLG919 and kynureninase (KYNase) are noncovalently loaded into the hydrogel to prepare NLG919 + KYNase@Gel. The obtained hydrogel can sustainably release NLG919 and KYNase to synergistically deplete Kyn, thereby reversing immunosuppression to enhance the antitumor immunity within TME through "Kynurenine Starvation Therapy". Moreover, a single implantation of NLG919 + KYNase@Gel not only effectively inhibits the postoperative recurrence and metastasis in 4 T1 tumor-bearing mice, but also restrains the growth in an orthotopic TNBC mouse model. These findings highlight an innovative strategy to reinforce the antitumor immune response for the treatment of postsurgical TNBC.
Collapse
Affiliation(s)
- Zengguang Liu
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jiaxin Yin
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Tianyuan Qiu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Aijiang Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yanan Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Shengcai Yang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Ziling Liu
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130012, China.
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; China-Singapore Belt and Road Joint Laboratory on Liver Disease Research, The First Hospital of Jilin University, Changchun 130012, China.
| |
Collapse
|
15
|
Jiang Y, Liang X, Sun H, Yin P, Zhou J, Yu C. Immunomodulatory role of RNA modifications in sex hormone-dependent cancers. Front Immunol 2025; 16:1513037. [PMID: 40406121 PMCID: PMC12095187 DOI: 10.3389/fimmu.2025.1513037] [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: 10/17/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Recent studies have identified that RNA epigenetic modifications, including m6A, m1A, m5C, etc, play pivotal roles in tumor progression. These modifications influence mRNA stability, RNA processing, translational efficiency, and decoding precision. However, comprehensive reviews detailing the connection between m6A RNA modifications and hormone-dependent cancers in both male and female populations remain scarce(breast cancer, ovarian cancer, and endometrial cancer, prostate cancer). In this article, we explore the cellular and molecular roles of various RNA modifications alongside the key elements of the tumor microenvironment. We examine how these RNA modifications influence the development of hormone-dependent cancers through their impact on immune mechanisms. By enhancing our understanding of the function of RNA modifications within the immune systems of four specific tumors, we offer fresh insights for their potential applications in diagnosis and treatment.
Collapse
Affiliation(s)
- Yujia Jiang
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaolan Liang
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hongyi Sun
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ping Yin
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhou
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chaoqin Yu
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
16
|
Edwards DN, Wang S, Kane K, Song W, Kim LC, Ngwa VM, Hwang Y, Ess K, Boothby MR, Chen J. Increased fatty acid delivery by tumor endothelium promotes metastatic outgrowth. JCI Insight 2025; 10:e187531. [PMID: 40198126 DOI: 10.1172/jci.insight.187531] [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: 09/27/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Metastatic outgrowth in distant microscopic niches requires sufficient nutrients, including fatty acids (FAs), to support tumor growth and to generate an immunosuppressive tumor microenvironment (TME). However, despite the important role of FAs in metastasis, the regulation of FA supply in metastatic niches has not been defined. In this report, we show that tumor endothelium actively promotes outgrowth and restricts antitumor cytolysis by transferring FAs into developing metastatic tumors. We describe a process of transendothelial FA delivery via endosomes that requires mTORC1 activity. Thus, endothelial cell-specific targeted deletion of Raptor (RptorECKO), a unique component of the mTORC1 complex, significantly reduced metastatic tumor burden that was associated with improved markers of T cell cytotoxicity. Low-dose everolimus that selectively inhibited endothelial mTORC1 improves immune checkpoint responses in metastatic disease models. This work reveals the importance of transendothelial nutrient delivery to the TME, highlighting a future target for therapeutic development.
Collapse
Affiliation(s)
- Deanna N Edwards
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Shan Wang
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kelby Kane
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Wenqiang Song
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Division of Epidemiology, and
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Laura C Kim
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Verra M Ngwa
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yoonha Hwang
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kevin Ess
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mark R Boothby
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
| | - Jin Chen
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Garemilla SSS, Gampa SC, Garimella S. Role of the tumor microenvironment in cancer therapy: unveiling new targets to overcome drug resistance. Med Oncol 2025; 42:202. [PMID: 40332723 DOI: 10.1007/s12032-025-02754-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Cancer is a leading cause of death globally, with resistance to therapy representing a major obstacle to effective treatment. The tumor microenvironment (TME), comprising a complex network to cellular and non-cellular components including cancer-associated fibroblasts, immune cells, the extracellular matrix and region of hypoxia, is integral to cancer progression and therapeutic resistance. This review delves into the multifaceted interactions within the TME that contribute to tumor growth, survival and immune evasion. Key elements such as the role of cancer- associated fibroblasts in remodeling the extracellular matrix and promoting angiogenesis, the influence of immune cells such as tumor-associated macrophages in creating an immunosuppressive milieu and the impact of hypoxia conditions on metabolic adaptation and therapy resistance are thoroughly examined. This review evaluates current and emerging TME-targeted therapeutic strategies, including inhibitors of extracellular matrix components, modulators of immune cell activity and approached to alleviate hypoxia. Combination therapies that integrate TME-targeted agents with conventional treatments such as chemotherapy and immunotherapy are also discussed for their potential to enhance treatment efficacy and circumvent resistance mechanisms. By synthesising recent advances in TME research and therapeutic innovation, this paper aims to underscore the importance of TME in cancer therapy and highlight promising avenues for improving patient outcomes through targeted intervention.
Collapse
Affiliation(s)
| | - Siri Chandana Gampa
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India
| | - Sireesha Garimella
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India.
| |
Collapse
|
18
|
Atas E, Berchtold K, Schlederer M, Prodinger S, Sternberg F, Pucci P, Steel C, Matthews JD, James ER, Philippe C, Trachtová K, Moazzami AA, Artamonova N, Melchior F, Redmer T, Timelthaler G, Pohl EE, Turner SD, Heidegger I, Krueger M, Resch U, Kenner L. The anti-diabetic PPARγ agonist Pioglitazone inhibits cell proliferation and induces metabolic reprogramming in prostate cancer. Mol Cancer 2025; 24:134. [PMID: 40320521 PMCID: PMC12051277 DOI: 10.1186/s12943-025-02320-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Prostate cancer (PCa) and Type 2 diabetes (T2D) often co-occur, yet their relationship remains elusive. While some studies suggest that T2D lowers PCa risk, others report conflicting data. This study investigates the effects of peroxisome proliferator-activated receptor (PPAR) agonists Bezafibrate, Tesaglitazar, and Pioglitazone on PCa tumorigenesis. Analysis of patient datasets revealed that high PPARG expression correlates with advanced PCa and poor survival. The PPARγ agonists Pioglitazone and Tesaglitazar notably reduced cell proliferation and PPARγ protein levels in primary and metastatic PCa-derived cells. Proteomic analysis identified intrinsic differences in mTORC1 and mitochondrial fatty acid oxidation (FAO) pathways between primary and metastatic PCa cells, which were further disrupted by Tesaglitazar and Pioglitazone. Moreover, metabolomics, Seahorse Assay-based metabolic profiling, and radiotracer uptake assays revealed that Pioglitazone shifted primary PCa cells' metabolism towards glycolysis and increased FAO in metastatic cells, reducing mitochondrial ATP production. Furthermore, Pioglitazone suppressed cell migration in primary and metastatic PCa cells and induced the epithelial marker E-Cadherin in primary PCa cells. In vivo, Pioglitazone reduced tumor growth in a metastatic PC3 xenograft model, increased phosho AMPKα and decreased phospho mTOR levels. In addition, diabetic PCa patients treated with PPAR agonists post-radical prostatectomy implied no biochemical recurrence over five to ten years compared to non-diabetic PCa patients. Our findings suggest that Pioglitazone reduces PCa cell proliferation and induces metabolic and epithelial changes, highlighting the potential of repurposing metabolic drugs for PCa therapy.
Collapse
Affiliation(s)
- Emine Atas
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, Vienna, Austria.
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria.
| | | | | | | | - Felix Sternberg
- Department of Biological Sciences and Pathobiology, Unit of Physiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
- Department of Nutritional Science, University of Vienna, Vienna, Austria
| | - Perla Pucci
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Christopher Steel
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Jamie D Matthews
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Emily R James
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Cécile Philippe
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Karolína Trachtová
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, Vienna, Austria
- Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic
| | - Ali A Moazzami
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, 75007, Uppsala, Sweden
| | | | - Felix Melchior
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Torben Redmer
- Unit of Laboratory Animal Pathology, Institute of Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gerald Timelthaler
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Elena E Pohl
- Department of Biological Sciences and Pathobiology, Unit of Physiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Isabel Heidegger
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marcus Krueger
- Institute for Genetics, Cologne Excellence Cluster of Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Ulrike Resch
- Center of Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Christian Doppler Laboratory for Applied Metabolomics (CDL-AM), Medical University of Vienna, Vienna, Austria.
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria.
- Unit of Laboratory Animal Pathology, Institute of Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.
- Center for Biomarker Research in Medicine GmbH (CBmed), Graz, Austria.
- Department of Molecular Biology, Umeå University, Umea, Sweden.
| |
Collapse
|
19
|
Han Y, Hu M, Wang Y, Xu S, Jiang F, Wang Y, Liu Z. A coagulation-related long non-coding RNA signature to predict prognosis and immune features of breast cancer. Discov Oncol 2025; 16:662. [PMID: 40317354 PMCID: PMC12049355 DOI: 10.1007/s12672-025-02316-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
Breast cancer (BC) remains one of the most common malignancies among women worldwide, with persistently poor prognosis despite advancements in diagnostics and therapies. Long non-coding RNAs (lncRNAs) and coagulation-related genes (CRGs) are increasingly recognized for their roles in prognosis and immune modulation. Using transcriptomic data from 1,045 BC patients in TCGA, we identified CRG-associated lncRNAs via coexpression analysis (Pearson |R|> 0.4, p < 0.001) and constructed a prognostic model through univariate Cox analysis, LASSO regression with tenfold cross-validation (λ = 0.05), and multivariate Cox analysis. The model stratified patients into high- and low-risk groups with distinct overall survival (HR = 3.21, p < 0.001) and demonstrated robust predictive accuracy (AUC = 0.795 at 1 year). Functional enrichment revealed immune-related pathways (e.g., cytokine signaling, PD-L1 regulation), and high-risk patients exhibited elevated tumor mutational burden (TMB) and PD-L1 expression, suggesting enhanced immunotherapy responsiveness. Drug sensitivity analysis identified 5 targeted agents (e.g., BIBW2992) with differential efficacy between risk groups. This CRG-lncRNA signature provides a novel tool for prognosis prediction and personalized immunotherapy in BC, illuminating crosstalk between coagulation and immune pathways.
Collapse
Affiliation(s)
- Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yanzhong Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China.
| |
Collapse
|
20
|
Jia X, Zhang Q, Wang Z, Cao J, Song A, Lan C, Hu Y. Integrative single-cell RNA sequencing and bulk RNA sequencing reveals the characteristics of glutathione metabolism and protective role of GSTA4 gene in pancreatic cancer. Front Immunol 2025; 16:1571431. [PMID: 40375987 PMCID: PMC12078301 DOI: 10.3389/fimmu.2025.1571431] [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: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/18/2025] Open
Abstract
Background Recent studies have increasingly reported abnormal glutathione (GSH) metabolism within the tumor microenvironment across various solid tumors. However, the specific mechanisms underlying aberrant GSH metabolism in pancreatic cancer (PC) remain unclear. This study aims to investigate the prognostic significance of GSH metabolism-related genes in PC and to identify key molecular targets, thereby providing novel perspectives for targeted PC therapy. Methods The GSH metabolism gene set was retrieved from the KEGG database. Utilizing single-cell transcriptomic data from the GSE205049 dataset, this study analyzed the variation in GSH metabolic signaling intensity across distinct cell types within the tumor microenvironment of PC. Additionally, transcriptomic data from multiple repositories, including TCGA, ICGC, and GEO, comprising a total of 930 patients with PC, were integrated to construct a prognostic molecular classifier related to GSH metabolism. Furthermore, the role of the key gene GSTA4 in PC was experimentally validated through a series of in vitro assays. Results Significant differences in GSH metabolic signaling intensity were observed across various cell types in both normal pancreatic and PC tissues. A prognostic signature comprising six GSH metabolism-related genes (GSTA5, PGD, IDH2, GSTA4, GPX2, and GPX3) was established, wherein a high-risk score was associated with a poorer patient prognosis. Notably, GSTA4 expression was significantly reduced in PC tissues, and higher GSTA4 levels were linked to a favorable prognosis. In vitro functional analyses demonstrated that GSTA4 overexpression markedly inhibited PC cell proliferation and migration. Conclusion The GSH metabolism-associated prognostic signature developed in this study effectively identifies high-risk patients with PC. As a prognostic protective factor, GSTA4 exhibits downregulated expression in PC tissues and suppresses tumor proliferation and migration, highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xinya Jia
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Wang
- Department of Urology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianliang Cao
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Anran Song
- Department of Physical Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chao Lan
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuepeng Hu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Guo S, Zhang L, Ren J, Lu Z, Ma X, Liu X, Jin H, Li J. The roles of enhancer, especially super-enhancer-driven genes in tumor metabolism and immunity. Int J Biol Macromol 2025; 308:142414. [PMID: 40132720 DOI: 10.1016/j.ijbiomac.2025.142414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Abnormal metabolism is a characteristic of malignant tumors. Numerous factors play roles in the regulation of tumor metabolism. As epigenetic regulators, enhancers, especially the super-enhancers (SEs), serve as platforms for transcription factors that regulate the expression of metabolism-related enzymes or transporters at the gene level. In this study, we review the effects of enhancer/ SE-driven genes on tumor metabolism and immunity. Enhancers/SEs play regulatory roles in glucose metabolism (glycolysis, gluconeogenesis, tricarboxylic acid (TCA) cycle, pyruvate, and pentose phosphate pathway, lipid metabolism (cholesterol, fatty acid, phosphatide, and sphingolipid), and amino acid metabolism (glutamine, tryptophan, arginine, and cystine). By regulating tumor metabolism, enhancers and SEs can reprogram tumor microenvironment, especially the status of various immune cells. Therefore, interfering enhancers/SEs that regulate the tumor metabolism is likely to enhance the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Songyue Guo
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Lu Zhang
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Jiao Ren
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Zhong Lu
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xiaolin Ma
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xinling Liu
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| | - Hongchuan Jin
- Department of Medical Oncology, Cancer Center of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China.
| | - Jiaqiu Li
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| |
Collapse
|
22
|
Kim YK, Song J. Metabolic imbalance and brain tumors: The interlinking metabolic pathways and therapeutic actions of antidiabetic drugs. Pharmacol Res 2025; 215:107719. [PMID: 40174814 DOI: 10.1016/j.phrs.2025.107719] [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: 01/08/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Brain tumors are complex, heterogeneous malignancies, often associated with significant morbidity and mortality. Emerging evidence suggests the important role of metabolic syndrome, such as that observed in diabetes mellitus, in the progression of brain tumors. Several studies indicated that hyperglycemia, insulin resistance, oxidative stress, and altered adipokine profiles influence tumor growth, proliferation, and treatment resistance. Intriguingly, antidiabetic drugs (e.g., metformin, sulfonylureas, dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, and thiazolidinediones) have shown promise as adjunctive or repurposed agents in managing brain tumors. Metformin can impair tumor cell proliferation, enhance treatment sensitivity, and modify the tumor microenvironment by activating AMP-activated protein kinase (AMPK) and inhibiting mammalian target of rapamycin (mTOR) signaling pathways. DPP-4 inhibitors and GLP-1 receptor agonists can target both metabolic and inflammatory aspects of brain tumors, while thiazolidinediones may induce apoptosis in tumor cells and synergize with other therapeutics. Consequently, further studies and clinical trials are needed to confirm the efficacy, safety, and utility of metabolic interventions in treating brain tumors. Here, we review the evidence for the metabolic interconnections between metabolic diseases and brain tumors and multiple actions of anti-diabetes drugs in brain tumors.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
23
|
Gao H, Yao Y, Li W, Xu Z, Hu W, Luo K, Chen P, Shang W, Luan S, Shi G, Cao M, Chen P. Caspase-6/Gasdermin C-Mediated Tumor Cell Pyroptosis Promotes Colorectal Cancer Progression Through CXCL2-Dependent Recruitment of Myeloid-Derived Suppressor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411375. [PMID: 40213993 PMCID: PMC12120772 DOI: 10.1002/advs.202411375] [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] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/24/2025] [Indexed: 06/01/2025]
Abstract
Gasdermin (GSDM) family proteins mediate inflammatory cell pyroptosis and exert critical contributions to the pathogenesis of gastrointestinal cancers, infections, and gut mucosal inflammation. Gasdermin C (GSDMC) is overexpressed in human colorectal cancer (CRC); however, the molecular mechanisms underlying GSDMC regulation of CRC tumorigenesis are largely elusive. Here, it is found that both GSDMC expression and activation are significantly elevated in human and mouse CRC tissues. Gsdmc2/3/4 deficiency attenuates tumor progression in both chemically induced CRC mouse model and spontaneous intestinal tumor model. Mechanistically, under hypoxia and low-glucose condition, GSDMC2/3/4 are directly activated by Caspase-6, but not by Caspase-8, as previously reported in other cancers. GSDMC2/3/4-mediated pyroptosis in tumor cells leads to the release of high mobility group protein B1 (HMGB1), which enhances the expression of chemokine attractant C-X-C motif chemokine 2 (CXCL2) in surrounding tumor cells. Subsequently, the elevated CXCL2 secretion from tumor cells promotes the recruitment of myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment (TME) through C-X-C chemokine receptor type 2 (CXCR2), thereby facilitating CRC progression. These findings reveal a mechanism by which Caspase-6/GSDMC-mediated tumor cell pyroptosis, in response to hypoxic and low-glucose conditions, remodels the immunosuppressive microenvironment through CXCL2-dependent recruitment of MDSCs. These results identify GSDMC as a potential drug target for CRC therapy.
Collapse
Affiliation(s)
- Hanchao Gao
- Department of NephrologyShenzhen Longhua District Central HospitalShenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney DiseaseShenzhenGuangdong518110China
| | - Yikun Yao
- Shanghai Institute of Nutrition & HealthChinese Academy of ScienceShanghai200031China
| | - Weilong Li
- Department of NephrologyShenzhen Longhua District Central HospitalShenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney DiseaseShenzhenGuangdong518110China
| | - Zigan Xu
- Department of NephrologyShenzhen Longhua District Central HospitalShenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney DiseaseShenzhenGuangdong518110China
| | - Wenjun Hu
- Department of AnesthesiologyThe 305 Hospital of Liberation Army of China (PLA)Beijing100036China
| | - Kewang Luo
- Department of Medical LaboratoryPeople's Hospital of LonghuaShenzhenGuangdong518110China
| | - Peishan Chen
- Department of NephrologyShenzhen Longhua District Central HospitalShenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney DiseaseShenzhenGuangdong518110China
| | - Wanjing Shang
- Lymphocyte Biology SectionLaboratory of Immune System BiologyNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMD20892USA
| | - Shaodong Luan
- Department of NephrologyShenzhen Longhua District Central HospitalShenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney DiseaseShenzhenGuangdong518110China
| | - Guojun Shi
- Department of Endocrinology and MetabolismMedical Center for Comprehensive Weight ControlThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
- State Key Laboratory of Oncology in South ChinaGuangzhou510630China
| | - Mengtao Cao
- Department of Respiratory MedicineShenzhen Longhua District Central HospitalShenzhenGuangdong518110China
| | - Pengfei Chen
- Department of Traumatic OrthopedicsShenzhen Longhua District Central HospitalShenzhenGuangdong518110China
| |
Collapse
|
24
|
Ji X, Qian X, Luo G, Yang W, Huang W, Lei Z, Zhou J, Zhong G, Zhou J, Liu N, Ma L, Li M, Liu X, Wu S, Zhang Y. Engineered macrophage nanoparticles enhance microwave ablation efficacy in osteosarcoma via targeting the CD47-SIRPα Axis: A novel Biomimetic immunotherapeutic approach. Bioact Mater 2025; 47:248-265. [PMID: 39925711 PMCID: PMC11803168 DOI: 10.1016/j.bioactmat.2025.01.012] [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: 10/13/2024] [Revised: 12/30/2024] [Accepted: 01/12/2025] [Indexed: 02/11/2025] Open
Abstract
Osteosarcoma (OS) is a lethal bone tumor that primarily affects adolescents. OS is characterized by a high incidence of recurrence following surgical intervention, which is attributed to the presence of residual microscopic disease. Tumor-associated macrophages, which dominate the tumor microenvironment, often suppress immune responses and facilitate tumor progression and recurrence. This study developed an innovative nanotherapeutic approach by utilizing genetically engineered macrophage membranes with M1 polarization, stably overexpressing signal regulatory protein alpha (SIRPα), to encapsulate microwave-responsive nano-Prussian blue (SIRPα-M@nanoPB) nanoparticles. These nanoparticles induce tumor cell death selectively through hyperthermia and microwave dynamic effects upon targeted microwave irradiation. It is of critical importance to note that the enhancement of SIRPα on the nanoparticle surface actively targets and binds CD47 of tumor cells, thereby disrupting the "don't-eat-me" signal and effectively countering the immunosuppressive tumor environment. This action restores macrophage phagocytosis with M1 polarization, triggering potent immune responses. Our strategy holds considerable promise when it comes to improving the efficacy of microwave ablation through immune modulation, while reducing thermal damage to adjacent normal tissue and minimizing the risk of tumor recurrence. Thus, it offers a significant advancement in microwave therapies for patients with OS.
Collapse
Affiliation(s)
- Xiongfa Ji
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Xin Qian
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No.168 Litang Road, Changping District, 100000, Beijing, China
| | - Guowen Luo
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Wenjie Yang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Wenhan Huang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Zehua Lei
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Jiaqi Zhou
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Guoqing Zhong
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Jielong Zhou
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Nan Liu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Limin Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Mei Li
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| | - Xiangmei Liu
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340, 300401, Tianjin, China
| | - Shuilin Wu
- School of Materials Science & Engineering, Peking University, Yi-He-Yuan Road 5, 100871, Beijing, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
- Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, 510080, Guangzhou, China
- Medical Research Center, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 510080, Guangzhou, China
| |
Collapse
|
25
|
Wang Y, Wu Q, Wei X, Huang G, Feng G, Xu H, Gou X. Increased Immune Infiltration and Improved Prognosis of Head and Neck Squamous Cell Carcinoma Associated with Reduced Ancient Ubiquitous Protein 1 Gene Expression. Mol Biotechnol 2025; 67:1826-1842. [PMID: 38862860 DOI: 10.1007/s12033-024-01161-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 04/08/2024] [Indexed: 06/13/2024]
Abstract
This study aimed to explore the molecular mechanism underlying the prognostic role of ancient ubiquitous protein 1 (AUP1) in head and neck squamous cell carcinoma (HNSCC) and its relationship with the tumor immune microenvironment. Various web resources were used to analyze the differential expression of AUP1 and its role in the HNSCC pathogenesis. A nomogram aimed at predicting 1-, 3-, and 5-year survival rates was developed based on the patient's clinicopathological characteristics and AUP1 expression pattern. Several algorithms and analytical tools were used to explore the correlation between AUP1 expression and sensitivity to immune checkpoint gene therapy by evaluating infiltrating immune cells in patients with HNSCC. Higher AUP1 mRNA and protein expression levels were observed in most tumors and HNSCC than in the normal tissues. High AUP1 expression was an independent predictive risk factor for the overall survival of patients as it was closely associated with the patients' T, M, clinical, and pathological stages and lymphovascular invasion in HNSCC. In conclusion, AUP1 is involved in the occurrence and progression of HNSCC, may be used as an independent prognostic factor in patients with HNSCC, and could serve as a potential intervention target to improve immunotherapy sensitivity in HNSCC.
Collapse
Affiliation(s)
- Yi Wang
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Wu
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiao Wei
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Gang Huang
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Guangyong Feng
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Hui Xu
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiaoxia Gou
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
26
|
Sun R, Liu R, Tian Y, Li Y, Fan B, Li S. Removing Barriers to Tumor 'Oxygenation': Depleting Glutathione Nanozymes in Cancer Therapy. Int J Nanomedicine 2025; 20:5613-5643. [PMID: 40331231 PMCID: PMC12051984 DOI: 10.2147/ijn.s515734] [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: 01/04/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
Nanozymes are nanomaterials capable of mimicking natural enzyme catalysis in the complex biological environment of the human body. Due to their good stability and strong catalytic properties, nanozymes are widely used in various fields of biomedicine. Among them, nanozymes that trigger intracellular reactive oxygen species (ROS) levels for cancer therapy have gained significant attention. However, the 'explosion' of ROS in tumor cells was prevented by the high levels of glutathione (GSH) in the tumor microenvironment (TME). GSH, a prominent endogenous antioxidant, increases the resistance of tumor cells to oxidative stress by scavenging ROS. Certain nanozymes can deplete intracellular GSH levels by mimicking GSH oxidase (GSHOx), GSH peroxidase (GPx) or by interfering with the reduction of oxidized glutathione (GSSG). On the one hand, elevated the level of intracellular ROS and induced lipid peroxidation reaction leading to ferroptosis. On the other hand, it creates favorable conditions for the treatment of tumors with photodynamic therapy (PDT), sonodynamic therapy (SDT), chemodynamical therapy (CDT) and targeted therapy. In this paper, we present a comprehensive analysis of GSH-depleting nanozymes reported in recent years, including classification, mechanism, responsiveness to TME and their roles in cancer therapy, and look forward to future applications and developments.
Collapse
Affiliation(s)
- Ruilong Sun
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
- Gansu Provincial Key Laboratory of Stem Cells and Gene Drugs, Lanzhou, People’s Republic of China
| | - Ruitang Liu
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Yongzheng Tian
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Yunfei Li
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Bo Fan
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Songkai Li
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| |
Collapse
|
27
|
Huang X, Li H, Wang Q, Liao Y, Xie X, Zhou Y, Wu D, Li H. Hypoxia-responsive albumin nanoparticles co-delivering banoxantrone and STING agonist enhance immunotherapy of high-intensity focused ultrasound. J Control Release 2025; 383:113789. [PMID: 40318808 DOI: 10.1016/j.jconrel.2025.113789] [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: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
High-intensity focused ultrasound (HIFU) has great potential as a noninvasive, breast-conserving treatment for patients with breast cancer. The HIFU-induced antitumor immune response plays a critical role in postoperative prognosis. However, after surgery, tumor cells exhibit low immunogenicity, and severe hypoxia exacerbates the immunosuppressive tumor microenvironment, thereby rendering HIFU-induced immune effects insufficient to prevent tumor recurrence and metastasis. Herein, hypoxia-responsive, crosslinked albumin-based nanoparticles (HACS NPs), comprising the hypoxia-activated prodrug AQ4N, the stimulator of interferon genes (STING) agonist SR-717, and calcium carbonate (CaCO3), were developed to augment the postoperative antitumor immune response. The HACS NPs are activated to "on" state in the postoperative hypoxic tumor microenvironment and release the loaded AQ4N to increase immunogenic cell death (ICD). Moreover, SR-717 promotes intratumoral infiltration of immune cells through STING activation, while CaCO3 creates immune-supportive tumor microenvironment by consuming lactate, thereby polarizing M2 macrophages to the M1 phenotype. Based on these improvements, the HACS NPs have been shown to significantly enhance the post-HIFU antitumor immune response, effectively suppress tumor recurrence and metastasis. This work provides a successful paradigm for improving the prognosis of surgical interventions, including HIFU.
Collapse
Affiliation(s)
- Xinchang Huang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China
| | - Hechun Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China
| | - Qi Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China
| | - Yan Liao
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China
| | - Xiaomei Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China
| | - Yinglin Zhou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China
| | - Dezhou Wu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China
| | - Huanan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, 400016 Chongqing, PR China.
| |
Collapse
|
28
|
Jiang Y, Shu Z, Cheng L, Wang H, He T, Fu L, Zhao C, Li X, Zeng W. MS4A7 based metabolic gene signature as a prognostic predictor in lung adenocarcinoma. Front Mol Biosci 2025; 12:1591446. [PMID: 40356720 PMCID: PMC12066319 DOI: 10.3389/fmolb.2025.1591446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) represents the most common form of lung cancer, contributing to significant global mortality. Metabolic reprogramming in tumor cells has been increasingly recognized as a hallmark of tumorigenesis, contributing to an immunosuppressive microenvironment. Given the promising prediction value of metabolism-related genes in LUAD, this study aims to explore the role of MS4A7, a member of the MS4A gene family, in LUAD prognosis and immune microenvironment dynamics. Methods A prognostic signature for LUAD was developed using the LASSO-Cox regression algorithm with RNA-seq data from 500 LUAD patients in The Cancer Genome Atlas database. Genes with differential expression linked to metabolic pathways were identified, and 20 genes were included to develop a risk signature. Further functional enrichment analysis was conducted to compare the biological pathways activated in high-risk versus low-risk groups. Single-cell RNA sequencing was employed to identify the expression profile and role of MS4A7 in different macrophage populations within the LUAD. Results The constructed prognostic model displayed high predictive accuracy, outperforming single gene-based predictions. High-risk patients exhibited significantly poorer survival outcomes. Pathway enrichment analysis revealed dysregulated metabolic pathways in high-risk patients, including activation of glycolysis, mTORC1 signaling, and ROS production. Single-cell RNA sequencing revealed that MS4A7 expression was predominantly found in macrophage populations, with high expression localized in MS4A7+ macrophages. These macrophages exhibited distinct metabolic reprogramming and key immune functions, particularly in crosstalk with T cells and neutrophils. Conclusion The MS4A7 gene plays a critical role in LUAD prognosis, particularly through its involvement in immune modulation within the TME. MS4A7+ macrophages, characterized by distinct metabolic reprogramming and immune interactions, are pivotal in shaping LUAD progression and immune response. The findings highlight the potential of MS4A7 as a novel prognostic biomarker and therapeutic target for LUAD. Further investigation into the metabolic and immune regulatory mechanisms of MS4A7+ macrophages could offer new insights into LUAD treatment strategies.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Reproductive Medicine Nursing, Key Laboratory of Birth Defects and Related Disease of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhengyu Shu
- Department of Reproductive Medicine Nursing, Key Laboratory of Birth Defects and Related Disease of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lei Cheng
- Jiangsu Province Hospital of Chinese Medicine Chongqing Hospital, Chongqing Yongchuan Hospital of Chinese Medicine, Chongqing, China
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haowei Wang
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Taiping He
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liwen Fu
- Department of Orthopedic Oncology, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weicheng Zeng
- Jiangsu Province Hospital of Chinese Medicine Chongqing Hospital, Chongqing Yongchuan Hospital of Chinese Medicine, Chongqing, China
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
29
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
30
|
Zhao Q, Wei Z, Yang G, Wei L, Chen H, Cui Z, Liao N, Qin M, Cheng J. KCNJ2 Facilitates Clear Cell Renal Cell Carcinoma Progression and Glucose Metabolism. Int J Genomics 2025; 2025:2210652. [PMID: 40314029 PMCID: PMC12045686 DOI: 10.1155/ijog/2210652] [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: 12/17/2024] [Accepted: 02/27/2025] [Indexed: 05/03/2025] Open
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) is marked by aggressive characteristics and a poor prognosis. The involvement of KCNJ2, an inward rectifying potassium channel, in the progression of ccRCC, along with its potential roles in immune modulation and metabolic pathways, remains unclear. Methods: The Cancer Genome Atlas (TCGA) database was utilized to analyze the gene expression, clinicopathological characteristics, and clinical relevance of KCNJ2. The prognostic value of KCNJ2 in ccRCC was evaluated with Kaplan-Meier survival analysis and receiver operating characteristic curve analyses. The TCGA-KIRC dataset was utilized to analyze tumor microenvironment (TME), focusing on tumor-infiltrating immune cells and immunomodulators. The biological functions of KCNJ2 were investigated in vitro using CCK-8, flow cytometry, wound healing, transwell, qRT-PCR, and Western blotting assays. Results: KCNJ2 expression was notably higher in ccRCC than in normal kidney tissues, with increased levels associated with advanced tumor stages. However, KCNJ2 did not exhibit obvious prognostic value. Coexpression analysis identified associations with genes implicated in energy metabolism. Analysis of the TME and immune profile indicated a link between KCNJ2 expression and immune cell infiltration, along with particular immune checkpoints. In vitro studies demonstrated that KCNJ2 overexpression enhanced cell proliferation, migration, invasion, glucose production, and ATP generation. Conclusion: KCNJ2 plays a crucial role in ccRCC progression through affecting glucose metabolism and immune responses. Our findings reveal the functional role of KCNJ2 in promoting tumor progression and metabolic reprogramming in ccRCC, highlighting its therapeutic potential as a novel target for ccRCC treatment. Further studies are essential to clarify the mechanisms by which KCNJ2 affects ccRCC biology and to evaluate its clinical relevance.
Collapse
Affiliation(s)
- Qiyue Zhao
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Urology, Liuzhou Workers' Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
| | - Zhengshu Wei
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Urology, Liuzhou Workers' Hospital, Guangxi Medical University, Liuzhou, Guangxi, China
| | - Guanglin Yang
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Liwei Wei
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hao Chen
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Zelin Cui
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Naikai Liao
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Min Qin
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiwen Cheng
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
31
|
Chen G, Zhang C, Zou J, Zhou Z, Zhang J, Yan Y, Liang Y, Tang G, Chen G, Xu X, Wang N, Feng Y. Coptidis rhizoma and berberine as anti-cancer drugs: A 10-year updates and future perspectives. Pharmacol Res 2025; 216:107742. [PMID: 40258505 DOI: 10.1016/j.phrs.2025.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025]
Abstract
Cancer continues to be among the most substantial health challenges globally. Among various natural compounds, berberine, an isoquinoline alkaloid obtained from Coptidis Rhizoma, has garnered considerable attention for its broad-spectrum biological activities, including anti-inflammatory, antioxidant, anti-diabetic, anti-obesity, and anti-microbial activities. Furthermore, berberine exhibits a broad spectrum of anti-cancer efficacy against various malignancies, such as ovarian, breast, lung, gastric, hepatic, colorectal, cervical, and prostate cancers. Its anti-cancer mechanisms are multifaceted, encompassing the inhibition of cancer cell proliferation, the prevention of metastasis, the induction of apoptosis, the facilitation of autophagy, the modulation of the tumor microenvironment and gut microbiota, and the enhancement of the efficacy of conventional therapeutic strategies. This paper offers an exhaustive overview of the cancer-fighting characteristics of Coptidis Rhizoma and berberine, while also exploring recent developments in nanotechnology aimed at enhancing the bioavailability of berberine. Furthermore, the side effects and safety of berberine are addressed as well. The potential role of artificial intelligence in optimizing berberine's therapeutic applications is also highlighted. This paper provides precious perspectives on the prospective application of Coptidis Rhizoma and berberine in the prevention and management of cancer.
Collapse
Affiliation(s)
- Guoming Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Jiayi Zou
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zitian Zhou
- The Fourth School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiayi Zhang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Yan
- The School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinglan Liang
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoyi Tang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Guang Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Xiaoyu Xu
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
32
|
Liu J, Liu J, Chen H, Zhang Q, Zhang P, Chen Z, Lu X, Xu Y. Prognostic value of combined nutritional and inflammatory markers in NSCLC patients receiving ICIs. Discov Oncol 2025; 16:571. [PMID: 40253541 PMCID: PMC12009260 DOI: 10.1007/s12672-025-02391-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/14/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND In the treatment of non-small cell lung cancer (NSCLC), immune checkpoint inhibitors (ICIs) have markedly improved patient survival, yet some patients do not benefit. The existing prognostic factors are limited, highlighting the development of reliable and convenient predictive indicators. METHODS A retrospective analysis was performed on 219 NSCLC patients treated with ICIs from June 2019 to January 2024. The nutritional risk screening (NRS 2002) and the neutrophil-to-lymphocyte ratio (NLR) were employed to evaluate the patients' nutritional status and inflammatory response, aiming to investigate the correlation between these markers and treatment outcomes. RESULTS The median follow-up duration for the overall population was 29 (IQR: 25.96-32.04) months. The analysis showed that the median progression-free survival (mPFS) and median overall survival (mOS) in the high nutritional risk group (NRS2002 ≥ 3, accounting for 23.74%) were significantly lower than those in the low nutritional risk group (NRS2002 < 3, accounting for 76.26%) (mPFS: 2.5 vs 16 months; mOS: 8 vs 16 months, both P < 0.001). Similarly, patients with high NLR values (> 4.92) had significantly shorter OS and PFS than those with low NLR (≤ 4.92) values (mOS: 7 vs 18 months; mPFS: 3 vs 17 months, both P < 0.001). Multivariate Cox analysis revealed that a high NRS 2002 score (HR = 2.76, 95% CI 1.68-4.54, P < 0.001) and high NLR (HR = 2.77, 95% CI 1.65-4.64, P < 0.001) were independent predictors of poor prognosis. Risk stratification was performed using a combined scoring system of NRS 2002 and NLR (0 points-low risk, 1 point-moderate risk, 2 points-high risk), and it was found that as the risk score increased, OS and PFS significantly decreased (mOS: 8 [2.61-13.39] vs 16 [13.04-18.96] vs NA [NA-NA] months; mPFS: 2.5 [0.99-4.02] vs 8.5 [5.47-11.53] vs 16 [11.41-20.59] months, respectively, both P < 0.001). The utility of the combined NLR and NRS2002 scoring model was assessed using a time-dependent receiver operating characteristic (ROC) curve, with results indicating that at 12 months, the AUC value of the combined scoring model was 0.81 (CI 0.72-0.90). At 24 and 36 months, the AUC values were 0.73 (CI 0.66-0.80) and 0.70 (CI 0.64-0.76), respectively. Moreover, the nomogram model exhibited high predictive accuracy in predicting survival prognosis, with AUC values of 0.84 (CI 0.77-0.91), 0.85 (CI 0.79-0.91), and 0.78 (CI 0.69-0.88) at 12, 24, and 36 months, respectively. CONCLUSION The combined NRS 2002 and NLR scoring can serve as an effective prognostic tool for NSCLC patients receiving ICIs treatment. This scoring system helps clinicians more accurately identify patients who will benefit from immunotherapy, thereby facilitating more personalized treatment plans. Further validation of this scoring system's applicability and reliability is warranted in future multicenter, large-sample prospective studies.
Collapse
Affiliation(s)
- Jianying Liu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Jiaxin Liu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Haiyan Chen
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Qingwei Zhang
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Peihong Zhang
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Zhisheng Chen
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Xuefeng Lu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
| | - Yijiao Xu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
| |
Collapse
|
33
|
Luo Y, Li J, Fu Q, Zhang P, Song X, Liu M, Mo R, Fu J, Tang S, Wu J, Yang X, Liu X, Wang T, Ni G. Caerin 1.1 and 1.9 peptides induce acute caspase 3/GSDME-mediated pyroptosis in epithelial cancer cells. Sci Rep 2025; 15:13377. [PMID: 40251208 PMCID: PMC12008296 DOI: 10.1038/s41598-025-96438-0] [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: 08/14/2024] [Accepted: 03/28/2025] [Indexed: 04/20/2025] Open
Abstract
Caerin peptides exhibit a dual role in cancer treatment by directly killing cancer cells and modulating the tumour microenvironment to enhance anti-tumour immunity. This study investigates the mechanisms underlying caerin 1.1/1.9-induced acute cell death in epithelial cancer cells and explores their therapeutic potential. HeLa, A549, and Huh-7 cancer cell lines were treated with caerin 1.1/1.9 peptides. Morphological observations, flow cytometry, lactate dehydrogenase (LDH) release, and IL-18 secretion assays revealed the occurrence of pyroptosis following treatment. Specifically, a 1-h treatment with caerin 1.1/1.9 induced pyroptosis in HeLa, A549, and Huh-7 cells, characterised by cell swelling, membrane bubbling, and the release of IL-18 and LDH. Western blotting confirmed the upregulation of pyroptosis markers, including caspase-3, cleaved caspase-3, and GSDME-N fragments. These findings highlight the significant role of caerin peptides in inducing acute pyroptosis, a form of programmed cell death that enhances the immunogenicity of dying cancer cells, thus potentially improving the effectiveness of immunotherapies. This research underscores the therapeutic potential of caerin 1.1/1.9 peptides in cancer treatment, providing a foundation for developing new anti-cancer strategies that leverage both direct cytotoxic effects and immune modulation to achieve more effective and sustained anti-tumour responses.
Collapse
Affiliation(s)
- Yuandong Luo
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Junjie Li
- Zhongao Biomedical Technology (Guangdong) Co. Ltd, Zhongshan, 528400, Guangdong, China
| | - Quanlan Fu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Pingping Zhang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Xinyi Song
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Mengqi Liu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Rongmi Mo
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Jiawei Fu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Shuxian Tang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Jialing Wu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Xiaodan Yang
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Xiaosong Liu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China.
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| | - Tianfang Wang
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
| | - Guoying Ni
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| |
Collapse
|
34
|
Guo H, Zhang L, Tang H, Liu P, Hu B, Gong Y, Hou R, Wu Z. Exploring the Role of T-Cell Metabolism in Modulating Immunotherapy Efficacy for Non-Small Cell Lung Cancer Based on Clustering. J Clin Lab Anal 2025:e25020. [PMID: 40244859 DOI: 10.1002/jcla.25020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/26/2024] [Accepted: 02/08/2024] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Immunotherapy, especially immune checkpoint blockade (ICB) therapy, has demonstrated noteworthy advancements in the realm of non-small cell lung cancer (NSCLC). However, the efficacy of ICB therapy is limited to a small subset of patients with NSCLC, and the underlying mechanisms remain poorly understood. STUDY DESIGN AND DISCOVERIES In this study, we conducted a comprehensive investigation of the metabolic profiles of infiltrating T cells in NSCLC tumors and revealed the metabolic heterogeneity, which associated with the prognosis of ICB therapy, in three T-cell subtypes. After metabolic clustering, we split these metabolic clusters into two groups: Nonresponse-associated (NR) clusters that enriched with cells from nonresponders, and response-associated (R) clusters that not belonging to NR clusters. Then, we elucidated their metabolic differences and specific functions. Notably, we discovered HSPA1A was significantly downregulated in NR clusters of all three T-cell subtypes. In addition, leveraging single-cell T-cell receptor sequencing data and pseudotime series analysis, we revealed the reciprocal interconversion between R and NR metabolic clusters within the same T-cell clone. This suggests a potential metabolic reprogramming capability of T cells. Furthermore, through the analysis of intercellular communication, we identified the specific intercellular signaling in the R clusters, which might promote the activation and regulation of signal transduction pathways that affect the prognosis of ICB therapy. CONCLUSION In conclusion, our study offers substantial insights into the mechanisms of relationships between T-cell metabolisms and ICB therapy outcomes, shedding light on the mechanism of immunotherapy efficacy in patients with NSCLC. Such investigations will contribute to overcoming treatment resistance.
Collapse
Affiliation(s)
- Hongzhe Guo
- School of Electrical and Information Engineering, Anhui University of Technology, Maanshan, China
| | - Liangyu Zhang
- Department of Medical Oncology, The General Hospital of Daqing Oil Field, Daqing, China
| | - Hu Tang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peiwen Liu
- School of Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yue Gong
- Geneis Beijing Co., Ltd., Beijing, China
| | - Rui Hou
- Geneis Beijing Co., Ltd., Beijing, China
| | - Ziheng Wu
- School of Electrical and Information Engineering, Anhui University of Technology, Maanshan, China
| |
Collapse
|
35
|
Dang Y, Ma M, Wang Y, Zhao M, Cao Y, Su H, Liu T, Zheng M, Gao J, Wu X, Xu J, Chen L, Xi JJ, Fei Y, Liu H. Carvedilol sensitizes chemotherapy by targeting STING to boost anti-tumor immunity. Cell Rep 2025; 44:115572. [PMID: 40249703 DOI: 10.1016/j.celrep.2025.115572] [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: 12/18/2024] [Revised: 02/16/2025] [Accepted: 03/26/2025] [Indexed: 04/20/2025] Open
Abstract
The stimulator of interferon genes (STING)-mediated type I interferon (IFN) response is critical for mounting anti-tumor immunity and sensitizing chemotherapy by remodeling the tumor immune microenvironment. However, no clinically available drugs have been applied for STING activation. Based on high-throughput screening of small-molecule microarrays, we found that carvedilol, an adrenergic receptor blocker used to treat essential hypertension and symptomatic heart failure, is a STING activator. Mechanistically, carvedilol interacts with STING at threonine 263 and enhances its dimerization. Importantly, carvedilol enhances the therapeutic effect of etoposide in both the allografted tumor model and patient-derived tumor-like cell clusters (PTCs) by promoting etoposide-induced STING activation. Our findings identify carvedilol as a STING activator and provide a theoretical basis for combining carvedilol and etoposide in cancer therapy.
Collapse
Affiliation(s)
- Yifang Dang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Department of Microbiology and Immunology, School of Medicine, Tongji University, Shanghai 200072, China; Central Laboratory, Taicang Hospital Affiliated to Soochow University, Taicang 215400, China
| | - Mingtong Ma
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yan Wang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Department of Microbiology and Immunology, School of Medicine, Tongji University, Shanghai 200072, China
| | - Mengmeng Zhao
- Research Center of Translational Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yajuan Cao
- Department of Integrated Traditional Chinese and Western Medicine, Tongji University School of Medicine, Shanghai 200433, China
| | - Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Tianhao Liu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Department of Microbiology and Immunology, School of Medicine, Tongji University, Shanghai 200072, China
| | - Mengge Zheng
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Department of Microbiology and Immunology, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jiani Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiangyang Wu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Junfang Xu
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Li Chen
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Jianzhong Jeff Xi
- State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yiyan Fei
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai 200433, China
| | - Haipeng Liu
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Department of Microbiology and Immunology, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
36
|
Zhang D, Wang W, Tang M, Qu C, Jiang Z, Li X, Luan Y. In Situ Gene Engineering Approach to Overcome Tumor Resistance and Enhance T Cell-Mediated Cancer Immunotherapy. NANO LETTERS 2025; 25:6200-6208. [PMID: 40180619 DOI: 10.1021/acs.nanolett.5c00488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
T cell-mediated cancer immunotherapy harnesses the power of cytotoxic T lymphocytes (CTLs) to target and eradicate tumor cells. However, tumor cells often evade immune attack through membrane repair mechanisms involving endosomal sorting complexes required for transport (ESCRT) and immune suppression within the tumor microenvironment. Here, we developed a robust TMV@PpCHIL nanomedicine to address these issues by reprogramming tumor cells via in situ gene editing. Using CRISPR/Cas9, we disrupted the Chmp4b gene, a key component of the ESCRT machinery, preventing tumor cells from repairing CTL-induced membrane damage. Simultaneously, we genetically engineered tumor cells to produce interleukin-12 (IL-12), a cytokine that enhances CTL activation. The TMV@PpCHIL nanomedicine, designed by coating tumor membrane vesicles (TMVs) onto polyamidoamine (PAMAM) dendrimer-condensed plasmid complexes, ensured efficient CRISPR/Cas9-based gene editing and sustained IL-12 production. This approach significantly enhanced CTL-mediated tumor cell cytotoxicity, suppressed tumor growth, reduced metastasis, and prolonged survival, providing a promising strategy for durable cancer treatment.
Collapse
Affiliation(s)
- Di Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Wenjuan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Mingtan Tang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Chenghao Qu
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, China
| | - Zeyu Jiang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Xuerui Li
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Yuxia Luan
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| |
Collapse
|
37
|
Ye W, Zhang X, Tang Z, Hu Y, Zheng Y, Yuan Y. Comprehensive analysis of glycometabolism-related genes reveals PLOD2 as a prognostic biomarker and therapeutic target in gastric cancer. BMC Gastroenterol 2025; 25:256. [PMID: 40229676 PMCID: PMC11998276 DOI: 10.1186/s12876-025-03878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide, with limited therapeutic options and a poor prognosis, particularly in advanced stages. Glycometabolism, a hallmark of cancer, plays a critical role in tumor progression, immune evasion, and response to therapy. However, the specific roles of glycometabolism-related genes and their prognostic and therapeutic implications in GC remain inadequately understood. METHODS Transcriptomic and clinical data from GC patients were retrieved from TCGA and GEO databases. Glycometabolism-related genes were identified and analyzed using machine learning algorithms to construct a prognostic model. Functional assays, immune profiling, and pathway enrichment analyses were performed to explore the roles of these genes in tumor progression, immune-modulatory effects, and drug resistance. PLOD2, the gene with the highest prognostic significance, was further investigated to uncover its underlying regulatory mechanisms, roles in immune modulation, and contribution to therapeutic resistance. RESULTS A glycometabolism-related prognostic model consisting of four genes (PLOD2, CHSY3, SLC2A3 and SLC5A1) was developed and validated, effectively stratifying GC patients into high- and low-risk subgroups with distinct survival outcomes. Among these, PLOD2 emerged as the most significant gene, exhibiting strong associations with tumor progression and poor survival. Functional analyses revealed that PLOD2 promotes glycolysis and tumor progression through activation of the PI3K/AKT/mTOR pathway. Immune profiling revealed that PLOD2 overexpression is associated with an immunosuppressive tumor microenvironment, characterized by increased M2 macrophage infiltration and reduced immune activity. Moreover, treatment with rapamycin, an mTOR inhibitor, significantly suppressed PLOD2-mediated proliferation and anchorage-independent growth in GC cells, highlighting the central role of the PI3K/AKT/mTOR pathway in PLOD2-driven oncogenic behaviors. CONCLUSIONS This study identifies PLOD2 as a key prognostic biomarker and therapeutic target in gastric cancer. As a central component in a glycometabolism-related model, PLOD2 promotes glycolysis, tumor progression, and immune evasion via the PI3K/AKT/mTOR pathway. The model effectively stratifies patient risk, offering both prognostic utility and therapeutic insight. Targeting PLOD2-mediated pathways may represent a promising strategy for precision therapy and improved clinical outcomes in gastric cancer.
Collapse
Affiliation(s)
- Wanchun Ye
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Xiaolei Zhang
- Department of Clinical Laboratory, Jinan Fourth People's Hospital, Jinan, China
| | - Zhongjie Tang
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yufeng Hu
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yuanliang Zheng
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yuping Yuan
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China.
| |
Collapse
|
38
|
Shao X, Yu L, Li C, Qian J, Yang X, Yang H, Liao J, Fan X, Xu X, Fan X. Extracellular vesicle-derived miRNA-mediated cell-cell communication inference for single-cell transcriptomic data with miRTalk. Genome Biol 2025; 26:95. [PMID: 40229908 PMCID: PMC11998287 DOI: 10.1186/s13059-025-03566-x] [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: 06/26/2023] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
MicroRNAs are released from cells in extracellular vesicles (EVs), representing an essential mode of cell-cell communication (CCC) via a regulatory effect on gene expression. Single-cell RNA-sequencing technologies have ushered in an era of elucidating CCC at single-cell resolution. Herein, we present miRTalk, a pioneering approach for inferring CCC mediated by EV-derived miRNA-target interactions (MiTIs). The benchmarking against simulated and real-world datasets demonstrates the superior performance of miRTalk, and the application to four disease scenarios reveals the in-depth MiTI-mediated CCC mechanisms. Collectively, miRTalk can infer EV-derived MiTI-mediated CCC with scRNA-seq data, providing new insights into the intercellular dynamics of biological processes.
Collapse
Affiliation(s)
- Xin Shao
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women'S Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- The Joint-Laboratory of Clinical Multi-Omics Research Between, Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo, 315012, China.
| | - Lingqi Yu
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women'S Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chengyu Li
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jingyang Qian
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyu Yang
- The Center for Integrated Oncology and Precision Medicine, School of Medicine, Affiliated Hangzhou First People'S Hospital, Westlake University, Hangzhou, 310006, China
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Haihong Yang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, 310027, China
| | - Jie Liao
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xueru Fan
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiao Xu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People'S Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310024, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
| | - Xiaohui Fan
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women'S Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- The Joint-Laboratory of Clinical Multi-Omics Research Between, Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo, 315012, China.
| |
Collapse
|
39
|
Ciombor KK, Bae SW, Whisenant JG, Ayers GD, Sheng Q, Peterson TE, Smith GT, Lin K, Chowdhury S, Kanikarla Marie P, Sorokin A, Cohen AS, Goff LW, Cardin DB, Shen JP, Kopetz S, Eng C, Shyr Y, Berlin J, Manning HC. Results of the Phase I/II Study and Preliminary B-cell Gene Signature of Combined Inhibition of Glutamine Metabolism and EGFR in Colorectal Cancer. Clin Cancer Res 2025; 31:1437-1448. [PMID: 39927885 PMCID: PMC11996605 DOI: 10.1158/1078-0432.ccr-24-3133] [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: 09/23/2024] [Revised: 11/20/2024] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
PURPOSE EGFR-targeting mAbs are essential for managing rat sarcoma virus wild-type metastatic colorectal cancer (mCRC), but their limited efficacy necessitates exploring immunologic and metabolic factors influencing response. This study evaluated glutamine metabolism targeting with EGFR inhibition to identify response biomarkers in patients with prior anti-EGFR treatment progression. PATIENTS AND METHODS We conducted a phase I/II trial in patients with KRAS wild-type mCRC, combining panitumumab (6 mg/kg) and CB-839 (600 mg/kg or 800 mg/kg), hypothesizing that the dual inhibition of glutamine metabolism and MAPK signaling would enhance outcomes. As study correlatives, we investigated the B-cell activation signature "B-score" and glutamine PET as potential treatment response biomarkers. RESULTS The combination of panitumumab and CB-839 was tolerable with manageable side effects, including grade 4 hypomagnesemia in four patients, a known panitumumab-related event. Two patients achieved partial response, and five had stable disease, with a 41% disease control rate. Median progression-free survival and overall survival were 1.84 and 8.87 months, respectively. A positive correlation between "B-score" and lesion size reduction suggested its association with clinical benefit (partial response and stable disease). Lower "B-score" correlated with greater tumor avidity for glutamine by PET, indicating B-cell activation sensitivity to glutamine depletion. CONCLUSIONS The combination of CB-839 and panitumumab showed safety and promising preliminary responses, but the study closed early due to CB-839 development termination. The B-cell activation signature "B-score" emerged as a potential biomarker for EGFR and glutaminase inhibition in mCRC, warranting further studies. These findings suggest opportunities to improve immune response and therapies in glutaminolysis-dependent tumors.
Collapse
Affiliation(s)
- Kristen K Ciombor
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Seong-Woo Bae
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer G Whisenant
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Todd E Peterson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gary T Smith
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kangyu Lin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Preeti Kanikarla Marie
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexey Sorokin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Allison S Cohen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Laura W Goff
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dana B Cardin
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cathy Eng
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jordan Berlin
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
40
|
Soleimani Mamalo A, Pashaei M, Valilo M, Ojarudi M. Cytokine-mediated regulation of immune cell metabolic pathways in the tumor microenvironment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04133-8. [PMID: 40220026 DOI: 10.1007/s00210-025-04133-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Cancer, an important global health problem, is defined by aberrant cell proliferation and continues to be the main cause of death globally. The tumor microenvironment (TME) plays an essential role in the development of cancer, resistance to therapy, and regulation of the immune response. Some immune cells in the TME, like T cells, B cells, macrophages, dendritic cells, and natural killer cells, can either stop or help tumor growth, depending on how metabolic and cytokine changes happen. Cytokines function as essential signaling molecules that modulate immune cell metabolism, altering their functionality. This review focuses on how cytokine-mediated metabolic reprogramming affects the activity of immune cells inside the TME, which can either make the immune response stronger or weaker. New ways of treating cancer that focus on metabolic pathways and cytokine signaling, such as using IL (Interleukin) - 15, IL- 10, and IL- 4, show promise in boosting immune cell activity and making cancer treatments more effective. Finding these pathways could lead to new ways to treat cancer with immunotherapy that focus on metabolic competition and immune resistance in the TME.
Collapse
Affiliation(s)
| | - Mohammadreza Pashaei
- Department of Internal Medicine, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoud Ojarudi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
41
|
Chang X, Huang Y, Qu Y, Guo Y, Fan W, Zhen H. Integrative analysis of mitochondrial-related gene profiling identifies prognostic clusters and drug resistance mechanisms in low-grade glioma. Discov Oncol 2025; 16:465. [PMID: 40186003 PMCID: PMC11971116 DOI: 10.1007/s12672-025-02201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Mitochondrial dysfunction has emerged as a critical factor in the progression and prognosis of low-grade glioma (LGG). In this study, we explored the role of mitochondrial-related genes through consensus clustering analysis using multi-omics data from the TCGA, CGGA, and other independent datasets. Patients were categorized into three clusters (Cluster A, B, and C), with Cluster B consistently associated with poorer prognosis. Mutation landscape analysis revealed distinct genetic alterations and copy number variations among clusters, particularly in Cluster B, which exhibited unique genetic signatures. Immune infiltration analysis showed that Cluster B had higher expression levels of immune checkpoint genes, stronger immune evasion activity, and greater immune cell infiltration, suggesting an immunosuppressive tumor microenvironment. Furthermore, we identified mitochondrial-related prognostic markers and developed a MITscore based on gene expression patterns, which stratified patients into high- and low-risk groups. High MITscore groups displayed stronger stemness characteristics, poorer survival outcomes, and differential responses to chemotherapy and immunotherapy. Cross-validation with drug sensitivity and immunotherapy cohorts indicated that high MITscore patients were more sensitive to certain chemotherapeutic agents and responded better to immunotherapy. Finally, using the SRGA method, we identified novel biomarkers (KDR, LRRK2, SQSTM1) closely associated with mitochondrial function, which may serve as potential targets for therapeutic intervention. These findings highlight the critical role of mitochondrial dysfunction in LGG prognosis, tumor microenvironment regulation, and treatment response, providing new avenues for precision oncology.
Collapse
Affiliation(s)
- Xiaozan Chang
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China
| | - Yingxuan Huang
- Pediatric Intensive Care Unit, The Affiliated Hospital of Youjiang Medical University for Nationalities; Key Laboratory of Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases of Baise, Baise, China
| | - Ying Qu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Guo
- Nanfang Hospital (ZengCheng Branch), Southern Medical University, No. 28, Innovation Avenue, Ningxi Street, Guangzhou, China.
| | - Wenwen Fan
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Nanli, Panjiayuan, Beijing, 100021, China.
| | - Haining Zhen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
42
|
Pan Y, Qiu Y, Zhou X, Mao W, Xu X. Cancer-associated fibroblasts: multidimensional players in liver cancer. Front Oncol 2025; 15:1454546. [PMID: 40248197 PMCID: PMC12003132 DOI: 10.3389/fonc.2025.1454546] [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: 06/25/2024] [Accepted: 02/19/2025] [Indexed: 04/19/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs), the most abundant stromal cells in the tumor microenvironment (TME), control tumor growth through production and organization of the extracellular matrix (ECM) for a long time. However, the results from different studies that have focused on targeting CAFs to disturb tumor progression are extremely controversial. Recent studies using advanced single-cell RNA sequencing technology (scRNAseq) combined with multiple genetically engineered mouse models have identified diverse CAF subpopulations in the premalignant liver microenvironment (PME) of hepatocellular carcinoma (HCC) and TME of intrahepatic cholangiocarcinoma (ICC), providing a deeper understanding of the exact roles of each CAF subpopulation in cancer development. This review focuses on the specific protein markers, signaling pathways, and functions of various emerging CAF subclusters that contribute to the development of ICC and HCC. Elucidating the role and regulation of CAF subpopulations under different pathophysiological conditions will facilitate the discovery of new therapeutics that modulate CAF activity.
Collapse
Affiliation(s)
- Yanyun Pan
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yuangang Qiu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xinbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Wei Mao
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Zhejiang Key Laboratory of Integrative Chinese and Western Medicine for Diagnosis and Treatment of Circulatory Diseases, Hangzhou, China
| | - Xiaoming Xu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
43
|
Roichman A, Reyes-Castellanos G, Chen Z, Chen Z, Mitchell SJ, MacArthur MR, Sawant A, Levett L, Powers J, Gomez M, Ibrahim M, Xu X, Tomlinson B, Hang X, Wei Y, Kang Y, White E, Rabinowitz JD. Lack of consistent effect of dietary fiber on immune checkpoint blockade efficacy across diverse murine tumor models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645975. [PMID: 40235983 PMCID: PMC11996298 DOI: 10.1101/2025.03.28.645975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Immune checkpoint blockade (ICB) has transformed cancer treatment, but success rates remain low in most cancers. Recent research suggest that dietary fiber enhances ICB response in melanoma patients and murine preclinical models through microbiome-dependent mechanisms. Yet, the robustness of this effect across cancer types and dietary contexts remains unclear. Specifically, prior literature compared grain-based chow (high fiber) to low-fiber purified diet, but these diets differ also on other dimensions including phytochemicals. Here we investigated, in mice fed grain-based chow or purified diets with differing quantities of isolated fibers (cellulose and inulin), metabolite levels and ICB activity in multiple tumor models. The blood and fecal metabolome were relatively similar between mice fed high- and low-fiber purified diets, but differed massively between mice fed purified diets or chow, identifying the factor as diet type, independent of fiber. Tumor growth studies in three implantable and two spontaneous genetically engineered tumor models revealed that fiber has a weaker impact on ICB (anti-PD-1) efficacy than previously reported. In some models, dietary modulation impacted ICB activity, but not in a consistent direction across models. In none of the models did we observe the pattern expected if fiber controlled ICB efficacy: strong efficacy in both chow and high-fiber purified diet but low efficacy in low-fiber purified diet. Thus, dietary fiber appears to have limited or inconsistent effect on ICB efficacy in mouse models, and other dietary factors that correlate with fiber intake may underlie the clinical correlations between fiber consumption and immunotherapy outcomes.
Collapse
|
44
|
Zhu Y, Zhou Z, Du X, Lin X, Liang ZM, Chen S, Sun Y, Wang Y, Na Z, Wu Z, Zhong J, Han B, Zhu X, Fu W, Li H, Luo ML, Hu H. Cancer cell-derived arginine fuels polyamine biosynthesis in tumor-associated macrophages to promote immune evasion. Cancer Cell 2025:S1535-6108(25)00116-3. [PMID: 40185095 DOI: 10.1016/j.ccell.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 01/08/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Arginine metabolism reshapes the tumor microenvironment (TME) into a pro-tumor niche through complex metabolic cross-feeding among various cell types. However, the key intercellular metabolic communication that mediates the collective effects of arginine metabolism within the TME remains unclear. Here, we reveal that the metabolic interplay between cancer cells and macrophages plays a dominant role in arginine-driven breast cancer progression. Within the TME, breast cancer cells serve as the primary source of arginine, which induces a pro-tumor polarization of tumor-associated macrophages (TAMs), thereby suppressing the anti-tumor activity of CD8+ T cells. Notably, this cancer cell-macrophage interaction overrides the arginine-mediated enhancement of CD8+ T cell anti-tumor activity. Mechanistically, polyamines derived from arginine metabolism enhance pro-tumor TAM polarization via thymine DNA glycosylase (TDG)-mediated DNA demethylation, regulated by p53 signaling. Importantly, targeting the arginine-polyamine-TDG axis between cancer cells and macrophages significantly suppresses breast cancer growth, highlighting its therapeutic potential.
Collapse
Affiliation(s)
- Yinghua Zhu
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Genetic Medicine, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Ziwei Zhou
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xin Du
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xiaorong Lin
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou 515031, China
| | - Zhi-Mei Liang
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Si Chen
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yiwei Sun
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310018, China; Experimental Research Center, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yue Wang
- Experimental Research Center, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhenkun Na
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310018, China
| | - Zhiyong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou 515031, China
| | - Jiaxin Zhong
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Beinan Han
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xiangping Zhu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Wenkui Fu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Hongde Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310018, China.
| | - Man-Li Luo
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Hai Hu
- Breast Cancer Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
45
|
Yuan X, Xiao Y, Yu D. Turn cold tumors hot by reprogramming the tumor microenvironment. Nat Biotechnol 2025; 43:466-470. [PMID: 40229361 DOI: 10.1038/s41587-025-02597-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Affiliation(s)
- Xiangliang Yuan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yi Xiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
46
|
Wu Y, Song W, Su M, He J, Hu R, Zhao Y. The Role of Cholesterol Metabolism and Its Regulation in Tumor Development. Cancer Med 2025; 14:e70783. [PMID: 40145543 PMCID: PMC11948085 DOI: 10.1002/cam4.70783] [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: 10/28/2024] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Within the tumor microenvironment, tumor cells undergo metabolic reprogramming of cholesterol due to intrinsic cellular alterations and changes in the extracellular milieu. Furthermore, cholesterol reprogramming within this microenvironment influences the immune landscape of tumors, facilitating immune evasion and consequently promoting tumorigenesis. These biological changes involve modifications in numerous enzymes associated with cholesterol uptake and synthesis, including NPC1L1, SREBP, HMGCR, SQLE, and PCSK9. REVIEW This review systematically summarizes the role of cholesterol metabolism and its associated enzymes in cancer progression, examines the mechanisms through which dysregulation of cholesterol metabolism affects immune cells within the tumor microenvironment, and discusses recent advancements in cancer therapies that target cholesterol metabolism. CONCLUSION Targeting cholesterol metabolism-related enzymes can inhibit tumor growth, reshape immune landscapes, and rejuvenate antitumor immunity, offering potential therapeutic avenues in cancer treatment.
Collapse
Affiliation(s)
- Yongmei Wu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Wenqian Song
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Min Su
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Jing He
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Rong Hu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Youbo Zhao
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| |
Collapse
|
47
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [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: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
48
|
Peng P, Shen F, Peng B, Chen Z, Zhou L, Hao X, Liu Y. Genetic Evidence Supporting the Repurposing of mTOR Inhibitors for Reducing BMI. Biomedicines 2025; 13:839. [PMID: 40299431 PMCID: PMC12025023 DOI: 10.3390/biomedicines13040839] [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: 03/10/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Although mTOR has long been regarded as a promising target for cancer treatment, the efficacy of mTOR inhibitors in most clinical trials has been rather limited. Nevertheless, their favorable safety profile has opened up opportunities for drug repurposing, even as their potential applications across various diseases remain largely unexplored. Methods: We performed an MR-PheWAS analysis across 1431 phenotypes to explore drug repurposing opportunities. We analyzed GWAS data of 452 plasma metabolites, 731 immune traits, and 412 gut microbiota to uncover potential mechanisms for the causal link between the mTOR gene and body mass index (BMI). Results: A causal link between mTOR gene expression and BMI has been established. Additionally, mTOR-related vulnerabilities associated with BMI, including alterations in metabolites, immune traits, and gut microbiota, were identified. Conclusions: The identified causal relationship between mTOR and BMI suggests novel potential non-cancer applications for mTOR inhibitors.
Collapse
Affiliation(s)
- Ping Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (B.P.); (Z.C.); (L.Z.)
| | - Fan Shen
- Nursing Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Bi Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (B.P.); (Z.C.); (L.Z.)
| | - Ziqi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (B.P.); (Z.C.); (L.Z.)
| | - Lei Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (B.P.); (Z.C.); (L.Z.)
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yuanhui Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (B.P.); (Z.C.); (L.Z.)
| |
Collapse
|
49
|
Wu Y, Shi Y, Luo Z, Zhou X, Chen Y, Song X, Liu S. Spatial multi-omics analysis of tumor-stroma boundary cell features for predicting breast cancer progression and therapy response. Front Cell Dev Biol 2025; 13:1570696. [PMID: 40206396 PMCID: PMC11979139 DOI: 10.3389/fcell.2025.1570696] [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: 02/04/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
Background The tumor boundary of breast cancer represents a highly heterogeneous region. In this area, the interactions between malignant and non-malignant cells influence tumor progression, immune evasion, and drug resistance. However, the spatial transcriptional profile of the tumor boundary and its role in the prognosis and treatment response of breast cancer remain unclear. Method Utilizing the Cottrazm algorithm, we reconstructed the intricate boundaries and identified differentially expressed genes (DEGs) associated with these regions. Cell-cell co-positioning analysis was conducted using SpaCET, which revealed key interactions between tumor-associated macrophage (TAMs) and cancer-associated fibroblasts (CAFs). Additionally, Lasso regression analysis was employed to develop a malignant body signature (MBS), which was subsequently validated using the TCGA dataset for prognosis prediction and treatment response assessment. Results Our research indicates that the tumor boundary is characterized by a rich reconstruction of the extracellular matrix (ECM), immunomodulatory regulation, and the epithelial-to-mesenchymal transition (EMT), underscoring its significance in tumor progression. Spatial colocalization analysis reveals a significant interaction between CAFs and M2-like tumor-associated macrophage (TAM), which contributes to immune exclusion and drug resistance. The MBS score effectively stratifies patients into high-risk groups, with survival outcomes for patients exhibiting high MBS scores being significantly poorer. Furthermore, drug sensitivity analysis demonstrates that high-MB tumors had poor response to chemotherapy strategies, highlighting the role of the tumor boundary in modulating therapeutic efficacy. Conclusion Collectively, we investigate the spatial transcription group and bulk data to elucidate the characteristics of tumor boundary molecules in breast cancer. The CAF-M2 phenotype emerges as a critical determinant of immunosuppression and drug resistance, suggesting that targeting this interaction may improve treatment responses. Furthermore, the MBS serves as a novel prognostic tool and offers potential strategies for guiding personalized treatment approaches in breast cancer.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Youyang Shi
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhanyang Luo
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiqiu Zhou
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghao Chen
- West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoyun Song
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
50
|
Li S, Yuan H, Yang XZ, Xu X, Yu W, Wu Y, Yao S, Xie J, He W, Guo Z, Chen Y. Synergistic Antitumor Immunotherapy via Mitochondria Regulation in Macrophages and Tumor Cells by an Iridium Photosensitizer. ACS CENTRAL SCIENCE 2025; 11:441-451. [PMID: 40161955 PMCID: PMC11950858 DOI: 10.1021/acscentsci.4c02156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Mitochondrial targeting has emerged as an attractive method for antitumor treatment. However, most of the mitochondria targeted drugs focused on inhibiting tumor cells, while their potential for activation of immune responses in the tumor microenvironment has rarely been described. In this study, we report a photosensitive iridium complex MitoIrL2, which enabled the simultaneous mitochondrial modulation of macrophages and tumor cells to achieve synergistic antitumor immunity. The adjustment of the mitochondrial respiratory chain, HIF-1α, and the NF-κB pathway in macrophages drove the metabolic reprogramming from oxidative phosphorylation (OXPHOS) to glycolysis, converting protumor M2 into the antitumor M1 phenotype. Downregulated expression of immunosuppressive checkpoint SIRPα has also been observed on macrophages. Meanwhile, the mitochondrial targeting MitoIrL2 enhanced the immunogenic cell death of tumor cells and reversed the immunosuppressive tumor microenvironment, which activated the systemic immune response and established long-term immune memory in vivo. This work illustrates a promising strategy to simultaneously regulate macrophages toward the antitumor phenotype and enhance immunogenic cell death in tumor cells for synergistic antitumor immunotherapy.
Collapse
Affiliation(s)
- Shumeng Li
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Hao Yuan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Xiu-Zhi Yang
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Xinyu Xu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Wenhao Yu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Yanping Wu
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Shankun Yao
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Jin Xie
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Weijiang He
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
| | - Yuncong Chen
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Chemistry and Biomedicine Innovation Center
(ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing 210023, Jiangsu, P.R. China
- Department
of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, Jiangsu, P.R. China
| |
Collapse
|