1
|
Barone S, Bello I, Guadagni A, Cerchia C, Filocamo G, Cassese E, Alfano AI, Esposito C, Feliz Morel ÁJ, Brunetti M, Lavecchia A, Summa V, Panza E, Brindisi M. Challenging triple negative breast cancer through HDAC6 selective inhibition: Novel cap-group identification, structure-activity relationships, computational and biological studies. Eur J Med Chem 2025; 292:117634. [PMID: 40288122 DOI: 10.1016/j.ejmech.2025.117634] [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: 02/13/2025] [Revised: 04/03/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Triple negative breast cancer (TNBC) stands out among breast cancers subtypes for its high aggressiveness and invasiveness. Compelling new evidence pointed out the role of epigenetic modifications in TNBC, with recent studies demonstrating that approximately 30 % of human breast cancers could potentially benefit from histone deacetylase 6 (HDAC6) inhibitor therapy. We herein disclose a novel class of spiro-fused compounds acting as potent and selective HDAC6 inhibitors. Structure-based optimization led to derivatives 23c and 24c with high potency and selectivity towards HDAC6 in vitro and in cell-based settings. Following our observation that mRNA expression level of HDAC6 was significantly higher in MDA-MB-231, we have evaluated the effect of the compounds on cell viability. Moreover, we have unveiled for compounds 23c and 24c the involvement of the autophagic machinery in cell death induction. Scratch assay revealed for the newly conceived compounds a very potent effect on inhibiting the migration process in MDA-MB-231 cells. Our results underscore the key role of HDAC6 in TNBC progression, providing a solid groundwork to reshape TNBC therapy.
Collapse
Affiliation(s)
- Simona Barone
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Ivana Bello
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Anna Guadagni
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Carmen Cerchia
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | | | - Emilia Cassese
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Antonella Ilenia Alfano
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Camilla Esposito
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | | | - Mirko Brunetti
- Exiris s.r.l., Via di Castel Romano, 100, 00128, Rome, Italy
| | - Antonio Lavecchia
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Vincenzo Summa
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Elisabetta Panza
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Margherita Brindisi
- Department of Pharmacy, Department of Excellence, 2023-2027, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| |
Collapse
|
2
|
Caliendo A, Camorani S, Ibarra LE, Pinto G, Agnello L, Albanese S, Caianiello A, Illiano A, Festa R, Ambrosio V, Scognamiglio G, Cantile M, Amoresano A, Fedele M, Zannetti A, Cerchia L. A novel CD44-targeting aptamer recognizes chemoresistant mesenchymal stem-like TNBC cells and inhibits tumor growth. Bioact Mater 2025; 50:443-460. [PMID: 40342488 PMCID: PMC12059597 DOI: 10.1016/j.bioactmat.2025.04.027] [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: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/20/2025] [Indexed: 05/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) represents a significant therapeutic challenge owing to the scarcity of targeted medicines and elevated recurrence rates. We previously reported the development of the nuclease-resistant RNA sTN58 aptamer, which selectively targets TNBC cells. Here, sTN58 aptamer was employed to capture and purify its binding target from the membrane protein fraction of cisplatin-resistant mesenchymal stem-like TNBC cells. Mass spectrometry in conjunction with aptamer binding assays across various cancer cell lines identified CD44 as the cellular target of sTN58. By binding to CD44, sTN58 inhibits the invasive growth and hyaluronic acid-dependent tube formation in chemoresistant TNBC cells, where CD44 serves as a key driver of tumor cell aggressiveness and stem-like plasticity. Moreover, in vivo studies demonstrated the aptamer's high tumor targeting efficacy and its capacity to significantly inhibit tumor growth and lung metastases following intravenous administration in mice with orthotopic TNBC. Overall, our findings reveal the striking potential of sTN58 as a targeting reagent for the recognition and therapy of cancers overexpressing CD44.
Collapse
Affiliation(s)
- Alessandra Caliendo
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Simona Camorani
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Luis Exequiel Ibarra
- Institute of Environmental Biotechnology and Health (INBIAS), National University of Rio Cuarto (UNRC), National Council for Scientific and Technological Research (CONICET), Río Cuarto, X5800BIA, Argentina
| | - Gabriella Pinto
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Lisa Agnello
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging, National Research Council, 80145, Naples, Italy
| | - Antonietta Caianiello
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Anna Illiano
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Rosaria Festa
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Vincenzo Ambrosio
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Giosuè Scognamiglio
- Institutional Biobank-Scientific Directorate, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Monica Cantile
- Institutional Biobank-Scientific Directorate, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Angela Amoresano
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Monica Fedele
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, National Research Council, 80145, Naples, Italy
| | - Laura Cerchia
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| |
Collapse
|
3
|
Wen R, Ma Y, Liu C, Feng R. Class-aware multi-level attention learning for semi-supervised breast cancer diagnosis under imbalanced label distribution. Med Biol Eng Comput 2025; 63:1997-2009. [PMID: 39907850 DOI: 10.1007/s11517-025-03291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 01/11/2025] [Indexed: 02/06/2025]
Abstract
Breast cancer affects a significant number of patients worldwide, and early diagnosis is critical for improving cure rates and prognosis. Deep learning-based breast cancer classification algorithms have substantially alleviated the burden on medical personnel. However, existing breast cancer diagnosis models face notable limitations which are challenging to obtain in clinical settings, such as reliance on a large volume of labeled samples, an inability to comprehensively extract features from breast cancer images, and susceptibility to overfitting on account of imbalanced class distribution. Therefore, we propose the class-aware multi-level attention learning model focused on semi-supervised breast cancer diagnosis to effectively reduce the dependency on extensive data annotation. Additionally, we develop the multi-level fusion attention learning module, which integrates multiple mutual attention components across different layers, allowing the model to precisely identify critical regions for lesion categorization. Finally, we design the class-aware adaptive pseudo-labeling module which adaptively predicts category distribution in unlabeled data, and directs the model to focus on underrepresented categories, ensuring a balanced learning process. Experimental results on the BACH dataset demonstrate that our proposed model achieves an accuracy of 86.7% with only 40% labeled microscopic data, showcasing its outstanding contribution to semi-supervised breast cancer diagnosis.
Collapse
Affiliation(s)
- Renjun Wen
- China Comservice Enrising Information Technology Co., Ltd., Chengdu, Sichuan, 610041, China
| | - Yufei Ma
- Sichuan Provincial Government Affairs Service and Public Resources Exchange Service Center, No.2, Caoshi Street, Qingyang District, Chengdu City, Sichuan Province, 610000, China.
| | - Changdong Liu
- China Comservice Enrising Information Technology Co., Ltd., Chengdu, Sichuan, 610041, China
| | - Renwei Feng
- China Comservice Enrising Information Technology Co., Ltd., Chengdu, Sichuan, 610041, China
| |
Collapse
|
4
|
Huang R, Zhang B, Chen G, Zhao Y, Wang R, Zhu H, Duan S, Mo L, Li Z, Zhang X, Zhang L. Ultrasound-mediated FAK-targeted nano-sapper for tumor extracellular matrix remodeling to potentiate cancer immunotherapy. CHEMICAL ENGINEERING JOURNAL 2025; 515:163837. [DOI: 10.1016/j.cej.2025.163837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2025]
|
5
|
Xu X, Zhou H, Hong R, Gong J, Wan Y, Fu Q, Huang K, Li Y, Wang N, Zhao P, Cai K, Li F. A self-accelerating 'copper bomb' strategy activated innate and adaptive immune response against triple-negative breast cancer. Bioact Mater 2025; 49:193-206. [PMID: 40130080 PMCID: PMC11931225 DOI: 10.1016/j.bioactmat.2025.02.019] [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: 10/29/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 03/26/2025] Open
Abstract
Triple-negative breast cancer (TNBC) presents therapeutic challenges due to its aggressive, drug-resistance, and low immunological reactivity. Cuproptosis, an emerging therapeutic modality, is a promising strategic intervention for treating TNBC. Nonetheless, the effectiveness of cuproptosis is compromised by tumor adaptations, including the Warburg effect, increased intracellular glutathione (GSH), and copper efflux, thus breaking the barrier of cuproptosis is the basis for developing cuproptosis-based clinical therapies. Herein, a self-accelerating strategy utilizing a pH-responsive copper framework encapsulating glucose oxidase (GOx), modified with polyethylene glycol (PEG) and tumor-penetrating peptide (tLyp1) has been developed. Upon reaching the acidic tumor microenvironment, the released GOx increases intracellular acidity and hydrogen peroxide (H2O2). The elevated intracellular GSH and H2O2 serve as "fuel" to amplify the copper-based catalytic within tumor cells. Concurrently, the reduction of copper efflux proteins (ATP7B) and the depletion of GSH lead to copper overload in tumor cells, leading to cuproptosis via copper overload, mitochondrial disruption, and Fe-S protein instability. This constellation of interrelated events constitutes a potent "Copper Bomb," which concurrently triggers the immune system and effectively kills the tumor. It robustly engages innate and adaptive immunity via the release of mitochondrial DNA, facilitating the cGAS-STING pathway and precipitating immunogenic cell death. This process reverses the immunosuppressive tumor microenvironment, eliminates tumor cells, and suppresses metastasis, thus offering a novel therapeutic modality for the comprehensive treatment of triple-negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Xinzhi Xu
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Hang Zhou
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ruixia Hong
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jiaqi Gong
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yujie Wan
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Qihuan Fu
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Kaifeng Huang
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ying Li
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Na Wang
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Peng Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Fang Li
- Department of Ultrasound, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
6
|
Ren H, Liu S, Ji D, Li X, Sun X, Wang W, Liu T, Li Y. Transcriptome analysis reveals the potential role of neural factor EN1 for long-terms survival in estrogen receptor-independent breast cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200965. [PMID: 40207200 PMCID: PMC11981748 DOI: 10.1016/j.omton.2025.200965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/27/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025]
Abstract
Breast cancer patients with estrogen receptor-negative (ERneg) status, encompassing triple negative breast cancer (TNBC) and human epidermal growth factor receptor 2 positive breast cancer, are confronted with a heightened risk of drug resistance, often leading to early recurrence; the biomarkers and biological processes associated with recurrence is still unclear. In this study, we analyzed bulk RNA sequencing (RNA-seq) data from 285 cancer and paracancerous samples from 155 TNBC patients, along with transcriptome data from 11 independent public cohorts comprising 7,449 breast cancer patients and 26 single-cell RNA-seq datasets. Our results revealed differential enrichment of nerve-related pathways between TNBC patients with and without 10-year recurrence-free survival. We developed an early recurrence index (ERI) using a machine learning model and constructed a nomogram that accurately predicts the 10-year survival of ERneg patients (area under the curve [AUC]Training = 0.79; AUCTest = 0.796). Further analysis linked ERI to enhanced neural function and immunosuppression. Additionally, we identified EN1, the most significant ERI gene, as a potential biomarker that may regulate the tumor microenvironment and sensitize patients to immunotherapy.
Collapse
Affiliation(s)
- He Ren
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Shan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Dongchen Ji
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Xue Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Xue Sun
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Wenzheng Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, China
| |
Collapse
|
7
|
Zeng L, You K, Lu M, Hu X, Zheng C, Yao L, Kang B, Lin S, Deng X, Yan J, Zhou X. A Multimodal Nanoplatform Integrating Photodynamic Therapy and Wnt/β-Catenin Inhibition Reprograms the Tumor Microenvironment to Potentiate Immune Checkpoint Therapy in Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40525603 DOI: 10.1021/acsami.5c04799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Abstract
The immunosuppressive tumor microenvironment (TME) poses a critical barrier to the efficacy of immune checkpoint inhibitors in triple-negative breast cancer (TNBC). Here, we report a self-assembled polymeric nanoplatform coloading the photosensitizer verteporfin and the Wnt/β-catenin inhibitor XAV-939. This dual-functional system enhanced cellular uptake and potentiated photodynamic therapy (PDT)-induced immunogenic cell death, while simultaneously downregulating β-catenin signaling to reverse immunosuppression. In vivo, the nanoplatform substantially improved therapeutic outcomes, converting "cold" tumors into immune-responsive phenotypes characterized by augmented dendritic cell maturation, increased cytotoxic T cell infiltration, reduced regulatory T cell abundance, and enhanced proinflammatory cytokine release. Combined with PD-L1 blockade, this strategy synergistically activated systemic antitumor immunity, resulting in primary tumor regression, metastasis reduction, and systemic abscopal effects against distal tumors. These results highlight the promise of targeted TME reprogramming as a strategy to overcome TNBC's recalcitrance to immunotherapy.
Collapse
Affiliation(s)
- Lingjun Zeng
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Kaiqin You
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Mingjian Lu
- Department of Pharmacy, Yongding District Hospital of Longyan City, Longyan 364100, PR China
| | - Xiaomu Hu
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Changqing Zheng
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Lingyan Yao
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Bingkun Kang
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Shuang Lin
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Xiaoliang Deng
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Jia Yan
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| | - Xin Zhou
- Department of Pharmacy, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou 350025, PR China
| |
Collapse
|
8
|
Tang Y, Xu A, Xu Z, Xie J, Huang W, Zhang L, Chen Y, Yang L, Du S, Wang K. Multi-omics analyses of the heterogenous immune microenvironment in triple-negative breast cancer implicate UQCRFS1 potentiates tumor progression. Exp Hematol Oncol 2025; 14:85. [PMID: 40524231 PMCID: PMC12172350 DOI: 10.1186/s40164-025-00672-1] [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: 11/12/2024] [Accepted: 05/14/2025] [Indexed: 06/19/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is commonly characterized by high-grade and aggressive features, resulting in an augmented likelihood of distant metastasis and inferior prognosis for patients. Tumor immune microenvironment (TME) has been recently considered to be tightly correlated with tumor progression and immunotherapy response. However, the actual heterogenous TME within TNBC remains more explorations. METHODS The thorough analyses of different cell types within TME were conducted on the self-tested single-cell RNA sequencing dataset which contained nine TNBC treatment-naïve patients, including subclusters classification, CellChat algorithm, transcription factors (TFs) expression, pseudotime analysis and functional enrichment assay. The malignant epithelial cluster was confirmed by copy number variations analysis, and subsequently LASSO-Cox regression was carried out to establish a Malignant Cell Index (MCI) model on the basis of five crucial genes (BGN, SDC1, IMPDH2, SPINT1, and UQCRFS1), which was validated in several TNBC cohorts through Kaplan-Meier survival and immunotherapy response analyses. The public spatial transcriptome, proteome data and qRT-PCR, western blotting experiments were exploited to corroborate UQCRFS1 expression in RNA and protein levels. Additionally, functional experiments were implemented to unravel the impacts of UQCRFS1 on TNBC cells. RESULTS The diverse subclusters of TME cells within TNBC were clarified to display distinct characteristics in cell-cell interactions, TFs expression, differentiation trajectory and functional pathways. Particularly, IL32high Treg imparted an essential effect on tumor evasion and predicted a worsened prognosis of TNBC patients. Furthermore, MCI model enabled to notify the inferior prognosis and immunotherapy resistance in TNBC. Ultimately, UQCRFS1 knockdown dampened the proliferative and migratory competence in vitro as well as tumor growth in vivo of TNBC cells. CONCLUSIONS Our study offers innovative perspectives on comprehending the heterogeneity within TME of TNBC, thereby facilitating the elucidation of TNBC biology and providing clinical recommendations for TNBC patients' prognosis, such as IL32high Treg infiltration, MCI evaluation, and UQCRFS1 expression.
Collapse
Affiliation(s)
- Yuhui Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Aiqi Xu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Zhongbiao Xu
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Jindong Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Wei Huang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Liulu Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Yitian Chen
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China
| | - Lu Yang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China.
| | - Shasha Du
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China.
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
9
|
Lu J, Yin A, Tan S, Zhuge R, Liu Y, Zhang P, Liu L, Xuan X, Li H, Wang W, Zhang X, Yin Q, Wang H. Design, Synthesis, and Evaluation of Pyrazolopyridine Derivatives as Novel Calreticulin (CALR) Ligands That Inhibit Triple-Negative Breast Cancer (TNBC) via Inducing Calcium Overloading. J Med Chem 2025; 68:11419-11436. [PMID: 40464282 DOI: 10.1021/acs.jmedchem.5c00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, posing significant therapeutic challenges due to the lack of effective targets. Elevating intracellular calcium levels is a promising strategy in cancer therapy, and highly expressed calreticulin (CALR) in tumors has emerged as a potential target for inducing calcium overload. However, few studies on CALR ligands have been reported. Herein, we designed, synthesized, and evaluated pyrazolopyridine derivatives as potential CALR ligands. Among them, the leading compound 2a was identified as a high binding affinity ligand (Kd = 2.6 μM) with potent antitumor activity (IC50 = 0.1 μM). Mechanistic studies demonstrated that 2a could interact with CALR, inducing calcium overload and leading to apoptosis in TNBC cells. Further in vivo pharmacodynamic evaluations confirmed the safety and antitumor activity of 2a. In conclusion, our findings developed a novel CALR ligand and provided a new anti-TNBC strategy via inducing calcium dysregulation.
Collapse
Affiliation(s)
- Jiaguo Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Anqi Yin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Shenpeng Tan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Ruihong Zhuge
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Yi Liu
- School of Chemistry and Chemical Engineering, Yantai University, Yantai 264005, China
| | - Peng Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Li Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Xuan Xuan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Huirong Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Wenyan Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Xiaolin Zhang
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, Sichuan 610051, China
| | - Qikun Yin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Basic Science Research Center Base (Pharmaceutical Science), Yantai University, Yantai 264005, China
| |
Collapse
|
10
|
Dai M, Tian Z, Xu F, Yao B, Liang H, Li D, Wang J, Rong J, Liu T, Tang H, Lu H, Zhang W. Radiation-Based Multi-Modal Therapy Combining with Immunotherapy to Develop a Vaccine-Like Effective Treatment for Triple-Negative Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:483-496. [PMID: 40524764 PMCID: PMC12169013 DOI: 10.2147/bctt.s518625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 05/21/2025] [Indexed: 06/19/2025]
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive malignancy with high metastasis and recurrence rates. Current treatments like chemotherapy and immunotherapy face challenges due to chemotherapy side effects, limited immunotherapy applicability, and TNBC's immunosuppressive microenvironment. Purpose To achieve a more effective treatment for TNBC, a novel therapeutic strategy has been developed, which uses X-ray excited photodynamic therapy (X-PDT) to activate the tumor immune microenvironment following with the immunotherapy of Anti-CTLA4. Methods Base on the 4T1 tumor mouse model, this study initially investigated the regulatory effects of X-PDT on the tumor immune microenvironment. Subsequently, the therapeutic efficacy of combining X-PDT with Anti-CTLA4 was evaluated for its inhibitory effects on primary, metastatic, and recurrent tumors. The underlying mechanisms were further elucidated through comprehensive techniques including flow cytometry, ELISA, and immunofluorescence assays. Results The synergistic strategy can effectively ablate the primary tumor while inhibiting metastasis and preventing recurrence like a vaccine. It enhances intratumoural dendritic cells (DCs) maturation (from 25.7% to 58.3%, P < 0.05) and immune T cell infiltration activating a strong anti-tumor immune response. The anti-tumor efficacy of synergistic therapy is enhanced by 2.5 times comparing with immunotherapy alone, while the tumor metastasis has been inhibited significantly. The maturation level of mature dendritic cells was increased from 26.7% to 86.3% (P < 0.01). The intratumoural CD8+/CD4+ T cells were increased from 0.51% and 1.54% to 15.4% and 23.1% (P < 0.0001), respectively. The synergistic therapy exerts a powerful vaccine-like long-term immune memory function to prevent tumor recurrence with the elevated level of effector memory T (Tem) cells (from 12.8% to 33.3%, P < 0.05). Conclusion Based on the 4T1 mouse model, developed an effective vaccine-like therapeutic strategy combining X-PDT with Anti-CTLA4, which can effectively ablate tumors, inhibit metastasis, and prevent tumor recurrence. This work may provide a novel effective therapeutic modality for the clinical treatment of TNBC.
Collapse
Affiliation(s)
- Mengyan Dai
- The College of Life Sciences, Northwest University, Xi’an City, 710069, People’s Republic of China
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Zuhong Tian
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Fanyuan Xu
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Bang Yao
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
- School of Materials Science and Engineering, Shaanxi University of Science and Technology, Xi’an, Shaanxi, 710021, People’s Republic of China
| | - Hongxia Liang
- The College of Life Sciences, Northwest University, Xi’an City, 710069, People’s Republic of China
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Dongyan Li
- The College of Life Sciences, Northwest University, Xi’an City, 710069, People’s Republic of China
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Jiangang Wang
- Department of General Surgery, The Second Affiliated Hospital of the Fourth Military Medical University, Xi’an, 710038, People’s Republic of China
| | - Junyan Rong
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Tianshuai Liu
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Haili Tang
- Department of General Surgery, The Second Affiliated Hospital of the Fourth Military Medical University, Xi’an, 710038, People’s Republic of China
| | - Hongbing Lu
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| | - Wenli Zhang
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University, Xi’an, Shaanxi, 710032, People’s Republic of China
| |
Collapse
|
11
|
Wang H, Yao L, Zhong L, Fang J, He Q, Busch TM, Cengel K, Qin L. Marrow adipogenic lineage precursors (MALPs) facilitate bone marrow recovery after chemotherapy. Bone 2025; 195:117446. [PMID: 40057216 DOI: 10.1016/j.bone.2025.117446] [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: 12/12/2024] [Revised: 02/22/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Chemotherapy-induced hematopoietic toxicity is a multifactorial challenge in the treatment of oncology patients. The resultant bone marrow suppression is a major dose-limiting side effect. In this study, we utilized 5-fluorouracil (5-FU), a commonly used chemotherapeutic agent, to investigate the mechanisms underlying bone marrow recovery following chemotherapy. A single injection of 5-FU did not alter mouse bone structure but caused acute damage to bone marrow cellularity and vasculature. Single-cell RNA-sequencing of bone marrow mesenchymal lineage cells revealed a substantial reduction in early mesenchymal progenitors and a marked expansion of marrow adipogenic lineage precursors (MALPs) five days post-treatment. Furthermore, 5-FU upregulated the expression of myofibroblast markers in MALPs, indicating a myofibroblast transformation. Using Adipoq-Cre to label MALPs in vivo, we observed that 5-FU transiently increases the number of MALPs in the bone marrow by promoting their proliferation. Immunostaining confirmed the elevated expression of myofibroblast markers in MALPs. By day 14 after 5-FU injection, bone marrow cellularity and vasculature were largely restored; however, the ablation of MALPs significantly impaired this recovery. Taken together, our study uncovers the critical role of MALPs in facilitating bone marrow repair following chemotherapy-induced injury and identifies them as a potential cellular target for treating chemotherapy-induced myelosuppression.
Collapse
Affiliation(s)
- Huan Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiankang Fang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qi He
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Keith Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Kim MJ, Kim HJ, Kim JY, Shin J, Park YH. Effectiveness of Adjuvant Capecitabine in Triple-Negative Breast Cancer Patients With Residual Disease After Neoadjuvant Treatment: A Real-World Evidence Study in Korea. Clin Breast Cancer 2025; 25:e431-e439.e4. [PMID: 39909791 DOI: 10.1016/j.clbc.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 02/07/2025]
Abstract
BACKGROUND Residual disease after neoadjuvant chemotherapy (NAC) has important role in triple negative breast cancer (TNBC). The CREATE-X study demonstrated a survival benefit from adjuvant capecitabine (adjC) in breast cancer patients, especially for TNBC populations. Because the landscape of early TNBC treatment has been changing rapidly, an optimal adjuvant strategy for real-world practice is needed. We evaluated the effectiveness of adjC in TNBC patients with residual disease after NAC. METHOD We used de-identified, anonymous data from an institutional clinical data warehouse to retrospectively analyze 934 TNBC patients who received NAC between 2017 and 2023. Among them, 405 patients received at least 1 cycle of adjC, and 77 received no adjuvant treatment. The primary outcomes of the study were distant-disease free survival (DDFS) rate and overall survival (OS) rate at 3 years. The secondary outcomes were subgroup analyses and Cox regression analyses of survival outcomes. RESULT The median follow up period was 34.3 months (range 1.8-71.5). The DDFS rate at 3 years was higher in the capecitabine group: 86.3% of the capecitabine group and 74.4% of the no adjuvant group (P = .019). The OS rates at 3 years were 93.3% and 83.8%, respectively (P = .032). Subgroup analyses indicated a greater benefit from adjC in patients aged 50 years or older and those who received platinum-based NAC, both in terms of DDFS and OS. CONCLUSION Our study showed that adjC was more effective than no adjuvant treatment for TNBC patients with residual disease in terms of DDFS and OS.
Collapse
Affiliation(s)
- Min Jeong Kim
- Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea; Research Institution for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyo Jung Kim
- Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea; Research Institution for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Junghoon Shin
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yeon Hee Park
- Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea; Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Nie HJ, Fu YJ, Long S, Wang JY, Zhao WS, Zhai LH, Yang YL, Tan MJ, Hu H, Chen XH. Chemoproteomics reveals proteome-wide covalent and non-covalent targets of withaferin A. Acta Pharmacol Sin 2025; 46:1782-1793. [PMID: 39900821 PMCID: PMC12098870 DOI: 10.1038/s41401-024-01468-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/22/2024] [Indexed: 02/05/2025]
Abstract
Withaferin A (WA), a natural product used in traditional medicine, has recently garnered attention because of its diverse pharmacological effects. However, the direct targets responsible for these effects remain elusive. The discovery of targets is usually serendipitous and research has predominantly concentrated on covalent interactions, overlooking non-covalent targets. The unbiased and proteome-wide mapping of WA-interacting proteins in living cells remains largely unexplored. We have developed a chemical proteomics platform that enabled profiling of the covalent/non-covalent interactome and target occupancy in disease-related cells, which was used to reveal the landscape of the targets of WA in triple-negative breast cancer (TNBC) cells. Analysis of the discovered high-occupancy targets suggested that WA was substantially involved in the RNA metabolism pathway, in addition to other biological processes. Moreover, we biochemically validated a selection of previously unknown high-occupancy targets from various important biological pathways, including the non-covalent target MVK and covalent targets HNRNPF and CKAP4, which all play critical roles in TNBC. Collectively, these findings provided a target map for comprehensive understanding of the anti-TNBC activity of WA, and present WA-targetable proteins as new avenues for pharmacological intervention in TNBC. We anticipate that this platform will be applicable for the unbiased profiling of the targets of WA in various other disease-related cell models, as well as for other bioactive electrophilic natural products in different pathophysiological systems.
Collapse
Affiliation(s)
- Hui-Jun Nie
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ying-Jie Fu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Shang Long
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia-Yu Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wen-Si Zhao
- School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lin-Hui Zhai
- School of Medicine, Tongji University, Shanghai, 200433, China
| | - Yin-Long Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Min-Jia Tan
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Hu
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Xiao-Hua Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
14
|
Giannone G, Lombardi P, Filetti M, Paparo J, Rognone C, Stefanizzi S, Valsecchi AA, Zumstein L, McNeish IA, Pinato DJ, Gennari A, Daniele G, Di Maio M. Integration of translational research in phase III trials: A systematic review of breast cancer studies in a 5-year period. Breast 2025; 81:104431. [PMID: 40132394 PMCID: PMC11984566 DOI: 10.1016/j.breast.2025.104431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Samples' collection for translational analyses in phase III trials requires a huge effort and there is no evidence on how it translates into new knowledge on tumour biology or optimization of patients' selection. We systematically reviewed phase III trials in breast cancer (BC) to evaluate how frequently a translational project has been integrated into their design and how this integration translated into new translational evidence. METHODS Interventional phase III trials evaluating anticancer drugs in BC published in 11 major journals between 2014 and 2018 were included. RESULTS 89 BC phase III trials were identified, 3 had no sample collection. Among the others, in 36 % the information on sample collection for research purposes was not clear while more than half of the samples had definitive evidence of it. After a median follow-up of 87.9 months, 55.8 % studies published translational data with a mean number of 1.31 (SD 1.7) and 1.07 (SD 1.8), congress abstracts and secondary papers, respectively. There was a higher probability of published translational results for studies with positive outcomes (68.6 % vs 47.1 %), clear evidence of sample collection (72.2 % vs 28.1 %), well-established translational endpoint (73 % vs 42.9 %) and higher impact factor journal (IF) for the clinical publications (64.5 % vs 33.3 %). Secondary translational papers were usually published in lower IF journals with a significant delay from the clinical publication. CONCLUSIONS Although extremely resource-demanding, sample collections for translational analyses in phase III trials are frequently not well defined, and only 50 % produce new translational evidence, which is delayed in time and published in lower IF journals.
Collapse
Affiliation(s)
- G Giannone
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, United Kingdom
| | - P Lombardi
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, United Kingdom; Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - M Filetti
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168, Rome, Italy
| | - J Paparo
- Department of Oncology, University of Turin, Italy
| | - C Rognone
- Department of Oncology, University of Turin, Italy
| | - S Stefanizzi
- Department of Oncology, University of Turin, Italy
| | - A A Valsecchi
- Department of Oncology, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, Ospedale Molinette, Turin, Italy
| | - L Zumstein
- Department of Oncology, University of Turin, Italy
| | - I A McNeish
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, United Kingdom
| | - D J Pinato
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, United Kingdom; Department of Translational Medicine, Università Del Piemonte Orientale, Novara, Italy
| | - A Gennari
- Department of Translational Medicine, Università Del Piemonte Orientale, Novara, Italy
| | - G Daniele
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168, Rome, Italy
| | - M Di Maio
- Department of Oncology, University of Turin, A.O.U. Città della Salute e della Scienza di Torino, Ospedale Molinette, Turin, Italy
| |
Collapse
|
15
|
Zhong N, Zu Z, Lu Y, Sha X, Li Y, Liu Y, Lu S, Luo X, Zhou Y, Tao J, Wu F, Teng Z, Tang Y, Wang S. Mitochondria-targeted manganese-based mesoporous silica nanoplatforms trigger cGAS-STING activation and sensitize anti PD-L1 therapy in triple-negative breast cancer. Acta Biomater 2025; 199:374-386. [PMID: 40294811 DOI: 10.1016/j.actbio.2025.04.040] [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: 01/23/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025]
Abstract
Activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway could effectively initiate antitumor immunity in triple-negative breast cancer. However, current nuclear DNA-mediated activation of STING pathway remains constrained by the tight protection of nuclear membrane and histones, highlighting the need for new strategies to enhance its efficacy. Mitochondrial DNA (mtDNA), in contrast, is more vulnerable to damage. Herein, our nanoplatforms exploited the high glutathione (GSH) environment characteristic of tumors to release abundant Mnb+, which induced mitochondrial dysfunction and the release of endogenous mtDNA. The released mtDNA, in conjunction with Mnb+ itself functioning as a strong cGAS agonist, effectively activated cGAS-STING pathway. Consequently, the cGAS-STING-dependent secretion of type I interferon successively enhanced the maturation of dendritic cells and cross-priming of CD8+ T cells. In a poorly immunogenic 4T1 tumor model, TPP-MMONs efficiently primed systemic antitumor immunity and significantly enhanced the therapeutic efficacy of αPD-L1 therapy, suppressing tumor growth in both localized and metastatic tumor models. These findings provided an innovative and straightforward strategy to enhance TNBC immunogenicity by targeting mitochondrial damage to induce mtDNA-mediated cGAS-STING activation, thereby sensitizing tumors to immune checkpoint inhibitor therapy. STATEMENT OF SIGNIFICANCE: The cGAS-STING pathway is a promising target for overcoming immunoresistance in TNBC. However, current nuclear DNA-based activation strategies are limited by the tight protection of nuclear membrane and histones. Herein, we reported novel manganese-rich, mitochondria-targeting nanoplatforms (TPP-MMONs), which can release abundant Mn²⁺ and significantly induce mitochondrial dysfunction, leading to the release of mtDNA. As a result, the nanoplatforms can effectively stimulate the cGAS-STING pathway, thereby enhancing immune responses and improving the therapeutic efficacy of αPD-L1 therapy, offering new insights into TNBC treatments.
Collapse
Affiliation(s)
- Nan Zhong
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ziyue Zu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yishi Lu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xuan Sha
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Shangyu Lu
- Department of Interventional Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xi Luo
- Department of Interventional Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yan Zhou
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jun Tao
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Feiyun Wu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yuxia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
16
|
Modi S, Zhang S, Byng D, Hunter S, Strübing A, Xiong Y, Dunton K, Mbanya Z, Jacot W. Treatment patterns and outcomes in HER2-low metastatic breast cancer patients previously treated with chemotherapy: a US real-world cohort study. Breast Cancer Res Treat 2025; 211:351-362. [PMID: 40131662 PMCID: PMC12006274 DOI: 10.1007/s10549-025-07649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/10/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE Real-world outcomes are poorly understood for patients with human epidermal growth factor receptor 2 (HER2)-low (immunohistochemistry 1+ or 2+ with negative in situ hybridization) metastatic breast cancer (mBC). METHODS Using for the first time a nationwide electronic health record-derived de-identified database, we assessed demographics, treatment patterns, and outcomes of patients with HER2-low mBC who previously received one line of chemotherapy in the metastatic setting. The post-chemotherapy line was termed the index line of therapy (LOT). RESULTS 3765 patients [hormone receptor (HR)-positive: 78.8%, HR-negative: 21.0%] met the inclusion criteria (1 January 2011-30 April 2023). 61.7% of HR-positive patients received endocrine therapy prior to the index LOT. The largest patient percentage received single-agent chemotherapy at the index and subsequent two LOTs. For the overall cohort, the median real-world time to treatment discontinuation/death was 4.1 months (95% CI: 3.9-4.2) and the median real-world time to next treatment/death was 5.1 months (95% CI: 4.8-5.3) from the index LOT. Median real-world overall survival (all patients) was 15.8 months (95% confidence interval: 15.2-16.5, median follow-up = 54.5 months) from the index LOT. CONCLUSION These data highlight the unmet clinical needs of patients with HER2-low mBC by characterizing the treatment patterns and poor outcomes in this population on the current standard of care.
Collapse
Affiliation(s)
- Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | - Yan Xiong
- Daiichi Sankyo, Inc, Basking Ridge, NJ, USA
| | | | | | - William Jacot
- Institut du Cancer de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| |
Collapse
|
17
|
Gao B, Schäfers C, Kuan SL, Weil T. A Supramolecular, Triple Negative Breast Cancer-Targeting Avidin-Photosensitizer. Macromol Biosci 2025; 25:e2400610. [PMID: 40129407 PMCID: PMC12169506 DOI: 10.1002/mabi.202400610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/25/2025] [Indexed: 03/26/2025]
Abstract
The potential of photodynamic therapy (PDT) in combination with chemotherapy to improve treatment outcomes for triple-negative breast cancer (TNBC), for which no targeted therapy is available, is the subject of considerable investigation. In PDT, photosensitizers (PSs) are frequently administered directly but do not selectively target cancer cells. To address the delivery of a PS to TNBC and enhance cellular uptake, the Ru-NH2-modified avidin bioconjugate (RuAvi) via Tyr-specific modification using the Mannich reaction is prepared. The RuAvi is further assembled with the cinnamoyl peptide-F(D)LF(D)LFK-NH2 (FK), which binds to formyl peptide receptor 1, overexpressed in TNBC. Notably, the modified Avi still possesses the ability to efficiently bind biotin for the assembly of up to four copies of the FK peptides. The resultant FK4-RuAvi exhibited an IC50 value of 0.36 ± 0.08 µM, which is ≈3.5-fold lower than that of RuAvi (1.25 ± 0.09 µM), upon irradiation in the triple-negative MDA-MB-231 breast cancer cells. FK4-RuAvi also shows efficient uptake in MDA-MB-231 tumor spheroids and exhibited significant toxicity after irradiation compared to the control RuAvi. The presented strategy has the potential to improve the efficacy of targeted PDT to meet the high demand for targeted therapies to treat TNBC, such as targeted adjuvant treatment after breast cancer surgery.
Collapse
Affiliation(s)
- Bingjie Gao
- Department of Synthesis of MacromoleculesMax Planck Institute for Polymer Research55128MainzGermany
| | - Charlotte Schäfers
- Department of Synthesis of MacromoleculesMax Planck Institute for Polymer Research55128MainzGermany
- Johannes Gutenberg UniversityDuesbergweg 10–1455128MainzGermany
| | - Seah Ling Kuan
- Department of Synthesis of MacromoleculesMax Planck Institute for Polymer Research55128MainzGermany
| | - Tanja Weil
- Department of Synthesis of MacromoleculesMax Planck Institute for Polymer Research55128MainzGermany
| |
Collapse
|
18
|
Zhou L, Zheng C, Ding S, Wang Z, Yang Y, Wang Y, He G, Fu S, Deng X. Lovastatin Targets the USP14-Survivin Axis to Suppress Triple-Negative Breast Cancer via Ubiquitin-Mediated Proteasomal Degradation. Cells 2025; 14:816. [PMID: 40497992 PMCID: PMC12154129 DOI: 10.3390/cells14110816] [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: 04/18/2025] [Revised: 05/26/2025] [Accepted: 05/27/2025] [Indexed: 06/19/2025] Open
Abstract
Triple-negative breast cancer (TNBC), characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor type 2 (HER2) expression, represents a therapeutic challenge due to its aggressive nature and limited treatment options. Here, we identified the cholesterol-lowering drug lovastatin (LV) as a potent apoptosis-inducing agent in TNBC. Mechanistically, LV disrupts the interaction between the deubiquitinating enzyme USP14 and Survivin, a key anti-apoptotic protein, enhancing polyubiquitination and the proteasomal degradation of Survivin. The overexpression of USP14 was found to stabilize Survivin and rescue LV-induced apoptosis and tumor suppression in vitro and in vivo, whereas USP14 silencing or inhibition with IU1 (a USP14-specific inhibitor) enhanced Survivin turnover and synergized with LV to suppress colony formation in TNBC cells. Clinical relevance was demonstrated through bioinformatic analysis and immunohistochemistry, revealing that elevated Survivin expression in TNBC tissues correlated with poor prognosis and is significantly upregulated in TNBC versus non-TNBC tissues. Our findings identify the USP14-Survivin axis as a potential therapeutic target and highlight LV as a promising candidate for TNBC treatment.
Collapse
Affiliation(s)
- Li Zhou
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chanjuan Zheng
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| | - Siyu Ding
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| | - Zhiyu Wang
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| | - Yiyuan Yang
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| | - Yian Wang
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| | - Guangchun He
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| | - Shujun Fu
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| | - Xiyun Deng
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha 410013, China; (L.Z.); (C.Z.); (S.D.); (Z.W.); (Y.Y.); (Y.W.); (G.H.); (S.F.)
| |
Collapse
|
19
|
Yang X, Xu C, Zeng Y, Wang C, Gao Y, Ding J, Chen S, Pan Y, Zhang X, Mao Z, Shi S. Pyroptosis-Inducing Platinum(IV) Prodrugs via GSDME Pathway for Chemoimmunotherapy and Metastasis Inhibition in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05567. [PMID: 40432601 DOI: 10.1002/advs.202505567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Pyroptosis has attracted significant attention for its role in cancer chemotherapy and immunotherapy. However, few drugs have been reported to induce pyroptosis via the Caspase-3/gasdermin E (GSDME) pathway. Herein, three novel PtIV prodrugs, MRP, DRP, and HRP are rationally designed by conjugating DNA methyltransferase (DNMT) inhibitor (RG108) and/or histone deacetylase (HDAC) inhibitor (PhB) to the PtIV center. These prodrugs can be easily reduced to cisplatin (CDDP) due to the high glutathione (GSH) levels in tumors, liberating the coordinated ligands. Released RG108 reactivates the GSDME gene and reduces pyroptosis in low GSDME-expressing tumor cells. Meanwhile, PhB-induced chromatin loosening enhances CDDP-DNA binding, which not only increases Caspase-3 expression, but also upregulates GSDME. HRP demonstrates superior ability to suppress tumor growth and metastasis while reducing systemic toxicity compared with CDDP. By reactivating GSDME and loosening chromatin, HRP effectively boosts tumor cell pyroptosis and exhibits the most pronounced anticancer performance. These findings highlight HRP's potential as a therapeutic agent for triple-negative breast cancer (TNBC) and offer innovative strategies for combining chemotherapy with immunotherapy. To the best of current knowledge, this is the first report of platinum complexes inducing pyroptosis via the Caspase-3/GSDME pathway in low GSDME-expressing tumor cells.
Collapse
Affiliation(s)
- Xinda Yang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Chuansheng Xu
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Youliang Zeng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Chunhui Wang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yan Gao
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Jie Ding
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Sirui Chen
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yuheng Pan
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Xin Zhang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Zongwan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Shuo Shi
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
20
|
Li S, Jin X, Zhang Y, Huang J, Wang H, Meng H, Li J, Zhu L. Concurrent induction of pyroptosis and immunogenic cell death by capsaicin/graphene nanocomplex for enhanced breast cancer immunotherapy. J Nanobiotechnology 2025; 23:386. [PMID: 40426230 PMCID: PMC12117793 DOI: 10.1186/s12951-025-03439-2] [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: 01/25/2025] [Accepted: 05/04/2025] [Indexed: 05/29/2025] Open
Abstract
Inducing immunogenic cell death (ICD) has emerged as a promising strategy for targeting immunologically "cold" tumors. However, most current therapies focus on a single mechanism, limiting their efficacy. In this study, we propose a nano-enabled approach that synergistically activates two complementary immunogenic killing mechanisms: pyroptosis, which elicits a potent inflammatory response, and ICD, characterized by the presentation of 'eat-me' signals and tumor antigens to the immune system. Capsaicin, a naturally occurring compound, was employed to induce pyroptosis via ROS-mediated gasdermin E (GSDME) cleavage, resulting in cell membrane blebbing and subsequent cell death. To simultaneously trigger ICD, we incorporated 2D graphene oxide (GO) engineered with optimized physicochemical properties to induce robust ICD under near-infrared irradiation. Our in vitro and in vivo experiments demonstrated that the combined treatment of capsaicin and GO not only enhanced cancer cell killing but also promoted immune cell infiltration and potentiated anti-tumor immunity, leading to significant tumor suppression. Moreover, the dual-trigger mechanism of pyroptosis and ICD yielded superior anti-tumor efficacy compared to single-modality treatments while maintaining a favorable biosafety profile. These findings highlight the potential of a synergistic nano-enabled strategy for improving cancer immunotherapy.
Collapse
Affiliation(s)
- Silu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, People's Republic of China
| | - Xin Jin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, People's Republic of China
| | - Yumo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, People's Republic of China
| | - Jidan Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, People's Republic of China
| | - Haiqiang Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, People's Republic of China
| | - Huan Meng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, People's Republic of China.
| | - Jiulong Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, People's Republic of China.
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
21
|
Hu J, Zhang KM, Wang X. Identification of circulating tumor cells marker genes as prognostic signature in triple-negative breast cancer. Discov Oncol 2025; 16:915. [PMID: 40413298 PMCID: PMC12103409 DOI: 10.1007/s12672-025-02604-1] [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: 01/08/2025] [Accepted: 05/06/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Breast cancer represents a significant contributor to cancer-related mortality among women worldwide, with triple-negative breast cancer (TNBC) often exhibiting more aggressive clinical features and a heightened lethality rate. The emergence of malignant progression, along with issues of drug resistance, poses substantial challenges in the clinical management of this disease. METHODS The analysis of gene expression profiles at the single-cell level was conducted on circulating tumor cells (CTCs) obtained from TNBC patients, with the objective of identifying specific marker genes associated with CTCs. The TCGA database served as the training cohort for the development of a prognostic CTCs signature model, while the METABRIC dataset was utilized as the validation cohort to assess the robustness of the CTCs signature model. Furthermore, we investigated the differences in prognosis, immune scores, tumor mutational burden, and responses to immunotherapy and chemotherapy across various risk groups established based on the CTCs signature model. Colony formation and transwell assays were conducted to assess the influence of CTCs signature genes on cellular proliferation and invasive capabilities. RESULTS Seven marker genes associated with CTCs (BLOC1S3, FOXD2, GZMB, KCNJ13, NTRK3, SOAT2, and ZNF589) were identified and incorporated into a CTCs signature model. The risk score derived from this model stratified TNBC patients into high-risk and low-risk groups. Notably, the overall survival (OS) rate for the low-risk group was significantly higher than that of the high-risk group. Furthermore, the low-risk cohort exhibited more favorable prognostic outcomes and demonstrated heightened sensitivity to both immunotherapy and chemotherapy. Finally, knockdown experiments conducted in TNBC cell lines demonstrated that CTCs signature genes play a crucial role in the regulation of cellular proliferation and invasion. CONCLUSION The CTCs signature model offers novel insights into the prognostic significance of CTC marker genes in TNBC. This understanding may serve as a valuable reference for predicting responses to immunotherapy and chemotherapy, as well as for revealing the molecular mechanisms and therapeutic targets of TNBC.
Collapse
Affiliation(s)
- Jia Hu
- Department of Breast Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, SunYat-sen University Cancer Center, Guangzhou, 510060, China
| | - Kai-Ming Zhang
- Department of Breast Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, SunYat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xi Wang
- Department of Breast Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, SunYat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
22
|
Ahmad B, Özgör E, Kavaz D, Shehu A. Synthesis of carob honey loaded chitosan nanoparticles and determination of its antimicrobial potential and cytotoxic effect on breast cancer cell line. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-21. [PMID: 40411780 DOI: 10.1080/09205063.2025.2505702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 05/08/2025] [Indexed: 05/26/2025]
Abstract
Embedding natural products into chitosan nanoparticles (CNP) is an effective way to produce a novel combination with better antimicrobial and anticancer activities. Therefore, this study aims to incorporate carob honey (CH) into CNP, determine its potential antimicrobial along with antiproliferative activities, by well diffusion and MTT cell viability assays, respectively. Successful loading of CH in CNP was confirmed after due characterization. The nanoparticles, synthesized by ionic gelation method, produced a small (101.3 ± 4.13 nm), stable (+27.27 ± 0.95 mV), and monodispersed (0.2265 ± 0.0027) CH-loaded CNP (CHCNP). The best antibacterial activity occurred in Klebsiella pneumoniae (K. pneumoniae) (23 ± 0 mm to 16 ± 1.7 mm) followed by Escherichia coli (E. coli) (18 ± 2.0 mm to 10 ± 1 mm). Meanwhile, Aspergillus niger (A. niger) and Aspergillus flavus (A. flavus) were evenly inhibited with inhibition zones in the range of 15 ± 3 mm to 7 ± 0.8 mm and 15 ± 5 mm to 9 ± 1.4 mm, respectively. CHCNP showed a remarkable cytotoxic effect on MDA-MB-231 according to concentration and time, with IC50 of 25 ± 5 to 18 ± 2.6 μg/mL within 24-72 h. These findings demonstrated the feasibility of loading CH in CNP to form a nanoformulation that could potentially serve as a target-specific therapeutic agent in the treatments of microbial infections and breast cancer. However, there is a need for further research on the safety, dosage optimization, in vivo studies and mechanisms of action of the nanoparticles.
Collapse
Affiliation(s)
- Bashir Ahmad
- Department of Bioengineering, Faculty of Engineering, Cyprus International University, Nicosia, Turkey
- Cyprus Bee and Bee Products Research Centre, Cyprus International University, Nicosia, Turkey
- Department of Biochemistry, Federal University Dutse, Jigawa State, Nigeria
| | - Erkay Özgör
- Cyprus Bee and Bee Products Research Centre, Cyprus International University, Nicosia, Turkey
| | - Doga Kavaz
- Department of Bioengineering, Faculty of Engineering, Cyprus International University, Nicosia, Turkey
| | - Ahmad Shehu
- Department of Bioengineering, Faculty of Engineering, Cyprus International University, Nicosia, Turkey
| |
Collapse
|
23
|
Zhang H, Gong L, Yu L, Xian C, Ma Z, Wang X, Xia R. Emerging roles of non-coding RNA derived from extracellular vesicles in regulating PD-1/PD-L1 pathway: insights into cancer immunotherapy and clinical applications. Cancer Cell Int 2025; 25:188. [PMID: 40410719 PMCID: PMC12103061 DOI: 10.1186/s12935-025-03809-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
Numerous studies have demonstrated that extracellular vesicles (EVs) carry a variety of noncoding RNAs (ncRNAs), which can be taken up by neighboring cells or transported to distant sites via bodily fluids, thereby facilitating intercellular communication and regulating multiple cellular functions. Within the tumor microenvironment, EV-ncRNA, on the one hand, regulate the expression of PD-L1, thereby influencing tumor immune evasion, promoting tumor cell proliferation, and enhancing tumor growth, invasion, and metastasis in vivo. On the other hand, these specific EV-ncRNAs can also modulate the functions of immune cells (such as CD8 + T cells, macrophages, and NK cells) through various molecular mechanisms, inducing an immunosuppressive microenvironment and promoting resistance to anti-PD-1 therapy. Therefore, delving into the molecular mechanisms underlying EV-ncRNA regulation of immune checkpoints presents compelling therapeutic prospects for strategies that selectively target EV-ncRNAs. In this review, we elaborate on the cutting-edge research progress related to EV-ncRNAs in the context of cancer and dissect their pivotal roles in the PD-1/PD-L1 immune checkpoint pathway. We also highlight the promising clinical applications of EV-ncRNAs in anti-PD-1/PD-L1 immunotherapy, bridging basic research with practical clinical applications.
Collapse
Affiliation(s)
- Haixia Zhang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Lianfeng Gong
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Li Yu
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
- Department of Urology, General Hospital of The Yangtze River Shipping, Wuhan, 430010, China
| | - Chenge Xian
- Naidong District People's Hospital, Shannan, 856004, Tibet Autonomous Region, China
| | - Zhaowu Ma
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| | - Xianwang Wang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
- Shannan Maternal and Child Health Hospital, Shannan, 856099, Tibet Autonomous Region, China.
| | - Ruohan Xia
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| |
Collapse
|
24
|
Ran R, Chen X, Yang J, Xu B. Immunotherapy in breast cancer: current landscape and emerging trends. Exp Hematol Oncol 2025; 14:77. [PMID: 40405250 PMCID: PMC12096519 DOI: 10.1186/s40164-025-00667-y] [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: 03/04/2025] [Accepted: 05/08/2025] [Indexed: 05/24/2025] Open
Abstract
Breast cancer remains one of the most prevalent malignancies worldwide, underscoring an urgent need for innovative therapeutic strategies. Immunotherapy has emerged as a transformative frontier in this context. In triple-negative breast cancer (TNBC), the combination of immunotherapy based on PD-1/PD-L1 immune checkpoint inhibitors (ICIs) with chemotherapy has proven efficacious in both early and advanced clinical trials. These encouraging results have led to the approval of ICIs for TNBC, opening up new therapeutic avenues for challenging-to-treat patient populations. Furthermore, a multitude of ongoing trials are actively investigating the efficacy of immunotherapy-based combinations, including ICIs in conjunction with chemotherapy, targeted therapy and radiation therapy, as well as other novel strategies such as bispecific antibodies, CAR-T cells and cancer vaccines across all breast cancer subtypes, including HR-positive/HER2-negative and HER2-positive disease. This review provides a comprehensive overview of current immunotherapeutic approaches in breast cancer, highlighting pivotal findings from recent clinical trials and the potential impact of these advancements on patient outcomes.
Collapse
Affiliation(s)
- Ran Ran
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Chen
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Yang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Binghe Xu
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
25
|
Ren TJ, Zhang YZ, Zhang Q, Tan M, Gu J, Tong Y, Wang Y, Yang C, Xu ZR. Accurate Cancer Diagnosis and Treatment Monitoring through Multiplexed Profiling of Protein Markers on Small Extracellular Vesicles. ACS NANO 2025; 19:18630-18643. [PMID: 40340378 DOI: 10.1021/acsnano.5c02864] [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: 05/10/2025]
Abstract
The detection of small extracellular vesicles (sEVs) is currently a pivotal liquid biopsy approach for noninvasive cancer diagnosis. However, the lack of adequate specificity and sensitivity, as well as labor-intensive purification and analysis procedures, present challenges in isolating and profiling sEVs. Here, we present a protein-specific enzymatic optical reporter deposition-based liquid biopsy assay for the rapid and efficient capture and ultrasensitive detection of sEVs using a minimal volume of initial biofluids (10 μL). Biotin aptamers were employed to label sEV proteins for peroxidase conjugation, catalyzing the conversion of fluorescein tyramine into highly reactive free radicals. Efficient signal conversion was achieved by depositing nanoheterolayers composed of covalent tyraminated complexes onto sEV surfaces. The present method offers a detection limit of 6.4 × 103 particles mL-1 with a linear range of 104-1010 particles mL-1 for sEVs. Two machine learning algorithms, principal coordinates analysis and principal component analysis, were subsequently applied for dimensionality reduction. In a clinical cohort of 84 patients, including 6 cancer types and noncancer cases, the assay achieved an overall accuracy of 100% (95% confidence interval) in distinguishing between cancer and noncancer controls and 96% in classifying cancer types. As drugs are frequently administered to patients to modulate the activity of tumor cells, we investigated the efficacy of this strategy in treatment monitoring, achieving an overall accuracy of 100%. This strategy demonstrates a cost-effective, rapid, and low sample volume consumption approach that holds significant potential for precise cancer diagnosis and auxiliary assessment of drug response in clinical settings.
Collapse
Affiliation(s)
- Ting-Ju Ren
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Ying-Zhi Zhang
- National Clinical Research Center for Laboratory Medicine, Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110819, China
| | - Qi Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Meilun Tan
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Jiahui Gu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Yuxiao Tong
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Yue Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Chunguang Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| | - Zhang-Run Xu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province 110819, China
| |
Collapse
|
26
|
Han JY, Rhee WJ, Shin JS. Cytoplasmic HMGB1 promotes the activation of JAK2-STAT3 signaling and PD-L1 expression in breast cancer. Mol Med 2025; 31:197. [PMID: 40389855 PMCID: PMC12090602 DOI: 10.1186/s10020-025-01235-0] [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: 09/30/2024] [Accepted: 04/28/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) plays various roles depending on its subcellular localization. Extracellular HMGB1 interacts with receptors, such as toll-like receptor 4 and receptor for advanced glycation end products (RAGE), promoting cell proliferation, survival, and migration in cancer cells. It also increases the expression of programmed death-ligand 1 (PD-L1) in cancer cells by binding to RAGE. However, the effect of intracellular HMGB1 on the regulation of immune checkpoints such as PD-L1 has not been well characterized. In this study, we aimed to investigate the effects of intracellular HMGB1 on PD-L1 expression in breast cancer cells. METHODS Human and mouse triple-negative breast cancer cells, MDA-MB-231 and 4T1, along with HMGB1-deficient mouse embryonic fibroblast cells, were cultured. HMGB1 overexpression was achieved using a Myc-tagged plasmid, while siHMGB1 constructs were used for gene silencing. Quantitative reverse-transcriptase PCR and western blot analysis were performed to assess gene and protein expressions. Confocal imaging, immunoprecipitation, and proximity ligation assays were used to investigate HMGB1 localization and Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) interactions. In vivo experiments were performed using tumor-bearing mice treated with STAT3 and HMGB1 inhibitors. Statistical analyses were performed using Student's t-tests, one-way analysis of variance, Pearson's correlation, and Kaplan-Meier survival analysis, with significance set at p < 0.05. RESULTS In breast cancer cells, HMGB1 translocation from the nucleus to the cytoplasm increased the JAK2-STAT3 interaction and induced STAT3 phosphorylation, leading to increased STAT3 target signaling, including the epithelial-mesenchymal transition (EMT) phenotype and PD-L1 expression. Inhibition of nucleo-cytoplasmic translocation of HMGB1 decreased STAT3 phosphorylation and PD-L1 expression. Furthermore, HMGB1 enhanced breast cancer cell migration, invasion, and EMT, contributing to tumor growth in an in vivo mouse model that were mitigated by the HMGB1-targeted approach. CONCLUSIONS These findings underscore the critical role of intracellular HMGB1 in modulating PD-L1 expression via the JAK2-STAT3 signaling pathways in breast cancer and suggest that targeting HMGB1 translocation is a promising strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Ju-Young Han
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Woo Joong Rhee
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea.
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
27
|
Zhu B, Wan H, Ling Z, Jiang H, Pei J. Machine learning and single-cell analysis uncover distinctive characteristics of CD300LG within the TNBC immune microenvironment: experimental validation. Clin Exp Med 2025; 25:167. [PMID: 40382513 PMCID: PMC12085369 DOI: 10.1007/s10238-025-01690-3] [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: 03/23/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
Investigating the essential function of CD300LG within the tumor microenvironment in triple-negative breast cancer (TNBC). Transcriptomic and single-cell data from TNBC were systematically collected and integrated. Four machine learning algorithms were employed to identify distinct target genes in TNBC patients. Specifically, CIBERSORT and ssGSEA algorithms were utilized to elucidate immune infiltration patterns, whereas TIDE and TCGA algorithms predicted immune-related outcomes. Moreover, single-cell sequencing data were analyzed to investigate the function of CD300LG-positive cells within the tumor microenvironment. Finally, immunofluorescence staining confirmed the significance of CD300LG in tumor phenotyping. After machine learning screening and independent dataset validation, CD300LG was identified as a unique prognostic biomarker for triple-negative breast cancer. Enrichment analysis revealed that CD300LG expression is strongly linked to immune infiltration and inflammation-related pathways, especially those associated with the cell cycle. The presence of CD8+ T cells and M1-type macrophages was elevated in the CD300LG higher group, whereas the abundance of M2-type macrophage infiltration showed a significant decrease. Immunotherapy prediction models indicated that individuals with low CD300LG expression exhibited better responses to PD-1 therapy. Additionally, single-cell RNA sequencing and immunofluorescence analyses uncovered a robust association between CD300LG and genes involved in tumor invasion. CD300LG plays a pivotal role in the tumor microenvironment of TNBC and represents a promising therapeutic target.
Collapse
Affiliation(s)
- Baoxi Zhu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Thyroid and Breast Surgery, Anhui No.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Hong Wan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zichen Ling
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Han Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Pei
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
28
|
Gao X, Wang T, Liu C, Li Y, Zhang W, Zhang M, Yao Y, Gao C, Liu R, Sun C. The integrated single-cell analysis interpret the lactate metabolism-driven immune suppression in triple-negative breast cancer. Discov Oncol 2025; 16:784. [PMID: 40377730 PMCID: PMC12084458 DOI: 10.1007/s12672-025-02605-0] [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: 01/28/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Individuals with triple-negative breast cancer (TNBC) exhibit elevated lactate levels, which offers a valuable lead for investigating the molecular mechanisms underlying the tumor microenvironment (TME) and identifying more efficacious treatments. METHODS TNBC samples were classified based on lactate-associated genes. A single-cell transcriptomic approach was employed to examine functional differences across cells with varying lactate metabolism. Immunohistochemistry was used to explore the relationship between lactate metabolism and the CXCL12/CXCR4 signaling axis. In addition, utilizing machine learning techniques, we constructed a prognostic model based on lactic acid phenotype genes. RESULTS Lactate-associated gene-based stratification revealed increased immune cell infiltration and immune checkpoint expression in Lactate Cluster 1. Elevated lactate metabolism scores were observed in both cancer-associated fibroblasts (CAFs) and malignant cells. CAFs with high lactate metabolism exhibited immune suppression through the CXCL12/CXCR4 axis. Immunohistochemistry confirmed elevated LDHA, LDHB, CXCL12, and CXCR4 levels in the high lactate group. CONCLUSION This study elucidates the complex interplay between lactate and immune cells in TNBC and highlights the CXCL12/CXCR4 axis as a key pathway through which lactate mediates immune suppression, offering new insights into metabolic regulation within the TME. Furthermore, we developed a prognostic model based on lactate metabolism phenotype genes to predict the prognosis of TNBC patients and guide immunotherapy.
Collapse
Affiliation(s)
- Xinhai Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Tianhua Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Wenfeng Zhang
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China
| | - Minpu Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, 999078, Macao, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, Shandong, China
| | - Chundi Gao
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China
| | - Ruijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, Shandong, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, 261000, Weifang, Shandong, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, Shandong, China.
| |
Collapse
|
29
|
Lin SP, Huang FY, Wu RH, Xie WJ, Chen MH, Dai SZ, Xu WT, Zheng WP, Tan GH. Toxicarioside H induces ferroptosis in triple-negative breast cancer cells through Nrf2/HO-1 pathway. Discov Oncol 2025; 16:772. [PMID: 40372576 PMCID: PMC12081807 DOI: 10.1007/s12672-025-02333-5] [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: 10/13/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Recent studies have identified novel cardiac glycosides from natural sources with potential anti-tumor properties. Toxicarioside H (ToxH) is a novel cardiac glycoside isolated by our collaborative research team. However, its ability to induce ferroptosis in triple-negative breast cancer (TNBC) cells has not been investigated. Therefore, this study evaluates whether ToxH has the capability of inducing ferroptosis and elucidates the underlying molecular mechanisms. Treatment with ToxH led to dose- and time-dependent growth inhibition in BT-549 and MDA-MB-468 cells. Flow cytometry analysis and lactate dehydrogenase assay revealed that ToxH induced various forms of cell death in both BT-549 and MDA-MB-468 cells. Examination through transmission electron microscopy, along with flow cytometry analysis of 7-AAD-stained dead cells and ferroptosis markers BODIPY-C11 and Fe2+ ions, identified various forms of cell death induced by ToxH, including apoptosis, autophagy, apoptotic necrosis, and ferroptosis. Co-treatment with the ferroptosis inhibitor Fer-1 significantly reduced ToxH-induced cell death, indicating that ToxH primarily inhibits TNBC cell growth by inducing ferroptosis. Further investigation into the molecular mechanisms revealed upregulation of Nrf2 and HO-1 expression by ToxH. Effective inhibition of ToxH-induced ferroptosis was achieved through shRNA-mediated knockdown of HO-1 expression. Animal experiments demonstrated that ToxH treatment markedly suppressed tumor growth compared to the control group, while co-administration of Fer-1 led to an increase in tumor growth. These findings suggest that ToxH suppresses TNBC cell growth by modulating the Nrf2/HO-1 signaling pathway to induce ferroptosis. ToxH presents itself as a promising cardiac glycoside compound for TNBC treatment, warranting further translational research.
Collapse
Affiliation(s)
- Sheng-Ping Lin
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China
| | - Feng-Ying Huang
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China
| | - Ri-Hong Wu
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China
| | - Wei-Jing Xie
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China
| | - Ming-Hui Chen
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China
| | - Shu-Zhen Dai
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China
| | - Wen-Tian Xu
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China.
| | - Wu-Ping Zheng
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China.
| | - Guang-Hong Tan
- NHC Key Laboratory of Tropical Disease Control & The Second Affiliated Hospital, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou City, 571199, Hainan Province, China.
| |
Collapse
|
30
|
Song EJ, Kim WS, Han Y, Lee C, Moon EJ, Kim HJ, Kang NS. Discovery of a transforming growth factor-β1 inhibitory peptide, Charis 1000 to enhance the therapeutic efficacy of paclitaxel in triple-negative breast cancer. Int J Biol Macromol 2025; 314:144216. [PMID: 40379179 DOI: 10.1016/j.ijbiomac.2025.144216] [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: 01/12/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and invasive subtype of breast cancer for which chemotherapy, such as paclitaxel (PTX), remains a primary treatment option. However, resistance to chemotherapy poses a significant challenge, necessitating the development of novel therapeutic strategies. This study aimed to address PTX resistance in TNBC by developing a peptide drug, Charis 1000 (C1K), designed to target transforming growth factor beta (TGF-β) signaling. C1K was synthesized using standard solid-phase peptide synthesis and optimized for enhanced stability. Molecular docking predicted the binding interactions between C1K and TGF-β1, and surface plasmon resonance (SPR) confirmed a moderate binding affinity. The therapeutic potential of C1K was evaluated in TNBC cell lines (4T1, MDA-MB-231, and PTX-resistant MDA-MB-231) and in vivo using a syngeneic 4T1 mouse model. Functional assays demonstrated that C1K inhibited TGF-β-mediated signaling, reduced autophagy, a key mechanism underlying PTX resistance, and significantly enhanced PTX-induced apoptosis. In vivo studies further revealed synergistic effects of C1K and PTX, resulting in enhanced apoptosis in both sensitive and PTX-resistant TNBC cells. These findings suggest that C1K effectively targets TGF-β to inhibit autophagy and potentiate the apoptotic effects of PTX, as a promising combinatorial therapeutic agent for improving treatment outcomes in TNBC patients.
Collapse
Affiliation(s)
- Eun Ju Song
- Graduate School of New Drug Discovery and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Won-Sam Kim
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Yunhee Han
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Cheolmin Lee
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea; Graduate School of Chemistry, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896 Republic of Korea
| | - Eun-Joung Moon
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Hae-Jin Kim
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea.
| | - Nam Sook Kang
- Graduate School of New Drug Discovery and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
31
|
Guo W, Wang M, Chen X, Wang M, Meng Y. A light-activated Fe 2+ release nanosystem for enhanced chemodynamic/chemo therapy via cascade amplification of ROS generation. Biomater Sci 2025; 13:2713-2727. [PMID: 40192435 DOI: 10.1039/d4bm01425a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Ferrous iron (Fe2+)-based chemodynamic therapy (CDT) shows great potential for improving chemotherapeutic efficacy and reducing side effects. However, spontaneous oxidation and biological matrixes can influence the catalytic reactive oxygen species (ROS) generation of Fe2+, thereby limiting the efficacy of CDT. Herein, we reported a simple and convenient method to construct hyaluronic acid (HA)-stabilized iron/zinc oxide nanoparticles (IZ@H NPs), which showed intrinsic peroxidase (POD)-like activity and excellent light-activated Fe2+ release performance. Moreover, we demonstrate that catalytic ROS generation follows a cascade amplification manner due to the light-activated release of Fe2+ from IZ@H NPs, leading to formation of iron-DNA complexes (IDCs). After loading doxorubicin (DOX), the nanosystem (termed IZD@H NPs) exhibits tumor cell targeting, robust ROS generation and high cytotoxicity, significantly suppressing tumor growth in xenograft mouse models while maintaining good biosafety. This work gives novel insight into amplifying Fe2+-mediated catalytic ROS generation and presents a new strategy for in vivo Fe2+ delivery to enhance chemodynamic/chemotherapy.
Collapse
Affiliation(s)
- Wei Guo
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- College of Pharmacy, Xinjiang Medical University, Xinjiang 830017, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Xinjiang Medical University, Xinjiang 830017, China
| | - Min Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xisha Chen
- Cancer Research Institute, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Mei Wang
- College of Pharmacy, Xinjiang Medical University, Xinjiang 830017, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Xinjiang Medical University, Xinjiang 830017, China
| | - Yingcai Meng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
32
|
Rosenbaum SR, Hughes CJ, Fields KM, Purdy SC, Gustafson AL, Wolin A, Hampton D, Shrivastava NM, Turner N, Danis E, Ebmeier C, Spoelstra N, Richer J, Jedlicka P, Costello JC, Zhao R, Ford HL. EYA3 regulation of NF-κB and CCL2 suppresses cytotoxic NK cells in the premetastatic niche to promote TNBC metastasis. SCIENCE ADVANCES 2025; 11:eadt0504. [PMID: 40333987 PMCID: PMC12057687 DOI: 10.1126/sciadv.adt0504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 04/02/2025] [Indexed: 05/09/2025]
Abstract
Triple-negative breast cancer cells must evade immune surveillance to metastasize to distant sites, yet this process is not well understood. The Eyes absent (EYA) family of proteins, which are crucial for embryonic development, become dysregulated in cancer, where they have been shown to mediate proliferation, migration, and invasion. Our study reveals an unusual mechanism by which EYA3 reduces the presence of cytotoxic natural killer (NK) cells in the premetastatic niche (PMN) to enhance metastasis, independent of its effects on the primary tumor. We find that EYA3 up-regulates nuclear factor κB signaling to enhance CCL2 expression, which, in contrast to previous findings, suppresses cytotoxic NK cell activation in vitro and their infiltration into the PMN in vivo. These findings uncover an unexpected role for CCL2 in inhibiting NK cell responses at the PMN and suggest that targeting EYA3 could be an effective strategy to reactivate antitumor immune responses to inhibit metastasis.
Collapse
Affiliation(s)
- Sheera R. Rosenbaum
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Connor J. Hughes
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Kaiah M. Fields
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Stephen Connor Purdy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Annika L. Gustafson
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Arthur Wolin
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Drake Hampton
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Natasha M. Shrivastava
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Nicholas Turner
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Etienne Danis
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Christopher Ebmeier
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Nicole Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Jennifer Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Paul Jedlicka
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - James C. Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Rui Zhao
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| |
Collapse
|
33
|
Huang HY, Lin LZY, Lu XY, Jiang Y. F-α-DDB-derivative, a novel synthetic of bifendate, plus epirubicin improves antitumor efficacy against triple negative breast cancer without additional cardiotoxicity. Discov Oncol 2025; 16:690. [PMID: 40338489 PMCID: PMC12061825 DOI: 10.1007/s12672-025-02545-9] [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: 01/17/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Triple negative breast cancer (TNBC) refers to a molecular subtype of breast cancers (BC) with high rate of distant metastases and poor prognosis. Epirubicin (EPI) is widely used for the therapy of TNBC but it's limited in clinical use due to cardiotoxicity and chemotherapy resistance. We previously identified F-α-DDB-derivative, a novel synthetic of bifendate, as a potential agent to improve the therapeutic efficacy of TNBC in combination with EPI since F-α-DDB-derivative inhibited MDA-MB-468, but not as much as EPI. In this study, we investigated the antitumor activity of F-α-DDB-derivative in combination with EPI against TNBC in vitro and in vivo and whether co-treatment would induce additional cardiotoxicity. In human TNBC MDA-MB-468 cells, application of F-α-DDB-derivative (11.5, 23.0, 46.0 μg/ml) in combination with EPI (1.5, 3.0, 6.0 μg/ml) exhibited great inhibition on cell viability and cell proliferation. Additionally, F-α-DDB-derivative (5.75, 11.5, 23.0, 46.0 μg/ml) interacted with EPI (0.75, 1.5, 3.0, 6.0 μg/ml) synergistically to induce apoptosis in MDA-MB-468 cells. This suggests that F-α-DDB-derivative may be more sensitive to apoptotic pathways. Furthermore, we revealed that co-administration of EPI and F-α-DDB-derivative (ip, once every other day for 14 days) significantly increased the therapeutic efficacy of EPI (2.0 mg/kg) or F-α-DDB-derivative (20.0 mg/kg) in mice harboring MDA-MB-468 cell xenografts without additional cardiotoxicity compared to that in EPI monotherapy group. These results implicate that co-treatment of EPI and F-α-DDB-derivative may be a potential therapeutic approach for the treatment of TNBC.
Collapse
Affiliation(s)
- Hai-Yi Huang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lisa Zong Yong Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin-Yi Lu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China.
| |
Collapse
|
34
|
Gomes GMDA, Xu M, Syeda AKR, Raudonis R, Almasi S, Vijayan VV, Gujar S, Dong X, Cheng Z, Pulinilkunnil T, El Hiani Y. Targeting TRPML3 inhibits proliferation and invasion, and enhances doxorubicin sensitivity by disrupting lysosomal acidification and mitochondrial function in triple-negative breast cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119979. [PMID: 40348344 DOI: 10.1016/j.bbamcr.2025.119979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 04/22/2025] [Accepted: 04/27/2025] [Indexed: 05/14/2025]
Abstract
TNBC remains the most aggressive and therapy-resistant type of breast cancer, for which efficient targeted therapies have not been developed yet. Here, we identified TRPML3 (ML3) as a potential therapeutic target in TNBC. Our data showed that ML3 is significantly upregulated in TNBC cells compared with nontumorigenic control cells. ML3 knockdown (KD) impairs TNBC cell proliferation by inducing cell cycle arrest and caspase-dependent apoptosis. ML3 KD also inhibits TNBC cell migration and invasion. Mechanistically, ML3 KD reduces lysosomal number and enhances lysosomal acidification, which in turn activates mTORC1, thereby inhibiting autophagy initiation and flux. This disruption negatively impacts mitochondrial function, as evidenced by reduced ATP production, increased ROS and NO production, and mitochondrial fragmentation. Importantly, ML3 KD enhances TNBC cell sensitivity to doxorubicin and paclitaxel. The finding suggests that targeting ML3 disrupts lysosomal and mitochondrial homeostasis and enhance chemosensitivity, presenting ML3 as a potential therapeutic vulnerability in TNBC enhancing chemosensitivity.
Collapse
Affiliation(s)
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada.
| | | | - Renee Raudonis
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | | | - Vishnu Vijay Vijayan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Shashi Gujar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Xianping Dong
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | | | - Yassine El Hiani
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
35
|
Le X, Chen Q, Wen Q, Cao S, Zhang L, Hu L, Hu G, Li Q, Chen Z. Design, synthesis and optimization of Apcin analogues as Cdc20 inhibitors for triple-negative breast cancer therapy. Eur J Med Chem 2025; 289:117434. [PMID: 40020424 DOI: 10.1016/j.ejmech.2025.117434] [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: 12/29/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Cell division cycle 20 homologue (Cdc20) is an essential mitotic regulator whose overexpression is closely associated with tumorigenesis and poor prognosis in triple-negative breast cancer (TNBC). Targeting Cdc20 has therefore emerged as a promising therapeutic avenue for this aggressive malignancy. In the present study, a receptor-based drug design approach was employed to optimize Apcin analogues as Cdc20 inhibitors. Through a two-step strategy-concept validation followed by structural optimization-we identified compound 14c, which demonstrated remarkable Cdc20 binding affinity (KD: 7.65 μM), potent antiproliferative effects against MDA-MB-231 TNBC cells (IC50: 3.28 μM), and a favorable selectivity index (4.22 for MCF-7 non-TNBC cells and 7.27 for MCF 10A normal cells). 14c effectively inhibited Cdc20 activity, induced G2/M phase arrest, promoted DNA damage accumulation, and stabilized key substrates such as Cyclin B1 and Bim, leading to enhanced apoptosis and suppression of tumor cell proliferation and migration. In vivo, 14c significantly inhibited tumor growth in an MDA-MB-231 xenograft model with a 90 % tumor inhibition rate and no observable toxicity. These results highlight the potential of 14c as a potent Cdc20 inhibitor, offering a promising therapeutic approach for TNBC.
Collapse
Affiliation(s)
- Xiangyang Le
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Department of Pharmacy, Yiyang Central Hospital, Yiyang, Hunan, 413000, China
| | - Qingsong Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Qiwan Wen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Shuyang Cao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Lei Zhang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Liqing Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Pharmaceutical Sciences, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| |
Collapse
|
36
|
Zhang X, Zhuang M, Zhang H, Zhu Y, Yang J, Wu X, Yu X, Tao J, Liu X. Melatonin-mediated cGAS-STING signal in senescent macrophages promote TNBC chemotherapy resistance and drive the SASP. J Biol Chem 2025; 301:108438. [PMID: 40127867 DOI: 10.1016/j.jbc.2025.108438] [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: 10/11/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
The build-up of senescent cells in tissues is a key indicator of aging, associated with negative prognosis and therapy resistance. Despite immune dysfunction related to aging, also known as immunosenescence, is recognized as a factor in this process, the exact mechanisms are still unclear. In this study, we reported that melatonin deficiency accelerated macrophage senescence in triple-negative breast cancer, whereas melatonin could defend macrophages against senescence through the Nfatc1-Trim26-cgas-Sting pathway. Mechanistically, melatonin enhanced the nuclear translocation of Nfatc1 and elevated Trim26 transcription levels. Trim26, functioning as an E3 ligase, ubiquitinates cgas, thereby inhibiting the activation of the cgas-Sing pathway and consequently preventing cell senescence. Conversely, melatonin deficiency induced cgas-Sting pathway activation to promote macrophage aging. Our results show that melatonin inhibited macrophage senescence and improved chemotherapy responsiveness, with further enhancement when combined with the cgas inhibitor (G150). Overall, our findings indicated that melatonin protects macrophages from immunosenescence, suggesting its therapeutic potential for enhancing chemotherapy response.
Collapse
Affiliation(s)
- Xiaoqiang Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Minyu Zhuang
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Hongfei Zhang
- Department of Ultrasound in Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yanhui Zhu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Junzhe Yang
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xian Wu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xiafei Yu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China.
| | - Jing Tao
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiaoan Liu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
37
|
Chen H, Hou S, Zhang H, Zhou B, Xi H, Li X, Lufeng Z, Guo Q. RETRACTED: MiR-375 impairs breast cancer cell stemness by targeting the KLF5/G6PD signaling axis. ENVIRONMENTAL TOXICOLOGY 2025; 40:E31-E43. [PMID: 38470012 DOI: 10.1002/tox.24204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/13/2024]
Abstract
Recurrence of breast cancer may be due to the presence of breast cancer stem cells (BCSC). Abnormal tumor cell growth is closely associated with increased reactive oxygen species (ROS) and disruption of redox homeostasis, and BCSCs exhibit low levels of ROS. The detailed mechanism between the low levels of ROS in BCSCs and their maintenance of stemness characteristics has not been reported. A growing number of studies have shown that tumor development is often accompanied by metabolic reprogramming, which is an important hallmark of tumor cells. As the first rate-limiting enzyme of pentose phosphate pathway (PPP), the expression of G6PD is precisely regulated in tumor cells, and there is a certain correlation between PPP and BCSCs. MiR-375 has been shown to inhibit stem cell-like properties in breast cancer, but the exact mechanism is not clear. Here, KLF5, as a transcription factor, was identified to bind to the promoter of G6PD to promote its expression, whereas miR-375 inhibited the expression of KLF5 by binding to the 3'UTR region of KLF5 mRNA and thus reduced the expression of G6PD expression, inhibits PPP to reduce NADPH, and increases ROS levels in breast cancer cells, thereby weakening breast cancer cell stemness. Our study reveals the specific mechanism by which miR-375 targets the KLF5/G6PD signaling axis to diminish the stemness of breast cancer cells, providing a therapeutic strategy against BCSCs.
Collapse
Affiliation(s)
- Haitao Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, People's Republic of China
| | - Hongwei Zhang
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Bing Zhou
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Huifang Xi
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Xiaofang Li
- Department of Anesthesiology, Hepatobiliary Surgery, Neonatology, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Zheng Lufeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
38
|
Liu Y, Hao Y, Lv X, Zhang Y, Chen J, Tian J, Ma X, Zhou Y, Feng L. A tetramethylpyrazine releasing hydrogel can potentiate CAR-T cell therapy against triple negative breast cancer by reprogramming tumor vasculatures. FUNDAMENTAL RESEARCH 2025; 5:1288-1297. [PMID: 40528964 PMCID: PMC12167868 DOI: 10.1016/j.fmre.2023.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/21/2023] [Accepted: 05/14/2023] [Indexed: 06/20/2025] Open
Abstract
Irregular vasculature of solid tumors has proven to be a pivotal factor restricting their response to chimeric antigen receptor-T (CAR-T) cell therapy because it is tightly associated with hypoxia and other biological barriers. Herein, an injectable hydrogel composed of poly (ethylene glycol) dimethacrylate (PEGDMA) and ferrous chloride (FeCl2) responding to endogenous hydrogen peroxides (H2O2) is developed to enable sustained intratumoral release of Chinese herbal extracts tetramethylpyrazine (TMP). TMP is selected due to its potency in activating vascular endothelial growth factor (VEGF) expression and the endothelial nitric oxide synthase/nitric oxide (eNOS/NO) axis inside vascular endothelial cells. Upon being fixed inside tumors with the PEGDMA based hydrogel, TMP can remodel tumor vasculature by simultaneously promoting angiogenesis and dilating tumor vasculature and thus attenuate tumor hypoxia in two murine xenografts bearing human triple negative breast cancer (TNBC). Resultantly, treatment with TMP fixation potentiates the tumor suppression effect of intravenously injected epidermal growth factor receptor expressing CAR-T (HER1-CAR-T) cells toward two TNBC tumor xenografts by promoting their tumor infiltration, survival, and effector function. This study highlights a concise yet effective approach to reinforce the therapeutic potency of CAR-T cells towards targeted solid tumors by simply remodeling tumor vasculature.
Collapse
Affiliation(s)
- Yan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Yu Hao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Xiang Lv
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Yefei Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jiahui Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jia Tian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Cancer Institute, Department of Biochemistry, College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Xinxing Ma
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yehui Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
39
|
Wang X, Wang L, Liu Y. Current Status of Immune Checkpoint Inhibitors and Treatment Responsive Biomarkers for Triple-Negative Breast Cancer. Thorac Cancer 2025; 16:e70072. [PMID: 40324951 PMCID: PMC12052518 DOI: 10.1111/1759-7714.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
Triple-negative breast cancer (TNBC), accounting for about 10%-20% of all breast cancer cases, is characterized by its aggressive nature, high recurrence rates, and poor prognosis. Unlike other breast cancer subtypes, TNBC lacks hormone receptors and specific molecular targets, limiting therapeutic options. In recent years, immune checkpoint inhibitors (ICIs) have shown promise in treating TNBC by targeting immune evasion mechanisms. Despite these advancements, several issues remain unresolved, including low response rates in programmed cell death ligand 1 (PD-L1) negative TNBC subtypes and the challenge of predicting which patients will benefit from ICIs. Consequently, there is growing interest in identifying reliable biomarkers beyond PD-L1 expression. This review synthesizes recent studies to provide a comprehensive perspective on ICI therapy in TNBC, clarifying the status of single-agent ICI therapies and combination strategies, emphasizing the need for further research into biomarkers. These insights provide clues for more personalized and effective treatment approaches, ultimately aiming to improve clinical outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Xinran Wang
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Lingxia Wang
- Value & Implementation, Global Medical & Scientific AffairsMSD ChinaShanghaiChina
| | - Yueping Liu
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
40
|
Tian M, Keshavarz M, Demircali AA, Han B, Yang G. Localized Microrobotic Delivery of Enzyme-Responsive Hydrogel-Immobilized Therapeutics to Suppress Triple-Negative Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408813. [PMID: 39692188 PMCID: PMC12051738 DOI: 10.1002/smll.202408813] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/09/2024] [Indexed: 12/19/2024]
Abstract
Triple-negative breast cancer (TNBC), characterized by its aggressive metastatic propensity and lack of effective targeted therapeutic options, poses a major challenge in oncological management. A proof-of-concept neoadjuvant strategy aimed at inhibiting TNBC tumor growth and mitigating metastasis through a localized delivery of chemotherapeutics is reported in this paper. This approach addresses the limitations in payload capacity and stimuli responsiveness commonly associated with microrobotics in oncology. A hydrogel-based system is developed for the immobilization of chemotherapeutic agents, subsequently encapsulated within magnetically responsive microrobots. This design leverages external magnetic fields to facilitate the precise navigation and localization of the therapeutic agents directly to the tumor site. The efficacy of this approach is demonstrated in an animal model, in which a significant 14-fold reduction in tumor size and suppression of metastasis to critical organs such as the liver and lungs are observed. Crucially, the drug release mechanism is engineered to be responsive to the tumor microenvironment and is regulated by the overexpression of the enzymatic activity of matrix metalloproteinases (MMP2 and MMP9) in TNBC tumors, triggering the degradation of the hydrogel matrix, leading to controlled release of the immobilized therapeutic drug. This ensures that the therapeutic action is localized, reducing systemic toxicity and enhancing treatment efficacy. These findings suggest that this neoadjuvant approach holds promise for broader applications in other cancer types.
Collapse
Affiliation(s)
- Mingzhen Tian
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Meysam Keshavarz
- The Hamlyn Centre, Institute of Global Health InnovationImperial College LondonLondonSouth KensingtonSW7 2AZUK
| | - Ali Anil Demircali
- Department of Metabolism, Digestion, and Reproduction, Faculty of MedicineImperial College LondonLondonSW7 2AZUK
| | - Bing Han
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Guang‐Zhong Yang
- Institute of Medical Robotics, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| |
Collapse
|
41
|
Chen N, Matossian M, Saha P, Rampurwala M, Kamaraju S, Hahn O, Howard FM, Fleming GF, Freeman JQ, Karrison T, Conzen S, Nanda R, Stringer-Reasor EM. A randomized phase II trial of nab-paclitaxel with or without mifepristone for advanced triple-negative breast cancer. Breast Cancer Res Treat 2025; 211:111-119. [PMID: 39928262 PMCID: PMC11952973 DOI: 10.1007/s10549-025-07626-5] [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: 09/03/2024] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
PURPOSE Glucocorticoid receptor (GR) activity may mediate chemoresistance in advanced triple-negative breast cancer (TNBC). Preclinical studies demonstrate that GR antagonism can augment the effect of taxanes in TNBC models. We hypothesized that pretreatment with mifepristone, a potent GR antagonist, would enhance nab-paclitaxel efficacy in advanced TNBC. METHODS This trial was terminated early due to poor accrual. 29 of 64 planned patients were enrolled. Patients were randomized to receive nab-paclitaxel with or without mifepristone; oral mifepristone 300 mg was administered the day prior and day of each dose of nab-paclitaxel. The primary endpoint was progression-free survival (PFS); secondary/exploratory endpoints included response rate and correlation of response with GR expression. RESULTS The addition of mifepristone to nab-paclitaxel did not improve PFS (3.0 m vs 3.0 m, p = 0.687) or overall response rate (23% vs 31.5%) compared to nab-paclitaxel alone. There was a trend towards improved overall survival in the combination group, primarily driven by one long-term responder. Increased rates of grade 3 neutropenia (46% vs 7%) and febrile neutropenia were observed in the combination arm, while other toxicities were similar in both groups. Increased GR expression was not correlated with clinical response in the combination arm. CONCLUSIONS While there were responders to the combination, the study was underpowered to meet the primary endpoint. Higher rates of neutropenia were observed in the combination, but overall it was well tolerated. Preclinical data in TNBC and clinical data in other malignancies support further investigation of GR modulators. Future studies should incorporate biomarkers to select patients who benefit from GR inhibition.
Collapse
Affiliation(s)
- Nan Chen
- Department of Medicine Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Margarite Matossian
- Department of Medicine Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Poornima Saha
- Department of Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Murtuza Rampurwala
- Department of Medicine Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Salaija Kamaraju
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Olwen Hahn
- Department of Medicine Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Frederick M Howard
- Department of Medicine Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Gini F Fleming
- Department of Medicine Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Jincong Q Freeman
- Department of Public Health, University of Chicago, Chicago, IL, USA
| | - Theodore Karrison
- Department of Biostatistics, University of Chicago, Chicago, IL, USA
| | - Suzanne Conzen
- Department of Medicine Section of Hematology/Oncology, University of Texas Southwestern, Dallas, TX, USA
| | - Rita Nanda
- Department of Medicine Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA.
| | | |
Collapse
|
42
|
Lee SR, Kim NR, Mukae M, Won YS, Hong EJ. Sex hormone-binding globulin dampens growth and metastasis of breast cancer in an estrogen-independent manner. Am J Physiol Cell Physiol 2025; 328:C1685-C1698. [PMID: 40241272 DOI: 10.1152/ajpcell.00747.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/30/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Early studies have shown that sex hormone-binding globulin (SHBG) suppresses breast cancer by decreasing estrogen activity. However, the sex hormone-independent role of SHBG in breast cancer has received limited attention. Building on our previous research linking SHBG with tumor-associated macrophage (TYRO3, AXL, and MerTK) receptors, we aimed to explore SHBG's sex hormone-independent involvement in breast cancer progression. Analysis of public datasets and tumor slides from patients with breast cancer revealed that invasive breast cancer was associated with a significant decrease in SHBG, and lower SHBG levels correlated with poor cancer prognosis. In the polyomavirus middle T antigen overexpression mouse model (MMTV-PyMT), SHBG-Tg mice exhibited extended survival both under naïve and ovariectomized conditions. Although SHBG-Tg tumors had an estrogenic environment, their growth was suppressed, which correlated with reduced AXL levels. SHBG plasma treatment inhibited proliferation, tumorsphere growth, and invasion in MDA-MB-231 cells, accompanied by a decrease in AXL levels. In subcutaneous allograft models, SHBG-Tg mice showed reduced tumor growth and metastasis, and intraperitoneal injection of SHBG plasma significantly delayed tumor progression in PyMT mice compared with WT plasma. In summary, our study highlights SHBG's inhibitory role in breast cancer growth and metastasis, which may be particularly relevant for estrogen-independent patients with triple-negative breast cancer.NEW & NOTEWORTHY Our study is the first in vivo experiment using polyomavirus middle T antigen-sex hormone-binding globulin (PyMT-SHBG) mouse model to assess the physiological role of SHBG in breast cancer development. We show that SHBG presence in PyMT model restrains breast cancer development and progression in sex hormone-independent manner.
Collapse
Affiliation(s)
- Sang R Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Physiology, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Na Rim Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Moeka Mukae
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Young Suk Won
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
43
|
Mao J, Wang K, Tong J, Zhang W, Shen G, Wang D, Liao Z, Zhang Z, Miao Q, Jiang S, Zhang K. Discovery of dual PARP/NAMPT inhibitors for the treatment of BRCA wild-type triple-negative breast cancer. Bioorg Med Chem Lett 2025; 120:130117. [PMID: 39889972 DOI: 10.1016/j.bmcl.2025.130117] [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: 12/21/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Simultaneous inhibition of poly(ADP-ribose) polymerase (PARP) and nicotinamide phosphoribosyltransferase (NAMPT) has been shown to be synergistically effective against breast cancer susceptibility (BRCA) wild-type triple-negative breast cancer (TNBC) through synthetic lethality, which may be explored to broaden the clinical utility of PARP inhibitors. Herein, we report the discovery of dual PARP/NAMPT inhibitors through a pharmacophore linking approach. The lead compound 13j with potent inhibitory activity against both PARP1 and NAMPT (IC50 = 0.8 and 18 nM, respectively) effectively inhibited the proliferation of TNBC MDA-MB-231 cells with wild-type BRCA at submicromolar level. Mechanically, 13j disrupted the homologous recombination repair (HRR) pathway, caused the accumulation of DNA double-strand breaks (DSBs) and ultimately induced apoptotic cell death. In addition, this compound exhibited potent inhibitory potency on the migration of MDA-MB-231 cells. This study demonstrates that compound 13j is a promising lead compound for the development of better PARP/NAMPT inhibitors to treat TNBC with wild-type BRCA.
Collapse
Affiliation(s)
- Jie Mao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Kaizhen Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Jun Tong
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Wanheng Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Guoqing Shen
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Dexiang Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Zepeng Liao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Zhiyi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Qi Miao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China
| | - Sheng Jiang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China.
| | - Kuojun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009 China.
| |
Collapse
|
44
|
Ding X, Wu Q, Du Y, Ji MM, Yang H, Hu Q, Ye Y. CDK16 + Luminal Progenitor Cell-Like Tumor Cells Interacted with POSTN + Cancer-Associated Fibroblasts Associate with Chemo-Resistance In Breast Cancer. SMALL METHODS 2025; 9:e2401192. [PMID: 39930931 DOI: 10.1002/smtd.202401192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/12/2025] [Indexed: 05/26/2025]
Abstract
Tumor heterogeneity and interaction with tumor microenvironment play a crucial role in neoadjuvant chemotherapy (NAC) resistance in breast cancer (BRCA). Unraveling this dynamic interaction may help uncover novel therapeutic targets. Here, dynamic changes in tumor states and cellular composition are systemically characterized using 175,825 single-cell transcriptomics from naïve and post-treatment biopsies of BRCA patients receiving NAC. CDK16+ tumors are identified featured with luminal progenitor cell (LPC)-like tumor cells enriched in the triple-negative subtype of BRCA, associated with chemo-resistance. Integrating single-cell RNA sequencing (scRNA-seq), spatial transcriptomics, and six independent public gene expression profiles that underwent chemotherapy revealed that POSTN+ cancer-associated fibroblasts (CAFs) are closely localized and interacted with CDK16+ LPC-like tumor cells to promote chemo-resistance. In vivo, CDK16 knockdown in tumor cells combined with chemotherapy significantly enhanced therapeutic efficacy. This in-house scRNA-seq from a mouse model validated that CDK16 knockdown reduced the LPC-like tumor cell signature, and the interaction of tumor featured with LPC-like tumor cells and POSTN+ CAFs. Together, the systematically integrated analyses uncovered an interaction network of CDK16+ tumor and POSTN+ CAFs that contributed to NAC- resistance, providing a new strategy for targeting CDK16 to enhance chemotherapy efficacy.
Collapse
Affiliation(s)
- Xinyu Ding
- Shanghai Institute of Immunology, Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, 230001, China
| | - Yanhua Du
- Shanghai Institute of Immunology, Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meng-Meng Ji
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, 071000, China
| | - Hua Yang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, 071000, China
| | - Qingsong Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, 230001, China
| | - Youqiong Ye
- Shanghai Institute of Immunology, Center for Immune-Related Diseases Research at Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
45
|
Chen L, Ma S, Wu H, Zheng L, Yi Y, Liu G, Li B, Sun J, Du Y, Wang B, Liu Y, Zhang C, Chang J, Pang Y, Wang W, Wang M, Zhu M. Zonated Copper-Driven Breast Cancer Progression Countered by a Copper-Depleting Nanoagent for Immune and Metabolic Reprogramming. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412434. [PMID: 40270472 PMCID: PMC12120698 DOI: 10.1002/advs.202412434] [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: 10/06/2024] [Revised: 02/14/2025] [Indexed: 04/25/2025]
Abstract
While studies of various carcinomas have reported aberrant metal metabolism, much remains unknown regarding their spatial accumulation and regulatory impacts in tumors. Here, elevated copper levels are detected in breast cancer tumors from patients and animal models, specifically exhibiting a zonate spatial pattern. Spatially resolved multiomics analyses reveal that copper zonation drives a tumor metabolic preference for oxidative phosphorylation (OXPHOS) over glycolysis and promotes tumor metastatic and immune-desert phenotypes. Then, a copper-depleting nanoagent is developed based on copper chelator tetrathiomolybdate (TM)-loaded hybridized bacterial outer membrane vesicles (hOMVs) from both Akkermansia muciniphila bacteria and CD326-targeting peptide-engineered Escherichia coli (TM@CD326hOMV). Systemic administration of TM@CD326hOMV reduces the labile copper level in tumors and inhibits both tumor growth and metastatic phenotypes, specifically through metabolic reprograming of OXPHOS toward glycolysis and restoration of antitumor immunity responses involving natural killer cells, CD4+ T cells, and cytotoxic CD8+ T cells in tumors. Assessing survival in murine breast cancer models, a combination of TM@CD326hOMV and a checkpoint blockade agent outperforms monotherapies. Notably, a copper-rich diet undermines the therapeutic efficacy of TM@CD326hOMV. Beyond demonstrating an effective nanoagent for treating breast cancer, this study deepens the understanding of how the pattern of copper accumulation in tumors affects pathophysiology and immunity.
Collapse
Affiliation(s)
- Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- School of Nanoscience and EngineeringUniversity of Chinese Academy of ScienceBeijing100049China
| | - Saibo Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- College of Marine Life ScienceOcean University of ChinaQingdao266003China
| | - Hao Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Lingna Zheng
- Key Laboratory of Nuclear Analytical Techniques and Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy PhysicsChinese Academy of SciencesBeijing100049China
| | - Yunpeng Yi
- Shandong Provincial Animal and Poultry Green Health Products Creation Engineering LaboratoryInstitute of Poultry ScienceShandong Academy of Agricultural ScienceJinan250100China
| | - Guangnian Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- Department of Hepatobiliary and Pancreatic SurgeryPeking University First HospitalBeijing100035China
| | - Baoyi Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- Department of Hepatobiliary and Pancreatic SurgeryPeking University First HospitalBeijing100035China
| | - Jiayi Sun
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Yang Du
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Bing Wang
- Key Laboratory of Nuclear Analytical Techniques and Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy PhysicsChinese Academy of SciencesBeijing100049China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- School of Nanoscience and EngineeringUniversity of Chinese Academy of ScienceBeijing100049China
| | - Cheng Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersBeijing Key Laboratory of Carcinogenesis and Translational ResearchDepartment of Gastrointestinal OncologyPeking University Cancer Hospital & InstituteBeijing100142China
| | - Jing Chang
- College of Marine Life ScienceOcean University of ChinaQingdao266003China
| | - Yuheng Pang
- Beijing YouAn HospitalCapital Medical UniversityBeijing Institute of HepatologyBeijing100069China
| | - Wenjing Wang
- Beijing YouAn HospitalCapital Medical UniversityBeijing Institute of HepatologyBeijing100069China
| | - Meng Wang
- Key Laboratory of Nuclear Analytical Techniques and Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy PhysicsChinese Academy of SciencesBeijing100049China
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
- School of Nanoscience and EngineeringUniversity of Chinese Academy of ScienceBeijing100049China
| |
Collapse
|
46
|
Li T, Meng H, Huang X, Yu Q, Sheng S, Jiang Y, Ren F. Photodynamic Biomimetic Liposomes Targeted to the Endoplasmic Reticulum Enhance Combined Immunotherapy for Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:25112-25127. [PMID: 40258178 DOI: 10.1021/acsami.5c03687] [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: 04/23/2025]
Abstract
Cancer immunotherapies, such as immune checkpoint inhibitors, have advanced rapidly and achieved notable success, yet they face significant challenges due to poor response rates and immune-related adverse effects, particularly in cases of triple-negative breast cancer (TNBC). Photodynamic therapy (PDT) can initiate immunogenic cell death (ICD) by inducing endoplasmic reticulum (ER) stress, thereby enhancing the effectiveness of tumor immunotherapy. Herein, we develop potent PDT biomimetic liposomes (PB Lipo) locating the ER to realize a synergistic immuno-photodynamic treatment. The PB Lipo is prepared using the optimal ratios of the phospholipids in the ER membrane. It is then loaded with indocyanine green (ICG), a photosensitizer approved for clinical use. PB Lipo has the unique ability to accumulate in the ER via membrane fusion, leading to severe ER stress when exposed to near-infrared (NIR) laser light, thus intensifying ICD. In combination with the antiprogrammed death-ligand 1 (PD-L1) antibody (αPD-L1), PB Lipo significantly improves efficiency against tumors in xenograft TNBC models. As a result, our combined treatment enhances mature dendritic cells, activates CD4+ T and CD8+ T cells, and promotes the secretion of cytotoxic cytokines. Collectively, our findings reveal that PB Lipo-mediated PDT presents a viable approach for effectively targeting the ER and enhancing ICD, thereby boosting antitumor efficacy in TNBC.
Collapse
Affiliation(s)
- Tianyang Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Haimei Meng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xinfeng Huang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qin Yu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Sizhe Sheng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yufei Jiang
- First clinical medicine college, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Fei Ren
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
47
|
Zhang J, Cheng X, Chen G, Chen X, Zhao X, Chen W, Du W, He Z, Yao X, Han B, Yao D. Discovery of a Novel Selective PAK1/HDAC6/HDAC10 Inhibitor ZMF-25 that Induces Mitochondrial Metabolic Breakdown and Autophagy-Related Cell Death in Triple-Negative Breast Cancer. RESEARCH (WASHINGTON, D.C.) 2025; 8:0670. [PMID: 40302782 PMCID: PMC12038163 DOI: 10.34133/research.0670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/16/2025] [Accepted: 03/22/2025] [Indexed: 05/02/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, and addressing its intrinsic heterogeneity has emerged as a valuable avenue for novel clinical treatment strategy. Here, we put forward an innovative strategy for TNBC treatment by simultaneously suppressing both p21-activated kinase 1 (PAK1) and histone deacetylase (HDAC) class IIb (HDAC6/10). A series of pyrido [2,3-d]pyrimidin-7(8H)-one moiety derivatives was successfully designed and synthesized to target PAK1/HDAC6/HDAC10 by utilizing structure-based screening and pharmacophore integration. ZMF-25 demonstrates marked inhibitory activity against PAK1, HDAC6, and HDAC10 with respective IC50 values of 33, 64, and 41 nM, remarkable selectivity over HDACs and PAKs, as well as prominent antiproliferative efficiency in MDA-MB-231 cells. Additionally, ZMF-25 effectively suppresses TNBC proliferation and migration by inhibiting PAK1/HDAC6/HDAC10. Moreover, it was found to impair glycolysis and trigger reactive oxygen species generation, resulting in autophagy-related cell death by inhibiting the AKT/mTOR/ULK1 signaling. Furthermore, ZMF-25 exhibits remarkable therapeutic potential with no obvious toxicity in vivo and good pharmacokinetics. In summary, these observations indicate that ZMF-25 is a novel and potent triple-targeting PAK1/HDAC6/HDAC10 inhibitor, which is expected to provide a novel and effective strategy for TNBC treatment.
Collapse
Affiliation(s)
- Jin Zhang
- School of Pharmaceutical Sciences, Health Science Center,
Shenzhen University, Shenzhen 518060, China
| | - Xiaoling Cheng
- School of Pharmaceutical Sciences, Health Science Center,
Shenzhen University, Shenzhen 518060, China
- School of Pharmaceutical Sciences,
Shenzhen Technology University, Shenzhen 518118, China
| | - Gang Chen
- School of Pharmaceutical Sciences, Health Science Center,
Shenzhen University, Shenzhen 518060, China
- School of Pharmaceutical Sciences,
Shenzhen Technology University, Shenzhen 518118, China
| | - Xiya Chen
- School of Pharmaceutical Sciences, Health Science Center,
Shenzhen University, Shenzhen 518060, China
- School of Pharmaceutical Sciences,
Shenzhen Technology University, Shenzhen 518118, China
| | - Xi Zhao
- School of Pharmaceutical Sciences, Health Science Center,
Shenzhen University, Shenzhen 518060, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital ofChengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Weiji Chen
- School of Pharmaceutical Sciences, Health Science Center,
Shenzhen University, Shenzhen 518060, China
- School of Pharmaceutical Sciences,
Shenzhen Technology University, Shenzhen 518118, China
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences,
Macao Polytechnic University, Macao 999078, China
| | - Wei Du
- West China School of Pharmacy,
Sichuan University, Chengdu 610000, China
| | - Zhendan He
- School of Pharmaceutical Sciences, Health Science Center,
Shenzhen University, Shenzhen 518060, China
- School of Pharmaceutical Sciences,
Shenzhen Technology University, Shenzhen 518118, China
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences,
Macao Polytechnic University, Macao 999078, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital ofChengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dahong Yao
- School of Pharmaceutical Sciences,
Shenzhen Technology University, Shenzhen 518118, China
| |
Collapse
|
48
|
Xu H, Yang A, Kang M, Lai H, Zhou X, Chen Z, Lin L, Zhou P, Deng H. Intratumoral and peritumoral radiomics signature based on DCE-MRI can distinguish between luminal and non-luminal breast cancer molecular subtypes. Sci Rep 2025; 15:14720. [PMID: 40289183 PMCID: PMC12034752 DOI: 10.1038/s41598-025-98155-0] [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: 12/29/2024] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Distinguishing the luminal subtypes of breast cancer (BC) remaining challenging. Thus, the aim of this study was to investigate the feasibility of radiomic signature using intratumoral and peritumoral features obtained from dynamic contrast-enhanced MRI (DCE-MRI) in preoperatively discriminating the luminal from non-luminal type in patients with BC. A total of 305 patients with pathologically confirmed BC from three hospitals were retrospectively enrolled. The LASSO method was then used for selecting features, and the radiomic score (radscore) for each patient was calculated. Based on the radscore, Radiomic signature of intratumoral, peritumoral, and combined intratumoral and peritumoral were established, respectively. The performances of the radiomic signatures were validated with receiver operator characteristic (ROC) curve and decision curve analysis. For predicting molecular subtypes, the AUC for intratumoral radiomic signature was 0.817, 0.838, and 0.883 in the training set, internal validation set, and external validation set, respectively. AUC for the peritumoral radiomic signature was 0.863, 0.895, and 0.889 in the training set, internal validation set, and external validation set, respectively. The AUC for combined intratumoral and peritumoral radiomic signature was 0.956, 0.945, and 0.896 in the training set, internal validation set, and external validation set, respectively. Additional contributing value of combined intratumoral and peritumoral radiomic signature to the intratumoral radiomic signature was statistically significant [NRI, 0.300 (95% CI: 0.117-0.482), P = 0.001 in internal validation set; NRI, 0.224 (95% CI: 0.038-0.410), P = 0.018 in external validation set]. These results indicated that the radiomic signature combining intratumoral and peritumoral features showed good performance in predicting the luminal type of breast cancer.
Collapse
Affiliation(s)
- Hao Xu
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Ao Yang
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Kang
- Department of Radiology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Hua Lai
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinzhu Zhou
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhe Chen
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Libo Lin
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Zhou
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Heping Deng
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
49
|
Zhao Q, Pramanik J, Lu Y, Homer NZM, Imianowski CJ, Zhang B, Iqbal M, Shaji SK, Morris AC, Roychoudhuri R, Okkenhaug K, Qiu P, Mahata B. Perturbing local steroidogenesis to improve breast cancer immunity. Nat Commun 2025; 16:3945. [PMID: 40287432 PMCID: PMC12033260 DOI: 10.1038/s41467-025-59356-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Breast cancer, particularly triple-negative breast cancer (TNBC), evades the body's immune defences, in part by cultivating an immunosuppressive tumour microenvironment. Here, we show that suppressing local steroidogenesis can augment anti-tumour immunity against TNBC. Through targeted metabolomics of steroids coupled with immunohistochemistry, we profiled the existence of immunosuppressive steroids in TNBC patient tumours and discerned the steroidogenic activity in immune-infiltrating regions. In mouse, genetic inhibition of immune cell steroidogenesis restricted TNBC tumour progression with a significant reduction in immunosuppressive components such as tumour associated macrophages. Steroidogenesis inhibition appears to bolster anti-tumour immune responses in dendritic and T cells by impeding glucocorticoid signalling. Undertaking metabolic modelling of the single-cell transcriptomics and targeted tumour-steroidomics, we pinpointed the predominant steroidogenic cells. Inhibiting steroidogenesis pharmacologically using a identified drug, posaconazole, curtailed tumour expansion in a humanised TNBC mouse model. This investigation paves the way for targeting steroidogenesis and its signalling pathways in breast cancer affected by immune-steroid maladaptation.
Collapse
Affiliation(s)
- Qiuchen Zhao
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Cancer Research UK Cambridge Centre and Department of Oncology, University of Cambridge, Cambridge, CB2 0XZ, UK
| | - Jhuma Pramanik
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Yongjin Lu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Baojie Zhang
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Muhammad Iqbal
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | | | | | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Pengfei Qiu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- The Precision Breast Cancer Institute, Addenbrookes Hospital, Department of Oncology, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.
| |
Collapse
|
50
|
Alharthi S, Alrashidi AA, Ziora Z, Ebrahimi Shahmabadi H, Alavi SE. Innovative PEGylated chitosan nanocarriers for co-delivery of doxorubicin and CpG in breast cancer therapy: Preparation, characterization, and immunotherapeutic potential. Med Oncol 2025; 42:176. [PMID: 40266471 DOI: 10.1007/s12032-025-02714-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/30/2025] [Indexed: 04/24/2025]
Abstract
This study aimed to design polyethylene glycol (PEG)ylated chitosan (CS, PEG-CS) nanoparticles for the co-delivery of doxorubicin (DOX), cytosine-phosphate-guanine oligodeoxynucleotide (CpG), and ovalbumin (OVA) to enhance breast cancer therapy. PEG-CS nanoparticles were synthesized using the ionotropic gelation method and characterized for size, zeta potential (ZP), entrapment efficiency, and drug release. In vitro and in vivo studies were conducted to assess cytotoxicity, immune activation, and antitumor efficacy. The optimized nanoparticles had a mean diameter of 156.4 ± 8.9 nm, a ZP of +18 mV, and demonstrated 75.3% DOX and 68.3% CpG release over 72 h. PEG-CS-DOX/CpG/OVA enhanced tumor reduction by 2.6-fold in vivo, with no significant toxicity. PEG-CS-DOX/CpG/OVA nanoparticles showed promise as a co-delivery platform for cancer therapy, combining cytotoxic and immune-stimulating effects with minimal toxicity.
Collapse
Affiliation(s)
- Sitah Alharthi
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, 11961, Al-Dawadmi, Saudi Arabia
| | - Amal Abdullah Alrashidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Zyta Ziora
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Qld, 4067, Australia
| | - Hasan Ebrahimi Shahmabadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, 7718175911, Iran.
| | - Seyed Ebrahim Alavi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, 7718175911, Iran.
| |
Collapse
|