1
|
Fines C, McCarthy H, Buckley N. The search for a TNBC vaccine: the guardian vaccine. Cancer Biol Ther 2025; 26:2472432. [PMID: 40089851 PMCID: PMC11913391 DOI: 10.1080/15384047.2025.2472432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/17/2025] Open
Abstract
Nearly 20 million people are diagnosed with cancer each year with breast cancer being the most common among women. Triple negative breast cancer (TNBC), defined by its no/low expression of ER and PR and lack of amplification of HER2, makes up 15-20% of all breast cancer cases. While patients overall have a higher response to chemotherapy, this subgroup is associated with the lowest survival rate indicating significant clinical and molecular heterogeneity demanding alternate treatment options. Therefore, new therapies have been explored, with a large focus on utilizing the immune system. A whole host of immunotherapies have been studied including immune checkpoint inhibitors, now standard of care for eligible patients, and possibly the most exciting and promising is that of a TNBC vaccine. While currently there are no approved TNBC vaccines, this review highlights many promising studies and points to an antigen, p53, which we believe is highly relevant for TNBC.
Collapse
Affiliation(s)
- Cory Fines
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Helen McCarthy
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Niamh Buckley
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| |
Collapse
|
2
|
Yang X, Liu S, Liu Y, Wang Y, Cui D, Lan T, Zhu D, Su Z, Hao E, Qin L, Guo H. Total flavonoids of litchi seed inhibit breast cancer metastasis by regulating the PI3K/AKT/mTOR and MAPKs signaling pathways. PHARMACEUTICAL BIOLOGY 2025; 63:229-249. [PMID: 40231974 PMCID: PMC12001861 DOI: 10.1080/13880209.2025.2488135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 03/07/2025] [Accepted: 03/30/2025] [Indexed: 04/16/2025]
Abstract
CONTEXT Total flavonoids from Litchi chinensis Sonn. (Sapindaceae) seeds (TFLS) effectively attenuate stem cell-like properties in breast cancer cells. However, their pharmacological effects and mechanisms in suppressing breast cancer metastasis remain unclear. OBJECTIVE This study aimed to elucidate the inhibitory effects and underlying mechanisms of TFLS on breast cancer metastasis. MATERIALS AND METHODS The antiproliferative, migratory, and invasive activities of breast cancer cells following TFLS treatment were evaluated using CCK-8, wound-healing, and transwell assays. The epithelial-mesenchymal transition (EMT) biomarkers were evaluated via Western blot analysis. The anti-metastatic effects of TFLS were further validated in vivo using zebrafish and mouse models. Network pharmacology methodology was utilized to predict potential targets and signaling pathways, which were subsequently corroborated by Western blot. Potential active compounds were identified through molecular docking, and the chemical constituents of TFLS were analyzed and characterized using UPLC-QTOF/MS. RESULTS TFLS suppressed the proliferation of MDA-MB-231 and MDA-MB-468 cells, with IC50 values of 44.47 μg/mL and 37.35 μg/mL at 72 h, respectively. It effectively suppressed breast cancer metastasis in vitro, demonstrated by a marked reduction in cellular motility and invasiveness, alongside the reversal of EMT. Consistent with pathway enrichment analysis, network pharmacology revealed that TFLS reduced the phosphorylation levels of PI3K, AKT, mTOR, JNK, ERK, and p38 in breast cancer cells. Molecular docking identified seven potential active ingredients, and UPLC-MS/MS confirmed the presence of key compounds, including procyanidin A2. DISCUSSION AND CONCLUSION TFLS effectively inhibits breast cancer cell proliferation, migration, and invasion in vitro by reversing the EMT phenotype, while suppressing metastasis in vivo. These effects are likely mediated via the attenuation of the PI3K/AKT/mTOR and MAPK signaling pathways.
Collapse
Affiliation(s)
- Xin Yang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| | - Shoushi Liu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Ying Liu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica & College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Yuanshuo Wang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Dianxin Cui
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Taijin Lan
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Dan Zhu
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhiheng Su
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica & College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Lilan Qin
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica & College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| |
Collapse
|
3
|
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
|
4
|
Márquez-Mendoza JM, Baranda-Ávila N, Lizano M, Langley E. Micro-RNAs targeting the estrogen receptor alpha involved in endocrine therapy resistance in breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167783. [PMID: 40057206 DOI: 10.1016/j.bbadis.2025.167783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Endocrine therapy resistance (ETR) in breast cancer (BC) is a multicausal phenomenon with diverse alterations in the tumor cell interactome. Within these alterations, non-coding RNAs (ncRNAs) such as micro-RNAs (miRNAs) modulate the expression of tumor suppressor genes and proto-oncogenes, such as the ESR1 gene encoding estrogen receptor alpha (ERα). This work aims to review the effects of miRNAs targeting ERα mRNA and their mechanisms related to ETR in BC. A thorough review of the literature and an in silico study were carried out to elucidate the involvement of each miRNA, thus contributing to the understanding of ETR in BC.
Collapse
Affiliation(s)
- J M Márquez-Mendoza
- Programa de Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - N Baranda-Ávila
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
| | - M Lizano
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - E Langley
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico.
| |
Collapse
|
5
|
Li L, Zhang Z, Huang N, Ren J, Qin Y, Luo Y. IGF1R activates FOXP3-β-catenin signaling to promote breast cancer development. Breast Cancer Res Treat 2025; 211:467-478. [PMID: 40055251 DOI: 10.1007/s10549-025-07663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/22/2025] [Indexed: 04/18/2025]
Abstract
PURPOSE Forkhead box P3 (FOXP3), a key marker of regulatory T cells (Tregs), is crucial for Treg differentiation and development. Emerging evidence suggests that FOXP3 is also expressed in various tumor cells; however, its role in tumor progression remains controversial. This study aimed to elucidate the impact of FOXP3 on breast cancer development. METHODS Breast cancer cell lines, including HCC1937, HCC1806, Hs 578T, MDA-MB-231, and MCF-7, along with xenograft mouse models, to assess the effects of FOXP3 on cell proliferation and tumor growth. FOXP3 expression in human breast cancer samples was analyzed using quantitative PCR and immunohistochemistry analyses. Cell proliferation and invasion were evaluated through MTS and transwell assays, respectively. Chromatin immunoprecipitation (ChIP) assays were performed to determine FOXP3 binding to the β-catenin gene promoter. RESULTS FOXP3 expression was elevated in advanced breast cancer and correlates with poor clinical outcomes. FOXP3 directly binds to β-catenin gene promoter - 986 to - 1168 region to facilitate β-catenin transcription, consequently resulting in increased breast cancer cell proliferation, migration, and invasion in vitro and tumor growth in vivo. Furthermore, IGF1R activated FOXP3-β-catenin signaling to promote breast tumor growth. Moreover, elesclomol, a potent copper ionophore, significantly inhibited FOXP3 expression to suppress breast tumor growth. CONCLUSION This study indicates that FOXP3 plays an oncogenic role in breast cancer development and suggests that targeting IGF1R-FOXP3-β-catenin signaling may be a putative therapeutic strategy for human breast cancer treatment.
Collapse
Affiliation(s)
- Lu Li
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Radiotherapy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Zhiming Zhang
- Key Laboratory of Environmental and Applied Microbiology, Key Laboratory of Environmental Microbiology of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Na Huang
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Radiotherapy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Jianlan Ren
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Radiotherapy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Yuan Qin
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Radiotherapy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Yangkun Luo
- Radiation Oncology Key Laboratory of Sichuan Province, Department of Radiotherapy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
6
|
Jeong H, Vacanti NM. A relative metabolic flux analysis model of glucose anaplerosis. Arch Biochem Biophys 2025; 768:110330. [PMID: 39922407 DOI: 10.1016/j.abb.2025.110330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 12/03/2024] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Glucose provides substrate for the predominant anaplerotic pathway which involves the activity of pyruvate carboxylase (PC). PC-mediated anaplerosis has been extensively studied as a metabolic regulator in glycolytic cells during tumorigenesis and metastasis. Herein, inaccuracies in established methods to measure relative intracellular flux through PC are highlighted and a compartmentalized condensed metabolic network (CCMN) is used to resolve the total malate pool into relative contributions from PC and other sources by metabolic flux analysis (MFA) with [U-13C6]glucose tracing. Performance of the CCMN method is evaluated in breast cancer cell lines that are exposed to small molecules targeting metabolism. Across conditions and cell lines, the CCMN approach yields results nearest to an accepted gold-standard methodology, using [3-13C]glucose, or even exposes the gold standard's limitations. The CCMN method does not require a separate experiment with a much more costly and generally less informative metabolic tracer, such as [3-13C]glucose, and in some cases, may outperform its application.
Collapse
Affiliation(s)
- Heesoo Jeong
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
7
|
Wang Y, Huang M, Zhou X, Li H, Ma X, Sun C. Potential of natural flavonoids to target breast cancer angiogenesis (review). Br J Pharmacol 2025; 182:2235-2258. [PMID: 37940117 DOI: 10.1111/bph.16275] [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: 07/03/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023] Open
Abstract
Angiogenesis is the process by which new blood vessels form and is required for tumour growth and metastasis. It helps in supplying oxygen and nutrients to tumour cells and plays a crucial role in the local progression and distant metastasis of, and development of treatment resistance in, breast cancer. Tumour angiogenesis is currently regarded as a critical therapeutic target; however, anti-angiogenic therapy for breast cancer fails to produce satisfactory results, owing to issues such as inconsistent efficacy and significant adverse reactions. As a result, new anti-angiogenic drugs are urgently needed. Flavonoids, a class of natural compounds found in many foods, are inexpensive, widely available, and exhibit a broad range of biological activities, low toxicity, and favourable safety profiles. Several studies find that various flavonoids inhibit angiogenesis in breast cancer, indicating great therapeutic potential. In this review, we summarize the role of angiogenesis in breast cancer and the potential of natural flavonoids as anti-angiogenic agents for breast cancer treatment. We discuss the value and significance of nanotechnology for improving flavonoid absorption and utilization and anti-angiogenic effects, as well as the challenges of using natural flavonoids as drugs. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Yuetong Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengge Huang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintong Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Xiaoran Ma
- Department of Oncology, Linyi People's Hospital, Linyi, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
8
|
Zhao C, Yu Y, Xiang P, Liao J, Yu B, Xing Y, Yin G. Association between radiotherapy and the risk of second primary malignancies in breast cancer patients with different estrogen receptor statuses. Eur J Cancer Prev 2025; 34:255-263. [PMID: 39230043 DOI: 10.1097/cej.0000000000000915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
BACKGROUND Breast cancer is the most common cancer among women. Second primary malignancies (SPMs) related to radiotherapy are significant complications. This study aims to investigate the correlation between radiotherapy and the occurrence of SPMs in breast cancer patients with different estrogen receptor statuses. METHODS We used data from the Surveillance, Epidemiology, and End Results (SEER) database, selecting estrogen receptor(+) and estrogen receptor(-) breast cancer patients from 1990 to 2015, with SPMs as the outcome measure. Fine-Gray competing risks regression and Poisson regression were employed to analyze the relationship between radiotherapy and the risk of SPMs in different estrogen receptor status groups. RESULTS Radiotherapy was associated with an increased risk of lung cancer, melanoma, non-Hodgkin lymphoma, and leukemia in estrogen receptor(+) patients. In estrogen receptor(-) patients, radiotherapy was linked to an increased risk of brain cancer and leukemia. The cumulative incidence, standardized incidence ratio, and subgroup analyses showed consistent results. In the dynamic assessment of radiotherapy-related risks, estrogen receptor(+) patients aged 50-70 exhibited a higher risk of leukemia and melanoma. Lung cancer risk was highest during a latency period of 20-30 years, while melanoma, non-Hodgkin lymphoma, and leukemia risks peaked within the first 10 years. For estrogen receptor(-) patients, brain cancer risk was higher between ages 50 and 70, and leukemia risk was elevated between ages 20 and 50. CONCLUSION Postoperative radiotherapy for breast cancer is associated with an increased risk of SPMs, with risks varying by estrogen receptor status and SPM type. Further research into the prevention of radiotherapy-related SPMs in different estrogen receptor status groups is crucial.
Collapse
Affiliation(s)
- Chengshan Zhao
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
9
|
Chu M, Zhang Y, Ye M, Yin Y, Chen H. Yi Shen Tiao Gan decoction alleviates aromatase inhibitor-related bone loss by promoting H-type vessel formation. Transl Oncol 2025; 55:102377. [PMID: 40168923 PMCID: PMC11999221 DOI: 10.1016/j.tranon.2025.102377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Aromatase inhibitors (AIs) are the main drugs used in the endocrine therapy of breast cancer. Given the continuous reduction in their estrogen levels, the quality of life and medication compliance of patients with osteopenia and osteoporosis, also called aromatase inhibitor-associated bone loss (AIBL), are seriously affected. Yi Shen Tiao Gan decoction (YSTG) is a traditional Chinese medicine prescription that is widely used in China to treat AIBL. Clinical studies have shown that YSTG can effectively decelerate AIBL. However, the molecular mechanisms underlying the effects of YSTG on AIBL remain unclear. In contrast to previous studies on the mechanism of AIBL, which focused on the relationship between estrogen and osteoclasts, this study adopts a novel perspective by focusing on H-type blood vessels and Slit guidance ligand 3 (SLIT3). H-type vessels, a recently identified subtype of capillaries, are predominantly located in the epiphysis and endosteum of long bones. These vessels play a crucial role in mediating the coupling of angiogenesis and osteogenesis within the bone microenvironment, with their abundance serving as a sensitive indicator of bone mass. Notably, SLIT3, a proangiogenic factor secreted by osteoblasts, is actively involved in the regulation of H-type vessel formation and bone formation processes. Here, we used the inoculation and resection of breast cancer xenografts, bilateral ovaritectomy (OVX), and gavage of letrozole (Femara) to construct a suitable AIBL animal model, which was verified by ELISA, microcomputed tomography (micro-CT), hematoxylin and eosin staining, and OCN and tartrate-resistant acid phosphatase (TRAP) immunohistochemical staining. H-type vessels were used as the entry point to study the mechanism of AIBL on the basis of the AIBL animal model. Among mice, the nude mice in the mastectomy (MX)+OVX+Le group showed the most severe bone loss after breast cancer cell implantation and resection, OVX, and Le gavage. Therefore, this group was suitable as an animal model of breast cancer AIBL than other groups. The animal experiment on the mechanism of YSTG in improving bone loss found that YSTG treatment significantly reduced the contents of C-terminal telopeptide of type I collagen, procollagen type I N-terminal propeptide, and growth hormone and increased those of serum estradiol and SLIT3. Micro-CT quantitative detection showed that after YSTG treatment, bone BMD, BV:TV, and Tb.N increased. The HE staining of bone tissue revealed that the thickness and number of trabecular bone and the number of cells in the cartilage layer of the femur remarkably increased after YSTG treatment. YSTG reduced TRAP content near the growth plate of the femoral metaphysis. YSTG significantly increased the OCN content at the margin of the trabecular bone in the metaphysis of the femur. Immunofluorescence results confirmed that YSTG could increase the amount of H-type vascular cells in bone. WB analysis confirmed that YSTG could significantly up-regulate the expression of SLIT3 protein in bone tissue. Therefore, YSTG may improve bone loss by up-regulating SLIT3 to induce H-type angiogenesis.
Collapse
Affiliation(s)
- Meiling Chu
- Department of Breast Surgery, Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhang
- Department of Breast Surgery, Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meina Ye
- Department of Breast Surgery, Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yulian Yin
- Department of Breast Surgery, Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Hongfeng Chen
- Department of Breast Surgery, Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
10
|
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
|
11
|
Shen Q, Yang K, Li Q, Qin T, Yu Y, Hong H, Yao H, Xu X. Nanoparticles (NPs)-mediated silencing of GSTP1 expression to reverse chemoresistance for effective breast cancer therapy. J Colloid Interface Sci 2025; 685:38-48. [PMID: 39827759 DOI: 10.1016/j.jcis.2025.01.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/11/2025] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
Chemotherapy remains the primary treatment modality for breast cancer (BCa) patients. However, chemoresistance commonly arises in clinical settings, contributing to poor prognosis. The development of chemoresistance is a dynamic and complex process involving the activation of oncogenes and inactivation of tumor suppressor genes. In this work, we utilized the RNA-sequencing (RNA-seq) technology to analyze the gene expression profiles of primary and recurrent tumor samples from BCa patients received the postoperative standard chemotherapy with doxorubicin (DOX), and identified glutathione S-transferase P1 (GSTP1) as a key factor in regulating chemoresistance. Molecular mechanistic studies revealed that high GSTP1 expression could not only impair the cytotoxicity of DOX by catalyzing the conjugation of reductive glutathione (GSH) with DOX, but also block the c-Jun NH2-terminal kinase (JNK) pathway to promote the proliferation via up-regulating anti-apoptotic B-cell lymphoma-2 (Bcl-2) expression. Given the severe side effects of DOX and the potential of RNA interference (RNAi) technology to silence target gene expression, we developed an endosomal pH-responsive nanoparticle (NP) platform for systemic co-delivery of DOX and GSTP1 siRNA (siGSTP1), and demonstrated its efficacy in reversing chemoresistance and suppressing the growth of DOX-resistant BCa tumors.
Collapse
Affiliation(s)
- Qian Shen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Ke Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Qingjian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Tao Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Huangming Hong
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, PR China.
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| |
Collapse
|
12
|
Masrour M, Moeinafshar A, Poopak A, Razi S, Rezaei N. The role of CXC chemokines and receptors in breast cancer. Clin Exp Med 2025; 25:128. [PMID: 40278951 DOI: 10.1007/s10238-025-01662-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
CXC chemokines are a class of cytokines possessing chemotactic properties. Studies indicate that CXC chemokines exhibit dysregulation in miscellaneous cancer categories and are significantly associated with the advancement of tumors. Breast cancer is a commonly diagnosed and fatal cancer among the female population. Breast cancer pathogenesis and progression involve various mechanisms, including invasion, metastasis, angiogenesis, and inflammation. Chemokines and their receptors are involved in all of these processes. The CXC chemokine receptors (CXCRs) and their related ligands have attracted considerable attention due to their multifaceted functions in facilitating and controlling tumor proliferation. CXCRs are expressed by both cancer cells and immune cells, and they play a crucial role in regulating the tumor microenvironment and the immune response. This review aims to assess the potential of CXCRs and CXC chemokines as therapeutic targets or biomarkers for personalized therapy. Additionally, it provides an overview of the current understanding of the expression, function, and prognostic relevance of CXCRs in breast cancer. Furthermore, the challenges and potential prospects pertaining to CXCR investigation in breast cancer are deliberated.
Collapse
Affiliation(s)
- Mahdi Masrour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Center for Orthopedic Trans-Disciplinary Applied Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Aysan Moeinafshar
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amirhossein Poopak
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific and Education Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
13
|
Wang JQ, Liu XM, Zhu ZS, Li Z, Xie CZ, Qiao X, Feng YK, Xu JY. Fluoxetine-Conjugated Platinum(IV) Prodrugs Targeting eEF2K and Conquering Multidrug Resistance against Triple-Negative Breast Cancer. J Med Chem 2025. [PMID: 40265570 DOI: 10.1021/acs.jmedchem.5c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Triple-negative breast cancer (TNBC) poses formidable challenges in the clinic owing to its particularly malignant and aggressive properties. Overexpression of eukaryotic elongation factor-2 kinase (eEF2K) is highly correlated with the poor prognosis of TNBC, representing a promising therapeutic target. Herein, Fluoxetine as eEF2K-inhibitor and chemosensitizer was conjugated with cisplatin/oxaliplatin to present two-in-one prodrugs 8-19. Multievaluation indicated that monosubstituted 8 and disubstituted 12 exhibited 407- and 174-fold higher cytotoxicity than cisplatin against MDA-MB-231 cells by elevating DNA damage-induced apoptosis and eEF2K-triggered autophagy. Moreover, 8 and 12 significantly overcame chemoresistance in A549cisR cells, evidenced by downregulating resistance-related key proteins P-gp, GST-π, ATM, and RAD51. Syngeneic and xenograft mouse models demonstrated that 8 and 12 could effectively inhibit tumor growth and metastasis, and reduce toxicity compared to cisplatin in vivo. Additionally, 8 and 12 stimulated immunomodulation including T-cell proliferation and Th1 cytokine production. All results hold the promise of 8 and 12 as multifunctional chemotherapeutic agents.
Collapse
Affiliation(s)
- Jia-Qian Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zi-Shuo Zhu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Li
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Cheng-Zhi Xie
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yu-Kuan Feng
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
14
|
Lv DM, Yang L, Fan C, Fang LH, Cheng SF. Identification of cell adhesion-related subtypes and construction of risk model to predict breast cancer prognostic and immunological properties. World J Surg Oncol 2025; 23:152. [PMID: 40264099 PMCID: PMC12013005 DOI: 10.1186/s12957-025-03802-5] [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: 02/18/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Breast invasive carcinoma is the most common form of breast cancer, often resulting in recurrence or metastasis in patients. Cell adhesion molecules play a crucial role in modulating the interactions between tumor cells and surrounding cells. The study aims to identify breast cancer subtypes related to cell adhesion and develop prognostic models that are essential for evaluating the prognostic risk and immunological profile of breast cancer. METHODS Transcriptome and clinical data were obtained from The Cancer Genome Atlas (TCGA) database, while cell adhesion-related genes (CARGs) from the MSigDB database. Molecular subtyping was performed using NMF clustering. Cox regression and Least absolute shrinkage and selection operator (LASSO) regression analyses were employed to construct a risk model for predicting patient prognosis. This model was validated in independent Gene Expression Omnibus (GEO) datasets, specifically GSE20685 and GSE42568. Immune cell infiltration was explored utilizing the CIBERSORT algorithm. Subsequently, we analyzed tumor mutation burden (TMB). Finally, potential drugs and drug sensitivity was evaluated using pRRobhetic algorithm. RESULTS Based on the expression levels of 39 genes related to cell adhesion, we identified 3 distinct subtypes, and LASSO regression analysis identified 8 genes that could be used as prognostic markers. Receiver operating characteristic (ROC) curves demonstrated that these cell adhesion genes were effective in predicting patient prognosis. Compared to the high-risk group, the low-risk group had a more favorable prognosis and a greater response to immunotherapy. These prognostic genes were found to be closely associated with immune cell infiltration and the response to immunotherapy. Furthermore, their significant associations with breast cancer sensitivities to anti-cancer drugs were revealed. CONCLUSION We developed a risk model focused on cell adhesion-related genes. This model accurately predicts the prognosis for breast cancer patients. It may also offer new insights for clinical decisions and immunotherapy.
Collapse
Affiliation(s)
- De-Ming Lv
- Department of Breast Thyroid Surgery, Jinhua Maternal And Child Health Care Hospital, Jinhua, 321000, China
| | - Li Yang
- Department of Breast Thyroid Surgery, Jinhua Maternal And Child Health Care Hospital, Jinhua, 321000, China
| | - Chen Fan
- Department of Breast Thyroid Surgery, Jinhua Maternal And Child Health Care Hospital, Jinhua, 321000, China
| | - Ling-Hui Fang
- Department of Breast Thyroid Surgery, Jinhua Maternal And Child Health Care Hospital, Jinhua, 321000, China
| | - Su-Fen Cheng
- Department of Breast Thyroid Surgery, Jinhua Maternal And Child Health Care Hospital, Jinhua, 321000, China.
| |
Collapse
|
15
|
Wang W, Yuan J, Zhu Y, Li R, Zhang J. Traditional Chinese medicine (TCM) enhances the therapeutic efficiency of a gemcitabine-loaded injectable hydrogel on postoperative breast cancer through modulating the microenvironment. J Mater Chem B 2025; 13:4864-4878. [PMID: 40171620 DOI: 10.1039/d4tb02776k] [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: 04/04/2025]
Abstract
Local injection of the drug-loaded hydrogel at the surgery site is promising for postoperative breast cancer. However, the postoperative changes in the tumor microenvironment, such as inflammation, abnormal angiogenesis and hypoxia, inhibit drug perfusion and contribute to breast cancer recurrence (BCR). Normalizing the abnormal blood vessels can effectively improve perfusion and reduce hypoxia. Here, we encapsulated gemcitabine (GEM) in a PLGA-PEG-PLGA hydrogel (GEM-hydrogel) for local treatment of postoperative breast cancer. The GEM-hydrogel can be injected into the surgery cavity allowing sustained release of the drug. Meanwhile, traditional Chinese medicine (TCM) Shexiang Baoxin Pill (SBP) was given to normalize the blood vessels to enhance drug perfusion. The results suggest that the combination of SBP enhances the therapeutic efficiency of the GEM-hydrogel, inhibiting tumor recurrence. Mechanism studies reveal that SBP works by promoting PDGFB expression in macrophages, subsequently recruiting pericytes, and normalizing blood vessels, finally alleviating hypoxia. This study demonstrates that the combination of TCM and chemotherapeutics is promising for suppressing postoperative tumor recurrence.
Collapse
Affiliation(s)
- Wenxu Wang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jixiang Yuan
- Urology Centre, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200000, China
| | - Yuying Zhu
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ruixiang Li
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jiange Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
16
|
Pang H, Badehnoosh B. Synergistic strength: unleashing exercise and polyphenols against breast cancer. Cancer Cell Int 2025; 25:144. [PMID: 40234950 PMCID: PMC11998149 DOI: 10.1186/s12935-025-03767-1] [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: 01/30/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025] Open
Abstract
Breast cancer remains a major global health challenge, necessitating innovative preventive and therapeutic strategies. Emerging evidence such as clinical trials suggests that the combination of exercise and polyphenol intake exerts synergistic effects in mitigating breast cancer progression by modulating key molecular pathways. Exercise enhances immune function, reduces inflammation, and regulates cellular metabolism, while polyphenols, natural compounds found in various plant-based foods, exhibit antioxidant, anti-inflammatory, and anti-carcinogenic properties. Together, these interventions influence apoptosis, oxidative stress, and ferroptosis which play crucial roles in breast cancer pathophysiology. This review explores the molecular mechanisms underlying the combined impact of exercise and polyphenols on breast cancer prevention and treatment. Understanding the interplay between exercise and polyphenols at the molecular level could pave the way for novel, non-invasive therapeutic strategies. Future research should focus on optimizing exercise regimens and dietary interventions to maximize their anti-cancer benefits. By bridging molecular insights with clinical applications, this review aims to provide a foundation for incorporating lifestyle-based interventions into breast cancer management. Our findings collectively highlight the promising potential of combining curcumin supplementation with exercise as a multifaceted approach to breast cancer treatment. The synergistic effects observed in various studies suggest that integrating lifestyle modifications with dietary interventions may enhance therapeutic efficacy and mitigate cancer progression. Further clinical investigations are warranted to validate these results and explore their applicability in human subjects. The evidence supports a holistic strategy for breast cancer management that could improve patient outcomes and quality of life during treatment.
Collapse
Affiliation(s)
- Haifan Pang
- Department of Physical Education, China University of Political Science and Law, Beijing, 102249, China.
| | - Bita Badehnoosh
- Department of Gynecology and Obstetrics, Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
17
|
Zhu G, Dong Y, Zhu R, Tan Y, Liu X, Tao J, Chen D. Dynamic contrast-enhanced magnetic resonance imaging parameters combined with diffusion-weighted imaging for discriminating malignant lesions, molecular subtypes, and pathological grades in invasive ductal carcinoma patients. PLoS One 2025; 20:e0320240. [PMID: 40233046 PMCID: PMC11999158 DOI: 10.1371/journal.pone.0320240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/15/2025] [Indexed: 04/17/2025] Open
Abstract
Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters or diffusion-weighted imaging (DWI) findings provide prognostic information on breast cancer. However, the accuracy of a single MRI technique is unsatisfactory. This study intended to explore the combination of DWI and DCE-MRI parameters in discriminating molecular subtypes in invasive ductal carcinoma (IDC) patients. Eighty-two IDC patients who underwent breast DWI and DCE-MRI examinations were retrospectively analyzed. Eighty-six patients with benign masses were retrieved as benign controls. The combination of ADC value, Ktrans, Kep, Ve, and iAUC had a good ability to discriminate IDC patients (vs. benign controls) with an area under the curve (AUC) [95% confidence interval (CI)] of 0.961 (0.935-0.987). A nomogram-based prediction model with the above combination showed a good predictive value for IDC probability. The combination of ADC value, Ktrans, Kep, and iAUC also had a certain ability to discriminate pathological grade III (vs. I or II) [AUC (95% CI): 0.698 (0.572-0.825)] in IDC patients. Notably, ADC value (P=0.010) and Kep (P=0.043) differed in IDC patients with different molecular subtypes. Besides, ADC value was increased (P<0.001), but Ktrans (P=0.037) and Kep (P=0.004) were decreased in IDC patients with Lumina A (vs. other molecular subtypes). The combination of ADC value, Ktrans, Kep, had an acceptable ability to discriminate Luminal A (vs. other molecular subtypes) [AUC (95% CI): 0.845 (0.748-0.941)] in IDC patients. DWI combined with DCE-MRI parameters discriminates IDC from benign masses; it also identifies Luminal A and pathological grade III in IDC patients.
Collapse
Affiliation(s)
- Gangming Zhu
- Department of radiology, Dongguan TungWah hospital, Dongguan, Guangdong, China
| | - Yongde Dong
- Department of radiology, Dongguan Songshan Lake TungWah hospital, Dongguan, Guangdong, China
| | - Ruiting Zhu
- Department of radiology, Dongguan Songshan Lake TungWah hospital, Dongguan, Guangdong, China
| | - Yuanman Tan
- Department of radiology, Dongguan Songshan Lake TungWah hospital, Dongguan, Guangdong, China
| | - Xiao Liu
- Department of radiology, Dongguan TungWah hospital, Dongguan, Guangdong, China
| | - Juan Tao
- Department of radiology, Dongguan TungWah hospital, Dongguan, Guangdong, China
| | - Decheng Chen
- Department of radiology, Dongguan Songshan Lake TungWah hospital, Dongguan, Guangdong, China
| |
Collapse
|
18
|
Ma Y, Zhu M, Zhang J, Jiao D, Hou Y, Chen X, Liu Z. Efficacy of adjuvant capecitabine in triple-negative breast cancer with residual disease after neoadjuvant therapy: a real-world study. Breast 2025; 81:104477. [PMID: 40250161 DOI: 10.1016/j.breast.2025.104477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE To determine the beneficiaries of capecitabine in patients with triple-negative breast cancer (TNBC) who failed to achieve pathological complete response (pCR) by analyzing the efficacy of the drug in different HER2 statuses and TNM stages. METHODS The Kaplan-Meier survival curve was plotted to estimate the effect of capecitabine therapy on disease-free survival (DFS) and overall survival (OS). Furthermore, the Cox proportional hazards model was used to analyze the factors that influence DFS and OS. RESULTS A total of 296 patients with TNBC who had non-pCR after undergoing neoadjuvant therapy (NAT) were included in this study. There were 152 patients (51.4 %) in the capecitabine group and 144 patients (48.6 %) in the no-capecitabine group. The 3-year DFS and OS rates of the capecitabine group were better than those of the no-capecitabine group (DFS 80.0 % vs. 68.0 % p = 0.012, OS 95.9 % vs. 86.9 % p = 0.011). In addition, the capecitabine group exhibited significantly better DFS and OS than the no-capecitabine group in the HER2-low (DFS p = 0.004, OS p = 0.009) and stage III (DFS p = 0.004, OS p = 0.008) populations but not in the HER2-0 or stage II population. CONCLUSION Adjuvant capecitabine therapy significantly improved the prognosis of patients with TNBC who had residual disease after NAT, and the improvements in the outcomes were significant in patients with HER2-low expression and stage III disease. Other effective treatment methods should be explored for patients with HER2-0 expression or stage II disease.
Collapse
Affiliation(s)
- Youzhao Ma
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Mingda Zhu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Jingyang Zhang
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Dechuang Jiao
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Yangyang Hou
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Xiuchun Chen
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China.
| |
Collapse
|
19
|
Wang S, Liang X, Li H, Zou J, Xu L, Zhang Y, Lin J, Zeng J, Zhong X, Liu X, Liu Z, Zheng Y, Nie M, Yang L. The NET-DNA-CCDC25 inhibitor di-Pal-MTO suppresses tumor progression and promotes the innate immune response. Cell Mol Immunol 2025:10.1038/s41423-025-01286-7. [PMID: 40229592 DOI: 10.1038/s41423-025-01286-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025] Open
Abstract
The DNA component of neutrophil extracellular traps (NET-DNA) is associated with cancer metastasis and chemotherapy resistance. However, recent studies have suggested that NET-DNA contributes to the activation of dendritic cells (DCs) and promotes the innate immune response to anticancer immunity. Therefore, exploring therapeutic approaches to inhibit NET-mediated tumor progression while maintaining antitumor immunity is essential. Our groups recently identified CCDC25 as a specific NET-DNA sensor on the cytoplasmic membrane of cancer cells that promotes cancer metastasis. In this study, we performed small-molecule compound screening and revealed that mitoxantrone (MTO) could block the interaction between NET-DNA and CCDC25. Molecular docking results indicated that MTO competed with NET-DNA by binding with the amino acid residues Tyr24 (Y24), Glu25 (E25), and Asp28 (D28) of the crystal structure of CCDC25. More importantly, we conjugated MTO with palmitoleic acids such as di-Pal-MTO to increase its residence time on the cytoplasmic membrane, which increased its inhibitory efficiency and decreased its cytotoxicity. In addition, di-Pal-MTO markedly inhibited the RAC1-CDC42 cascade to alleviate the NET-induced cytoskeleton arrangement and chemotactic migration of cancer cells. In multiple mouse models, di-Pal-MTO can suppress breast cancer metastasis and have synergistic effects with chemotherapeutics. Moreover, di-Pal-MTO promotes NET-DNA-dependent DC activation, leading to the subsequent expression of various chemokines that facilitate the infiltration of CD8+ T cells. Overall, we successfully identified a small molecule inhibitor, di-Pal-MTO, with dual effects on tumor repression and the antitumor immune response, which provides a novel therapeutic strategy against breast cancer.
Collapse
Affiliation(s)
- Shun Wang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xinyan Liang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Heliang Li
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Junying Zou
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Linxi Xu
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yetong Zhang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jianghua Lin
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiayi Zeng
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoming Zhong
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xu Liu
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zhou Liu
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yue Zheng
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China.
| | - Man Nie
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China.
| | - Linbin Yang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Atuati SF, Becker G, Oliveira SM. Diosmetin Attenuates Painful Symptoms Caused by Antineoplastics Paclitaxel and Anastrozole in Mice. Mol Neurobiol 2025:10.1007/s12035-025-04939-w. [PMID: 40229455 DOI: 10.1007/s12035-025-04939-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Cancer patients often experience painful symptoms as an adverse effect resulting from various factors, including antineoplastic therapy. Paclitaxel is a chemotherapeutic agent used to treat solid tumours and, unfortunately, causes acute and chronic peripheral neuropathic pain in patients. Anastrozole is an antineoplastic used to treat hormone receptor-positive breast cancer and causes musculoskeletal pain symptoms. The therapies used to control these pain symptoms have limited efficacy and cause adverse effects, highlighting the importance of finding new treatments. Diosmetin is a flavonoid that has shown efficacy in neuropathic, nociplastic, and inflammatory pain models. Therefore, we verified the antinociceptive effects of diosmetin on nociceptive parameters induced by paclitaxel and anastrozole. We investigated the effects of diosmetin (0.015, 0.15 and 1.5 mg/kg; oral route (p.o.)) on nociceptive parameters (mechanical and cold allodynia and pain affective‑motivational behaviour) induced by a single and repeated paclitaxel (1 mg/kg, intraperitoneal (i.p.)) administrations, as well as on the pain-like symptoms (mechanical allodynia, muscle strength, and pain affective‑motivational behaviour) of a single administration of anastrozole (0.2 mg/kg, p.o.) and the combination of both antineoplastics in mice. Diosmetin reduced the nociceptive parameters induced by paclitaxel and anastrozole. In addition, we observed that anastrozole exacerbated the mechanical allodynia induced by paclitaxel, an effect that was also reversed by diosmetin. These results suggest that diosmetin may be an effective therapeutic alternative for the treatment of painful symptoms caused by antineoplastic drugs, such as paclitaxel and anastrozole.
Collapse
Affiliation(s)
- Samuel Felipe Atuati
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriela Becker
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Department of Biochemistry and Molecular Biology, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
21
|
Goel S, Jovanović B, Chu X, Hughes M, Erick TK, Russo D, DiLullo M, Wrabel E, Jeselsohn R, Lin NU, Tayob N, Mittendorf E, Schnitt S, Tolaney SM. A Phase II Study of Abemaciclib for Patients with Retinoblastoma-Positive, Metastatic Triple-Negative Breast Cancer. Clin Cancer Res 2025; 31:1427-1436. [PMID: 39908010 PMCID: PMC11995003 DOI: 10.1158/1078-0432.ccr-24-2647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/26/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Cyclin-dependent kinase 4/6 inhibitors can significantly extend survival when given in combination with endocrine therapy in patients with hormone receptor-positive metastatic breast cancer. However, their activity has been relatively underexplored in patients with metastatic triple-negative breast cancer (mTNBC). PATIENTS AND METHODS We conducted a single-arm phase II study of abemaciclib monotherapy in patients with Rb-positive mTNBC. Patients were treated with abemaciclib 200 mg orally twice daily until disease progression, unacceptable toxicity, or withdrawal of consent. The primary endpoint was the objective response rate; secondary endpoints included progression-free survival (PFS), overall survival (OS), clinical benefit rate, disease control rate, and safety and tolerability. RESULTS A total of 27 patients were enrolled before the trial was closed early because of slow accrual. Patients had received a median of two lines of systemic therapy in the metastatic setting prior to enrollment. After a median follow-up of 28.5 months, the objective response rate was 0%, the clinical benefit rate was 14.8%, and the disease control rate was 22.2%. The median PFS was 1.94 months (95% confidence interval, 1.84-11.47), and the median OS was 8.44 months (95% confidence interval, 4.57-15.57). Median PFS and OS did not differ significantly based on androgen receptor and PD-L1 status. Pretreatment gene expression profiling of tumor tissue provided some hypothesis-generating insights into biological features associated with clinical benefit in this study. The most common treatment-related adverse events of grade 2 or higher were diarrhea (40.7%), neutropenia (40.7%), anemia (29.6%), and nausea (29.6%). CONCLUSIONS Abemaciclib monotherapy did not show clinical activity in patients with pretreated Rb-positive mTNBC.
Collapse
Affiliation(s)
- Shom Goel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
| | - Bojana Jovanović
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Xiangying Chu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Melissa Hughes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Timothy K. Erick
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Douglas Russo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Molly DiLullo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eileen Wrabel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Rinath Jeselsohn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Nancy U. Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Nabihah Tayob
- Harvard Medical School, Boston, Massachusetts
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elizabeth Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Stuart Schnitt
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Wu W, Yu M, Li Q, Zhao Y, Zhang L, Sun Y, Wang Z, Gong Y, Wang W, Liu C, Zhang J, Tang Y, Xu X, Guo X, Zhan J, Zhang H. Loss function of tumor suppressor FRMD8 confers resistance to tamoxifen therapy via a dual mechanism. eLife 2025; 13:RP101888. [PMID: 40213945 PMCID: PMC11991697 DOI: 10.7554/elife.101888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025] Open
Abstract
Approximately 40% ERα-positive breast cancer patients suffer from therapeutic resistance to tamoxifen. Although reduced ERα level is the major cause of tamoxifen resistance, the underlying mechanisms remain elusive. Here, we report that FRMD8 raises the level of ERα at both transcriptional and post-translational layers. FRMD8 deficiency in MMTV-Cre+; Frmd8fl/fl; PyMT mice accelerates mammary tumor growth and loss of luminal phenotype, and confers tamoxifen resistance. Single-cell RNA profiling reveals that Frmd8 loss decreases the proportion of hormone-sensing differentiated epithelial cells and downregulates the levels of ERα. Mechanically, on one hand, loss of FRMD8 inhibits ESR1 transcription via suppressing the expression of FOXO3A, a transcription factor of ESR1. On the other hand, FRMD8 interacts both with ERα and UBE3A, and disrupts the interaction of UBE3A with ERα, thereby blocking UBE3A-mediated ERα degradation. In breast cancer patients, FRMD8 gene promoter is found hypermethylated and low level of FRMD8 predicts poor prognosis. Therefore, FRMD8 is an important regulator of ERα and may control therapeutic sensitivity to tamoxifen in ERα-positive breast cancer patients.
Collapse
Affiliation(s)
- Weijie Wu
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Miao Yu
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Qianchen Li
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Yiqian Zhao
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Lei Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Yi Sun
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Zhenbin Wang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Yuqing Gong
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Wenjing Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory for Digestive Health; National Clinical Research Center for Digestive DiseasesBeijingChina
| | - Chenying Liu
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy of Ministry of Education of China, Tianjin Medical University, Tianjin’s Clinical Research Center for CancerTianjinChina
| | - Jing Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Yan Tang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Xiaojie Xu
- Department of Genetic Engineering, Beijing Institute of BiotechnologyBeijingChina
| | - Xiaojing Guo
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy of Ministry of Education of China, Tianjin Medical University, Tianjin’s Clinical Research Center for CancerTianjinChina
| | - Jun Zhan
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| | - Hongquan Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science CenterBeijingChina
| |
Collapse
|
23
|
Dong Z, Chen H, Yang Y, Hao H. Research on the optimization model of anti-breast cancer candidate drugs based on machine learning. Front Genet 2025; 16:1523015. [PMID: 40276676 PMCID: PMC12018315 DOI: 10.3389/fgene.2025.1523015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Breast cancer is one of the most common malignancies among women globally, with its incidence rate continuously increasing, posing a serious threat to women's health. Although current treatments, such as drugs targeting estrogen receptor alpha (ERα), have extended patient survival, issues such as drug resistance and severe side effects remain widespread. This study proposes a machine learning-based optimization model for anti-breast cancer candidate drugs, aimed at enhancing biological activity and optimizing ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) properties through multi-objective optimization. Initially, grey relational analysis and Spearman correlation analysis were performed on the molecular descriptors of 1,974 compounds, identifying 91 key descriptors. A Random Forest model combined with Shapley Additive Explanations (SHAP) values was then used to further select the top 20 descriptors with the greatest impact on biological activity. The constructed Quantitative Structure-Activity Relationship (QSAR) model, using algorithms such as LightGBM, Random Forest, and XGBoost, achieved an R2 value of 0.743 for biological activity prediction, demonstrating strong predictive performance. Additionally, a multi-model fusion strategy and Particle Swarm Optimization (PSO) algorithm were employed to optimize both biological activity and ADMET properties, thereby improving the prediction of Caco-2, CYP3A4, hERG, HOB, and MN properties. For example, the best model for predicting Caco-2 achieved an F1 score of 0.8905, while the model for predicting CYP3A4 reached an F1 score of 0.9733. This multi-objective optimization model provides a novel and efficient tool for drug development, offering significant improvements in both biological activity and pharmacokinetic properties, with practical implications for the optimization of future anti-breast cancer drugs.
Collapse
Affiliation(s)
- Zhou Dong
- School of Information Engineering, Xi’an Eurasia University, Xi’an, China
| | | | | | | |
Collapse
|
24
|
Wang X, Wang Z, Liao Q, Yuan P, Mei J, Zhang Y, Wu C, Kang X, Zheng S, Yang C, Liu J, Shang Q, Li J, Wang B, Li L, Liu H, Hu W, Dong Z, Zhao J, Wang L, Liu T, Den Y, Wang C, Han L, Chen Q, Yang H, Xu X, He J, Yue Z, Sun N, Fang X, Ying J. Spatially resolved atlas of breast cancer uncovers intercellular machinery of venular niche governing lymphocyte extravasation. Nat Commun 2025; 16:3348. [PMID: 40199901 PMCID: PMC11978809 DOI: 10.1038/s41467-025-58511-0] [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: 03/02/2024] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancers present intricate microenvironments comprising heterotypic cellular interactions, yet a comprehensive spatial map remained to be established. Here, we employed the DNA nanoball-based genome-wide in situ sequencing (Stereo-seq) to visualize the geospatial architecture of 30 primary breast tumors and metastatic lymph nodes across different molecular subtypes. This unprecedented high-resolution atlas unveils the fine structure of the tumor vasculature, highlighting heterogeneity in phenotype, spatial distribution, and intercellular communication within both endothelial and perivascular cells. In particular, venular smooth muscle cells are identified as the primary source of CCL21/CCL19 within the microenvironment. In collaboration with ACKR1-positive endothelial cells, they create a chemokine-rich venular niche to synergistically promote lymphocyte extravasation into tumors. High venule density predicts increased immune infiltration and improved clinical outcomes. This study provides a detailed spatial landscape of human breast cancer, offering key insights into the venular regulation of tumor immune infiltration.
Collapse
Affiliation(s)
- Xin Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Zhanyu Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Qijun Liao
- BGI Research, Shenzhen, 518083, P. R. China
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen, 518083, P. R. China
| | - Pei Yuan
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Junpu Mei
- BGI Research, Sanya, 572025, P. R. China
| | - Yin Zhang
- BGI Research, Shenzhen, 518083, P. R. China
| | - Chao Wu
- BGI Research, Shenzhen, 518083, P. R. China
| | - Xiyu Kang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Sufei Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
- Office for Cancer Diagnosis and Treatment Quality Control, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Chenxuan Yang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Jiaxiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Qingyao Shang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Jiangtao Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Bingning Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Liangyu Li
- BGI Research, Shenzhen, 518083, P. R. China
| | - Hui Liu
- BGI Research, Shenzhen, 518083, P. R. China
| | - Weining Hu
- BGI Research, Shenzhen, 518083, P. R. China
| | | | - Jie Zhao
- BGI Research, Shenzhen, 518083, P. R. China
| | | | - Tao Liu
- BGI Research, Shenzhen, 518083, P. R. China
| | - Yusheng Den
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, P. R. China
| | - Chengrui Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, P. R. China
| | - Lijuan Han
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, P. R. China
| | - Qianjun Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, P. R. China
| | | | - Xun Xu
- BGI Research, Shenzhen, 518083, P. R. China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Zhen Yue
- BGI Research, Sanya, 572025, P. R. China.
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China.
| | | | - Jianming Ying
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China.
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China.
| |
Collapse
|
25
|
Liu J, Liu Z, Hu J, Fan B, Zhang S, Chang K, Mao X, Huang G, Liu Z, Ma L. Anti-breast cancer activity of a novel genetically engineered fusion protein composed of HER2 affibody and proapoptotic peptide R8-KLA. Med Oncol 2025; 42:155. [PMID: 40205290 DOI: 10.1007/s12032-025-02707-3] [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: 11/27/2024] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
HER2-positive breast cancer is an aggressive subtype with unfavorable prognoses. Although HER2-targeted agents represented by monoclonal antibodies have achieved remarkable success in the clinic, there are still a substantial number of patients with disease relapse. Recently, multifunctional fusion proteins obtained via genetic engineering technology have received much attention in targeted tumor therapy, especially in breast cancer. In this study, we genetically engineered a novel recombinant fusion protein, named HMK, which was designed as a bifunctional construct including the HER2-specific affibody ZHER2:342 for targeted receptor recognition, and a proapoptotic module featuring a cell-penetrating octa-arginine (R8) motif conjugated to an antimicrobial peptide KLA. High-purity HMK proteins were successfully obtained using E. coli expression system and Ni-Nitrilotriacetic acid affinity purification method. HMK exhibited higher cytotoxicity in HER2-positive breast cancer cells SK-BR-3 (IC50 of 8.36 ± 0.62 μM) compared to normal breast epithelial cells MCF-10A (IC50 of 32.40 ± 2.93 μM), demonstrating favorable selectivity. HMK induced apoptosis in SK-BR-3 cells via activating both endogenous and exogenous apoptotic pathways, as evidenced by the cleavage of Caspase 8, Caspase 9, Caspase 3, and PARP. Caspase inhibitor Z-VAD significantly reversed the function of HMK in SK-BR-3 cells, suggesting that caspase-dependent apoptosis was crucial for the anti-breast cancer activity of HMK. Our results suggested that HMK protein may have the potential to become a candidate molecule for HER2-positive breast cancer treatment.
Collapse
Affiliation(s)
- Jian Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Zi Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China.
| | - Junfeng Hu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Binru Fan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Shizhun Zhang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Kaili Chang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Xiuping Mao
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Guozheng Huang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Zhi Liu
- Department of Pathology, Ma'anshan Municipal People's Hospital, Ma'anshan, Anhui, 243000, China
| | - Liang Ma
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China.
| |
Collapse
|
26
|
Lughmani ARK, Ibrahim N, Ali W, Bibi Y, Afzal A, Javed M, Hameed A, Shahzadi W, Saddique S, Ahmed M. Impact of Intermittent Fasting with a Ketogenic Diet on AMPK Levels in Breast Cancer Patients Receiving Chemotherapy. Nutr Cancer 2025:1-7. [PMID: 40198149 DOI: 10.1080/01635581.2025.2488065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK), a metabolic sensor activated by nutrient starvation, plays a multifaceted role in cancer. Whether AMPK is beneficial or malevolent is controversial. This study aimed to investigate AMPK levels in breast cancer patients receiving chemotherapy and compare the effects of intermittent fasting combined with different diets on these levels. Forty-five breast cancer patients were divided into three groups: a control, a group practicing 23:1-h intermittent fasting (IF) with a routine diet (RD), and another with a ketogenic diet (KD) over 4 weeks. Body mass index (BMI), Carbohydrate Antigen 15-3 (CA 15-3) levels, and serum AMPK levels were measured pre and post-intervention. Results showed a significant increase in AMPK levels in both the fasting groups and no significant difference in the non-fasting group, with the keto diet group showing the most significant growth. CA 15-3 levels were reduced in all the groups but significantly reduced in the KD group as compared to the RD group. This study shows that intermittent fasting with the keto diet improves AMPK levels and may serve as a valuable non-pharmacological complementary strategy for reducing or eliminating the tumor and, simultaneously, preventing the healthy cells from the toxic side effects of chemotherapy.
Collapse
Affiliation(s)
| | - Nouman Ibrahim
- Department of Physiology, Frontier Medical and Dental College, Abbottabad, Pakistan
| | - Wasiq Ali
- Department of Physiology, Frontier Medical and Dental College, Abbottabad, Pakistan
| | - Yasmeen Bibi
- Department of Physiology, Frontier Medical and Dental College, Abbottabad, Pakistan
| | - Adnan Afzal
- Department of Microbiology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Mizna Javed
- Department of Microbiology, GC University Faisalabad, Pakistan
| | - Ammara Hameed
- Department of Biochemistry, The Islamia University of Bahawalpur, Pakistan
| | - Wania Shahzadi
- Department of Microbiology, The Islamia University of Bahawalpur, Pakistan
| | - Saba Saddique
- Department of Microbiology, The Islamia University of Bahawalpur, Pakistan
| | - Maria Ahmed
- Department of Microbiology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| |
Collapse
|
27
|
Cui QF, Liu C, Dong XM, Liu ZQ. Exploring the biological functions and disease implications of OSGINs: A journey from discovery to clinical relevance. Biochem Pharmacol 2025; 237:116921. [PMID: 40199404 DOI: 10.1016/j.bcp.2025.116921] [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: 02/11/2025] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
Oxidative stress-induced growth inhibitors (OSGINs) represent a new category of proteins that respond to oxidative stress and modulate redox balance. Growing evidence indicates that OSGINs have extensive physiological and pathological functions by regulating essential cellular processes, including proliferation, autophagy, apoptosis, and ferroptosis, thus influencing the progression of various diseases such as cancer, atherosclerosis, and pulmonary fibrosis. Moreover, research indicates that some contaminants, biomaterials, active compounds, and drugs can induce the expression of OSGINs, thereby exerting toxicity or therapeutic effects on the organism. These many functions make OSGINs attractive targets. However, a thorough analysis of the topic is still lacking. This paper presents a systematic review of current OSGINs research, with an emphasis on their molecular functions, regulatory mechanisms, disease roles, and environmental stressors. Furthermore, using virtual screening tools, we identified a series of active molecules with potential inhibitory effects on OSGINs, providing valuable references for further drug development. Our review presents novel insights and guidance for the ongoing investigation of the biological significance and potential clinical applications of OSGINs.
Collapse
Affiliation(s)
- Qian-Fei Cui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Chong Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xue-Man Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhao-Qian Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
28
|
Habibi S, Bahramian S, Saeedeh ZJ, Mehri S, Ababzadeh S, Kavianpour M. Novel strategies in breast cancer management: From treatment to long-term remission. Crit Rev Oncol Hematol 2025; 211:104715. [PMID: 40187709 DOI: 10.1016/j.critrevonc.2025.104715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy among women and a leading cause of cancer-related mortality worldwide. Although improvements in early detection and therapy have been made, metastatic breast cancer (mBC) continues to be an incurable disease. Although existing treatments can prolong survival and enhance quality of life, they do not provide a definitive cure. Targeted therapies have significantly improved outcomes, particularly for subtypes such as human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor (HR)-positive (HR+) BC. Key innovations include antibodydrug conjugates (ADCs) and next-generation endocrine therapies. ADCs combine monoclonal antibodies with cytotoxic agents, allowing targeted delivery to tumor cells while minimizing systemic toxicity. Immunotherapy is emerging as a promising approach for aggressive subtypes, such as triple-negative breast cancer (TNBC). Strategies under investigation include chimeric antigen receptor T-cell (CAR-T) therapy, tumor-infiltrating lymphocyte (TIL) therapies, and natural killer (NK) cell treatments, all aimed at enhancing the ability of the immune system to target and eliminate resistant tumor cells. Tissue engineering, particularly hydrogel-based delivery systems, offers the potential for localized treatment. These systems enable the controlled release of therapeutic agents or immune cells directly to the tumor site, supporting tissue regeneration and enhancing immune surveillance to reduce recurrence. Despite these advancements, challenges remain, including treatment resistance, the immunosuppressive tumor microenvironment, and high costs. Overcoming these barriers requires further innovation in drug delivery systems and a deeper understanding of tumor biology.
Collapse
Affiliation(s)
- Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabbou Bahramian
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Zare Jalise Saeedeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sara Mehri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Guilan, Iran
| | - Shima Ababzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Maria Kavianpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
29
|
Zhang H, Lian J, Ma Y. FET-UNet: Merging CNN and transformer architectures for superior breast ultrasound image segmentation. Phys Med 2025; 133:104969. [PMID: 40184647 DOI: 10.1016/j.ejmp.2025.104969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
PURPOSE Breast cancer remains a significant cause of mortality among women globally, highlighting the critical need for accurate diagnosis. Although Convolutional Neural Networks (CNNs) have shown effectiveness in segmenting breast ultrasound images, they often face challenges in capturing long-range dependencies, particularly for lesions with similar intensity distributions, irregular shapes, and blurred boundaries. To overcome these limitations, we introduce FET-UNet, a novel hybrid framework that integrates CNNs and Swin Transformers within a UNet-like architecture. METHODS FET-UNet features parallel branches for feature extraction: one utilizes ResNet34 blocks, and the other employs Swin Transformer blocks. These branches are fused using an advanced feature aggregation module (AFAM), enabling the network to effectively combine local details and global context. Additionally, we include a multi-scale upsampling mechanism in the decoder to ensure precise segmentation outputs. This design enhances the capture of both local details and long-range dependencies. RESULTS Extensive evaluations on the BUSI, UDIAT, and BLUI datasets demonstrate the superior performance of FET-UNet compared to state-of-the-art methods. The model achieves Dice coefficients of 82.9% on BUSI, 88.9% on UDIAT, and 90.1% on BLUI. CONCLUSION FET-UNet shows great potential to advance breast ultrasound image segmentation and support more precise clinical diagnoses. Further research could explore the application of this framework to other medical imaging modalities and its integration into clinical workflows.
Collapse
Affiliation(s)
- Huaikun Zhang
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Lian
- School of Electronic and Information Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu, China
| | - Yide Ma
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
30
|
Yuan L, Cai Y, Wang G, Liu X, Chen B, Zhou D, Wu Y, Qu N, Li X, Zhou W. SGK3 promotes estrogen receptor-positive breast cancer proliferation by activating STAT3/ZMIZ2 pathway to stabilise β-catenin. Br J Pharmacol 2025; 182:1856-1875. [PMID: 39876548 DOI: 10.1111/bph.17453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND PURPOSE Breast cancer is a leading threat to women's health, with approximately 70% of cases being estrogen receptor-positive. SGK3 is regulated by estrogen and is positively associated with estrogen receptor expression, although its molecular role remains unclear. EXPERIMENTAL APPROACH Proteomics was used to identify SGK3's downstream targets. Tissue microarray immunofluorescence evaluated SGK3 and ZMIZ2 expression in ER+ breast cancer. Lentiviral-mediated knockdown and overexpression of SGK3 and/or ZMIZ2 assessed their effects on cell proliferation in vitro and in vivo. Chromatin immunoprecipitation (ChIP) analyzed p-STAT3 binding to the ZMIZ2 promoter, and Co-immunoprecipitation (Co-IP) examined ZMIZ2-β-catenin interaction. KEY RESULTS SGK3 expression was elevated in breast tumour tissues correlating with reduced patient survival. Proteomic analysis identified ZMIZ2 as a downstream target of SGK3. Overexpression of SGK3 promoted the proliferation of estrogen receptor-positive breast cancer in MCF-7 and T47D cells. Inhibition had the opposite effects. ZMIZ2 overexpression rescued the proliferation deficit in SGK3 knockdown cells. ZMIZ2 was found to bind and stabilises β-catenin. Knockdown of SGK3 led to β-catenin degradation via polyubiquitination, a process reversed by ZMIZ2 overexpression. STAT3 was identified as a downstream effector of SGK3 and its knockdown reduced cytoplasmic and nuclear p-STAT3 and STAT3, and inhibited ZMIZ2 and β-catenin expression. Celastrol suppressed estrogen receptor-positive breast cancer cell proliferation by inhibiting the SGK3/STAT3/ZMIZ2/β-catenin pathway. CONCLUSIONS AND IMPLICATIONS SGK3 expression is associated with poorer survival rates, thus SGK3 is a potential therapeutic target. As celastrol can inhibit SGK3 expression it could be an effective therapeutic agent.
Collapse
Affiliation(s)
- Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Gang Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xu Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Department of Pharmacy, The Third People's Hospital of Chengdu, Sichuan, China
| | - Bo Chen
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Duanfang Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yuanli Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Na Qu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Weiying Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular, College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Ferraioli D, Borella F, Meurer M, Lebreton C, Penel N, Bertucci F, Bompas E, Italiano A, Firmin N, Bertero L, Peyrottes I, Mancini J, Isambert N, Chevreau C, Ferron G, Perrin C, Piperno-Neumann S, Cosma S, Pautier P, Loaec C, Aust S, Chopin N, Meeus P, Blay JY, Gonin J, Duffaud F, Colombo PE, Ray-Coquard I. Primary ovarian leiomyosarcoma: results from an analysis by the French Sarcoma Group (Ovarian SArcoma MAnagement - OSAMA Study). Int J Gynecol Cancer 2025; 35:101692. [PMID: 40044476 DOI: 10.1016/j.ijgc.2025.101692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVES Primary ovarian leiomyosarcomas are exceptionally rare, constituting less than 1% of ovarian tumors, and they typically have a poor prognosis. The available data on the management of these tumors are sparse, with limited publications mainly comprising small retrospective series that include multiple histologic types. The aim is to evaluate the clinical, surgical, pathologic characteristics and clinical outcome of patient affected by primary ovarian leiomyosarcomas. METHODS Using the national database (NetSarc), we conducted a retrospective study of the outcomes of primary ovarian leiomyosarcomas at 18 French Sarcoma Group centers. Patients with any International Federation of Gynecology and Obstetrics stage of primary ovarian leiomyosarcoma at first diagnosis and available follow-up were included. RESULTS A total of 39 patients with primary ovarian leiomyosarcomas were included: 35 had localized disease and 4 had metastatic disease. The median tumor size was 134 mm. Radical and wide surgery was performed on 21 (62%) and 13 patients (38%), respectively. Tumor grade 3, presence of necrosis, mitoses ≥20 high-power field, and high Ki-67 expression >30% were reported in 17 of 34 (50%), 29 of 34 (85%), 17 of 34 (50%), and 17 of 27 patients (63%), respectively. Positive estrogen receptor expression was reported in 14 of 27 patients (52%), whereas progesterone receptor expression was observed in 10 of 27 patients (37%). Adjuvant chemotherapy was administered in 12 of 34 patients (35%), whereas pelvic adjuvant radiotherapy in 8 of 34 (23%). Of the early-stage primary ovarian leiomyosarcomas, 9 had isolated pelvic recurrence, whereas 18 had parenchymal distant metastases. A total of 15 patients (44%) died of disease. In early-stage primary ovarian leiomyosarcomas, high mitotic counts and progesterone receptor negativity were variables associated with worse survival. CONCLUSIONS Surgery is the cornerstone of treatment for early-stage primary ovarian leiomyosarcoma, whereas the role of adjuvant treatment remains unclear. Some pathologic features were associated with poorer survival. Owing to the rarity of ovarian leiomyosarcomas, referring patients to expert sarcoma centers is highly recommended.
Collapse
Affiliation(s)
- Domenico Ferraioli
- Centre Leon Berard and Claude Bernard University, Department of Surgical Oncology, Lyon, France.
| | - Fulvio Borella
- University of Turin, Sant'Anna Hospital, "Città della Salute e della Scienza," Department of Surgical Sciences, Turin, Italy; University of Turin, Sant'Anna Hospital, "Città della Salute e della Scienza," Gynecology and Obstetrics 1U, Turin, Italy
| | - Marie Meurer
- AP-HM and Aix-Marseille University, CHU Timone, Department of Medical Oncology, Marseille, France
| | | | - Nicolas Penel
- Oscar Lambret Center, Department of General Cancerology, Lille, France
| | - Francois Bertucci
- Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, France
| | - Emmanuelle Bompas
- Institut de Cancérologie de l'Ouest, Department of Medical Oncology, Angers-Nantes, France
| | - Antoine Italiano
- Institut Bergonié, Department of Medical Oncology, Bordeaux, France
| | - Nelly Firmin
- ICM, Department of Medical Oncology, Montpellier, France
| | - Luca Bertero
- University of Turin and City of Health and Science University Hospital, Department of Medical Sciences, Pathology Unit, Turin, Italy
| | | | - Julien Mancini
- Aix-Marseille Univ, INSERM, IRD, UMR 1252, SESSTIM, Cancers, Biomedicine ans Society group, APHM, Timone Hospital, Public Health Department (BIOSTIC), Marseille, France
| | - Nicolas Isambert
- Georges-François Leclerc Center, Department of Medical Oncology, Dijon, France
| | | | | | - Cristophe Perrin
- Eugene Marquis Center, Department of Medical Oncology, Rennes, France
| | | | - Stefano Cosma
- University of Turin, Sant'Anna Hospital, "Città della Salute e della Scienza," Department of Surgical Sciences, Turin, Italy; University of Turin, Sant'Anna Hospital, "Città della Salute e della Scienza," Gynecology and Obstetrics 1U, Turin, Italy
| | - Patricia Pautier
- Gustave Roussy, Department of Medicine, Gynecology Unit, Villejuif, France
| | - Cécile Loaec
- Boulevard Professor Monod, Institut de Cancerologie de l'Ouest, Department of Surgery, Saint Herblain, France
| | - Stephanie Aust
- Centre Leon Berard, and Claude Bernard University, Department of Adult Medical Oncology, Lyon, France
| | - Nicolas Chopin
- Centre Leon Berard and Claude Bernard University, Department of Surgical Oncology, Lyon, France
| | - Pierre Meeus
- Centre Leon Berard and Claude Bernard University, Department of Surgical Oncology, Lyon, France
| | - Jean-Yves Blay
- Centre Leon Berard, and Claude Bernard University, Department of Adult Medical Oncology, Lyon, France
| | | | - Florence Duffaud
- AP-HM and Aix-Marseille University, CHU Timone, Department of Medical Oncology, Marseille, France
| | | | - Isabelle Ray-Coquard
- Centre Leon Berard, and Claude Bernard University, Department of Adult Medical Oncology, Lyon, France
| |
Collapse
|
32
|
Shen J, Ye X, Hou H, Wang Y. Efficacy and Safety of Immunochemotherapy in Advanced Triple-negative Breast Cancer: A Meta-analysis of Randomised Clinical Trials. Clin Oncol (R Coll Radiol) 2025; 40:103783. [PMID: 39955967 DOI: 10.1016/j.clon.2025.103783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 02/18/2025]
Abstract
AIMS Based on the existing controversial clinical research results, this study conducted a comprehensive meta-analysis of relevant literature to clarify the benefits of immunochemotherapy (ICT)-which combines immune checkpoint inhibitors and chemotherapy (CT)-for patients with advanced triple-negative breast cancer (aTNBC). MATERILAS AND METHODS A thorough literature search was conducted up to February 15, 2024. Subsequently, meta-analyses were performed to aggregate hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS), odds ratios (ORs) for objective response rate (ORR) and relative risks (RRs) for adverse events (AEs). RESULTS Six randomised clinical trials (RCTs) involving 3,105 patients met the inclusion criteria. In comparison with CT, ICT yielded significant enhancements in PFS (HR, 0.80; 95%CI: 0.73-0.87), OS (HR, 0.87; 95%CI: 0.80-0.96), and ORR (OR, 1.34; 95%CI: 1.15-1.55) in the intention-to-treat population. However, ICT also exhibited an increase in grade ≥3 AEs (RR, 1.11; 95%CI: 1.04-1.19) and severe AEs (RR, 1.40; 95%CI: 1.18-1.66). Subgroup analyses revealed that ICT significantly improved PFS (HR, 0.67; 95%CI: 0.58-0.77), OS (HR, 0.75; 95%CI: 0.64-0.87), and ORR (OR, 1.47; 95%CI: 1.16-1.84) within the PD-L1-positive subgroup, whereas no statistically significant differences were detected for PD-L1-negative population. CONCLUSION ICT demonstrates superior efficacy over conventional CT in the treatment of aTNBC, albeit accompanied by heightened toxicity. Notably, the assessment of PD-L1 status may serve as a valuable biomarker in discerning aTNBC patients who are particularly predisposed to derive benefit from ICT. PROSPERO NUMBER CRD42024513270.
Collapse
Affiliation(s)
- J Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - X Ye
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China
| | - H Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| | - Y Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, PR China.
| |
Collapse
|
33
|
Song Y, Zhang P, Bhushan S, Wu X, Zheng H, Yang Y. The Critical Role of Inhibitor of Differentiation 4 in Breast Cancer: From Mammary Gland Development to Tumor Progression. Cancer Med 2025; 14:e70856. [PMID: 40186425 PMCID: PMC11971571 DOI: 10.1002/cam4.70856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Inhibitor of differentiation 4 (ID4) is a highly conserved DNA-binding inhibitory protein of mammals, and its main role is to bind basic helix-loop-helix (b-HLH) so that it loses its DNA-binding activity, which in turn regulates the transcription of key genes, regulating cell differentiation and proliferation as the physiological function. Breast tissue is a highly heterogeneous tissue organ with a strong capacity for remodeling and differentiation, and studies of breast carcinogenesis suggest that the mechanisms regulating the differentiation of breast tissue interact critically with tumorigenesis. The expression level of ID4 and its regulatory mechanism play a crucial role in the study of breast cancer, but its oncogenic or oncostatic role has not yet been unanimously identified, and its regulatory mechanism in breast cancer still needs to be further elucidated. This review summarizes and analyzes the relevant studies of ID4 and the research progress in breast cancer, integrating the development of breast tissue and tumorigenesis with the regulatory role of ID4, to provide some insights into develop new treatment strategies and diagnostic biomarkers.
Collapse
Affiliation(s)
- Yuhang Song
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| | - Panshi Zhang
- Department of Thyroid and Breast SurgeryTongji Hospital of Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell BiologyUnit of Reproductive Biology, Justus‐Liebig‐University GiessenGiessenGermany
| | - Xinhong Wu
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| | - Hongmei Zheng
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| | - Yalong Yang
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| |
Collapse
|
34
|
Zhou M, Zhang G, Hou J, Chen L, Luo C, Mou S, Guo K, Wang R, Xiong L, Wang Z, Guo N, Sun J. 4D printed Stimuli Responsive Scaffold with Tissue Expansion and Photothermal Tumor Ablation Property for Post-Mastectomy Breast Reconstruction. Adv Healthc Mater 2025; 14:e2404575. [PMID: 40095339 DOI: 10.1002/adhm.202404575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/26/2025] [Indexed: 03/19/2025]
Abstract
Breast tissue engineering is a promising alternative to post-mastectomy breast reconstruction. However, the high relapse rate (about 20%) of this malignant tumor negatively affects its long-term prognosis. Moreover, the shortage of recipient site volume often hampers the reconstruction of large-sized breast. Here, this work reports on the additive manufacturing of a novel breast tissue engineering scaffold with photothermal shape memory and tumor ablation properties to solve the aforementioned issues. Graphene nanosheets (GN) are used to functionalize the surface of 3D-printed polyurethane scaffolds (GfPU) without compromising the biocompatibility of polyurethane. Subsequently, the GfPU scaffolds are remodeled into temporary shapes using established procedures. After exposure to an 808 nm laser, the GfPU scaffolds are heated to 47.1 °C, and they converted from a temporary shape to their original shape in a light intensity-dependent manner. With an accurate structural design, the shape memory scaffold could act as a stimuli-responsive tissue expander in vivo. Meanwhile, the laser-irradiated GfPU could specifically ablate multiple breast cancer cell lines in vitro and suppress both tumor growth and tumor recurrence in vivo. This dual-functional scaffold has the potential to be used in breast tissue engineering applications to optimize both the oncological and reconstructive effect.
Collapse
Affiliation(s)
- Muran Zhou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Guo Zhang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Jinfei Hou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Lifeng Chen
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Chao Luo
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Shan Mou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Ke Guo
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Rongrong Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Lingyun Xiong
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Nengqiang Guo
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, China
| |
Collapse
|
35
|
Wan X, Gong R, Zhao X, Li Y, Shan T, Zhong C, Zhu R, Chen Z, Jiang S, He L, Cao S, Tian S, Yang J, Ye N, Yi W, Cheng Y. Identification of a Novel Substrate for eEF2K and the AURKA-SOX8 as the Related Pathway in TNBC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412985. [PMID: 39950798 PMCID: PMC11984844 DOI: 10.1002/advs.202412985] [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: 10/15/2024] [Revised: 12/27/2024] [Indexed: 04/12/2025]
Abstract
Eukaryotic elongation factor 2 kinase (eEF2K) has been considered as a putative target for cancer therapy; however, the underlying mechanisms of eEF2K in triple-negative breast cancer (TNBC) progression remain to be fully elucidated. In this study, it is shown that eEF2K is highly expressed in TNBC and is associated with poor prognosis. In vitro, in vivo, and patient-derived organoid experiments demonstrate that knockdown of eEF2K significantly impedes progression of TNBC. Proteomic analysis and confirmation experiments reveal that eEF2K positively regulates the mRNA and protein expressions of sex-determining region Y-box 8 (SOX8). Mechanistically, eEF2K binds to and phosphorylates aurora kinase A (AURKA) at S391, a newly identified phosphorylation site critical for maintaining AURKA protein stability and kinase activity. Moreover, the compound C1, a molecular glue to degrade eEF2K, is optimized by designing and synthesizing its derivatives using reasonable structure-based optimization approach. The new compound C4 shows better ability to degrade eEF2K and stronger anti-cancer activity than C1. These findings not only uncover the pivotal role of the eEF2K/AURKA/SOX8 axis in TNBC progression, but also provide a promising lead compound for developing novel drug for treatment of TNBC.
Collapse
Affiliation(s)
- Xiaoya Wan
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
| | - Rong Gong
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
| | - Xiaobao Zhao
- Department of Medicinal ChemistryJiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical SciencesSoochow UniversitySuzhou215123China
| | - Yizhi Li
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
| | - Tianjiao Shan
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
| | - Changxin Zhong
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
| | - Rongfeng Zhu
- Department of Medicinal ChemistryJiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical SciencesSoochow UniversitySuzhou215123China
| | - Zonglin Chen
- Department of General SurgeryThe Second Xiangya HospitalCentral South UniversityChangsha410011China
| | - Shilong Jiang
- Department of PharmacyXiangya HospitalCentral South UniversityChangsha410028China
| | - Linhao He
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
| | - Shijun Cao
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
| | - Sheng Tian
- Department of Medicinal ChemistryJiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical SciencesSoochow UniversitySuzhou215123China
| | - Jinming Yang
- Department of Cancer Biology and ToxicologyDepartment of PharmacologyCollege of Medicine and Markey Cancer CenterUniversity of KentuckyLexingtonKY40536USA
| | - Na Ye
- Department of Medicinal ChemistryJiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical SciencesSoochow UniversitySuzhou215123China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics DevelopmentSoochow UniversitySuzhou215123China
| | - Wenjun Yi
- Department of General SurgeryThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Clinical Research Center For Breast Disease In Hunan ProvinceChangsha410011China
| | - Yan Cheng
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative DrugChangsha410011China
- FuRong LaboratoryChangshaHunan410078China
- Key Laboratory of Diabetes ImmunologyCentral South UniversityMinistry of EducationChangsha410011China
- NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer DrugsXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
36
|
Yang C, Zhang Y, Yan L, Liu A, Li F, Li Y, Zhang Y. Comprehensive Analysis of GPSM2: From Pan-Cancer Analysis to Experimental Validation. J Cell Mol Med 2025; 29:e70527. [PMID: 40208185 PMCID: PMC11984320 DOI: 10.1111/jcmm.70527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/04/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
G-protein signalling modulator 2 (GPSM2) plays an important role in maintaining cell polarisation and regulating the cell cycle; however, a systematic and comprehensive analysis of GPSM2 in cancer is still lacking. Using extensive multi-omics data, we explored the pan-cancer expression levels of GPSM2 from multiple perspectives and its association with prognosis, diagnosis, tumour stemness, immune-related genes, immune cell infiltration, genomic instability, and response to immunotherapy. We also elucidated the potential pan-cancer biological functions of GPSM2 using gene set enrichment analysis (GSEA) and searched for targeted drugs that affect GPSM2 expression using connectivity map analysis. To elucidate the effect of GPSM2 on colon cancer, we evaluated its effect on the biological behaviour of two colon cancer cell lines. In this study, GPSM2 was systematically analysed and shown to have satisfactory performance in disease diagnosis and prognostic prediction of various cancers. G-protein signalling modulator 2 plays an important role in the genesis and development of various tumours and is a potential tumour diagnostic and prognostic biomarker as well as an anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Chunjiao Yang
- Department of OncologyThe Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of NanningNanningChina
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Yuzhe Zhang
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Lirong Yan
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Aoran Liu
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Fang Li
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Yanke Li
- Department of Anorectal SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Ye Zhang
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
37
|
Chen P, Qiao D, Xiao L, Deng G, Yang Q, Tian R. Cost-effectiveness analysis of toripalimab combined with nab-paclitaxel as a first-line treatment for advanced TNBC in the US. PLoS One 2025; 20:e0320727. [PMID: 40168440 PMCID: PMC11960867 DOI: 10.1371/journal.pone.0320727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
INTRODUCTION Initial immunotherapy outcomes with toripalimab suggest a potential paradigm shift in the treatment of advanced triple-negative breast cancer (TNBC), promising extended survival for patients. However, its cost-effectiveness in the treatment of TNBC within the US health care context remains to be determined. METHODS A 5-year Markov model was developed using data from the TORCHLIGHT study to evaluate the cost-effectiveness of toripalimab plus nab-paclitaxel as a first-line therapy for metastatic or recurrent TNBC in the US. The model incorporated efficacy and safety data, literature-derived costs and utilities, and calculated ICERs. Sensitivity analyses were conducted to assess the impact of variable uncertainties on the outcomes. RESULTS Toripalimab combined with nab-P chemotherapy for TNBC patients resulted in an additional 2.68 life years (LYs) and 1.72 quality-adjusted life years (QALYs), with an ICER of $593,750 per QALY. Sensitivity analyses indicated that the cost and survival utility of toripalimab significantly influence patient outcomes. At a $100,000/QALY WTP threshold, combination therapy was not cost-effective compared with nab-P alone. CONCLUSIONS Our analysis suggests that, from a US health care system perspective, toripalimab in combination with chemotherapy does not demonstrate a significant cost-effective advantage over nab-P chemotherapy as a first-line treatment for patients with TNBC at a WTP threshold of $100,000/QALY and has a limited impact on US health care policy and clinical practice.
Collapse
Affiliation(s)
- Ping Chen
- Department of Nursing, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China,
| | - Dan Qiao
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Liping Xiao
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Guiya Deng
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Qing Yang
- Department of Nursing, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China,
| | - Rendi Tian
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
38
|
Qureshi Z, Fatima E, Safi A, Khanzada M, Altaf F. Talazoparib for the Treatment of Metastatic Castration-resistant Prostate Cancer: A Narrative Review. Am J Clin Oncol 2025; 48:206-214. [PMID: 39761644 DOI: 10.1097/coc.0000000000001159] [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: 03/26/2025]
Abstract
Breast and prostate cancer are among the most commonly diagnosed cancers worldwide. Recent advances in tumor sequencing and gene studies have led to a paradigm shift from treatment centered on the type of tumor to therapy more focused on specific immune phenotype markers and molecular alterations. In this review, we discuss the utility and function of talazoparib concerning prostate cancer treatment and summarize recent and planned clinical trials on talazoparib. We searched medical databases for articles relating to the use of talazoparib in prostate cancer from inception. Poly ADP ribose polymerase (PARP) is a family of 17 necessary DNA repair enzymes responsible for base excision repair, single-strand break repair, and double-strand break repair. PARP inhibitors are a class of oral targeted therapies that compete for the NAD + binding site on PARP molecules. Talazoparib, a potent PARP inhibitor, has emerged as a significant therapeutic option in the treatment of metastatic castration-resistant prostate cancer (mCRPC), particularly for patients with specific genetic alterations. Its role as a PARP inhibitor makes it a targeted therapy, focusing on cancer cells with DNA repair deficiencies. Talazoparib's role as a biomarker-directed therapy in advanced prostate cancer has been increasingly recognized. The TALAPRO-1 demonstrated durable antitumor activity in mCRPC patients. TALAPRO-2 is a notable clinical trial, specifically examining the effectiveness of Talazoparib when used in combination therapies. Current investigations demonstrate a significant improvement in survival outcomes for the patients of mCRPC, making Talazoparib a promising intervention.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences
| | - Adnan Safi
- Department of Medicine, Lahore General Hospital
| | - Mikail Khanzada
- Department of Internal Medicine, Lahore Medical and Dental College, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| |
Collapse
|
39
|
Debbi K, Benderra MA, Medioni J, Durdux C, To NH, Boukhobza C, Grellier N, Benmaziane A, Monnier L, Gligorov J, Assaf E, Belkacemi Y. Safety and efficacy of combined trastuzumab-deruxtecan and concurrent radiation therapy in breast cancer. The TENDANCE multicentric French study. Breast 2025; 80:104421. [PMID: 39983437 PMCID: PMC11893322 DOI: 10.1016/j.breast.2025.104421] [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: 12/17/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
PURPOSE Trastuzumab Deruxtecan (T DXD), a new antibody drug conjugate is a new treatment option for 2nd line metastatic breast cancer (MBC) for HER2 + or HER2 low tumors. Palliative or ablative radiotherapy (RT) may be required in patients who are being treated with T-DXd. However there is a lack of evidence regarding the safety profile of combining T-DXd with RT. TENDANCE study aimed to evaluate safety and efficacy of combined T-DXd and RT. MATERIALS AND METHODS This retrospective multicenter study included 54 patients treated concurrently with T-DXd and RT for HER2+ and HER2 low MBC between February 2021 and December 2023. All data were collected from a web-questionnaire, centralized after medical records and validation of the protocol by the local ethical committee. Primary endpoint was the safety of combined therapy. RESULTS Median age was 60 years. Patients who received T-DXD were further categorized into HER2+ (40.7 %), HER2 low/hormonal receptors HR+ (40.8 %) or HER2 low/HR- (18.5 %). In the HER2+ patients, T-DXd was administered as 2nd (18.2 %) or 3rd (31.8 %) or 4th (50 %) line therapy. RT was delivered using palliative (72.2 %) or ablative doses (27.8 %). Indications consisted mostly of palliative bone irradiation (46.3 %) and stereotactic radiosurgery (SRS) (25.9 %). With the median follow-up of 9 months, 22.2 % of patients had a complete response and 77.8 % had either a partial response or stable disease. Grade 1 or 2 asthenia was observed in 51.8 % of patients, while only 16.6 % experienced other grade 1 or 2 adverse effects. There was no T-DXd therapy discontinuation related to RT. CONCLUSION To our knowledge, TENDANCE is the largest study evaluating concurrent T-DXd and RT. This preliminary report suggests the feasibility of the combination of RT and T-DXd with manageable toxicity rate. Longer follow-up and further prospective studies are required to confirm these results.
Collapse
Affiliation(s)
- K Debbi
- Department of Radiation Oncology and Henri Mondor Breast Center. Henri Mondor University Hospital, APHP, UPEC, Créteil, France; Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, i-Biot, UPEC, Créteil, France.
| | - M A Benderra
- Medical Oncology Department, Institut Universitaire de Cancérologie Assistance Publique-Hôpitaux de Paris-Sorbonne Université, Paris, France
| | - J Medioni
- Centre of Early Clinical Trials in Cancer, Hôpital Européen Georges-Pompidou, Université Paris Cité, Paris, France
| | - C Durdux
- Department of Radiation Oncology, European Hospital Georges Pompidou, University of Paris Descartes, Paris, France
| | - N H To
- Department of Radiation Oncology and Henri Mondor Breast Center. Henri Mondor University Hospital, APHP, UPEC, Créteil, France; Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, i-Biot, UPEC, Créteil, France
| | - C Boukhobza
- Department of Radiation Oncology and Henri Mondor Breast Center. Henri Mondor University Hospital, APHP, UPEC, Créteil, France
| | - N Grellier
- Department of Radiation Oncology and Henri Mondor Breast Center. Henri Mondor University Hospital, APHP, UPEC, Créteil, France
| | - A Benmaziane
- Medical Oncology Department, Hôpital Foch, Paris, France
| | - L Monnier
- Radiation Oncology Department, DMU Orphé, Hôpital Tenon, AP-HP.Sorbonne Université, France
| | - J Gligorov
- Medical Oncology Department, Institut Universitaire de Cancérologie Assistance Publique-Hôpitaux de Paris-Sorbonne Université, Paris, France
| | - E Assaf
- Department of Medical Oncology, The Henri Mondor University Hospital, Creteil, France
| | - Y Belkacemi
- Department of Radiation Oncology and Henri Mondor Breast Center. Henri Mondor University Hospital, APHP, UPEC, Créteil, France; Institut Mondor de Recherche Biomédicale (IMRB), INSERM U955, i-Biot, UPEC, Créteil, France
| |
Collapse
|
40
|
Corvino A, Schneider T, Vu‐Bezin J, Loap P, Kirova Y, Prezado Y. Photon mini-GRID therapy for preoperative breast cancer tumor treatment: A treatment plan study. Med Phys 2025; 52:2493-2506. [PMID: 39873910 PMCID: PMC11972043 DOI: 10.1002/mp.17634] [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: 07/09/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Breast cancer is the leading cause of female cancer mortality worldwide, accounting for 1 in 6 cancer deaths. Surgery, radiation, and systemic therapy are the three pillars of breast cancer treatment, with several strategies developed to combine them. The association of preoperative radiotherapy with immunotherapy may improve breast cancer tumor control by exploiting the tumor radio-induced immune priming. However, this requires the use of hypofractionated radiotherapy (3 × 8 Gy), increasing the risk of toxicity. Mini-GRID therapy (mini-GRT) is an innovative form of spatially fractionated radiation therapy (SFRT) characterized by narrow beam widths between 1 to 2 mm that promises a significant increase in normal tissue dose tolerances and could thereby represent a new alternative for preoperative breast cancer treatment. Mini-GRT has been successfully implemented at the Hospital de Santiago de Compostela (Spain) with a flattening filter-free LINAC (megavoltage x-rays). PURPOSE In this dosimetry proof-of-concept study, we evaluate the feasibility of photon mini-GRT for preoperative breast cancer treatment. We also assess the clinical potential of mini-GRT and compare it with the current treatment standard of intensity-modulated radiotherapy (IMRT). METHODS Seven unbiased breast cancer dosimetries of patients treated with stereotactic body radiotherapy (SBRT) (3 × 8 Gy, IMRT) were selected for the study. Photon mini-GRT was compared with SBRT using three main criteria: (i) the dose to organs at risk (OARs), (ii) the dose constraints dictated by normal tissue tolerance, and (iii) the lateral penumbra in OARs. Tumor coverage was evaluated in terms of normalized total dose at 8 Gy-fractions. The optimized SBRT by IMRT was realized at the Institut Curie, Paris, France. The dose in mini-GRT was calculated by means of Monte Carlo simulations based on the mini-GRT implementation realized at the University Hospital in Santiago de Compostela. RESULTS Compared to SBRT plans, mini-GRT resulted in a reduction of the mean dose to the lungs, heart, chest wall, and lymph nodes in the studied cases by a factor ranging from 50% to 100%. Additionally, valley, mean, and peak doses to normal tissues meet the dose tolerance limits for the considered OARs, the most challenging of all being the skin. The mean dose to the skin was reduced (20%-60% less) for most of the studied cases. Mini-GRT also yielded sharper lateral penumbras in the skin and lungs (size reduced by at least 50%). Similar tumor integral doses were obtained for the two treatment modalities. CONCLUSION Mini-GRT with megavoltage x-rays is an innovative treatment approach already implemented in a clinical context. In this proof-of-concept study, we evaluated mini-GRT for partial breast cancer irradiation, demonstrating its potential for preoperative treatment thanks to the high skin and normal tissue-sparing capabilities. These initial results represent a first step towards clinical use and encourage further prospective clinical studies.
Collapse
Affiliation(s)
- Angela Corvino
- Institut CurieUniversité PSLCNRS UMR3347Inserm U1021Signalisation Radiobiologie et CancerOrsayFrance
- Université Paris‐SaclayCNRS UMR3347Inserm U1021Signalisation Radiobiologie et CancerOrsayFrance
| | - Tim Schneider
- Laboratoire d'Imagerie Biomédicale MultimodaleBIOMAPSUniversité Paris‐SaclayService Hospitalier Frédéric JoliotOrsayFrance
| | | | - Pierre Loap
- Department of Radiation Oncology, Institut CurieParisFrance
| | - Youlia Kirova
- Department of Radiation Oncology, Institut CurieParisFrance
- University Versailles St. QuentinSt. QuentinFrance
| | - Yolanda Prezado
- Institut CurieUniversité PSLCNRS UMR3347Inserm U1021Signalisation Radiobiologie et CancerOrsayFrance
- Université Paris‐SaclayCNRS UMR3347Inserm U1021Signalisation Radiobiologie et CancerOrsayFrance
- New Approaches in Radiotherapy LabCenter for Research in Molecular Medicine and Chronic Diseases (CIMUS)Instituto de Investigacion Sanitaria de Santiago de Compostela (IDIS)University of Santiago de Compostela, Santiago de CompostelaA CorunaSpain
- Oportunius ProgramGalician Agency of Innovation (GAIN)Xunta de Galicia, Santiago de CompostelaA CorunaSpain
| |
Collapse
|
41
|
Lai X, Zhang Y, Li M, Yu S, Wang S, Zhang S, Niu H, Chen L, Lan X, Zhang J, Chen S. HGF/c-Met Promotes Breast Cancer Tamoxifen Resistance Through the EZH2/HOTAIR-miR-141/200a Feedback Signaling Pathway. Mol Carcinog 2025; 64:769-783. [PMID: 39853766 DOI: 10.1002/mc.23878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025]
Abstract
Tamoxifen is one of the most frequently used endocrine medications for the treatment of estrogen receptor-positive (ER + ) breast cancer (BC). Unfortunately, tamoxifen resistance (TR) brings more challenges to the clinical treatment, and the mechanisms of TR have not yet been fully clarified. HGF/c-Met is closely associated with cancer metastasis, but whether it is involved in TR remains unclear. In our study, we found that the activation of HGF/c-Met was crucial for TR maintenance. Synergistic interaction with HOTAIR and EZH2 accelerated HGF expression by repressing miR-141/200a. Additionally, HGF/c-Met activated NF-κB, forming a positive feedback loop of EZH2/HOTAIR-miR-141/200a-HGF/c-Met-NF-κB. Our findings indicated that HGF/c-Met functioned as an important biomarker for TR, and HGF/c-Met inhibition provided a novel approach to TR treatment.
Collapse
Affiliation(s)
- Xiaofeng Lai
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, China
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Yuan Zhang
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Mengyang Li
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shentong Yu
- Department of Pathology, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Shuiliang Wang
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Shenghang Zhang
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Huimin Niu
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Li Chen
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Xiaopeng Lan
- Department of Clinical Laboratory Medicine, Fuzhou General Clinical Medical School (The 900TH Hospital), Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Aptamers Technology, Affiliated Dongfang Hospital of School of Medicine, Xiamen University, Fuzhou, China
| | - Jian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, China
| | - Suning Chen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
42
|
Alshajrawi OM, Tengku Din TADAATD, Marzuki SSB, Maulidiani M, Mohd Rusli NARB, Badrol Hisham NFAB, Hui Ying L, Yahya MMB, Wan Azman WNB, Ramli RA, Wan Abdul Rahman WF. Exploring the complex relationship between metabolomics and breast cancer early detection (Review). Mol Clin Oncol 2025; 22:35. [PMID: 40083862 PMCID: PMC11905217 DOI: 10.3892/mco.2025.2830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/08/2024] [Indexed: 03/16/2025] Open
Abstract
An overview of metabolomics in cancer research, focusing on the identification of biomarkers, pharmacological targets and therapeutic agents, is provided in the present review. The fundamentals of metabolomics, the role of metabolites in cancer emergence and the methods used in metabolomic analysis, are reviewed. The applications of metabolomics in cancer therapy and diagnostics, as well as the challenges encountered in metabolomic research, are discussed. Finally, the potential clinical uses of metabolomics in cancer research and its future possibilities are explored, emphasising the importance of non-invasive diagnostic and monitoring techniques. The present review highlights the significance of metabolite-based metabolomics as a specialised tool for illuminating disease processes and identifying treatment potentials. The malfunctioning of metabolomic pathways and metabolite accumulation or depletion is caused by metabolomics abnormalities. Metabolite signatures close to a subject's phenotypic informative dimension can be used to monitor therapies and disease prediction diagnosis and prognosis. Non-invasive diagnostic and monitoring techniques with high specificity and selectivity are urgently needed. Metabolite-based metabolomics is a specialised metabolic biomarker and pathway-analysis technique, illuminating the putative processes of numerous human illnesses and determining treatment potentials. Locating biochemical pathway modifications that are early warning signs of pathological malfunction and illness is possible by identifying functional biomarkers linked to phenotypic variance. Scientists generated numerous metabolomics profiles to disclose the underlying processes and metabolomics networks for therapeutic target research in biomedicine. The metabolomic analysis of the potential utility of metabolites as biomarkers for clinical events is summarised in the present review. The significance of metabolite-based metabolomics as a specialised tool for illuminating disease processes and identifying treatment potentials is highlighted.
Collapse
Affiliation(s)
- Omar Mahmoud Alshajrawi
- Department of Chemical Pathology, School of Medical Science, Health Campus, University Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | | | - Shahira Sofea Binti Marzuki
- Department of Chemical Pathology, School of Medical Science, Health Campus, University Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | - Maulidiani Maulidiani
- Faculty of Science and Marine Environment, University Malaysia Terengganu, Kuala Nerus, Terengganu 21030, Malaysia
| | | | | | - Lim Hui Ying
- Faculty of Science and Marine Environment, University Malaysia Terengganu, Kuala Nerus, Terengganu 21030, Malaysia
| | - Maya Mazuwin Binti Yahya
- Department of Surgery, School of Medical Science, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | - Wan Norlina Binti Wan Azman
- Department of Chemical Pathology, School of Medical Science, Health Campus, University Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
- Hospital University Sains Malaysia, Health Campus, University Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | - Ras A. Ramli
- Faculty of Medicine, University Sultan Zainal Abidin, Kuala Terengganu, Terengganu 20400, Malaysia
| | - Wan Faiziah Wan Abdul Rahman
- Department of Pathology, School of Medical Science, Health Campus, University Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| |
Collapse
|
43
|
Zhang Q, Wang X, Shao Z, Zhang Y, Zhang L, Chen M, Zhou X, Zhu H, Zhou Y, Lu X, Li P, Chi W, Li L, Shao Z, Huang S, Xue J, Chi Y, Wu J, Xiu B. LINC01235 Promotes Clonal Evolution through DNA Replication Licensing-Induced Chromosomal Instability in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413527. [PMID: 39950924 PMCID: PMC11984920 DOI: 10.1002/advs.202413527] [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: 10/23/2024] [Revised: 01/19/2025] [Indexed: 04/12/2025]
Abstract
Despite the development of HER2-targeting drugs such as trastuzumab and T-DXd, treatment resistance is a substantial challenge, often leading to relapse and distant metastasis. Tumor heterogeneity in HER2-positive breast cancer drives the evolution of resistant clones following therapeutic stress. However, the targetable drivers of anti-HER2 treatment resistance are not thoroughly identified. This study aims to use neoadjuvant-targeted therapy cohorts and a patient-derived organoid in vitro treatment model to uncover the potential targetable drivers of anti-HER2 treatment resistance. it is found that LINC01235 significantly enhances DNA replication licensing and chromosomal instability, fostering clonal expansion and evolution, and ultimately increasing resistance to therapeutic interventions. LINC01235 regulates global H3K27ac, H3K9ac, and H3K36me3 modifications, promotes H2A.Z expression in regulatory regions, and increases the accessibility of DNA licensing factors to their promoter regions. XRCC5 is identified as a key component for maintaining genomic stability, crucial for LINC01235's role in replication licensing. Furthermore, therapeutic strategies targeting LINC01235, including the use of antisense oligonucleotides or ATR inhibitors, which showed promise in overcoming treatment resistance are explored. These findings underscore the pivotal role of LINC01235 in driving resistance mechanisms and highlight novel avenues for targeted therapies to improve the outcomes of patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xuliren Wang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Zhibo Shao
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Yi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Liyi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Ming Chen
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xujie Zhou
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Han Zhu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
| | - Yue Zhou
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xinya Lu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Pei Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Weiru Chi
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Lun Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Department of General SurgeryThe Second Xiangya HospitalCentral South UniversityHunan410011China
| | - Zhi‐Ming Shao
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Shenglin Huang
- Fudan University Shanghai Cancer CenterKey Laboratory of Medical Epigenetics and MetabolismInstitutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Jingyan Xue
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Yayun Chi
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Pathology CenterShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200080China
| | - Jiong Wu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Collaborative Innovation Center for Cancer MedicineShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Bingqiu Xiu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| |
Collapse
|
44
|
Dongrong Y, Xinyi L, Sua Y, Rachel B, Susan M, Sarah S, Paul S, Q Jackie W, Yang S. Breast radiation therapy fluence painting with multi-agent deep reinforcement learning. Med Phys 2025; 52:2015-2024. [PMID: 39853548 DOI: 10.1002/mp.17615] [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: 06/08/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND The electronic compensation (ECOMP) technique for breast radiation therapy provides excellent dose conformity and homogeneity. However, the manual fluence painting process presents a challenge for efficient clinical operation. PURPOSE To facilitate the clinical treatment planning automation of breast radiation therapy, we utilized reinforcement learning (RL) to develop an auto-planning tool that iteratively edits the fluence maps under the guidance of clinically relevant objectives. METHODS With institutional review board (IRB) approval, 70 patients treated with 6MV tangential photon beams with ECOMP technique were retrospectively collected and included in this study (20/50 for training/testing). Each pixel in the fluence map was assigned a reinforcement learning agent to perform independent action. Beam-eye-view projected dose profiles were generated to form state information as the input of the RL network. By predicting the Q value, pixel-wise actions were selected to modify specific pixel value in the fluence maps to improve overall plan quality. After dose calculation, reward signal calculated from the variation of target coverage and dose homogeneity was fed back to the RL framework and used to update network parameters. The RL generated plans were evaluated with dose distribution and dosimetric endpoints (i.e., Breast PTV V90%, Breast PTV V95%, Breast PTV V105%, Lung V20 Gy, Heart V5 Gy, Dmax) and compared with clinical plans. RESULTS The RL agent took around 90 s to generate a ECOMP treatment plan. The RL plans exhibited plan quality comparable to clinical plans in terms of isodose distribution and dosimetric endpoints. The mean Breast PTV V95%, Breast PTV V105% of RL plans are77.759 % ( ± 8.904 % ) $77.759{\mathrm{\ \% }}( { \pm 8.904{\mathrm{\ \% }}} )$ and8.522 cc ( ± 11.469 cc ) $8.522{\mathrm{\ cc\ }}( { \pm 11.469{\mathrm{\ cc}}} )$ , compared to78.568 % ( ± 9.094 % ) $78.568{\mathrm{\ \% }}( { \pm 9.094{\mathrm{\ \% }}} )$ and34.298 cc ( ± 36.297 cc ) $34.298\ {\mathrm{cc}}\ ( { \pm 36.297{\mathrm{\ cc}}} )$ cc of clinical plans. CONCLUSION The developed RL framework efficiently generates breast ECOMP plans with clinical acceptable plan quality.
Collapse
Affiliation(s)
- Yang Dongrong
- Department of Radiation Oncology, Duke University, North Carolina, USA
| | - Li Xinyi
- Department of Radiation Oncology, Duke University, North Carolina, USA
| | - Yoo Sua
- Department of Radiation Oncology, Duke University, North Carolina, USA
| | - Blitzblau Rachel
- Department of Radiation Oncology, Duke University, North Carolina, USA
| | - McDuff Susan
- Department of Radiation Oncology, Duke University, North Carolina, USA
| | - Stephens Sarah
- Department of Radiation Oncology, Duke University, North Carolina, USA
| | - Segars Paul
- Department of Radiology, Duke University, North Carolina, USA
| | - Wu Q Jackie
- Department of Radiation Oncology, Duke University, North Carolina, USA
| | - Sheng Yang
- Department of Radiation Oncology, Duke University, North Carolina, USA
| |
Collapse
|
45
|
Jiang Y, Cao Y, Yao Y, Zhang D, Wang Y. Chitosan and hyaluronic acid in breast cancer treatment: Anticancer efficacy and nanoparticle and hydrogel development. Int J Biol Macromol 2025; 301:140144. [PMID: 39848359 DOI: 10.1016/j.ijbiomac.2025.140144] [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: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The pervasive global health concern of breast cancer necessitates the development of innovative therapeutic interventions to enhance efficacy and mitigate adverse effects. Chitosan and hyaluronic acid, recognized for their biocompatibility and biodegradability, present compelling options for the novel drug delivery systems and therapeutic platforms in the context of breast cancer management. This review will delineate the distinctive attributes of chitosan and hyaluronic acid, encompassing their inherent anticancer properties, targeting capabilities, and suitability for chemical modifications along with nanoparticle development. These characteristics render them exceptionally well-suited for the fabrication of nanoparticles and hydrogels. The intrinsic anticancer potential of chitosan, in conjunction with its mucoadhesive properties, and the robust binding affinity of hyaluronic acid to CD44 receptors, facilitate specific drug delivery to the malignant cells, thus circumventing the limitations inherent in traditional treatment modalities such as chemotherapy. The incorporation of these materials into nanocarriers allows for the co-delivery of therapeutic agents, thereby potentiating synergistic effects, while hydrogel systems provide localized, controlled drug release and facilitate tissue regeneration. An analysis of advancements in their synthesis, functionalization, and application is presented, while also acknowledging challenges pertaining to scalability and clinical translation.
Collapse
Affiliation(s)
- Yanlin Jiang
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China
| | - Yu Cao
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yiqun Yao
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China
| | - Dianlong Zhang
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China.
| | - Yuying Wang
- Department of Breast Surgery, The Cancer Hospital of China Medical University Liaoning Cancer Hospital & Institute, China.
| |
Collapse
|
46
|
De Palma FDE, Pol JG, Carbonnier V, Scuderi SA, Mannino D, Montégut L, Sauvat A, Perez-Lanzon M, Uribe-Carretero E, Guarracino M, Granata I, Calogero R, Del Monaco V, Montanaro D, Stoll G, Botti G, D'Aiuto M, Baldi A, D'Argenio V, Guigó R, Rezsohazy R, Kroemer G, Maiuri MC, Salvatore F. Epigenetic regulation of HOXA2 expression affects tumor progression and predicts breast cancer patient survival. Cell Death Differ 2025; 32:730-744. [PMID: 39833374 PMCID: PMC11982354 DOI: 10.1038/s41418-024-01430-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Accumulating evidence suggests that genetic and epigenetic biomarkers hold potential for enhancing the early detection and monitoring of breast cancer (BC). Epigenetic alterations of the Homeobox A2 (HOXA2) gene have recently garnered significant attention in the clinical management of various malignancies. However, the precise role of HOXA2 in breast tumorigenesis has remained elusive. To address this point, we conducted high-throughput RNA sequencing and DNA methylation array studies on laser-microdissected human BC samples, paired with normal tissue samples. Additionally, we performed comprehensive in silico analyses using large public datasets: TCGA and METABRIC. The diagnostic performance of HOXA2 was calculated by means of receiver operator characteristic curves. Its prognostic significance was assessed through immunohistochemical studies and Kaplan-Meier Plotter database interrogation. Moreover, we explored the function of HOXA2 and its role in breast carcinogenesis through in silico, in vitro, and in vivo investigations. Our work revealed significant hypermethylation and downregulation of HOXA2 in human BC tissues. Low HOXA2 expression correlated with increased BC aggressiveness and unfavorable patient survival outcomes. Suppression of HOXA2 expression significantly heightened cell proliferation, migration, and invasion in BC cells, and promoted tumor growth in mice. Conversely, transgenic HOXA2 overexpression suppressed these cellular processes and promoted apoptosis of cancer cells. Interestingly, a strategy of pharmacological demethylation successfully restored HOXA2 expression in malignant cells, reducing their neoplastic characteristics. Bioinformatics analyses, corroborated by in vitro experimentations, unveiled a novel implication of HOXA2 in the lipid metabolism of BC. Specifically, depletion of HOXA2 leaded to a concomitantly decreased expression of PPARγ and its target CIDEC, a master regulator of lipid droplet (LD) accumulation, thereby resulting in reduced LD abundance in BC cells. In summary, our study identifies HOXA2 as a novel prognosis-relevant tumor suppressor in the mammary gland.
Collapse
Affiliation(s)
- Fatima Domenica Elisa De Palma
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Jonathan G Pol
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Vincent Carbonnier
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sarah Adriana Scuderi
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Deborah Mannino
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Léa Montégut
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Allan Sauvat
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maria Perez-Lanzon
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Elisabet Uribe-Carretero
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Depto. Bioquimica y Biologia Molecular y Genetica, Facultad de Enfermeria y Terapia Ocupacional, Caceres, Spain
| | - Mario Guarracino
- University of Cassino and Southern Lazio, Cassino, Italy
- National Research University Higher School of Economics, Moscow, Russia
| | - Ilaria Granata
- National Research Council, Inst. for High-Performance Computing and Networking, Naples, Italy
| | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | | | | | - Gautier Stoll
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Gerardo Botti
- Department of Senology, Istituto Nazionale Tumori-IRCCS Fondazione Pascale, Naples, Italy
| | - Massimiliano D'Aiuto
- Department of Senology, Istituto Nazionale Tumori-IRCCS Fondazione Pascale, Naples, Italy
| | - Alfonso Baldi
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Valeria D'Argenio
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy
| | - Roderic Guigó
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Bioinformatics and Genomics, Centre for Genomic Regulation (CRG), Barcelona, Catalonia, Spain
| | - René Rezsohazy
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, Belgium
| | - Guido Kroemer
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Maria Chiara Maiuri
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.
- Team «Metabolism, Cancer & Immunity », Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, Université de Paris, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.
- Inter-University Center for multifactorial and multi genetic chronic human diseases, "Federico II"- Naples, Tor Vergata- Roma II and Chieti-Pescara Universities, Chieti-Pescara, Italy.
| |
Collapse
|
47
|
Henning JW, Boyne DJ, Brenner DR, Carbonell C, Shokar S, Granados DP, Parackal A, Cheung WY. Real World Evidence Study to Assess Incidence, Treatment Patterns, Clinical Outcomes, and Health Care Resource Utilization in Early-Stage, High-Risk HER2-Negative Breast Cancer in Alberta, Canada. Clin Breast Cancer 2025; 25:e220-e228. [PMID: 39547858 DOI: 10.1016/j.clbc.2024.10.008] [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: 01/10/2024] [Revised: 06/24/2024] [Accepted: 10/13/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Data are needed to improve the current understanding of the epidemiology of patients with high-risk, HER2-negative, early breast cancer (eBC) (hormone receptor positive [HR+]/HER2-negative BC and triple-negative BC [TNBC]). PATIENTS AND METHODS This retrospective longitudinal cohort study used real-world, population-level data that included all individuals newly diagnosed with high-risk, HER2-negative eBC in Alberta, Canada, between 2010 and 2019. Data on treatment, laboratory results and pathology findings were collected through electronic health records and administrative databases. RESULTS The annual cumulative incidence of high-risk, HER2-negative eBC ranged from 6% to 9% of all incident BC cases. Individuals with TNBC were more likely to be younger, had stage II disease, grade 3 histology and received systemic therapy at a community centre (P < .05) compared to individuals with HR+/HER2-negative eBC. Only 14% of individuals diagnosed in 2010-2017 underwent germline BRCA testing postdiagnosis. Neoadjuvant systemic therapy was given to 37% of individuals. Adjuvant systemic therapy use increased from 77% (2012-2015) to 84% (2019). The 5-year overall survival (OS) from initiation of adjuvant systemic therapy or date of surgery (for individuals who did not receive adjuvant systemic therapy) was 77% (95% CI: 75-79). OS was significantly worse among individuals who were older, had grade 3 histology, had stage III disease, or had nodal involvement (P < .05). OS among individuals with TNBC between 2016 and 2019 who initiated adjuvant capecitabine was markedly worse compared to the overall cohort (2-year OS: 70% vs. 89%). CONCLUSION Outcomes analyses in this high-risk, HER2-negative eBC population suggest a continued unmet clinical need.
Collapse
Affiliation(s)
| | - Devon J Boyne
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Darren R Brenner
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | - Winson Y Cheung
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
48
|
Tu SN, Hu F, Zhang J, Cai H, Yang J. Research progress on the signaling pathway mechanism of terpenoids against breast cancer. Discov Oncol 2025; 16:433. [PMID: 40163255 PMCID: PMC11958888 DOI: 10.1007/s12672-025-01881-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Breast cancer is the most common malignant tumor in women worldwide. Current treatments include chemotherapy, hormone therapy, radiotherapy and surgery. Terpenoids have great anti-cancer potential due to their anti-inflammatory, antioxidant, anti-tumor, antiviral and other biological activities. They have become the central drug for the prevention and treatment of breast cancer. However, their low bioavailability and stability are urgent issues that need to be addressed. This article aims to sort out the mechanism of action of terpenoids in the treatment of breast cancer. By reviewing different signal transduction pathways, it is hoped that new ideas for the joint action of multiple pathways and multiple targets will be provided, and a theoretical basis will be provided for improving basic research and clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Sheng-Nan Tu
- School of Clinical Medicine, North China University of Science and Technology, Tangshan, 063210, China
| | - Fen Hu
- College of Life Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Juan Zhang
- Second Department of Mammary Gland, Tangshan People's Hospital, Tangshan, 063000, China
| | - Haifeng Cai
- Second Department of Mammary Gland, Tangshan People's Hospital, Tangshan, 063000, China.
| | - Junquan Yang
- School of Clinical Medicine, North China University of Science and Technology, Tangshan, 063210, China.
| |
Collapse
|
49
|
Im C, Hasan H, Stene E, Monick S, Rader RK, Sheade J, Wolfe H, Lu Z, Spector LG, McDonald AJ, Nolan V, Arnold MA, Conces MR, Moskowitz CS, Henderson TO, Robison LL, Armstrong GT, Yasui Y, Nanda R, Oeffinger KC, Neglia JP, Blaes A, Turcotte LM. Treatment, toxicity, and mortality after subsequent breast cancer in female survivors of childhood cancer. Nat Commun 2025; 16:3088. [PMID: 40164623 PMCID: PMC11958683 DOI: 10.1038/s41467-025-58434-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
Childhood cancer survivors, particularly those who received chest radiotherapy, are at high risk for developing subsequent breast cancer. Minimizing long-term toxicity risks associated with additional radiotherapy and chemotherapy is a priority, but therapeutic tradeoffs have not been comprehensively characterized and their impact on survival is unknown. In this study, 431 female childhood cancer survivors with subsequent breast cancer from a multicenter retrospective cohort study were evaluated. Compared with one-to-one matched females with first primary breast cancer, survivors are as likely to be prescribed guideline-concordant treatment (N = 344 pairs; survivors: 94%, controls: 93%), but more frequently undergo mastectomy (survivors: 81%, controls: 60%) and are less likely to be treated with anthracyclines (survivors: 47%, controls: 66%) or radiotherapy (survivors: 18%, controls: 61%). Despite this, survivors have nearly 3.5-fold (95% CI = 2.17-5.57) greater mortality risk. Here, we show survivors with subsequent breast cancer face excess mortality despite therapeutic tradeoffs and require specialized treatment guidelines.
Collapse
Affiliation(s)
- Cindy Im
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hasibul Hasan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Emily Stene
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sarah Monick
- Department of Hematology/Oncology, Mayo Clinic Arizona, Phoenix, AZ, 85054, USA
| | - Ryan K Rader
- Department of Medicine, University of Kansas, Westwood, KS, 66205, USA
| | - Jori Sheade
- Department of Hematology/Oncology, Northwestern Medicine Lake Forest Hospital, Lurie Cancer Center Affiliate Network, Lake Forest, IL, 60045, USA
| | - Heather Wolfe
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zhanni Lu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Logan G Spector
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Aaron J McDonald
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Vikki Nolan
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael A Arnold
- Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, and Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Miriam R Conces
- Department of Pathology & Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Chaya S Moskowitz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10017, USA
| | - Tara O Henderson
- Department of Pediatrics, University of Chicago, Chicago, IL, 60637, USA
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Rita Nanda
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | | | - Joseph P Neglia
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anne Blaes
- Department of Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Lucie M Turcotte
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
50
|
Chen H, Chen M, Yang C, Tang T, Wang W, Xue W. Machine learning based intratumor heterogeneity related signature for prognosis and drug sensitivity in breast cancer. Sci Rep 2025; 15:10828. [PMID: 40155597 PMCID: PMC11953232 DOI: 10.1038/s41598-025-92695-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025] Open
Abstract
Intratumor heterogeneity (ITH) is involved in tumor evolution and drug resistance. Drug sensitivity shows discrepancy in different breast cancer (BRCA) patients due to ITH. The genes mediating ITH in BRCA and their role in predicting prognosis and drug sensitivity is not yet elucidated. An ITH-related signature (IRS) was built by ten methods-based integrative machine learning programs using TCGA, METABRIC and five GEO datasets. Several indicating scores were employed to evaluate the correlation between IRS score and immune microenvironment. The biological role of PINK1 was investigated using CCK-8 assay. The optimal prognostic signature for BRCA cases was the IRS developed using StepCox(both) + Enet(alpha = 0.9) method, which had the highest average C-index of 0.79. IRS acted as a prognostic biomarker and showed good performance in predicting the prognosis of BRCA patients. Lower IRS score indicated high levels of immuno-activated cells, higher TMB score, higher PD1&CTLA4 immunophenoscore, lower ITH score, lower TIDE score and lower tumor escape score in BRCA. The gene set scores correlated with glycolysis, angiogenesis, NOTCH signaling and hypoxia were higher in BRCA with high IRS score. PINK1 knockdown significantly inhibited the proliferation of BRCA cells. Our study developed a novel IRS for BRCA, which could predict the clinical outcome and immunotherapy benefits of BRCA patients.
Collapse
Affiliation(s)
- Hongcai Chen
- Department of Internal Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Minna Chen
- Department of Internal Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Cui Yang
- Department of Gynaecology and Obstetrics, Shantou Central Hospital, Shantou, China
| | - Tingting Tang
- Department of Internal Medicine, Jinping District People's Hospital of Shantou, Shantou, China
| | - Wende Wang
- Department of Internal Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China.
| | - Wenwu Xue
- Department of Internal Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|